501
|
Forro C, Caron D, Angotzi GN, Gallo V, Berdondini L, Santoro F, Palazzolo G, Panuccio G. Electrophysiology Read-Out Tools for Brain-on-Chip Biotechnology. MICROMACHINES 2021; 12:124. [PMID: 33498905 PMCID: PMC7912435 DOI: 10.3390/mi12020124] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 02/07/2023]
Abstract
Brain-on-Chip (BoC) biotechnology is emerging as a promising tool for biomedical and pharmaceutical research applied to the neurosciences. At the convergence between lab-on-chip and cell biology, BoC couples in vitro three-dimensional brain-like systems to an engineered microfluidics platform designed to provide an in vivo-like extrinsic microenvironment with the aim of replicating tissue- or organ-level physiological functions. BoC therefore offers the advantage of an in vitro reproduction of brain structures that is more faithful to the native correlate than what is obtained with conventional cell culture techniques. As brain function ultimately results in the generation of electrical signals, electrophysiology techniques are paramount for studying brain activity in health and disease. However, as BoC is still in its infancy, the availability of combined BoC-electrophysiology platforms is still limited. Here, we summarize the available biological substrates for BoC, starting with a historical perspective. We then describe the available tools enabling BoC electrophysiology studies, detailing their fabrication process and technical features, along with their advantages and limitations. We discuss the current and future applications of BoC electrophysiology, also expanding to complementary approaches. We conclude with an evaluation of the potential translational applications and prospective technology developments.
Collapse
Affiliation(s)
- Csaba Forro
- Tissue Electronics, Fondazione Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci, 53-80125 Naples, Italy; (C.F.); (F.S.)
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Davide Caron
- Enhanced Regenerative Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego, 30-16163 Genova, Italy; (D.C.); (V.G.)
| | - Gian Nicola Angotzi
- Microtechnology for Neuroelectronics, Fondazione Istituto Italiano di Tecnologia, Via Morego, 30-16163 Genova, Italy; (G.N.A.); (L.B.)
| | - Vincenzo Gallo
- Enhanced Regenerative Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego, 30-16163 Genova, Italy; (D.C.); (V.G.)
| | - Luca Berdondini
- Microtechnology for Neuroelectronics, Fondazione Istituto Italiano di Tecnologia, Via Morego, 30-16163 Genova, Italy; (G.N.A.); (L.B.)
| | - Francesca Santoro
- Tissue Electronics, Fondazione Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci, 53-80125 Naples, Italy; (C.F.); (F.S.)
| | - Gemma Palazzolo
- Enhanced Regenerative Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego, 30-16163 Genova, Italy; (D.C.); (V.G.)
| | - Gabriella Panuccio
- Enhanced Regenerative Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego, 30-16163 Genova, Italy; (D.C.); (V.G.)
| |
Collapse
|
502
|
Wang L, Ye Z, Jang YY. Convergence of human pluripotent stem cell, organoid, and genome editing technologies. Exp Biol Med (Maywood) 2021; 246:861-875. [PMID: 33467883 DOI: 10.1177/1535370220985808] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The last decade has seen many exciting technological breakthroughs that greatly expanded the toolboxes for biological and biomedical research, yet few have had more impact than induced pluripotent stem cells and modern-day genome editing. These technologies are providing unprecedented opportunities to improve physiological relevance of experimental models, further our understanding of developmental processes, and develop novel therapies. One of the research areas that benefit greatly from these technological advances is the three-dimensional human organoid culture systems that resemble human tissues morphologically and physiologically. Here we summarize the development of human pluripotent stem cells and their differentiation through organoid formation. We further discuss how genetic modifications, genome editing in particular, were applied to answer basic biological and biomedical questions using organoid cultures of both somatic and pluripotent stem cell origins. Finally, we discuss the potential challenges of applying human pluripotent stem cell and organoid technologies for safety and efficiency evaluation of emerging genome editing tools.
Collapse
Affiliation(s)
- Lin Wang
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Zhaohui Ye
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Yoon-Young Jang
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, John Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
503
|
Song E, Zhang C, Israelow B, Lu-Culligan A, Prado AV, Skriabine S, Lu P, Weizman OE, Liu F, Dai Y, Szigeti-Buck K, Yasumoto Y, Wang G, Castaldi C, Heltke J, Ng E, Wheeler J, Alfajaro MM, Levavasseur E, Fontes B, Ravindra NG, Van Dijk D, Mane S, Gunel M, Ring A, Kazmi SAJ, Zhang K, Wilen CB, Horvath TL, Plu I, Haik S, Thomas JL, Louvi A, Farhadian SF, Huttner A, Seilhean D, Renier N, Bilguvar K, Iwasaki A. Neuroinvasion of SARS-CoV-2 in human and mouse brain. J Exp Med 2021; 218:211674. [PMID: 33433624 PMCID: PMC7808299 DOI: 10.1084/jem.20202135] [Citation(s) in RCA: 660] [Impact Index Per Article: 165.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 11/23/2020] [Accepted: 12/10/2020] [Indexed: 12/28/2022] Open
Abstract
Although COVID-19 is considered to be primarily a respiratory disease, SARS-CoV-2 affects multiple organ systems including the central nervous system (CNS). Yet, there is no consensus on the consequences of CNS infections. Here, we used three independent approaches to probe the capacity of SARS-CoV-2 to infect the brain. First, using human brain organoids, we observed clear evidence of infection with accompanying metabolic changes in infected and neighboring neurons. However, no evidence for type I interferon responses was detected. We demonstrate that neuronal infection can be prevented by blocking ACE2 with antibodies or by administering cerebrospinal fluid from a COVID-19 patient. Second, using mice overexpressing human ACE2, we demonstrate SARS-CoV-2 neuroinvasion in vivo. Finally, in autopsies from patients who died of COVID-19, we detect SARS-CoV-2 in cortical neurons and note pathological features associated with infection with minimal immune cell infiltrates. These results provide evidence for the neuroinvasive capacity of SARS-CoV-2 and an unexpected consequence of direct infection of neurons by SARS-CoV-2.
Collapse
Affiliation(s)
- Eric Song
- Department of Immunobiology, Yale School of Medicine, New Haven, CT
| | - Ce Zhang
- Department of Genetics, Yale School of Medicine, New Haven, CT.,Department of Neuroscience, Yale School of Medicine, New Haven, CT
| | - Benjamin Israelow
- Department of Immunobiology, Yale School of Medicine, New Haven, CT.,Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, CT
| | | | - Alba Vieites Prado
- Sorbonne Université, INSERM U1127, French National Centre for Scientific Research, Joint Research Unit 7225, Paris Brain Institute, Institut du Cerveau et de la Moelle Épinière, Paris, France
| | - Sophie Skriabine
- Sorbonne Université, INSERM U1127, French National Centre for Scientific Research, Joint Research Unit 7225, Paris Brain Institute, Institut du Cerveau et de la Moelle Épinière, Paris, France
| | - Peiwen Lu
- Department of Immunobiology, Yale School of Medicine, New Haven, CT
| | - Orr-El Weizman
- Department of Immunobiology, Yale School of Medicine, New Haven, CT
| | - Feimei Liu
- Department of Immunobiology, Yale School of Medicine, New Haven, CT.,Department of Biomedical Engineering, Yale University, New Haven, CT
| | - Yile Dai
- Department of Immunobiology, Yale School of Medicine, New Haven, CT
| | - Klara Szigeti-Buck
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT
| | - Yuki Yasumoto
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT
| | - Guilin Wang
- Yale Center for Genome Analysis, West Haven, CT
| | | | | | - Evelyn Ng
- Yale Center for Genome Analysis, West Haven, CT
| | | | - Mia Madel Alfajaro
- Department of Immunobiology, Yale School of Medicine, New Haven, CT.,Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT
| | - Etienne Levavasseur
- Sorbonne Université, INSERM U1127, French National Centre for Scientific Research, Joint Research Unit 7225, Paris Brain Institute, Institut du Cerveau et de la Moelle Épinière, Paris, France
| | - Benjamin Fontes
- Yale Environmental Health and Safety, Yale University, New Haven, CT
| | - Neal G Ravindra
- Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT.,Department of Computer Science, Yale University, New Haven, CT
| | - David Van Dijk
- Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT.,Department of Computer Science, Yale University, New Haven, CT
| | - Shrikant Mane
- Department of Genetics, Yale School of Medicine, New Haven, CT.,Yale Center for Genome Analysis, West Haven, CT
| | - Murat Gunel
- Department of Genetics, Yale School of Medicine, New Haven, CT.,Department of Neuroscience, Yale School of Medicine, New Haven, CT.,Department of Neurosurgery, Yale School of Medicine, New Haven, CT
| | - Aaron Ring
- Department of Immunobiology, Yale School of Medicine, New Haven, CT
| | | | - Kai Zhang
- Department of Laboratory Medicine, Geisinger Medical Center, Danville, PA
| | - Craig B Wilen
- Department of Immunobiology, Yale School of Medicine, New Haven, CT.,Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT
| | - Tamas L Horvath
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT
| | - Isabelle Plu
- Sorbonne Université, INSERM U1127, French National Centre for Scientific Research, Joint Research Unit 7225, Paris Brain Institute, Institut du Cerveau et de la Moelle Épinière, Paris, France.,Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Département de Neuropathologie, Paris, France
| | - Stephane Haik
- Sorbonne Université, INSERM U1127, French National Centre for Scientific Research, Joint Research Unit 7225, Paris Brain Institute, Institut du Cerveau et de la Moelle Épinière, Paris, France.,Yale Environmental Health and Safety, Yale University, New Haven, CT.,Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Département de Neuropathologie, Paris, France.,Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Cellule nationale de référence des maladies de Creutzfeldt-Jakob, Paris, France
| | - Jean-Leon Thomas
- Sorbonne Université, INSERM U1127, French National Centre for Scientific Research, Joint Research Unit 7225, Paris Brain Institute, Institut du Cerveau et de la Moelle Épinière, Paris, France.,Department of Neurology, Yale School of Medicine, New Haven, CT
| | - Angeliki Louvi
- Department of Neuroscience, Yale School of Medicine, New Haven, CT.,Department of Neurosurgery, Yale School of Medicine, New Haven, CT
| | - Shelli F Farhadian
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, CT.,Department of Neurology, Yale School of Medicine, New Haven, CT
| | - Anita Huttner
- Department of Pathology, Yale School of Medicine, New Haven, CT
| | - Danielle Seilhean
- Sorbonne Université, INSERM U1127, French National Centre for Scientific Research, Joint Research Unit 7225, Paris Brain Institute, Institut du Cerveau et de la Moelle Épinière, Paris, France.,Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Département de Neuropathologie, Paris, France
| | - Nicolas Renier
- Sorbonne Université, INSERM U1127, French National Centre for Scientific Research, Joint Research Unit 7225, Paris Brain Institute, Institut du Cerveau et de la Moelle Épinière, Paris, France
| | - Kaya Bilguvar
- Department of Genetics, Yale School of Medicine, New Haven, CT.,Yale Center for Genome Analysis, West Haven, CT
| | - Akiko Iwasaki
- Department of Immunobiology, Yale School of Medicine, New Haven, CT.,Department of Molecular, Cellular, and Developmental Biology, Yale School of Medicine, New Haven, CT.,Howard Hughes Medical Institute, Chevy Chase, MD
| |
Collapse
|
504
|
Passaro AP, Stice SL. Electrophysiological Analysis of Brain Organoids: Current Approaches and Advancements. Front Neurosci 2021; 14:622137. [PMID: 33510616 PMCID: PMC7835643 DOI: 10.3389/fnins.2020.622137] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 12/11/2020] [Indexed: 12/23/2022] Open
Abstract
Brain organoids, or cerebral organoids, have become widely used to study the human brain in vitro. As pluripotent stem cell-derived structures capable of self-organization and recapitulation of physiological cell types and architecture, brain organoids bridge the gap between relatively simple two-dimensional human cell cultures and non-human animal models. This allows for high complexity and physiological relevance in a controlled in vitro setting, opening the door for a variety of applications including development and disease modeling and high-throughput screening. While technologies such as single cell sequencing have led to significant advances in brain organoid characterization and understanding, improved functional analysis (especially electrophysiology) is needed to realize the full potential of brain organoids. In this review, we highlight key technologies for brain organoid development and characterization, then discuss current electrophysiological methods for brain organoid analysis. While electrophysiological approaches have improved rapidly for two-dimensional cultures, only in the past several years have advances been made to overcome limitations posed by the three-dimensionality of brain organoids. Here, we review major advances in electrophysiological technologies and analytical methods with a focus on advances with applicability for brain organoid analysis.
Collapse
Affiliation(s)
- Austin P. Passaro
- Regenerative Bioscience Center, University of Georgia, Athens, GA, United States
- Division of Neuroscience, Biomedical & Health Sciences Institute, University of Georgia, Athens, GA, United States
| | - Steven L. Stice
- Regenerative Bioscience Center, University of Georgia, Athens, GA, United States
- Division of Neuroscience, Biomedical & Health Sciences Institute, University of Georgia, Athens, GA, United States
- Department of Animal and Dairy Science, University of Georgia, Athens, GA, United States
| |
Collapse
|
505
|
Daly AC, Prendergast ME, Hughes AJ, Burdick JA. Bioprinting for the Biologist. Cell 2021; 184:18-32. [PMID: 33417859 DOI: 10.1016/j.cell.2020.12.002] [Citation(s) in RCA: 158] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 10/29/2020] [Accepted: 12/01/2020] [Indexed: 12/30/2022]
Abstract
Building tissues from scratch to explore entirely new cell configurations could revolutionize fundamental understanding in biology. Bioprinting is an emerging technology to do this. Although typically applied to engineer tissues for therapeutic tissue repair or drug screening, there are many opportunities for bioprinting within biology, such as for exploring cellular crosstalk or cellular morphogenesis. The overall goals of this Primer are to provide an overview of bioprinting with the biologist in mind, outline the steps in extrusion bioprinting (the most widely used and accessible technology), and discuss alternative bioprinting technologies and future opportunities for bioprinting in biology.
Collapse
Affiliation(s)
- Andrew C Daly
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Alex J Hughes
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jason A Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
506
|
PsychENCODE and beyond: transcriptomics and epigenomics of brain development and organoids. Neuropsychopharmacology 2021; 46:70-85. [PMID: 32659782 PMCID: PMC7689467 DOI: 10.1038/s41386-020-0763-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/24/2020] [Accepted: 06/25/2020] [Indexed: 12/13/2022]
Abstract
Crucial decisions involving cell fate and connectivity that shape the distinctive development of the human brain occur in the embryonic and fetal stages-stages that are difficult to access and investigate in humans. The last decade has seen an impressive increase in resources-from atlases and databases to biological models-that is progressively lifting the curtain on this critical period. In this review, we describe the current state of genomic, transcriptomic, and epigenomic datasets charting the development of normal human brain with a particular focus on recent single-cell technologies. We discuss the emergence of brain organoids generated from pluripotent stem cells as a model to compensate for the limited availability of fetal tissue. Indeed, comparisons of neural lineages, transcriptional dynamics, and noncoding element activity between fetal brain and organoids have helped identify gene regulatory networks functioning at early stages of brain development. Altogether, we argue that large multi-omics investigations have pushed brain development into the "big data" era, and that current and future transversal approaches needed to leverage both fetal brain and organoid resources promise to answer major questions of brain biology and psychiatry.
Collapse
|
507
|
Caffrey TM, Button EB, Robert J. Toward three-dimensional in vitro models to study neurovascular unit functions in health and disease. Neural Regen Res 2021; 16:2132-2140. [PMID: 33818484 PMCID: PMC8354124 DOI: 10.4103/1673-5374.310671] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The high metabolic demands of the brain require an efficient vascular system to be coupled with neural activity to supply adequate nutrients and oxygen. This supply is coordinated by the action of neurons, glial and vascular cells, known collectively as the neurovascular unit, which temporally and spatially regulate local cerebral blood flow through a process known as neurovascular coupling. In many neurodegenerative diseases, changes in functions of the neurovascular unit not only impair neurovascular coupling but also permeability of the blood-brain barrier, cerebral blood flow and clearance of waste from the brain. In order to study disease mechanisms, we need improved physiologically-relevant human models of the neurovascular unit. Advances towards modeling the cellular complexity of the neurovascular unit in vitro have been made using stem-cell derived organoids and more recently, vascularized organoids, enabling intricate studies of non-cell autonomous processes. Engineering and design innovations in microfluidic devices and tissue engineering are progressing our ability to interrogate the cerebrovasculature. These advanced models are being used to gain a better understanding of neurodegenerative disease processes and potential therapeutics. Continued innovation is required to build more physiologically-relevant models of the neurovascular unit encompassing both the cellular complexity and designed features to interrogate neurovascular unit functionality.
Collapse
Affiliation(s)
- Tara M Caffrey
- Djavad Mowafaghian Center for Brain Health; Department of Pathology, University of British Columbia, Vancouver, BC, Canada
| | - Emily B Button
- Djavad Mowafaghian Center for Brain Health; Department of Pathology, University of British Columbia, Vancouver, BC, Canada
| | - Jerome Robert
- Institute of Clinical Chemistry, University Hospital of Zurich, Zurich, Switzerland
| |
Collapse
|
508
|
Choi JS, Lee HJ, Rajaraman S, Kim DH. Recent advances in three-dimensional microelectrode array technologies for in vitro and in vivo cardiac and neuronal interfaces. Biosens Bioelectron 2021; 171:112687. [PMID: 33059168 PMCID: PMC7665982 DOI: 10.1016/j.bios.2020.112687] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/24/2020] [Accepted: 10/03/2020] [Indexed: 12/13/2022]
Abstract
Three-dimensional microelectrode arrays (3D MEAs) have emerged as promising tools to detect electrical activities of tissues or organs in vitro and in vivo, but challenges in achieving fast, accurate, and versatile monitoring have consistently hampered further advances in analyzing cell or tissue behaviors. In this review, we discuss emerging 3D MEA technologies for in vitro recording of cardiac and neural cellular electrophysiology, as well as in vivo applications for heart and brain health diagnosis and therapeutics. We first review various forms of recent 3D MEAs for in vitro studies in context of their geometry, materials, and fabrication processes as well as recent demonstrations of 3D MEAs to monitor electromechanical behaviors of cardiomyocytes and neurons. We then present recent advances in 3D MEAs for in vivo applications to the heart and the brain for monitoring of health conditions and stimulation for therapy. A brief overview of the current challenges and future directions of 3D MEAs are provided to conclude the review.
Collapse
Affiliation(s)
- Jong Seob Choi
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, United States
| | - Heon Joon Lee
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, United States
| | - Swaminathan Rajaraman
- NanoScience Technology Center (NSTC), University of Central Florida, Orlando, FL, 32826-0120, United States; Department of Electrical & Computer Engineering, University of Central Florida, Orlando, FL, 32816, United States; Department of Materials Science & Engineering, University of Central Florida, Orlando, FL, 32816, United States
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, United States; Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, United States.
| |
Collapse
|
509
|
Current and future applications of induced pluripotent stem cell-based models to study pathological proteins in neurodegenerative disorders. Mol Psychiatry 2021; 26:2685-2706. [PMID: 33495544 PMCID: PMC8505258 DOI: 10.1038/s41380-020-00999-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/02/2020] [Accepted: 12/09/2020] [Indexed: 12/13/2022]
Abstract
Neurodegenerative disorders emerge from the failure of intricate cellular mechanisms, which ultimately lead to the loss of vulnerable neuronal populations. Research conducted across several laboratories has now provided compelling evidence that pathogenic proteins can also contribute to non-cell autonomous toxicity in several neurodegenerative contexts, including Alzheimer's, Parkinson's, and Huntington's diseases as well as Amyotrophic Lateral Sclerosis. Given the nearly ubiquitous nature of abnormal protein accumulation in such disorders, elucidating the mechanisms and routes underlying these processes is essential to the development of effective treatments. To this end, physiologically relevant human in vitro models are critical to understand the processes surrounding uptake, release and nucleation under physiological or pathological conditions. This review explores the use of human-induced pluripotent stem cells (iPSCs) to study prion-like protein propagation in neurodegenerative diseases, discusses advantages and limitations of this model, and presents emerging technologies that, combined with the use of iPSC-based models, will provide powerful model systems to propel fundamental research forward.
Collapse
|
510
|
Sidhaye J, Knoblich JA. Brain organoids: an ensemble of bioassays to investigate human neurodevelopment and disease. Cell Death Differ 2021; 28:52-67. [PMID: 32483384 PMCID: PMC7853143 DOI: 10.1038/s41418-020-0566-4] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 05/07/2020] [Accepted: 05/15/2020] [Indexed: 12/12/2022] Open
Abstract
Understanding etiology of human neurological and psychiatric diseases is challenging. Genomic changes, protracted development, and histological features unique to human brain development limit the disease aspects that can be investigated using model organisms. Hence, in order to study phenotypes associated with human brain development, function, and disease, it is necessary to use alternative experimental systems that are accessible, ethically justified, and replicate human context. Human pluripotent stem cell (hPSC)-derived brain organoids offer such a system, which recapitulates features of early human neurodevelopment in vitro, including the generation, proliferation, and differentiation of neural progenitors into neurons and glial cells and the complex interactions among the diverse, emergent cell types of the developing brain in three-dimensions (3-D). In recent years, numerous brain organoid protocols and related techniques have been developed to recapitulate aspects of embryonic and fetal brain development in a reproducible and predictable manner. Altogether, these different organoid technologies provide distinct bioassays to unravel novel, disease-associated phenotypes and mechanisms. In this review, we summarize how the diverse brain organoid methods can be utilized to enhance our understanding of brain disorders. FACTS: Brain organoids offer an in vitro approach to study aspects of human brain development and disease. Diverse brain organoid techniques offer bioassays to investigate new phenotypes associated with human brain disorders that are difficult to study in monolayer cultures. Brain organoids have been particularly useful to study phenomena and diseases associated with neural progenitor morphology, survival, proliferation, and differentiation. OPEN QUESTION: Future brain organoid research needs to aim at later stages of neurodevelopment, linked with neuronal activity and connections, to unravel further disease-associated phenotypes. Continued improvement of existing organoid protocols is required to generate standardized methods that recapitulate in vivo-like spatial diversity and complexity.
Collapse
Affiliation(s)
- Jaydeep Sidhaye
- Institute of Molecular Biotechnology of Austrian academy of sciences (IMBA), Vienna BioCenter (VBC), Dr. Bohr-Gasse 3, 1030, Vienna, Austria
| | - Jürgen A Knoblich
- Institute of Molecular Biotechnology of Austrian academy of sciences (IMBA), Vienna BioCenter (VBC), Dr. Bohr-Gasse 3, 1030, Vienna, Austria.
| |
Collapse
|
511
|
Abstract
Engineered human mini-brains, made possible by knowledge from the convergence of precision microengineering and cell biology, permit systematic studies of complex neurological processes and of pathogenesis beyond what can be done with animal models. By culturing human brain cells with physiological microenvironmental cues, human mini-brain models reconstitute the arrangement of structural tissues and some of the complex biological functions of the human brain. In this Review, we highlight the most significant developments that have led to microphysiological human mini-brain models. We introduce the history of mini-brain development, review methods for creating mini-brain models in static conditions, and discuss relevant state-of-the-art dynamic cell-culture systems. We also review human mini-brain models that reconstruct aspects of major neurological disorders under static or dynamic conditions. Engineered human mini-brains will contribute to advancing the study of the physiology and aetiology of neurological disorders, and to the development of personalized medicines for them.
Collapse
|
512
|
Cao Y, Li S, Chen J. Modeling better in vitro models for the prediction of nanoparticle toxicity: a review. Toxicol Mech Methods 2021; 31:1-17. [PMID: 32972312 DOI: 10.1080/15376516.2020.1828521] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/22/2020] [Accepted: 09/21/2020] [Indexed: 02/07/2023]
Abstract
Exposure to nanoparticles (NPs) is plausible in real life due to ambient particulate exposure or development of nanotechnologies, hence the evaluation of NP toxicity as well as mechanism-based studies are necessary. The in vitro models allow rapid testing of NP toxicity, but it is required that the developed in vitro models are reliable to reflect the toxicity of NPs. In this review, we discussed the principles to model better in vitro models to predict the toxicity of NPs based on our own experiences and works of literature. We suggested that in vitro nanotoxicological studies should consider (1) using normal cells because the commonly used cancer cell lines might not reflect the toxicity of NPs to normal tissues; (2) the possible influence of biological molecules to reflect the toxicity of NPs in a biological microenvironment; (3) the influence of pathophysiological conditions to mimic the responses of NPs under different in vivo conditions; and (4) developing advanced tissue-based models to reflect the responses of tissues/organs to NPs. It is our hope that this review may provide useful information for the future design of in vitro nanotoxicological studies.
Collapse
Affiliation(s)
- Yi Cao
- Key Laboratory of Environment-Friendly Chemistry and Applications of Ministry Education, Laboratory of Biochemistry, College of Chemistry, Xiangtan University, Xiangtan, P. R. China
| | - Shuang Li
- Key Laboratory of Environment-Friendly Chemistry and Applications of Ministry Education, Laboratory of Biochemistry, College of Chemistry, Xiangtan University, Xiangtan, P. R. China
| | - Jiamao Chen
- Key Laboratory of Environment-Friendly Chemistry and Applications of Ministry Education, Laboratory of Biochemistry, College of Chemistry, Xiangtan University, Xiangtan, P. R. China
| |
Collapse
|
513
|
Weitz J, Menegaz D, Caicedo A. Deciphering the Complex Communication Networks That Orchestrate Pancreatic Islet Function. Diabetes 2021; 70:17-26. [PMID: 33355306 PMCID: PMC7881851 DOI: 10.2337/dbi19-0033] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 10/01/2020] [Indexed: 12/27/2022]
Abstract
Pancreatic islets are clusters of hormone-secreting endocrine cells that rely on intricate cell-cell communication mechanisms for proper function. The importance of multicellular cooperation in islet cell physiology was first noted nearly 30 years ago in seminal studies showing that hormone secretion from endocrine cell types is diminished when these cells are dispersed. These studies showed that reestablishing cellular contacts in so-called pseudoislets caused endocrine cells to regain hormone secretory function. This not only demonstrated that cooperation between islet cells is highly synergistic but also gave birth to the field of pancreatic islet organoids. Here we review recent advances related to the mechanisms of islet cell cross talk. We first describe new developments that revise current notions about purinergic and GABA signaling in islets. Then we comment on novel multicellular imaging studies that are revealing emergent properties of islet communication networks. We finish by highlighting and discussing recent synthetic approaches that use islet organoids of varied cellular composition to interrogate intraislet signaling mechanisms. This reverse engineering of islets not only will shed light on the mechanisms of intraislet signaling and define communication networks but also may guide efforts aimed at restoring islet function and β-cell mass in diabetes.
Collapse
Affiliation(s)
- Jonathan Weitz
- Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, FL
- Diabetes Research Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL
| | - Danusa Menegaz
- Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, FL
| | - Alejandro Caicedo
- Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, FL
- Diabetes Research Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL
- Department of Physiology and Biophysics, University of Miami Leonard M. Miller School of Medicine, Miami, FL
- Program in Neuroscience, University of Miami Leonard M. Miller School of Medicine, Miami, FL
| |
Collapse
|
514
|
Tran F, Klein C, Arlt A, Imm S, Knappe E, Simmons A, Rosenstiel P, Seibler P. Stem Cells and Organoid Technology in Precision Medicine in Inflammation: Are We There Yet? Front Immunol 2020; 11:573562. [PMID: 33408713 PMCID: PMC7779798 DOI: 10.3389/fimmu.2020.573562] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 11/19/2020] [Indexed: 12/12/2022] Open
Abstract
Individualised cellular models of disease are a key tool for precision medicine to recapitulate chronic inflammatory processes. Organoid models can be derived from induced pluripotent stem cells (iPSCs) or from primary stem cells ex vivo. These models have been emerging over the past decade and have been used to reconstruct the respective organ-specific physiology and pathology, at an unsurpassed depth. In cancer research, patient-derived cancer organoids opened new perspectives in predicting therapy response and provided novel insights into tumour biology. In precision medicine of chronic inflammatory disorders, stem-cell based organoid models are currently being evaluated in pre-clinical pharmacodynamic studies (clinical studies in a dish) and are employed in clinical studies, e.g., by re-transplanting autologous epithelial organoids to re-establish intestinal barrier integrity. A particularly exciting feature of iPSC systems is their ability to provide insights into organ systems and inflammatory disease processes, which cannot be monitored with clinical biopsies, such as immune reactions in neurodegenerative disorders. Refinement of differentiation protocols, and next-generation co-culturing methods, aimed at generating self-organised, complex tissues in vitro, will be the next logical steps. In this mini-review, we critically discuss the current state-of-the-art stem cell and organoid technologies, as well as their future impact, potential and promises in combating immune-mediated chronic diseases.
Collapse
Affiliation(s)
- Florian Tran
- Institute of Clinical Molecular Biology, University of Kiel, Kiel, Germany.,Klinik für Innere Medizin I, Universitätsklinikum Schleswig-Holstein, Kiel, Germany
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Alexander Arlt
- Klinik für Innere Medizin I, Universitätsklinikum Schleswig-Holstein, Kiel, Germany.,University Department for Gastroenterology, Klinikum Oldenburg AöR, European Medical School (EMS), Oldenburg, Germany
| | - Simon Imm
- Institute of Clinical Molecular Biology, University of Kiel, Kiel, Germany
| | - Evelyn Knappe
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Alison Simmons
- MRC Human Immunology Unit (MRC), University of Oxford, Oxford, United Kingdom.,Translational Gastroenterology Unit, University of Oxford, Oxford, United Kingdom
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, University of Kiel, Kiel, Germany
| | - Philip Seibler
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| |
Collapse
|
515
|
Giandomenico SL, Sutcliffe M, Lancaster MA. Generation and long-term culture of advanced cerebral organoids for studying later stages of neural development. Nat Protoc 2020; 16:579-602. [PMID: 33328611 DOI: 10.1038/s41596-020-00433-w] [Citation(s) in RCA: 149] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 10/02/2020] [Indexed: 11/09/2022]
Abstract
Cerebral organoids, or brain organoids, can be generated from a wide array of emerging technologies for modeling brain development and disease. The fact that they are cultured in vitro makes them easily accessible both genetically and for live assays such as fluorescence imaging. In this Protocol Extension, we describe a modified version of our original protocol (published in 2014) that can be used to reliably generate cerebral organoids of a telencephalic identity and maintain long-term viability for later stages of neural development, including axon outgrowth and neuronal maturation. The method builds upon earlier cerebral organoid methodology, with modifications of embryoid body size and shape to increase surface area and slice culture to maintain nutrient and oxygen access to the interior regions of the organoid, enabling long-term culture. We also describe approaches for introducing exogenous plasmid constructs and for sparse cell labeling to image neuronal axon outgrowth and maturation over time. Together, these methods allow for modeling of later events in cortical development, which are important for neurodevelopmental disease modeling. The protocols described can be easily performed by an experimenter with stem cell culture experience and take 2-3 months to complete, with long-term maturation occurring over several months.
Collapse
Affiliation(s)
| | - Magdalena Sutcliffe
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Madeline A Lancaster
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK. .,Wellcome Trust-MRC Cambridge Stem Cell Institute, Cambridge Biomedical Campus, Cambridge, UK.
| |
Collapse
|
516
|
van der Valk WH, Steinhart MR, Zhang J, Koehler KR. Building inner ears: recent advances and future challenges for in vitro organoid systems. Cell Death Differ 2020; 28:24-34. [PMID: 33318601 PMCID: PMC7853146 DOI: 10.1038/s41418-020-00678-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 02/07/2023] Open
Abstract
While inner ear disorders are common, our ability to intervene and recover their sensory function is limited. In vitro models of the inner ear, like the organoid system, could aid in identifying new regenerative drugs and gene therapies. Here, we provide a perspective on the status of in vitro inner ear models and guidance on how to improve their applicability in translational research. We highlight the generation of inner ear cell types from pluripotent stem cells as a particularly promising focus of research. Several exciting recent studies have shown how the developmental signaling cues of embryonic and fetal development can be mimicked to differentiate stem cells into “inner ear organoids” containing otic progenitor cells, hair cells, and neurons. However, current differentiation protocols and our knowledge of embryonic and fetal inner ear development in general, have a bias toward the sensory epithelia of the inner ear. We propose that a more holistic view is needed to better model the inner ear in vitro. Moving forward, attention should be made to the broader diversity of neuroglial and mesenchymal cell types of the inner ear, and how they interact in space or time during development. With improved control of epithelial, neuroglial, and mesenchymal cell fate specification, inner ear organoids would have the ability to truly recapitulate neurosensory function and dysfunction. We conclude by discussing how single-cell atlases of the developing inner ear and technical innovations will be critical tools to advance inner ear organoid platforms for future pre-clinical applications.
Collapse
Affiliation(s)
- Wouter H van der Valk
- Department of Otorhinolaryngology and Head & Neck Surgery, Leiden University Medical Center, Leiden, Netherlands.,Department of Otolaryngology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Matthew R Steinhart
- Department of Otolaryngology, Boston Children's Hospital, Boston, MA, 02115, USA.,Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Medical Neuroscience Graduate Program, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jingyuan Zhang
- Department of Otolaryngology, Boston Children's Hospital, Boston, MA, 02115, USA.,Department of Plastic and Oral Surgery, Boston Children's Hospital, Boston, MA, 02115, USA.,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, 02115, USA.,Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Karl R Koehler
- Department of Otolaryngology, Boston Children's Hospital, Boston, MA, 02115, USA. .,Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA. .,Department of Plastic and Oral Surgery, Boston Children's Hospital, Boston, MA, 02115, USA. .,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, 02115, USA. .,Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
517
|
Current and Future Perspectives of the Use of Organoids in Radiobiology. Cells 2020; 9:cells9122649. [PMID: 33317153 PMCID: PMC7764598 DOI: 10.3390/cells9122649] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/04/2020] [Accepted: 12/08/2020] [Indexed: 02/07/2023] Open
Abstract
The majority of cancer patients will be treated with radiotherapy, either alone or together with chemotherapy and/or surgery. Optimising the balance between tumour control and the probability of normal tissue side effects is the primary goal of radiation treatment. Therefore, it is imperative to understand the effects that irradiation will have on both normal and cancer tissue. The more classical lab models of immortal cell lines and in vivo animal models have been fundamental to radiobiological studies to date. However, each of these comes with their own limitations and new complementary models are required to fill the gaps left by these traditional models. In this review, we discuss how organoids, three-dimensional tissue-resembling structures derived from tissue-resident, embryonic or induced pluripotent stem cells, overcome the limitations of these models and thus have a growing importance in the field of radiation biology research. The roles of organoids in understanding radiation-induced tissue responses and in moving towards precision medicine are examined. Finally, the limitations of organoids in radiobiology and the steps being made to overcome these limitations are considered.
Collapse
|
518
|
Klein E, Hau AC, Oudin A, Golebiewska A, Niclou SP. Glioblastoma Organoids: Pre-Clinical Applications and Challenges in the Context of Immunotherapy. Front Oncol 2020; 10:604121. [PMID: 33364198 PMCID: PMC7753120 DOI: 10.3389/fonc.2020.604121] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 11/09/2020] [Indexed: 12/13/2022] Open
Abstract
Malignant brain tumors remain uniformly fatal, even with the best-to-date treatment. For Glioblastoma (GBM), the most severe form of brain cancer in adults, the median overall survival is roughly over a year. New therapeutic options are urgently needed, yet recent clinical trials in the field have been largely disappointing. This is partially due to inappropriate preclinical model systems, which do not reflect the complexity of patient tumors. Furthermore, clinically relevant patient-derived models recapitulating the immune compartment are lacking, which represents a bottleneck for adequate immunotherapy testing. Emerging 3D organoid cultures offer innovative possibilities for cancer modeling. Here, we review available GBM organoid models amenable to a large variety of pre-clinical applications including functional bioassays such as proliferation and invasion, drug screening, and the generation of patient-derived orthotopic xenografts (PDOX) for validation of biological responses in vivo. We emphasize advantages and technical challenges in establishing immunocompetent ex vivo models based on co-cultures of GBM organoids and human immune cells. The latter can be isolated either from the tumor or from patient or donor blood as peripheral blood mononuclear cells (PBMCs). We also discuss the challenges to generate GBM PDOXs based on humanized mouse models to validate efficacy of immunotherapies in vivo. A detailed characterization of such models at the cellular and molecular level is needed to understand the potential and limitations for various immune activating strategies. Increasing the availability of immunocompetent GBM models will improve research on emerging immune therapeutic approaches against aggressive brain cancer.
Collapse
Affiliation(s)
- Eliane Klein
- NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Ann-Christin Hau
- NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Anaïs Oudin
- NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Anna Golebiewska
- NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Simone P. Niclou
- NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
- Department of Biomedicine, University of Bergen, Bergen, Norway
| |
Collapse
|
519
|
Albanese A, Swaney JM, Yun DH, Evans NB, Antonucci JM, Velasco S, Sohn CH, Arlotta P, Gehrke L, Chung K. Multiscale 3D phenotyping of human cerebral organoids. Sci Rep 2020; 10:21487. [PMID: 33293587 PMCID: PMC7723053 DOI: 10.1038/s41598-020-78130-7] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 10/27/2020] [Indexed: 01/28/2023] Open
Abstract
Brain organoids grown from human pluripotent stem cells self-organize into cytoarchitectures resembling the developing human brain. These three-dimensional models offer an unprecedented opportunity to study human brain development and dysfunction. Characterization currently sacrifices spatial information for single-cell or histological analysis leaving whole-tissue analysis mostly unexplored. Here, we present the SCOUT pipeline for automated multiscale comparative analysis of intact cerebral organoids. Our integrated technology platform can rapidly clear, label, and image intact organoids. Algorithmic- and convolutional neural network-based image analysis extract hundreds of features characterizing molecular, cellular, spatial, cytoarchitectural, and organoid-wide properties from fluorescence microscopy datasets. Comprehensive analysis of 46 intact organoids and ~ 100 million cells reveals quantitative multiscale "phenotypes" for organoid development, culture protocols and Zika virus infection. SCOUT provides a much-needed framework for comparative analysis of emerging 3D in vitro models using fluorescence microscopy.
Collapse
Affiliation(s)
- Alexandre Albanese
- Institute for Medical Engineering and Science, MIT, Cambridge, MA, USA
- Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA
| | | | - Dae Hee Yun
- Institute for Medical Engineering and Science, MIT, Cambridge, MA, USA
- Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | - Nicholas B Evans
- Institute for Medical Engineering and Science, MIT, Cambridge, MA, USA
- Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA
| | - Jenna M Antonucci
- Institute for Medical Engineering and Science, MIT, Cambridge, MA, USA
| | - 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
| | - Chang Ho Sohn
- Institute for Medical Engineering and Science, MIT, Cambridge, MA, USA
- Picower Institute for Learning and Memory, MIT, 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
| | - Lee Gehrke
- Institute for Medical Engineering and Science, MIT, Cambridge, MA, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, 02115, USA
- Harvard-MIT Program in Health Sciences and Technology, Cambridge, MA, 02139, USA
| | - Kwanghun Chung
- Institute for Medical Engineering and Science, MIT, Cambridge, MA, USA.
- Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA.
- Department of Chemical Engineering, MIT, Cambridge, MA, USA.
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA.
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea.
- Yonsei-IBS Institute, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
520
|
Matsui TK, Tsuru Y, Kuwako KI. Challenges in Modeling Human Neural Circuit Formation via Brain Organoid Technology. Front Cell Neurosci 2020; 14:607399. [PMID: 33362473 PMCID: PMC7756199 DOI: 10.3389/fncel.2020.607399] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 11/12/2020] [Indexed: 01/12/2023] Open
Abstract
Human brain organoids are three-dimensional self-organizing tissues induced from pluripotent cells that recapitulate some aspects of early development and some of the early structure of the human brain in vitro. Brain organoids consist of neural lineage cells, such as neural stem/precursor cells, neurons, astrocytes and oligodendrocytes. Additionally, brain organoids contain fluid-filled ventricle-like structures surrounded by a ventricular/subventricular (VZ/SVZ) zone-like layer of neural stem cells (NSCs). These NSCs give rise to neurons, which form multiple outer layers. Since these structures resemble some aspects of structural arrangements in the developing human brain, organoid technology has attracted great interest in the research fields of human brain development and disease modeling. Developmental brain disorders have been intensely studied through the use of human brain organoids. Relatively early steps in human brain development, such as differentiation and migration, have also been studied. However, research on neural circuit formation with brain organoids has just recently began. In this review, we summarize the current challenges in studying neural circuit formation with organoids and discuss future perspectives.
Collapse
Affiliation(s)
- Takeshi K Matsui
- Department of Neural and Muscular Physiology, Shimane University School of Medicine, Izumo, Japan
| | - Yuichiro Tsuru
- Department of Neural and Muscular Physiology, Shimane University School of Medicine, Izumo, Japan
| | - Ken-Ichiro Kuwako
- Department of Neural and Muscular Physiology, Shimane University School of Medicine, Izumo, Japan
| |
Collapse
|
521
|
Trends and challenges in modeling glioma using 3D human brain organoids. Cell Death Differ 2020; 28:15-23. [PMID: 33262470 PMCID: PMC7707134 DOI: 10.1038/s41418-020-00679-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/06/2020] [Accepted: 11/06/2020] [Indexed: 12/21/2022] Open
Abstract
The human brain organoids derived from pluripotent cells are a new class of three-dimensional tissue systems that recapitulates several neural epithelial aspects. Brain organoids have already helped efficient modeling of crucial elements of brain development and disorders. Brain organoids’ suitability in modeling glioma has started to emerge, offering another usefulness of brain organoids in disease modeling. Although the current state-of-the organoids mostly reflect the immature state of the brain, with their vast cell diversity, human brain-like cytoarchitecture, feasibility in culturing, handling, imaging, and tractability can offer enormous potential in reflecting the glioma invasion, integration, and interaction with different neuronal cell types. Here, we summarize the current trend of employing brain organoids in glioma modeling and discuss the immediate challenges. Solving them might lay a foundation for using brain organoids as a pre-clinical 3D substrate to dissect the glioma invasion mechanisms in detail.
Collapse
|
522
|
Lee CS, Leong KW. Advances in microphysiological blood-brain barrier (BBB) models towards drug delivery. Curr Opin Biotechnol 2020; 66:78-87. [PMID: 32711361 PMCID: PMC7744339 DOI: 10.1016/j.copbio.2020.06.009] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/12/2020] [Accepted: 06/15/2020] [Indexed: 12/22/2022]
Abstract
Though the blood-brain barrier (BBB) is vital for the maintenance of brain homeostasis, it also accounts for a high attrition rate of therapies targeting the central nervous system (CNS). The challenge of delivery across the BBB is attributed to a combination of low permeability through an endothelium closely knit by tight and adherens junctions, extremely low rates of endothelial transcytosis, and efflux transporters. In the past decade, enormous research efforts have been spent to develop BBB penetration strategies using biochemical or physical stimuli, aided by BBB-on-chips or microphysiological BBB models to facilitate in vitro studies. Here, we discuss recent advances in BBB-chip technology that have enabled effective preclinical screenings of brain targeting therapeutics and external stimulation, such as sonoporation and electroporation, for improved BBB penetration.
Collapse
Affiliation(s)
- Caleb S Lee
- Columbia University, NY, NY, 10027, United States
| | - Kam W Leong
- Columbia University, NY, NY, 10027, United States.
| |
Collapse
|
523
|
Papaioannou MD, Sangster K, Sajid RS, Djuric U, Diamandis P. Cerebral organoids: emerging ex vivo humanoid models of glioblastoma. Acta Neuropathol Commun 2020; 8:209. [PMID: 33261657 PMCID: PMC7706050 DOI: 10.1186/s40478-020-01077-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/11/2020] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma is an aggressive form of brain cancer that has seen only marginal improvements in its bleak survival outlook of 12-15 months over the last forty years. There is therefore an urgent need for the development of advanced drug screening platforms and systems that can better recapitulate glioblastoma's infiltrative biology, a process largely responsible for its relentless propensity for recurrence and progression. Recent advances in stem cell biology have allowed the generation of artificial tridimensional brain-like tissue termed cerebral organoids. In addition to their potential to model brain development, these reagents are providing much needed synthetic humanoid scaffolds to model glioblastoma's infiltrative capacity in a faithful and scalable manner. Here, we highlight and review the early breakthroughs in this growing field and discuss its potential future role for glioblastoma research.
Collapse
Affiliation(s)
- Michail-Dimitrios Papaioannou
- Princess Margaret Cancer Centre, 101 College Street, Toronto, ON, M5G 1L7, Canada
- Laboratory Medicine Program, Department of Pathology, University Health Network, 200 Elizabeth Street, Toronto, ON, M5G 2C4, Canada
| | - Kevin Sangster
- Princess Margaret Cancer Centre, 101 College Street, Toronto, ON, M5G 1L7, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Rifat Shahriar Sajid
- Princess Margaret Cancer Centre, 101 College Street, Toronto, ON, M5G 1L7, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Ugljesa Djuric
- Princess Margaret Cancer Centre, 101 College Street, Toronto, ON, M5G 1L7, Canada
- Laboratory Medicine Program, Department of Pathology, University Health Network, 200 Elizabeth Street, Toronto, ON, M5G 2C4, Canada
| | - Phedias Diamandis
- Princess Margaret Cancer Centre, 101 College Street, Toronto, ON, M5G 1L7, Canada.
- Laboratory Medicine Program, Department of Pathology, University Health Network, 200 Elizabeth Street, Toronto, ON, M5G 2C4, Canada.
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G 1L7, Canada.
| |
Collapse
|
524
|
Muckom RJ, Sampayo RG, Johnson HJ, Schaffer DV. Advanced Materials to Enhance Central Nervous System Tissue Modeling and Cell Therapy. ADVANCED FUNCTIONAL MATERIALS 2020; 30:2002931. [PMID: 33510596 PMCID: PMC7840150 DOI: 10.1002/adfm.202002931] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Indexed: 05/04/2023]
Abstract
The progressively deeper understanding of mechanisms underlying stem cell fate decisions has enabled parallel advances in basic biology-such as the generation of organoid models that can further one's basic understanding of human development and disease-and in clinical translation-including stem cell based therapies to treat human disease. Both of these applications rely on tight control of the stem cell microenvironment to properly modulate cell fate, and materials that can be engineered to interface with cells in a controlled and tunable manner have therefore emerged as valuable tools for guiding stem cell growth and differentiation. With a focus on the central nervous system (CNS), a broad range of material solutions that have been engineered to overcome various hurdles in constructing advanced organoid models and developing effective stem cell therapeutics is reviewed. Finally, regulatory aspects of combined material-cell approaches for CNS therapies are considered.
Collapse
Affiliation(s)
- Riya J Muckom
- Department of Chemical and Biomolecular Engineering, UC Berkeley, Berkeley, CA 94704, USA
| | - Rocío G Sampayo
- Department of Chemical and Biomolecular Engineering, UC Berkeley, Berkeley, CA 94704, USA
| | - Hunter J Johnson
- Department of Bioengineering, UC Berkeley, Berkeley, CA 94704, USA
| | - David V Schaffer
- Department of Chemical and Biomolecular Engineering, UC Berkeley, Berkeley, CA 94704, USA
| |
Collapse
|
525
|
Learning about cell lineage, cellular diversity and evolution of the human brain through stem cell models. Curr Opin Neurobiol 2020; 66:166-177. [PMID: 33246264 DOI: 10.1016/j.conb.2020.10.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/19/2020] [Accepted: 10/22/2020] [Indexed: 02/07/2023]
Abstract
Here, we summarize the current knowledge on cell diversity in the cortex and other brain regions from in vivo mouse models and in vitro models based on pluripotent stem cells. We discuss the mechanisms underlying cell proliferation and temporal progression that leads to the sequential generation of neurons dedicated to different layers of the cortex. We highlight models of corticogenesis from stem cells that recapitulate specific transcriptional and connectivity patterns from different cortical areas. We overview state-of-the art of human brain organoids modeling different brain regions, and we discuss insights into human cortical evolution from stem cells. Finally, we interrogate human brain organoid models for their competence to recapitulate the essence of human brain development.
Collapse
|
526
|
Upgrading the Physiological Relevance of Human Brain Organoids. Neuron 2020; 107:1014-1028. [PMID: 32970996 PMCID: PMC10042151 DOI: 10.1016/j.neuron.2020.08.029] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/17/2020] [Accepted: 08/27/2020] [Indexed: 02/06/2023]
Abstract
The recent advent of human pluripotent stem cell (PSC)-derived 3D brain organoids has opened a window into aspects of human brain development that were not accessible before, allowing tractable monitoring and assessment of early developmental processes. However, their broad and effective use for modeling later stages of human brain development and disease is hampered by the lack of a stereotypic anatomical organization, which limits maturation processes dependent upon formation of unique cellular interactions and short- and long-range network connectivity. Emerging methods and technologies aimed at tighter regulatory control through bioengineering approaches, along with newer unbiased organoid analysis readouts, should resolve several of the current limitations. Here, we review recent advances in brain organoid generation and characterization with a focus on highlighting future directions utilizing interdisciplinary strategies that will be important for improving the physiological relevance of this model system.
Collapse
|
527
|
Robert J, Weilinger NL, Cao LP, Cataldi S, Button EB, Stukas S, Martin EM, Seibler P, Gilmour M, Caffrey TM, Rowe EM, Fan J, MacVicar B, Farrer MJ, Wellington CL. An in vitro bioengineered model of the human arterial neurovascular unit to study neurodegenerative diseases. Mol Neurodegener 2020; 15:70. [PMID: 33213497 PMCID: PMC7678181 DOI: 10.1186/s13024-020-00418-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 11/03/2020] [Indexed: 01/04/2023] Open
Abstract
INTRODUCTION The neurovascular unit (NVU) - the interaction between the neurons and the cerebrovasculature - is increasingly important to interrogate through human-based experimental models. Although advanced models of cerebral capillaries have been developed in the last decade, there is currently no in vitro 3-dimensional (3D) perfusible model of the human cortical arterial NVU. METHOD We used a tissue-engineering technique to develop a scaffold-directed, perfusible, 3D human NVU that is cultured in native-like flow conditions that mimics the anatomy and physiology of cortical penetrating arteries. RESULTS This system, composed of primary human vascular cells (endothelial cells, smooth muscle cells and astrocytes) and induced pluripotent stem cell (iPSC) derived neurons, demonstrates a physiological multilayer organization of the involved cell types. It reproduces key characteristics of cortical neurons and astrocytes and enables formation of a selective and functional endothelial barrier. We provide proof-of-principle data showing that this in vitro human arterial NVU may be suitable to study neurovascular components of neurodegenerative diseases such as Alzheimer's disease (AD), as endogenously produced phosphorylated tau and beta-amyloid accumulate in the model over time. Finally, neuronal and glial fluid biomarkers relevant to neurodegenerative diseases are measurable in our arterial NVU model. CONCLUSION This model is a suitable research tool to investigate arterial NVU functions in healthy and disease states. Further, the design of the platform allows culture under native-like flow conditions for extended periods of time and yields sufficient tissue and media for downstream immunohistochemistry and biochemistry analyses.
Collapse
Affiliation(s)
- Jerome Robert
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V6T 1Z3 Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, British Columbia V6T 1Z3 Canada
- Institute of Clinical Chemistry, University hospital Zurich, 8000 Zurich, Wagistrasse 14, CH-8952 Schlieren, Switzerland
| | - Nicholas L. Weilinger
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, British Columbia V6T 1Z3 Canada
| | - Li-Ping Cao
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V6T 1Z3 Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, British Columbia V6T 1Z3 Canada
- Centre for Applied Neurogenetics, University of British Columbia, Vancouver, British Columbia V6T 1Z3 Canada
| | - Stefano Cataldi
- Centre for Applied Neurogenetics, University of British Columbia, Vancouver, British Columbia V6T 1Z3 Canada
| | - Emily B. Button
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V6T 1Z3 Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, British Columbia V6T 1Z3 Canada
| | - Sophie Stukas
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V6T 1Z3 Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, British Columbia V6T 1Z3 Canada
| | - Emma M. Martin
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V6T 1Z3 Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, British Columbia V6T 1Z3 Canada
| | - Philip Seibler
- Institute of Neurogenetics, University of Luebeck, 23562 Luebeck, Germany
| | - Megan Gilmour
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V6T 1Z3 Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, British Columbia V6T 1Z3 Canada
| | - Tara M. Caffrey
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V6T 1Z3 Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, British Columbia V6T 1Z3 Canada
| | - Elyn M. Rowe
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V6T 1Z3 Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, British Columbia V6T 1Z3 Canada
| | - Jianjia Fan
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V6T 1Z3 Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, British Columbia V6T 1Z3 Canada
| | - Brian MacVicar
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, British Columbia V6T 1Z3 Canada
| | - Matthew J. Farrer
- Centre for Applied Neurogenetics, University of British Columbia, Vancouver, British Columbia V6T 1Z3 Canada
- Laboratory for Neurogenetics & Neuroscience, McKnight and Fixel Institutes, University of Florida, Gainesville, 32610 USA
| | - Cheryl L. Wellington
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V6T 1Z3 Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, British Columbia V6T 1Z3 Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia V6T 1Z3 Canada
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, British Columbia V5Z 1M9 Canada
| |
Collapse
|
528
|
Parvatam S, Bharadwaj S, Radha V, Rao M. The need to develop a framework for human-relevant research in India: Towards better disease models and drug discovery. J Biosci 2020. [DOI: 10.1007/s12038-020-00112-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
529
|
Venkataraman L, Fair SR, McElroy CA, Hester ME, Fu H. Modeling neurodegenerative diseases with cerebral organoids and other three-dimensional culture systems: focus on Alzheimer's disease. Stem Cell Rev Rep 2020; 18:696-717. [PMID: 33180261 PMCID: PMC7658915 DOI: 10.1007/s12015-020-10068-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2020] [Indexed: 12/11/2022]
Abstract
Many neurodegenerative diseases (NDs) such as Alzheimer’s disease, Parkinson’s disease, frontotemporal dementia, amyotrophic lateral sclerosis and Huntington’s disease, are characterized by the progressive accumulation of abnormal proteinaceous assemblies in specific cell types and regions of the brain, leading to cellular dysfunction and brain damage. Although animal- and in vitro-based studies of NDs have provided the field with an extensive understanding of some of the mechanisms underlying these diseases, findings from these studies have not yielded substantial progress in identifying treatment options for patient populations. This necessitates the development of complementary model systems that are better suited to recapitulate human-specific features of ND pathogenesis. Three-dimensional (3D) culture systems, such as cerebral organoids generated from human induced pluripotent stem cells, hold significant potential to model NDs in a complex, tissue-like environment. In this review, we discuss the advantages of 3D culture systems and 3D modeling of NDs, especially AD and FTD. We also provide an overview of the challenges and limitations of the current 3D culture systems. Finally, we propose a few potential future directions in applying state-of-the-art technologies in 3D culture systems to understand the mechanisms of NDs and to accelerate drug discovery. Graphical abstract ![]()
Collapse
Affiliation(s)
- Lalitha Venkataraman
- Department of Neuroscience, The Ohio State University Wexner Medical Center, 616 Biomedical Research Tower, 460 W. 12th Ave, Columbus, OH, 43210, USA
| | - Summer R Fair
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, 575 Children's Crossroad, Columbus, OH, 43215, USA
- College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Craig A McElroy
- College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Mark E Hester
- Department of Neuroscience, The Ohio State University Wexner Medical Center, 616 Biomedical Research Tower, 460 W. 12th Ave, Columbus, OH, 43210, USA.
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, 575 Children's Crossroad, Columbus, OH, 43215, USA.
- Department of Pediatrics, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| | - Hongjun Fu
- Department of Neuroscience, The Ohio State University Wexner Medical Center, 616 Biomedical Research Tower, 460 W. 12th Ave, Columbus, OH, 43210, USA.
| |
Collapse
|
530
|
Li YCE, Jodat YA, Samanipour R, Zorzi G, Zhu K, Hirano M, Chang K, Arnaout A, Hassan S, Matharu N, Khademhosseini A, Hoorfar M, Shin SR. Toward a neurospheroid niche model: optimizing embedded 3D bioprinting for fabrication of neurospheroid brain-like co-culture constructs. Biofabrication 2020; 13:10.1088/1758-5090/abc1be. [PMID: 33059333 PMCID: PMC8387028 DOI: 10.1088/1758-5090/abc1be] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 10/15/2020] [Indexed: 12/24/2022]
Abstract
A crucial step in creating reliablein vitroplatforms for neural development and disorder studies is the reproduction of the multicellular three-dimensional (3D) brain microenvironment and the capturing of cell-cell interactions within the model. The power of self-organization of diverse cell types into brain spheroids could be harnessed to study mechanisms underlying brain development trajectory and diseases. A challenge of current 3D organoid and spheroid models grown in petri-dishes is the lack of control over cellular localization and diversity. To overcome this limitation, neural spheroids can be patterned into customizable 3D structures using microfabrication. We developed a 3D brain-like co-culture construct using embedded 3D bioprinting as a flexible solution for composing heterogenous neural populations with neurospheroids and glia. Specifically, neurospheroid-laden free-standing 3D structures were fabricated in an engineered astrocyte-laden support bath resembling a neural stem cell niche environment. A photo-crosslinkable bioink and a thermal-healing supporting bath were engineered to mimic the mechanical modulus of soft tissue while supporting the formation of self-organizing neurospheroids within elaborate 3D networks. Moreover, bioprinted neurospheroid-laden structures exhibited the capability to differentiate into neuronal cells. These brain-like co-cultures could provide a reproducible platform for modeling neurological diseases, neural regeneration, and drug development and repurposing.
Collapse
Affiliation(s)
- Yi-Chen Ethan Li
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02139, United States of America
- Department of Chemical Engineering, Feng Chia University, Taichung 40724, Taiwan
| | - Yasamin A Jodat
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02139, United States of America
- Department of Mechanical Engineering, Stevens Institute of Technology, New Jersey 07030, United States of America
| | - Roya Samanipour
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02139, United States of America
- School of Engineering, University of British Columbia, Kelowna V1V 1V7, BC, Canada
| | - Giulio Zorzi
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02139, United States of America
| | - Kai Zhu
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02139, United States of America
- Department of Cardiac Surgery, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Minoru Hirano
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02139, United States of America
- Future Vehicle Research Department, Toyota Research Institute North America, Toyota Motor North America Inc. 1555 Woodridge Ave, Ann Arbor, MI 48105, United States of America
| | - Karen Chang
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taiwan
| | - Adnan Arnaout
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02139, United States of America
| | - Shabir Hassan
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02139, United States of America
| | - Navneet Matharu
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California 94158, United States of America
- Institute for Human Genetics, University of California, San Francisco, CA 94158, United States of America
| | - Ali Khademhosseini
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02139, United States of America
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, California 90095, United States of America
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, California 90095, United States of America
- Department of Chemical and Biomolecular Engineering, Henry Samueli School of Engineering and Applied Sciences, University of California–Los Angeles, Los Angeles, California 90095, United States of America
- Department of Radiology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA 90095, United States of America
| | - Mina Hoorfar
- School of Engineering, University of British Columbia, Kelowna V1V 1V7, BC, Canada
| | - Su Ryon Shin
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02139, United States of America
| |
Collapse
|
531
|
Generation and biobanking of patient-derived glioblastoma organoids and their application in CAR T cell testing. Nat Protoc 2020; 15:4000-4033. [PMID: 33169003 DOI: 10.1038/s41596-020-0402-9] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 08/20/2020] [Indexed: 12/21/2022]
Abstract
Glioblastoma tumors exhibit extensive inter- and intratumoral heterogeneity, which has contributed to the poor outcomes of numerous clinical trials and continues to complicate the development of effective therapeutic strategies. Most in vitro models do not preserve the cellular and mutational diversity of parent tumors and often require a lengthy generation time with variable efficiency. Here, we describe detailed procedures for generating glioblastoma organoids (GBOs) from surgically resected patient tumor tissue using a chemically defined medium without cell dissociation. By preserving cell-cell interactions and minimizing clonal selection, GBOs maintain the cellular heterogeneity of parent tumors. We include details of how to passage and cryopreserve GBOs for continued use, biobanking and long-term recovery. In addition, we describe procedures for investigating patient-specific responses to immunotherapies by co-culturing GBOs with chimeric antigen receptor (CAR) T cells. It takes ~2-4 weeks to generate GBOs and 5-7 d to perform CAR T cell co-culture using this protocol. Competence with human cell culture, tissue processing, immunohistology and microscopy is required for optimal results.
Collapse
|
532
|
Zhang I, Lépine P, Han C, Lacalle-Aurioles M, Chen CXQ, Haag R, Durcan TM, Maysinger D. Nanotherapeutic Modulation of Human Neural Cells and Glioblastoma in Organoids and Monocultures. Cells 2020; 9:cells9112434. [PMID: 33171886 PMCID: PMC7695149 DOI: 10.3390/cells9112434] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 12/17/2022] Open
Abstract
Inflammatory processes in the brain are orchestrated by microglia and astrocytes in response to activators such as pathogen-associated molecular patterns, danger-associated molecular patterns and some nanostructures. Microglia are the primary immune responders in the brain and initiate responses amplified by astrocytes through intercellular signaling. Intercellular communication between neural cells can be studied in cerebral organoids, co-cultures or in vivo. We used human cerebral organoids and glioblastoma co-cultures to study glia modulation by dendritic polyglycerol sulfate (dPGS). dPGS is an extensively studied nanostructure with inherent anti-inflammatory properties. Under inflammatory conditions, lipocalin-2 levels in astrocytes are markedly increased and indirectly enhanced by soluble factors released from hyperactive microglia. dPGS is an effective anti-inflammatory modulator of these markers. Our results show that dPGS can enter neural cells in cerebral organoids and glial cells in monocultures in a time-dependent manner. dPGS markedly reduces lipocalin-2 abundance in the neural cells. Glioblastoma tumoroids of astrocytic origin respond to activated microglia with enhanced invasiveness, whereas conditioned media from dPGS-treated microglia reduce tumoroid invasiveness. Considering that many nanostructures have only been tested in cancer cells and rodent models, experiments in human 3D cerebral organoids and co-cultures are complementary in vitro models to evaluate nanotherapeutics in the pre-clinical setting. Thoroughly characterized organoids and standardized procedures for their preparation are prerequisites to gain information of translational value in nanomedicine. This study provides data for a well-characterized dendrimer (dPGS) that modulates the activation state of human microglia implicated in brain tumor invasiveness.
Collapse
Affiliation(s)
- Issan Zhang
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir-William-Osler, Montreal, QC H3G 1Y6, Canada;
| | - Paula Lépine
- The Neuro’s Early Drug Discovery Unit (EDDU), McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada; (P.L.); (C.H.); (M.L.-A.); (C.X.-Q.C.); (T.M.D.)
| | - Chanshuai Han
- The Neuro’s Early Drug Discovery Unit (EDDU), McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada; (P.L.); (C.H.); (M.L.-A.); (C.X.-Q.C.); (T.M.D.)
| | - María Lacalle-Aurioles
- The Neuro’s Early Drug Discovery Unit (EDDU), McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada; (P.L.); (C.H.); (M.L.-A.); (C.X.-Q.C.); (T.M.D.)
| | - Carol X.-Q. Chen
- The Neuro’s Early Drug Discovery Unit (EDDU), McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada; (P.L.); (C.H.); (M.L.-A.); (C.X.-Q.C.); (T.M.D.)
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustraße 3, 14195 Berlin, Germany;
| | - Thomas M. Durcan
- The Neuro’s Early Drug Discovery Unit (EDDU), McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada; (P.L.); (C.H.); (M.L.-A.); (C.X.-Q.C.); (T.M.D.)
| | - Dusica Maysinger
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir-William-Osler, Montreal, QC H3G 1Y6, Canada;
- Correspondence: ; Tel.: +1-514-398-1264
| |
Collapse
|
533
|
Chiaradia I, Lancaster MA. Brain organoids for the study of human neurobiology at the interface of in vitro and in vivo. Nat Neurosci 2020; 23:1496-1508. [PMID: 33139941 DOI: 10.1038/s41593-020-00730-3] [Citation(s) in RCA: 194] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 09/29/2020] [Indexed: 02/07/2023]
Abstract
Brain development is an extraordinarily complex process achieved through the spatially and temporally regulated release of key patterning factors. In vitro neurodevelopmental models seek to mimic these processes to recapitulate the steps of tissue fate acquisition and morphogenesis. Classic two-dimensional neural cultures present higher homogeneity but lower complexity compared to the brain. Brain organoids instead have more advanced cell composition, maturation and tissue architecture. They can thus be considered at the interface of in vitro and in vivo neurobiology, and further improvements in organoid techniques are continuing to narrow the gap with in vivo brain development. Here we describe these efforts to recapitulate brain development in neural organoids and focus on their applicability for disease modeling, evolutionary studies and neural network research.
Collapse
Affiliation(s)
- Ilaria Chiaradia
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Madeline A Lancaster
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK.
| |
Collapse
|
534
|
Shi Y, Wu Q, Wang X. Modeling brain development and diseases with human cerebral organoids. Curr Opin Neurobiol 2020; 66:103-115. [PMID: 33130409 DOI: 10.1016/j.conb.2020.09.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 09/03/2020] [Accepted: 09/13/2020] [Indexed: 12/12/2022]
Abstract
Understanding the mechanisms that underlie human brain development and neurological and neuropsychiatric disorders is one of the key topics of neurobiology. Because of the poor accessibility of human and non-human primate brain tissues, the current perception and understanding of human brain development have been mainly derived from studies of rodents. However, some human-specific features of neural development cannot be well characterized by these animal models. Thanks to the advances in stem cell technologies, brain organoids are being under rapid development, showing the promising applications in decoding the human brain development and uncovering the pathology of brain diseases. In this review, we mainly summarized the recent advances in the development of brain organoid technology and discussed the limitations, applications and future prospects of this promising field.
Collapse
Affiliation(s)
- Yingchao Shi
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Brain-Intelligence Technology (Shanghai), Bioland Laboratory (Guangzhou), Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Qian Wu
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875, China; IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100875, China
| | - Xiaoqun Wang
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Brain-Intelligence Technology (Shanghai), Bioland Laboratory (Guangzhou), Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Advanced Innovation Center for Human Brain Protection, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
535
|
Jia N, Chong J, Sun L. Application of stem cell biology in treating neurodegenerative diseases. Int J Neurosci 2020; 132:815-825. [PMID: 33081549 DOI: 10.1080/00207454.2020.1840376] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
BACKGROUND The appropriate strategies are needed for stimulating the endogenous neurogenesis or introducing extrinsic neural progenitors, which could be harnessed as the regenerative resources for cueing the neurodegenerations. Adult neurogenesis is the endogenous continuing physiology in limited brain regions such as hippocampus, olfactory system, and hypothalamus. Besides adult neurogenesis, induced pluripotent stem cells (iPSCs) induced functional neurons could be another option for regenerative therapies. OBJECTIVE Current studies are trying to improve the adult neurogenesis and enable the iPSCs induced neurons into neural regeneration. Methods: Here in this review, we mainly introduced the recent progress in neural stem cell biology and its application in the treatment of the neurodegenerations. We main separated the strategy in summarizing the mediators and potential targets to promoting endogenous neural regeneration and transplantation of neural progenitors. CONCLUSION By collecting and comparing the advantages disadvantages between above-mentioned two strategies, we will offer the insight on future development of stem cell therapy in treating neurodegenerative patients.
Collapse
Affiliation(s)
- Na Jia
- Beijing University of Posts and Telecommunications, Beijing, China
| | - Jingping Chong
- Beijing University of Posts and Telecommunications, Beijing, China.,Shanghai University of Engineering Science, Shanghai, China
| | - Lina Sun
- Beijing University of Posts and Telecommunications, Beijing, China.,College of PE and Sport, Beijing Normal University, Beijing, China
| |
Collapse
|
536
|
Pamies D, Zurich MG, Hartung T. Organotypic Models to Study Human Glioblastoma: Studying the Beast in Its Ecosystem. iScience 2020; 23:101633. [PMID: 33103073 PMCID: PMC7569333 DOI: 10.1016/j.isci.2020.101633] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma is a very aggressive primary brain tumor in adults, with very low survival rates and no curative treatments. The high failure rate of drug development for this cancer is linked to the high-cost, time-consuming, and inefficient models used to study the disease. Advances in stem cell and in vitro cultures technologies are promising, however, and here we present the advantages and limitations of available organotypic culture models and discuss their possible applications for studying glioblastoma.
Collapse
Affiliation(s)
- David Pamies
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
- Swiss Centre for Applied Human Toxicology (SCAHT), Switzerland
| | - Marie-Gabrielle Zurich
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
- Swiss Centre for Applied Human Toxicology (SCAHT), Switzerland
| | - Thomas Hartung
- Center for Alternatives to Animal Testing (CAAT) Europe, University of Konstanz, Konstanz, Germany
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
537
|
Shou Y, Liang F, Xu S, Li X. The Application of Brain Organoids: From Neuronal Development to Neurological Diseases. Front Cell Dev Biol 2020; 8:579659. [PMID: 33195219 PMCID: PMC7642488 DOI: 10.3389/fcell.2020.579659] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 09/17/2020] [Indexed: 12/13/2022] Open
Abstract
Brain organoids are derived from induced pluripotent stem cells and embryonic stem cells under three-dimensional culture condition. The generation of an organoid requires the self-assembly of stem cells, progenitor cells, and multiple types of differentiated cells. Organoids display structures that resemble defined brain regions and simulate specific changes of neurological disorders; thus, organoids have become an excellent model for investigating brain development and neurological diseases. In the present review, we have summarized recent advances of the methods of culturing brain organoids and the applications of brain organoids in investigating neurodevelopmental and neurodegenerative diseases.
Collapse
Affiliation(s)
- Yikai Shou
- The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, China.,National Clinical Research Center for Child Health, Hangzhou, China
| | - Feng Liang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shunliang Xu
- Department of Neurology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xuekun Li
- The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, China.,National Clinical Research Center for Child Health, Hangzhou, China
| |
Collapse
|
538
|
Pasqualini C, Kozaki T, Bruschi M, Nguyen THH, Minard-Colin V, Castel D, Grill J, Ginhoux F. Modeling the Interaction between the Microenvironment and Tumor Cells in Brain Tumors. Neuron 2020; 108:1025-1044. [PMID: 33065047 DOI: 10.1016/j.neuron.2020.09.018] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 08/26/2020] [Accepted: 09/09/2020] [Indexed: 12/18/2022]
Abstract
Despite considerable recent advances in understanding and treating many other cancers, malignant brain tumors remain associated with low survival or severe long-term sequelae. Limited progress, including development of immunotherapies, relates in part to difficulties in accurately reproducing brain microenvironment with current preclinical models. The cellular interactions among resident microglia, recruited tumor-associated macrophages, stromal cells, glial cells, neurons, and cancer cells and how they affect tumor growth or behavior are emerging, yet many questions remain. The role of the blood-brain barrier, extracellular matrix components, and heterogeneity among tumor types and within different regions of a single tumor further complicate the matter. Here, we focus on brain microenvironment features impacted by tumor biology. We also discuss limits of current preclinical models and how complementary models, such as humanized animals and organoids, will allow deeper mechanistic insights on cancer biology, allowing for more efficient testing of therapeutic strategies, including immunotherapy, for brain cancers.
Collapse
Affiliation(s)
- Claudia Pasqualini
- Children and Adolescent Oncology Department, Gustave Roussy, Paris-Saclay University, Villejuif, France
| | - Tatsuya Kozaki
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Marco Bruschi
- Genomics & Oncogenesis of Pediatric Brain Tumors, INSERM U981, Gustave Roussy, Paris-Saclay University, Villejuif, France
| | - Thi Hai Hoa Nguyen
- Genomics & Oncogenesis of Pediatric Brain Tumors, INSERM U981, Gustave Roussy, Paris-Saclay University, Villejuif, France
| | - Véronique Minard-Colin
- Children and Adolescent Oncology Department, Gustave Roussy, Paris-Saclay University, Villejuif, France; INSERM U1015, Gustave Roussy, Paris-Saclay University, Villejuif, France
| | - David Castel
- Genomics & Oncogenesis of Pediatric Brain Tumors, INSERM U981, Gustave Roussy, Paris-Saclay University, Villejuif, France
| | - Jacques Grill
- Children and Adolescent Oncology Department, Gustave Roussy, Paris-Saclay University, Villejuif, France; Genomics & Oncogenesis of Pediatric Brain Tumors, INSERM U981, Gustave Roussy, Paris-Saclay University, Villejuif, France.
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore; Shanghai Institute of Immunology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore.
| |
Collapse
|
539
|
Baldassari S, Musante I, Iacomino M, Zara F, Salpietro V, Scudieri P. Brain Organoids as Model Systems for Genetic Neurodevelopmental Disorders. Front Cell Dev Biol 2020; 8:590119. [PMID: 33154971 PMCID: PMC7586734 DOI: 10.3389/fcell.2020.590119] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 09/18/2020] [Indexed: 12/18/2022] Open
Abstract
Neurodevelopmental disorders (NDDs) are a group of disorders in which the development of the central nervous system (CNS) is disturbed, resulting in different neurological and neuropsychiatric features, such as impaired motor function, learning, language or non-verbal communication. Frequent comorbidities include epilepsy and movement disorders. Advances in DNA sequencing technologies revealed identifiable genetic causes in an increasingly large proportion of NDDs, highlighting the need of experimental approaches to investigate the defective genes and the molecular pathways implicated in abnormal brain development. However, targeted approaches to investigate specific molecular defects and their implications in human brain dysfunction are prevented by limited access to patient-derived brain tissues. In this context, advances of both stem cell technologies and genome editing strategies during the last decade led to the generation of three-dimensional (3D) in vitro-models of cerebral organoids, holding the potential to recapitulate precise stages of human brain development with the aim of personalized diagnostic and therapeutic approaches. Recent progresses allowed to generate 3D-structures of both neuronal and non-neuronal cell types and develop either whole-brain or region-specific cerebral organoids in order to investigate in vitro key brain developmental processes, such as neuronal cell morphogenesis, migration and connectivity. In this review, we summarized emerging methodological approaches in the field of brain organoid technologies and their application to dissect disease mechanisms underlying an array of pediatric brain developmental disorders, with a particular focus on autism spectrum disorders (ASDs) and epileptic encephalopathies.
Collapse
Affiliation(s)
- Simona Baldassari
- Medical Genetics Unit, IRCSS Giannina Gaslini Institute, Genoa, Italy
| | - Ilaria Musante
- Medical Genetics Unit, IRCSS Giannina Gaslini Institute, Genoa, Italy.,Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI), University of Genoa, Genoa, Italy
| | - Michele Iacomino
- Medical Genetics Unit, IRCSS Giannina Gaslini Institute, Genoa, Italy
| | - Federico Zara
- Medical Genetics Unit, IRCSS Giannina Gaslini Institute, Genoa, Italy.,Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI), University of Genoa, Genoa, Italy
| | - Vincenzo Salpietro
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI), University of Genoa, Genoa, Italy.,Pediatric Neurology and Muscular Diseases Unit, IRCSS Giannina Gaslini Institute, Genoa, Italy.,Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Paolo Scudieri
- Medical Genetics Unit, IRCSS Giannina Gaslini Institute, Genoa, Italy.,Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI), University of Genoa, Genoa, Italy
| |
Collapse
|
540
|
Obal D, Wu JC. Induced pluripotent stem cells as a platform to understand patient-specific responses to opioids and anaesthetics. Br J Pharmacol 2020; 177:4581-4594. [PMID: 32767563 PMCID: PMC7520445 DOI: 10.1111/bph.15228] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 07/22/2020] [Accepted: 07/27/2020] [Indexed: 12/26/2022] Open
Abstract
Recent advances in human induced pluripotent stem cell (iPSC) technology may provide unprecedented opportunities to study patient-specific responses to anaesthetics and opioids. In this review, we will (1) examine the advantages and limitations of iPSC technology, (2) summarize studies using iPSCs that have contributed to our current understanding of anaesthetics and opioid action on the cardiovascular system and central nervous system (CNS), and (3) describe how iPSC technology can be used to further develop personalized analgesic and sedative pharmacotherapies with reduced or minimal detrimental cardiovascular effects.
Collapse
Affiliation(s)
- Detlef Obal
- Stanford Cardiovascular InstituteStanford UniversityStanfordCaliforniaUSA
- Department of Anesthesiology, Pain, and Perioperative MedicineStanford UniversityStanfordCaliforniaUSA
- Outcomes Research ConsortiumClevelandOhioUSA
| | - Joseph C. Wu
- Stanford Cardiovascular InstituteStanford UniversityStanfordCaliforniaUSA
- Department of Medicine, Division of Cardiovascular MedicineStanford UniversityStanfordCaliforniaUSA
- Department of RadiologyStanford UniversityStanfordCaliforniaUSA
| |
Collapse
|
541
|
Sahu S, Sharan SK. Translating Embryogenesis to Generate Organoids: Novel Approaches to Personalized Medicine. iScience 2020; 23:101485. [PMID: 32864586 PMCID: PMC7441954 DOI: 10.1016/j.isci.2020.101485] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The astounding capacity of pluripotent stem cells (PSCs) to differentiate and self-organize has revolutionized the development of 3D cell culture models. The major advantage is its ability to mimic in vivo microenvironments and cellular interactions when compared with the classical 2D cell culture models. Recent innovations in generating embryo-like structures (including blastoids and gastruloids) from PSCs have advanced the experimental accessibility to understand embryogenesis with immense potential to model human development. Taking cues on how embryonic development leads to organogenesis, PSCs can also be directly differentiated to form mini-organs or organoids of a particular lineage. Organoids have opened new avenues to augment our understanding of stem cell and regenerative biology, tissue homeostasis, and disease mechanisms. In this review, we provide insights from developmental biology with a comprehensive resource of signaling pathways that in a coordinated manner form embryo-like structures and organoids. Moreover, the advent of assembloids and multilineage organoids from PSCs opens a new dimension to study paracrine function and multi-tissue interactions in vitro. Although this led to an avalanche of enthusiasm to utilize organoids for organ transplantation studies, we examine the current limitations and provide perspectives to improve reproducibility, scalability, functional complexity, and cell-type characterization. Taken together, these 3D in vitro organ-specific and patient-specific models hold great promise for drug discovery, clinical management, and personalized medicine.
Collapse
Affiliation(s)
- Sounak Sahu
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Building 560, Room 32-04, 1050 Boyles Street, Frederick, MD 21702, USA
| | - Shyam K. Sharan
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Building 560, Room 32-33, 1050 Boyles Street, Frederick, MD 21702, USA
| |
Collapse
|
542
|
Garcia-Leon JA, Caceres-Palomo L, Sanchez-Mejias E, Mejias-Ortega M, Nuñez-Diaz C, Fernandez-Valenzuela JJ, Sanchez-Varo R, Davila JC, Vitorica J, Gutierrez A. Human Pluripotent Stem Cell-Derived Neural Cells as a Relevant Platform for Drug Screening in Alzheimer's Disease. Int J Mol Sci 2020; 21:ijms21186867. [PMID: 32962164 PMCID: PMC7558359 DOI: 10.3390/ijms21186867] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/15/2020] [Accepted: 09/16/2020] [Indexed: 12/13/2022] Open
Abstract
Extracellular amyloid-beta deposition and intraneuronal Tau-laden neurofibrillary tangles are prime features of Alzheimer's disease (AD). The pathology of AD is very complex and still not fully understood, since different neural cell types are involved in the disease. Although neuronal function is clearly deteriorated in AD patients, recently, an increasing number of evidences have pointed towards glial cell dysfunction as one of the main causative phenomena implicated in AD pathogenesis. The complex disease pathology together with the lack of reliable disease models have precluded the development of effective therapies able to counteract disease progression. The discovery and implementation of human pluripotent stem cell technology represents an important opportunity in this field, as this system allows the generation of patient-derived cells to be used for disease modeling and therapeutic target identification and as a platform to be employed in drug discovery programs. In this review, we discuss the current studies using human pluripotent stem cells focused on AD, providing convincing evidences that this system is an excellent opportunity to advance in the comprehension of AD pathology, which will be translated to the development of the still missing effective therapies.
Collapse
Affiliation(s)
- Juan Antonio Garcia-Leon
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (L.C.-P.); (E.S.-M.); (M.M.-O.); (C.N.-D.); (J.J.F.-V.); (R.S.-V.); (J.C.D.)
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain;
- Correspondence: (J.A.G.-L.); (A.G.); Tel.: +34-952131935 (J.A.G.-L.); +34-952133344 (A.G.)
| | - Laura Caceres-Palomo
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (L.C.-P.); (E.S.-M.); (M.M.-O.); (C.N.-D.); (J.J.F.-V.); (R.S.-V.); (J.C.D.)
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain;
| | - Elisabeth Sanchez-Mejias
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (L.C.-P.); (E.S.-M.); (M.M.-O.); (C.N.-D.); (J.J.F.-V.); (R.S.-V.); (J.C.D.)
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain;
| | - Marina Mejias-Ortega
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (L.C.-P.); (E.S.-M.); (M.M.-O.); (C.N.-D.); (J.J.F.-V.); (R.S.-V.); (J.C.D.)
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain;
| | - Cristina Nuñez-Diaz
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (L.C.-P.); (E.S.-M.); (M.M.-O.); (C.N.-D.); (J.J.F.-V.); (R.S.-V.); (J.C.D.)
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain;
| | - Juan Jose Fernandez-Valenzuela
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (L.C.-P.); (E.S.-M.); (M.M.-O.); (C.N.-D.); (J.J.F.-V.); (R.S.-V.); (J.C.D.)
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain;
| | - Raquel Sanchez-Varo
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (L.C.-P.); (E.S.-M.); (M.M.-O.); (C.N.-D.); (J.J.F.-V.); (R.S.-V.); (J.C.D.)
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain;
| | - Jose Carlos Davila
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (L.C.-P.); (E.S.-M.); (M.M.-O.); (C.N.-D.); (J.J.F.-V.); (R.S.-V.); (J.C.D.)
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain;
| | - Javier Vitorica
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain;
- Departamento Bioquimica y Biologia Molecular, Facultad de Farmacia, Universidad de Sevilla, Instituto de Biomedicina de Sevilla (IBiS)-Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla, 41012 Sevilla, Spain
| | - Antonia Gutierrez
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (L.C.-P.); (E.S.-M.); (M.M.-O.); (C.N.-D.); (J.J.F.-V.); (R.S.-V.); (J.C.D.)
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain;
- Correspondence: (J.A.G.-L.); (A.G.); Tel.: +34-952131935 (J.A.G.-L.); +34-952133344 (A.G.)
| |
Collapse
|
543
|
Zhang Y, Yang X. The Roles of TGF-β Signaling in Cerebrovascular Diseases. Front Cell Dev Biol 2020; 8:567682. [PMID: 33072751 PMCID: PMC7530326 DOI: 10.3389/fcell.2020.567682] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 08/31/2020] [Indexed: 12/13/2022] Open
Abstract
Cerebrovascular diseases are one of the leading causes of death worldwide, however, little progress has been made in preventing or treating these diseases to date. The transforming growth factor-β (TGF-β) signaling pathway plays crucial and highly complicated roles in cerebrovascular development and homeostasis, and dysregulated TGF-β signaling contributes to cerebrovascular diseases. In this review, we provide an updated overview of the functional role of TGF-β signaling in the cerebrovascular system under physiological and pathological conditions. We discuss the current understanding of TGF-β signaling in cerebral angiogenesis and the maintenance of brain vessel homeostasis. We also review the mechanisms by which disruption of TGF-β signaling triggers or promotes the progression of cerebrovascular diseases. Finally, we briefly discuss the potential of targeting TGF-β signaling to treat cerebrovascular diseases.
Collapse
Affiliation(s)
- Yizhe Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| | - Xiao Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| |
Collapse
|
544
|
Habib A, Kodovali VC, Zinn PO. Commentary: A Primer on Human Brain Organoids for the Neurosurgeon. Neurosurgery 2020; 87:E443-E444. [PMID: 32542381 DOI: 10.1093/neuros/nyaa243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 04/15/2020] [Indexed: 11/12/2022] Open
Affiliation(s)
- Ahmed Habib
- Department of Neurosurgery, University of Pittsburgh and Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Venkata C Kodovali
- Department of Neurosurgery, University of Pittsburgh and Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Pascal O Zinn
- Department of Neurosurgery, University of Pittsburgh and Hillman Cancer Center, Pittsburgh, Pennsylvania
| |
Collapse
|
545
|
Cheroni C, Caporale N, Testa G. Autism spectrum disorder at the crossroad between genes and environment: contributions, convergences, and interactions in ASD developmental pathophysiology. Mol Autism 2020; 11:69. [PMID: 32912338 PMCID: PMC7488083 DOI: 10.1186/s13229-020-00370-1] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 08/07/2020] [Indexed: 12/12/2022] Open
Abstract
The complex pathophysiology of autism spectrum disorder encompasses interactions between genetic and environmental factors. On the one hand, hundreds of genes, converging at the functional level on selective biological domains such as epigenetic regulation and synaptic function, have been identified to be either causative or risk factors of autism. On the other hand, exposure to chemicals that are widespread in the environment, such as endocrine disruptors, has been associated with adverse effects on human health, including neurodevelopmental disorders. Interestingly, experimental results suggest an overlap in the regulatory pathways perturbed by genetic mutations and environmental factors, depicting convergences and complex interplays between genetic susceptibility and toxic insults. The pervasive nature of chemical exposure poses pivotal challenges for neurotoxicological studies, regulatory agencies, and policy makers. This highlights an emerging need of developing new integrative models, including biomonitoring, epidemiology, experimental, and computational tools, able to capture real-life scenarios encompassing the interaction between chronic exposure to mixture of substances and individuals' genetic backgrounds. In this review, we address the intertwined roles of genetic lesions and environmental insults. Specifically, we outline the transformative potential of stem cell models, coupled with omics analytical approaches at increasingly single cell resolution, as converging tools to experimentally dissect the pathogenic mechanisms underlying neurodevelopmental disorders, as well as to improve developmental neurotoxicology risk assessment.
Collapse
Affiliation(s)
- Cristina Cheroni
- High Definition Disease Modelling Lab, Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology, IRCCS, Milan, Italy.
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy.
| | - Nicolò Caporale
- High Definition Disease Modelling Lab, Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology, IRCCS, Milan, Italy.
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy.
- Human Technopole, Via Cristina Belgioioso 171, Milan, Italy.
| | - Giuseppe Testa
- High Definition Disease Modelling Lab, Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology, IRCCS, Milan, Italy.
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy.
- Human Technopole, Via Cristina Belgioioso 171, Milan, Italy.
| |
Collapse
|
546
|
Sen D, Keung AJ. Capturing complex epigenetic phenomena through human multicellular systems. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2020; 16:34-41. [PMID: 32905378 DOI: 10.1016/j.cobme.2020.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Epigenetic states inherently define a wide range of complex biological phenotypes and processes in development and disease. Accurate cellular modeling would ideally capture the epigenetic complexity of these processes as well as the accompanying molecular changes in chromatin biochemistry including in DNA and histone modifications. Here we highlight recent work that demonstrate how multicellular systems provide a natural approach to capture complex epigenetic phenomena. They accomplish this through more closely matching the in vivo environment and through the intrinsic nature of multicellular systems being able to generate and model multiple distinct cellular states, all within one system. We also discuss challenges and limitations of such systems, efforts to tune and modulate epigenetics directly in multicellular systems, and how molecular interventional approaches could advance and improve the utility of these models.
Collapse
Affiliation(s)
- Dilara Sen
- North Carolina State University, Raleigh, NC 27606
| | | |
Collapse
|
547
|
Song E, Zhang C, Israelow B, Lu-Culligan A, Prado AV, Skriabine S, Lu P, Weizman OE, Liu F, Dai Y, Szigeti-Buck K, Yasumoto Y, Wang G, Castaldi C, Heltke J, Ng E, Wheeler J, Alfajaro MM, Levavasseur E, Fontes B, Ravindra NG, Van Dijk D, Mane S, Gunel M, Ring A, Kazmi SAJ, Zhang K, Wilen CB, Horvath TL, Plu I, Haik S, Thomas JL, Louvi A, Farhadian SF, Huttner A, Seilhean D, Renier N, Bilguvar K, Iwasaki A. Neuroinvasion of SARS-CoV-2 in human and mouse brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.06.25.169946. [PMID: 32935108 PMCID: PMC7491522 DOI: 10.1101/2020.06.25.169946] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Although COVID-19 is considered to be primarily a respiratory disease, SARS-CoV-2 affects multiple organ systems including the central nervous system (CNS). Yet, there is no consensus whether the virus can infect the brain, or what the consequences of CNS infection are. Here, we used three independent approaches to probe the capacity of SARS-CoV-2 to infect the brain. First, using human brain organoids, we observed clear evidence of infection with accompanying metabolic changes in the infected and neighboring neurons. However, no evidence for the type I interferon responses was detected. We demonstrate that neuronal infection can be prevented either by blocking ACE2 with antibodies or by administering cerebrospinal fluid from a COVID-19 patient. Second, using mice overexpressing human ACE2, we demonstrate in vivo that SARS-CoV-2 neuroinvasion, but not respiratory infection, is associated with mortality. Finally, in brain autopsy from patients who died of COVID-19, we detect SARS-CoV-2 in the cortical neurons, and note pathologic features associated with infection with minimal immune cell infiltrates. These results provide evidence for the neuroinvasive capacity of SARS-CoV2, and an unexpected consequence of direct infection of neurons by SARS-CoV-2.
Collapse
Affiliation(s)
- Eric Song
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Ce Zhang
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Benjamin Israelow
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, CT 06510, USA
| | - Alice Lu-Culligan
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Alba Vieites Prado
- Sorbonne Université, INSERM U1127, CNRS UMR 7225, Paris Brain Institute - ICM, Paris, France
| | - Sophie Skriabine
- Sorbonne Université, INSERM U1127, CNRS UMR 7225, Paris Brain Institute - ICM, Paris, France
| | - Peiwen Lu
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Orr-El Weizman
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Feimei Liu
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT 06510, USA
| | - Yile Dai
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Klara Szigeti-Buck
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT 06510, USA
| | - Yuki Yasumoto
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Guilin Wang
- Yale Center for Genome Analysis, West Haven, CT 06510, USA
| | | | - Jaime Heltke
- Yale Center for Genome Analysis, West Haven, CT 06510, USA
| | - Evelyn Ng
- Yale Center for Genome Analysis, West Haven, CT 06510, USA
| | - John Wheeler
- Yale Center for Genome Analysis, West Haven, CT 06510, USA
| | - Mia Madel Alfajaro
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Etienne Levavasseur
- Sorbonne Université, INSERM U1127, CNRS UMR 7225, Paris Brain Institute - ICM, Paris, France
| | - Benjamin Fontes
- Yale Environmental Health and Safety, Yale University, New Haven, CT 06510, USA
| | - Neal G. Ravindra
- Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Computer Science, Yale University, New Haven, CT 06510, USA
| | - David Van Dijk
- Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Computer Science, Yale University, New Haven, CT 06510, USA
| | - Shrikant Mane
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
- Yale Center for Genome Analysis, West Haven, CT 06510, USA
| | - Murat Gunel
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06510, USA
| | - Aaron Ring
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Syed A. Jaffar Kazmi
- Department of Laboratory Medicine, Geisinger Medical Center, Danville, Pennsylvania
| | - Kai Zhang
- Department of Laboratory Medicine, Geisinger Medical Center, Danville, Pennsylvania
| | - Craig B Wilen
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Tamas L. Horvath
- Department of Biomedical Engineering, Yale University, New Haven, CT 06510, USA
- Department of Molecular Biophysics and Biochemistry, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Molecular, Cellular, and Developmental Biology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Isabelle Plu
- Sorbonne Université, INSERM U1127, CNRS UMR 7225, Paris Brain Institute - ICM, Paris, France
- Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Département de Neuropathologie, Paris, France
| | - Stephane Haik
- Sorbonne Université, INSERM U1127, CNRS UMR 7225, Paris Brain Institute - ICM, Paris, France
- Yale Environmental Health and Safety, Yale University, New Haven, CT 06510, USA
- Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Département de Neuropathologie, Paris, France
- Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Cellule nationale de référence des maladies de Creutzfeldt-Jakob, Paris, France
| | - Jean-Leon Thomas
- Sorbonne Université, INSERM U1127, CNRS UMR 7225, Paris Brain Institute - ICM, Paris, France
- Department of Neurology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Angeliki Louvi
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06510, USA
| | - Shelli F. Farhadian
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Neurology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Anita Huttner
- Department of Pathology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Danielle Seilhean
- Sorbonne Université, INSERM U1127, CNRS UMR 7225, Paris Brain Institute - ICM, Paris, France
- Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Département de Neuropathologie, Paris, France
| | - Nicolas Renier
- Sorbonne Université, INSERM U1127, CNRS UMR 7225, Paris Brain Institute - ICM, Paris, France
| | - Kaya Bilguvar
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
- Yale Center for Genome Analysis, West Haven, CT 06510, USA
| | - Akiko Iwasaki
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Molecular, Cellular, and Developmental Biology, Yale School of Medicine, New Haven, CT 06510, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Lead Contact
| |
Collapse
|
548
|
Galet B, Cheval H, Ravassard P. Patient-Derived Midbrain Organoids to Explore the Molecular Basis of Parkinson's Disease. Front Neurol 2020; 11:1005. [PMID: 33013664 PMCID: PMC7500100 DOI: 10.3389/fneur.2020.01005] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/30/2020] [Indexed: 12/20/2022] Open
Abstract
Induced pluripotent stem cell-derived organoids offer an unprecedented access to complex human tissues that recapitulate features of architecture, composition and function of in vivo organs. In the context of Parkinson's Disease (PD), human midbrain organoids (hMO) are of significant interest, as they generate dopaminergic neurons expressing markers of Substantia Nigra identity, which are the most vulnerable to degeneration. Combined with genome editing approaches, hMO may thus constitute a valuable tool to dissect the genetic makeup of PD by revealing the effects of risk variants on pathological mechanisms in a representative cellular environment. Furthermore, the flexibility of organoid co-culture approaches may also enable the study of neuroinflammatory and neurovascular processes, as well as interactions with other brain regions that are also affected over the course of the disease. We here review existing protocols to generate hMO, how they have been used so far to model PD, address challenges inherent to organoid cultures, and discuss applicable strategies to dissect the molecular pathophysiology of the disease. Taken together, the research suggests that this technology represents a promising alternative to 2D in vitro models, which could significantly improve our understanding of PD and help accelerate therapeutic developments.
Collapse
Affiliation(s)
- Benjamin Galet
- Molecular Pathophysiology of Parkinson's Disease Group, Paris Brain Institute (ICM), INSERM U, CNRS UMR 7225, Sorbonne University, Paris, France
| | | | | |
Collapse
|
549
|
Semertzidou A, Brosens JJ, McNeish I, Kyrgiou M. Organoid models in gynaecological oncology research. Cancer Treat Rev 2020; 90:102103. [PMID: 32932156 DOI: 10.1016/j.ctrv.2020.102103] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 08/29/2020] [Accepted: 08/31/2020] [Indexed: 02/06/2023]
Abstract
Cell culture and animal models represent experimental cornerstones for the investigation of tissue, organ and body physiology in the context of gynaecological research. However, their ability to accurately reflect human mechanisms in vivo is limited. The development of organoid technologies has begun to address this limitation by providing platforms ex vivo that resemble the phenotype and genotype of the multi-cellular tissue from which they were derived more accurately. In this review, we discuss advances in organoid derivation from endometrial, ovarian, fallopian tube and cervical tissue, both benign and malignant, the manipulation of organoid microenvironment to preserve stem cell populations and achieve long-term expansion and we explore the morphological and molecular kinship of organoids to parent tissue. Apart from providing new insight into mechanisms of carcinogenesis, gynaecological cancer-derived organoids can be utilised as tools for drug screening of chemotherapeutic and hormonal compounds where they exhibit interpatient variability consistent with states in vivo and xenografted tumours allowing for patient-tailored treatment strategies. Bridging organoid with bioengineering accomplishments is clearly the way forward to the generation of organoid-on-a-chip technologies enhancing the robustness of the model and its translational potential. Undeniably, organoids are expected to stand their ground in the years to come and revolutionize development and disease modelling studies.
Collapse
Affiliation(s)
- Anita Semertzidou
- Department of Surgery and Cancer & Department of Digestion, Metabolism and Reproduction, Faculty of Medicine, Imperial College London, London W12 0NN, UK; Queen Charlotte's and Chelsea - Hammersmith Hospital, Imperial College Healthcare NHS Trust, London W12 0HS, UK
| | - Jan J Brosens
- Division of Biomedical Sciences, Clinical Science Research Laboratories, Warwick Medical School, University of Warwick, Coventry CV2 2DX, UK; Tommy's National Centre for Miscarriage Research, University Hospitals Coventry & Warwickshire, Coventry CV2 2DX, UK
| | - Iain McNeish
- Department of Surgery and Cancer & Department of Digestion, Metabolism and Reproduction, Faculty of Medicine, Imperial College London, London W12 0NN, UK
| | - Maria Kyrgiou
- Department of Surgery and Cancer & Department of Digestion, Metabolism and Reproduction, Faculty of Medicine, Imperial College London, London W12 0NN, UK; Queen Charlotte's and Chelsea - Hammersmith Hospital, Imperial College Healthcare NHS Trust, London W12 0HS, UK.
| |
Collapse
|
550
|
Mateos-Aparicio P, Bello SA, Rodríguez-Moreno A. Challenges in Physiological Phenotyping of hiPSC-Derived Neurons: From 2D Cultures to 3D Brain Organoids. Front Cell Dev Biol 2020; 8:797. [PMID: 32984317 PMCID: PMC7479826 DOI: 10.3389/fcell.2020.00797] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/28/2020] [Indexed: 12/31/2022] Open
Abstract
Neurons derived from human induced pluripotent stem cells (hiPSC-derived neurons) offer novel opportunities for the development of preclinical models of human neurodegenerative diseases (NDDs). Recent advances in the past few years have increased substantially the potential of these techniques and have uncovered new challenges that the field is facing. Here, we outline and discuss challenges related to the functional characterization of hiPSC-derived neurons and propose ways to overcome current difficulties. In particular, the enormous variability among studies in the electrical properties of hiPSC-derived neurons and broad differences in cell maturation are factors that impair reproducibility. Furthermore, we discuss how the use of 3D brain organoids are of help in resolving some difficulties posed by 2D cultures. Finally, we elaborate on recent and future advances that may help to overcome the discussed challenges and speed-up progress in the field.
Collapse
Affiliation(s)
| | | | - Antonio Rodríguez-Moreno
- Laboratory of Cellular Neuroscience and Plasticity, Department of Physiology, Anatomy and Cell Biology, Universidad Pablo de Olavide, Seville, Spain
| |
Collapse
|