951
|
Liu YN, Lu SY, Yao J. Application of induced pluripotent stem cells to understand neurobiological basis of bipolar disorder and schizophrenia. Psychiatry Clin Neurosci 2017; 71:579-599. [PMID: 28393474 DOI: 10.1111/pcn.12528] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/04/2017] [Indexed: 12/12/2022]
Abstract
The etiology of neuropsychiatric disorders, such as schizophrenia and bipolar disorder, usually involves complex combinations of genetic defects/variations and environmental impacts, which hindered, for a long time, research efforts based on animal models and patients' non-neuronal cells or post-mortem tissues. However, the development of human induced pluripotent stem cell (iPSC) technology by the Yamanaka group was immediately applied to establish cell research models for neuronal disorders. Since then, techniques to achieve highly efficient differentiation of different types of neural cells following iPSC modeling have made much progress. The fast-growing iPSC and neural differentiation techniques have brought valuable insights into the pathology and neurobiology of neuropsychiatric disorders. In this article, we first review the application of iPSC technology in modeling neuronal disorders and discuss the progress in the accompanying neural differentiation. Then, we summarize the progress in iPSC-based research that has been accomplished so far regarding schizophrenia and bipolar disorder.
Collapse
Affiliation(s)
- Yao-Nan Liu
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Si-Yao Lu
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Jun Yao
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| |
Collapse
|
952
|
Abstract
Three-dimensional brain organoid models have come into the spotlight as in vitro tools to recapitulate complex features of the brain. Four recent papers now leverage current technologies to generate new flavours of brain organoids and address aspects of brain biology which, to date, have been challenging to explore.
Collapse
Affiliation(s)
- Lin Yang
- Genome Institute of Singapore, 60 Biopolis Street, Singapore 138672, Singapore
| | - Huck-Hui Ng
- Genome Institute of Singapore, 60 Biopolis Street, Singapore 138672, Singapore
| |
Collapse
|
953
|
Abstract
Human astrocytes are increasingly appreciated as important contributors to brain function in health and disease, but techniques to study them are limited. In this issue of Neuron, Sloan and colleagues (2017) describe a new 3D culture model that generates mature human astrocytes, opening the door to future studies of their function.
Collapse
Affiliation(s)
- Nicola J Allen
- Hearst Foundation Development Chair, Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
954
|
Sloan SA, Darmanis S, Huber N, Khan TA, Birey F, Caneda C, Reimer R, Quake SR, Barres BA, Paşca SP. Human Astrocyte Maturation Captured in 3D Cerebral Cortical Spheroids Derived from Pluripotent Stem Cells. Neuron 2017; 95:779-790.e6. [PMID: 28817799 DOI: 10.1016/j.neuron.2017.07.035] [Citation(s) in RCA: 400] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 06/15/2017] [Accepted: 07/28/2017] [Indexed: 12/21/2022]
Abstract
There is significant need to develop physiologically relevant models for investigating human astrocytes in health and disease. Here, we present an approach for generating astrocyte lineage cells in a three-dimensional (3D) cytoarchitecture using human cerebral cortical spheroids (hCSs) derived from pluripotent stem cells. We acutely purified astrocyte-lineage cells from hCSs at varying stages up to 20 months in vitro using immunopanning and cell sorting and performed high-depth bulk and single-cell RNA sequencing to directly compare them to purified primary human brain cells. We found that hCS-derived glia closely resemble primary human fetal astrocytes and that, over time in vitro, they transition from a predominantly fetal to an increasingly mature astrocyte state. Transcriptional changes in astrocytes are accompanied by alterations in phagocytic capacity and effects on neuronal calcium signaling. These findings suggest that hCS-derived astrocytes closely resemble primary human astrocytes and can be used for studying development and modeling disease.
Collapse
Affiliation(s)
- Steven A Sloan
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Spyros Darmanis
- Departments of Bioengineering and Applied Physics, Stanford University and Chan Zuckerberg Biohub, Stanford, CA 94305, USA
| | - Nina Huber
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Themasap A Khan
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Fikri Birey
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Christine Caneda
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Richard Reimer
- Department of Neurology and Neurological Science, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Stephen R Quake
- Departments of Bioengineering and Applied Physics, Stanford University and Chan Zuckerberg Biohub, Stanford, CA 94305, USA
| | - Ben A Barres
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sergiu P Paşca
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
955
|
Lee MO, Jeon H, Son MY, Lee SC, Cho YS. Clump-passaging-based efficient 3D culture of human pluripotent stem cells under chemically defined conditions. Biochem Biophys Res Commun 2017; 493:723-730. [PMID: 28859981 DOI: 10.1016/j.bbrc.2017.08.124] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 08/28/2017] [Indexed: 12/27/2022]
Abstract
Large-scale production of human pluripotent stem cells (hPSCs) in an efficient and safe manner is crucial to the successful application of hPSCs in biomedical research and regenerative medicine. Three-dimensional culture methods for hPSCs have been extensively studied using single-cell passaging approaches; however, these techniques have been challenged by the induction of massive cell death and accumulation of genomic abnormalities. In this work, we developed and optimized a novel, simple clump-passaging method for in vitro hPSCs 3-dimensional (3D) culture that can be exploited for large-scale production. Fully grown hPSC spheroids were dissociated into smaller-sized spheroid clumps by simple treatment with enzyme-free dissociation buffer, and clumped hPSCs were inoculated and maintained for 3D suspension culture. Our clump-passaging method effectively increased the hPSCs survival rate after subculture and supported scalable hPSCs 3D expansion. We also tested and selected chemically defined media formulations that are suitable for 3D culture and commercially available. Overall, our clump-passaging and expansion method demonstrated high survival and expansion rates for hPSC spheroids compared with conventional methods and may also have the advantage of maintaining genomic stability.
Collapse
Affiliation(s)
- Mi-Ok Lee
- Stem Cell Research Laboratory, Immunotherapy Convergence Research Center, KRIBB, Daejeon, Republic of Korea.
| | - Hyejin Jeon
- Stem Cell Research Laboratory, Immunotherapy Convergence Research Center, KRIBB, Daejeon, Republic of Korea.
| | - Mi-Young Son
- Stem Cell Research Center, KRIBB, Daejeon, Republic of Korea; Department of Bioscience, KRIBB School, University of Science & Technology, Daejeon, Republic of Korea.
| | - Sang Chul Lee
- Research Center for Metabolic Regulation, KRIBB, Daejeon, Republic of Korea.
| | - Yee Sook Cho
- Stem Cell Research Laboratory, Immunotherapy Convergence Research Center, KRIBB, Daejeon, Republic of Korea; Department of Bioscience, KRIBB School, University of Science & Technology, Daejeon, Republic of Korea.
| |
Collapse
|
956
|
Liu B, Teschemacher AG, Kasparov S. Neuroprotective potential of astroglia. J Neurosci Res 2017; 95:2126-2139. [PMID: 28836687 DOI: 10.1002/jnr.24140] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 07/14/2017] [Accepted: 07/24/2017] [Indexed: 12/13/2022]
Abstract
Astroglia are the homoeostatic cells of the central nervous system, which participate in all essential functions of the brain. Astrocytes support neuronal networks by handling water and ion fluxes, transmitter clearance, provision of antioxidants, and metabolic precursors and growth factors. The critical dependence of neurons on constant support from the astrocytes confers astrocytes with intrinsic neuroprotective properties. On the other hand, loss of astrocytic support or their pathological transformation compromises neuronal functionality and viability. Manipulating neuroprotective functions of astrocytes is thus an important strategy to enhance neuronal survival and improve outcomes in disease states. © 2017 The Authors Journal of Neuroscience Research Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Beihui Liu
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, United Kingdom
| | - A G Teschemacher
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, United Kingdom
| | - Sergey Kasparov
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, United Kingdom.,Institute of Living Systems, School of Life Sciences, Immanuel Kant Baltic Federal University, Kaliningrad, Russian Federation
| |
Collapse
|
957
|
Zhu Y, Wang L, Yu H, Yin F, Wang Y, Liu H, Jiang L, Qin J. In situ generation of human brain organoids on a micropillar array. LAB ON A CHIP 2017; 17:2941-2950. [PMID: 28752164 DOI: 10.1039/c7lc00682a] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Brain organoids derived from human induced pluripotent stem cells can recapitulate the early stages of brain development, representing a powerful in vitro system for modeling brain development and diseases. However, the existing methods for brain organoid formation often require time-consuming procedures, including the initial formation of embryoid bodies (EBs) from hiPSCs, and subsequent neural induction and differentiation companied by multi-steps of cell transfer and encapsulation in a 3D matrix. Herein, we propose a simple strategy to enable in situ formation of massive brain organoids from hiPSCs on a micropillar array without tedious manual procedures. The optimized micropillar configurations allow for controlled EB formation, neural induction and differentiation, and generation of functional human brain organoids in 3D culture on a single device. The generated brain organoids were examined to imitate brain organogenesis in vivo at early stages of gestation with specific features of neuronal differentiation, brain regionalization, and cortical organization. By combining microfabrication techniques with stem cells and developmental biology principles, the proposed method can greatly simplify brain organoid formation protocols as compared to conventional methods, overcoming the potential limitations of cell contamination, lower throughput and variance of organoid morphology. It can also provide a useful platform for the engineering of stem cell organoids with improved functions and extending their applications in developmental biology, drug testing and disease modeling.
Collapse
Affiliation(s)
- Yujuan Zhu
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, China.
| | | | | | | | | | | | | | | |
Collapse
|
958
|
Lee CT, Bendriem RM, Wu WW, Shen RF. 3D brain Organoids derived from pluripotent stem cells: promising experimental models for brain development and neurodegenerative disorders. J Biomed Sci 2017; 24:59. [PMID: 28822354 PMCID: PMC5563385 DOI: 10.1186/s12929-017-0362-8] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 08/09/2017] [Indexed: 02/07/2023] Open
Abstract
Three-dimensional (3D) brain organoids derived from human pluripotent stem cells (hPSCs), including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), appear to recapitulate the brain's 3D cytoarchitectural arrangement and provide new opportunities to explore disease pathogenesis in the human brain. Human iPSC (hiPSC) reprogramming methods, combined with 3D brain organoid tools, may allow patient-derived organoids to serve as a preclinical platform to bridge the translational gap between animal models and human clinical trials. Studies using patient-derived brain organoids have already revealed novel insights into molecular and genetic mechanisms of certain complex human neurological disorders such as microcephaly, autism, and Alzheimer's disease. Furthermore, the combination of hiPSC technology and small-molecule high-throughput screening (HTS) facilitates the development of novel pharmacotherapeutic strategies, while transcriptome sequencing enables the transcriptional profiling of patient-derived brain organoids. Finally, the addition of CRISPR/Cas9 genome editing provides incredible potential for personalized cell replacement therapy with genetically corrected hiPSCs. This review describes the history and current state of 3D brain organoid differentiation strategies, a survey of applications of organoids towards studies of neurodevelopmental and neurodegenerative disorders, and the challenges associated with their use as in vitro models of neurological disorders.
Collapse
Affiliation(s)
- Chun-Ting Lee
- Facility for Biotechnology Resources, Center for Biologics Evaluation and Research, FDA, Silver Spring, MD 20993 USA
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Building 52, Rm 1121, 10903 New Hampshire Avenue, Silver Spring, MD 20993 USA
| | - Raphael M. Bendriem
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021 USA
| | - Wells W. Wu
- Facility for Biotechnology Resources, Center for Biologics Evaluation and Research, FDA, Silver Spring, MD 20993 USA
| | - Rong-Fong Shen
- Facility for Biotechnology Resources, Center for Biologics Evaluation and Research, FDA, Silver Spring, MD 20993 USA
| |
Collapse
|
959
|
Three-Dimensional Organoid System Transplantation Technologies in Future Treatment of Central Nervous System Diseases. Stem Cells Int 2017; 2017:5682354. [PMID: 28904534 PMCID: PMC5585580 DOI: 10.1155/2017/5682354] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 05/24/2017] [Accepted: 06/08/2017] [Indexed: 02/06/2023] Open
Abstract
In recent years, scientists have made great achievements in understanding the development of human brain and elucidating critical elements of stepwise spatiotemporal control strategies in neural stem cell specification lineage, which facilitates successful induction of neural organoid in vitro including the cerebral cortex, cerebellar, neural tube, hippocampus cortex, pituitary, and optic cup. Besides, emerging researches on neural organogenesis promote the application of 3D organoid system transplantation in treating central nervous system (CNS) diseases. Present review will categorize current researches on organogenesis into three approaches: (a) stepwise, direct organization of region-specific or population-enriched neural organoid; (b) assemble and direct distinct organ-specific progenitor cells or stem cells to form specific morphogenesis organoid; and (c) assemble embryoid bodies for induction of multilayer organoid. However, the majority of these researches focus on elucidating cellular and molecular mechanisms involving in brain organogenesis or disease development and only a few of them conducted for treating diseases. In this work, we will compare three approaches and also analyze their possible indications for diseases in future treatment on the basis of their distinct characteristics.
Collapse
|
960
|
Sloan SA, Darmanis S, Huber N, Khan TA, Birey F, Caneda C, Reimer R, Quake SR, Barres BA, Paşca SP. Human Astrocyte Maturation Captured in 3D Cerebral Cortical Spheroids Derived from Pluripotent Stem Cells. Neuron 2017. [PMID: 28817799 DOI: 10.1016/j.neuron.2017.07.035.human] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023]
Abstract
There is significant need to develop physiologically relevant models for investigating human astrocytes in health and disease. Here, we present an approach for generating astrocyte lineage cells in a three-dimensional (3D) cytoarchitecture using human cerebral cortical spheroids (hCSs) derived from pluripotent stem cells. We acutely purified astrocyte-lineage cells from hCSs at varying stages up to 20 months in vitro using immunopanning and cell sorting and performed high-depth bulk and single-cell RNA sequencing to directly compare them to purified primary human brain cells. We found that hCS-derived glia closely resemble primary human fetal astrocytes and that, over time in vitro, they transition from a predominantly fetal to an increasingly mature astrocyte state. Transcriptional changes in astrocytes are accompanied by alterations in phagocytic capacity and effects on neuronal calcium signaling. These findings suggest that hCS-derived astrocytes closely resemble primary human astrocytes and can be used for studying development and modeling disease.
Collapse
Affiliation(s)
- Steven A Sloan
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Spyros Darmanis
- Departments of Bioengineering and Applied Physics, Stanford University and Chan Zuckerberg Biohub, Stanford, CA 94305, USA
| | - Nina Huber
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Themasap A Khan
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Fikri Birey
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Christine Caneda
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Richard Reimer
- Department of Neurology and Neurological Science, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Stephen R Quake
- Departments of Bioengineering and Applied Physics, Stanford University and Chan Zuckerberg Biohub, Stanford, CA 94305, USA
| | - Ben A Barres
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sergiu P Paşca
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
961
|
Marton RM, Paşca SP. Neural Differentiation in the Third Dimension: Generating a Human Midbrain. Cell Stem Cell 2017; 19:145-146. [PMID: 27494668 DOI: 10.1016/j.stem.2016.07.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
In recent years, technological improvements in three-dimensional (3D) culture systems have enabled the generation of organoids or spheroids representing a variety of tissues, including the brain. In this issue of Cell Stem Cell, Jo et al. (2016) describe a 3D culture model of the human midbrain containing dopaminergic neurons and neuromelanin.
Collapse
Affiliation(s)
- Rebecca M Marton
- Graduate Program in Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Psychiatry and Behavioral Sciences, Center for Sleep Sciences and Medicine, Stanford Neurosciences Institute and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sergiu P Paşca
- Department of Psychiatry and Behavioral Sciences, Center for Sleep Sciences and Medicine, Stanford Neurosciences Institute and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
962
|
Sart S, Bejoy J, Li Y. Characterization of 3D pluripotent stem cell aggregates and the impact of their properties on bioprocessing. Process Biochem 2017. [DOI: 10.1016/j.procbio.2016.05.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
963
|
Xiang Y, Tanaka Y, Patterson B, Kang YJ, Govindaiah G, Roselaar N, Cakir B, Kim KY, Lombroso AP, Hwang SM, Zhong M, Stanley EG, Elefanty AG, Naegele JR, Lee SH, Weissman SM, Park IH. Fusion of Regionally Specified hPSC-Derived Organoids Models Human Brain Development and Interneuron Migration. Cell Stem Cell 2017; 21:383-398.e7. [PMID: 28757360 DOI: 10.1016/j.stem.2017.07.007] [Citation(s) in RCA: 462] [Impact Index Per Article: 57.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 06/06/2017] [Accepted: 07/11/2017] [Indexed: 01/18/2023]
Abstract
Organoid techniques provide unique platforms to model brain development and neurological disorders. Whereas several methods for recapitulating corticogenesis have been described, a system modeling human medial ganglionic eminence (MGE) development, a critical ventral brain domain producing cortical interneurons and related lineages, has been lacking until recently. Here, we describe the generation of MGE and cortex-specific organoids from human pluripotent stem cells that recapitulate the development of MGE and cortex domains, respectively. Population and single-cell RNA sequencing (RNA-seq) profiling combined with bulk assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq) analyses revealed transcriptional and chromatin accessibility dynamics and lineage relationships during MGE and cortical organoid development. Furthermore, MGE and cortical organoids generated physiologically functional neurons and neuronal networks. Finally, fusing region-specific organoids followed by live imaging enabled analysis of human interneuron migration and integration. Together, our study provides a platform for generating domain-specific brain organoids and modeling human interneuron migration and offers deeper insight into molecular dynamics during human brain development.
Collapse
Affiliation(s)
- Yangfei Xiang
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Yoshiaki Tanaka
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Benjamin Patterson
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Young-Jin Kang
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Gubbi Govindaiah
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Naomi Roselaar
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Bilal Cakir
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Kun-Yong Kim
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Adam P Lombroso
- Department of Biology, Program in Neuroscience and Behavior, Hall-Atwater Laboratory, Wesleyan University, Middletown, CT 06459, USA
| | - Sung-Min Hwang
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Mei Zhong
- Department of Cell Biology, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Edouard G Stanley
- Murdoch Childrens Research Institute, The Royal Children's Hospital, Parkville, VIC 3052, Australia; Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC 3052, Australia; Department of Anatomy and Developmental Biology, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC 3800, Australia
| | - Andrew G Elefanty
- Murdoch Childrens Research Institute, The Royal Children's Hospital, Parkville, VIC 3052, Australia; Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC 3052, Australia; Department of Anatomy and Developmental Biology, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC 3800, Australia
| | - Janice R Naegele
- Department of Biology, Program in Neuroscience and Behavior, Hall-Atwater Laboratory, Wesleyan University, Middletown, CT 06459, USA
| | - Sang-Hun Lee
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Sherman M Weissman
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - In-Hyun Park
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
964
|
Stem cell therapy for abrogating stroke-induced neuroinflammation and relevant secondary cell death mechanisms. Prog Neurobiol 2017; 158:94-131. [PMID: 28743464 DOI: 10.1016/j.pneurobio.2017.07.004] [Citation(s) in RCA: 179] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 07/18/2017] [Accepted: 07/18/2017] [Indexed: 12/13/2022]
Abstract
Ischemic stroke is a leading cause of death worldwide. A key secondary cell death mechanism mediating neurological damage following the initial episode of ischemic stroke is the upregulation of endogenous neuroinflammatory processes to levels that destroy hypoxic tissue local to the area of insult, induce apoptosis, and initiate a feedback loop of inflammatory cascades that can expand the region of damage. Stem cell therapy has emerged as an experimental treatment for stroke, and accumulating evidence supports the therapeutic efficacy of stem cells to abrogate stroke-induced inflammation. In this review, we investigate clinically relevant stem cell types, such as hematopoietic stem cells (HSCs), mesenchymal stem cells (MSCs), endothelial progenitor cells (EPCs), very small embryonic-like stem cells (VSELs), neural stem cells (NSCs), extraembryonic stem cells, adipose tissue-derived stem cells, breast milk-derived stem cells, menstrual blood-derived stem cells, dental tissue-derived stem cells, induced pluripotent stem cells (iPSCs), teratocarcinoma-derived Ntera2/D1 neuron-like cells (NT2N), c-mycER(TAM) modified NSCs (CTX0E03), and notch-transfected mesenchymal stromal cells (SB623), comparing their potential efficacy to sequester stroke-induced neuroinflammation and their feasibility as translational clinical cell sources. To this end, we highlight that MSCs, with a proven track record of safety and efficacy as a transplantable cell for hematologic diseases, stand as an attractive cell type that confers superior anti-inflammatory effects in stroke both in vitro and in vivo. That stem cells can mount a robust anti-inflammatory action against stroke complements the regenerative processes of cell replacement and neurotrophic factor secretion conventionally ascribed to cell-based therapy in neurological disorders.
Collapse
|
965
|
Obiorah IV, Muhammad H, Stafford K, Flaherty EK, Brennand KJ. THC Treatment Alters Glutamate Receptor Gene Expression in Human Stem Cell-Derived Neurons. MOLECULAR NEUROPSYCHIATRY 2017; 3:73-84. [PMID: 29230395 DOI: 10.1159/000477762] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 05/23/2017] [Indexed: 12/21/2022]
Abstract
Given the cognitive and behavioral effects following in utero Δ9-tetrahydrocannabinol (THC) exposure that have been reported in humans and rodents, it is critical to understand the precise consequences of THC on developing human neurons. Here, we utilize excitatory neurons derived from human-induced pluripotent stem cells (hiPSCs), and report that in vitro THC exposure reduced expression of glutamate receptor subunit genes (GRIA1, GRIA2, GRIN2A, and GRIN2B). By expanding these studies across hiPSC-derived neurons from individuals with a variety of genotypes, we believe that a hiPSC-based model will facilitate studies of the interaction of THC exposure and the genetic risk factors underlying neuropsychiatric disease vulnerability.
Collapse
Affiliation(s)
- Ifeanyi V Obiorah
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Hamza Muhammad
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Khalifa Stafford
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Erin K Flaherty
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kristen J Brennand
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
966
|
Phillips AW, Nestor JE, Nestor MW. Developing HiPSC Derived Serum Free Embryoid Bodies for the Interrogation of 3-D Stem Cell Cultures Using Physiologically Relevant Assays. J Vis Exp 2017. [PMID: 28784957 DOI: 10.3791/55799] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Although a number of in vitro disease models have been developed using hiPSCs, one limitation is that these two-dimensional (2-D) systems may not represent the underlying cytoarchitectural and functional complexity of the affected individuals carrying suspected disease variants. Conventional 2-D models remain incomplete representations of in vivo-like structures and do not adequately capture the complexity of the brain. Thus, there is an emerging need for more 3-D hiPSC-based models that can better recapitulate the cellular interactions and functions seen in an in vivo system. Here we report a protocol to develop a 3-D system from undifferentiated hiPSCs based on the serum free embryoid body (SFEB). This 3-D model mirrors aspects of a developing ventralized neocortex and allows for studies into functions integral to living neural cells and intact tissue such as migration, connectivity, communication, and maturation. Specifically, we demonstrate that the SFEBs using our protocol can be interrogated using physiologically relevant and high-content cell based assays such as calcium imaging, and multi-electrode array (MEA) recordings without cryosectioning. In the case of MEA recordings, we demonstrate that SFEBs increase both spike activity and network-level bursting activity during long-term culturing. This SFEB protocol provides a robust and scalable system for the study of developing network formation in a 3-D model that captures aspects of early cortical development.
Collapse
|
967
|
Abstract
Recent breakthroughs in pluripotent stem cell technologies have enabled a new class of in vitro systems for functional modeling of human brain development. These advances, in combination with improvements in neural differentiation methods, allow the generation of in vitro systems that reproduce many in vivo features of the brain with remarkable similarity. Here, we describe advances in the development of these methods, focusing on neural rosette and organoid approaches, and compare their relative capabilities and limitations. We also discuss current technical hurdles for recreating the cell-type complexity and spatial architecture of the brain in culture and offer potential solutions.
Collapse
|
968
|
Jacobson EF, Tzanakakis ES. Human pluripotent stem cell differentiation to functional pancreatic cells for diabetes therapies: Innovations, challenges and future directions. J Biol Eng 2017; 11:21. [PMID: 28680477 PMCID: PMC5494890 DOI: 10.1186/s13036-017-0066-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 06/02/2017] [Indexed: 12/18/2022] Open
Abstract
Recent advances in the expansion and directed pancreatogenic differentiation of human pluripotent stem cells (hPSCs) have intensified efforts to generate functional pancreatic islet cells, especially insulin-secreting β-cells, for cell therapies against diabetes. However, the consistent generation of glucose-responsive insulin-releasing cells remains challenging. In this article, we first present basic concepts of pancreatic organogenesis, which frequently serves as a basis for engineering differentiation regimens. Next, past and current efforts are critically discussed for the conversion of hPSCs along pancreatic cell lineages, including endocrine β-cells and α-cells, as well as exocrine cells with emphasis placed on the later stages of commitment. Finally, major challenges and future directions are examined, such as the identification of factors for in vivo maturation, large-scale culture and post processing systems, cell loss during differentiation, culture economics, efficiency, and efficacy and exosomes and miRNAs in pancreatic differentiation.
Collapse
Affiliation(s)
- Elena F Jacobson
- Department of Chemical and Biological Engineering, Tufts University, 4 Colby St., Room 276A, Medford, MA 02155 USA
| | - Emmanuel S Tzanakakis
- Department of Chemical and Biological Engineering, Tufts University, 4 Colby St., Room 276A, Medford, MA 02155 USA.,Tufts Clinical and Translational Science Institute, Tufts Medical Center, Boston, MA 02111 USA
| |
Collapse
|
969
|
Russell JJ, Theriot JA, Sood P, Marshall WF, Landweber LF, Fritz-Laylin L, Polka JK, Oliferenko S, Gerbich T, Gladfelter A, Umen J, Bezanilla M, Lancaster MA, He S, Gibson MC, Goldstein B, Tanaka EM, Hu CK, Brunet A. Non-model model organisms. BMC Biol 2017; 15:55. [PMID: 28662661 PMCID: PMC5492503 DOI: 10.1186/s12915-017-0391-5] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Model organisms are widely used in research as accessible and convenient systems to study a particular area or question in biology. Traditionally only a handful of organisms have been widely studied, but modern research tools are enabling researchers to extend the set of model organisms to include less-studied and more unusual systems. This Forum highlights a range of 'non-model model organisms' as emerging systems for tackling questions across the whole spectrum of biology (and beyond), the opportunities and challenges, and the outlook for the future.
Collapse
Affiliation(s)
- James J Russell
- Department of Biology, Howard Hughes Medical Institute Stanford University, Stanford, CA, 94305, USA
| | - Julie A Theriot
- Departments of Biochemistry and of Microbiology & Immunology, Howard Hughes Medical Institute Stanford University, Stanford, CA, 94305, USA.
| | - Pranidhi Sood
- Department of Biochemistry & Biophysics, University of California San Francisco, 600 16th St, San Francisco, CA, 94158, USA
| | - Wallace F Marshall
- Department of Biochemistry & Biophysics, University of California San Francisco, 600 16th St, San Francisco, CA, 94158, USA.
| | - Laura F Landweber
- Departments of Biochemistry & Molecular Biophysics and Biological Sciences, Columbia University, New York, NY, 10032, USA
| | | | - Jessica K Polka
- Visiting Scholar, Whitehead Institute, 9 Cambridge Center, Cambridge, MA, 02142, USA
| | - Snezhana Oliferenko
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
- Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, London, SE1 1UL, UK
| | - Therese Gerbich
- 516 Fordham Hall, University of North Carolina Chapel Hill, Chapel Hill, NC, 27514, USA
| | - Amy Gladfelter
- 516 Fordham Hall, University of North Carolina Chapel Hill, Chapel Hill, NC, 27514, USA
| | - James Umen
- Donald Danforth Plant Science Center, 975 N. Warson Rd, St. Louis, MO, 63132, USA
| | | | - Madeline A Lancaster
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, CB2 0QH, Cambridge, UK
| | - Shuonan He
- Stowers Institute for Medical Research, Kansas City, MO, 64110, USA
| | - Matthew C Gibson
- Stowers Institute for Medical Research, Kansas City, MO, 64110, USA
- Department of Anatomy and Cell Biology, The University of Kansas School of Medicine, Kansas City, KS, 66160, USA
| | - Bob Goldstein
- Biology Department, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Elly M Tanaka
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Campus Vienna Biocenter 1, 1030, Vienna, Austria
| | - Chi-Kuo Hu
- Department of Genetics, Stanford University, Stanford, CA, 94305, USA
| | - Anne Brunet
- Department of Genetics, Stanford University, Stanford, CA, 94305, USA
- Glenn Laboratories for the Biology of Aging at Stanford, Stanford, CA, 94305, USA
| |
Collapse
|
970
|
Brawner AT, Xu R, Liu D, Jiang P. Generating CNS organoids from human induced pluripotent stem cells for modeling neurological disorders. INTERNATIONAL JOURNAL OF PHYSIOLOGY, PATHOPHYSIOLOGY AND PHARMACOLOGY 2017; 9:101-111. [PMID: 28694921 PMCID: PMC5498882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 05/21/2017] [Indexed: 06/07/2023]
Abstract
Understanding human brain development and disease is largely hampered by the relative inaccessibility of human brain tissues. Recent advances in human induced pluripotent stem cells (hiPSCs) have led to the generation of unlimited human neural cells and thereby facilitate the investigation of human brain development and pathology. Compared with traditional 2-dimensional (2D) culture methods, culturing the hiPSC-derived neural cells in a three-dimensional (3D) free-floating manner generates human central nervous system (CNS) organoids. These 3D CNS organoids possess the unique advantage of recapitulating multi-regional or region-specific cytoarchitecture seen in the early human fetal brain development. The CNS organoids are becoming a strong complement to the animal model in studying brain development and pathology, and developing new therapies to treat neurodevelopmental diseases. Further improvements to the long-term maintenance and neural maturation of the organoids may allow them to model neurodegenerative diseases. In this review, we will summarize the current development of hiPSCs to generate CNS organoids for modeling neurological disorders and future perspective.
Collapse
Affiliation(s)
- Andrew T Brawner
- Department of Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical CenterOmaha 68198, NE, USA
| | - Ranjie Xu
- Department of Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical CenterOmaha 68198, NE, USA
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical CenterOmaha 68198, NE, USA
| | - Dingfeng Liu
- Department of Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical CenterOmaha 68198, NE, USA
| | - Peng Jiang
- Department of Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical CenterOmaha 68198, NE, USA
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical CenterOmaha 68198, NE, USA
| |
Collapse
|
971
|
Arber C, Lovejoy C, Wray S. Stem cell models of Alzheimer's disease: progress and challenges. ALZHEIMERS RESEARCH & THERAPY 2017; 9:42. [PMID: 28610595 PMCID: PMC5470327 DOI: 10.1186/s13195-017-0268-4] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 05/22/2017] [Indexed: 02/08/2023]
Abstract
A major challenge to our understanding of the molecular mechanisms of Alzheimer’s disease (AD) has been the lack of physiologically relevant in vitro models which capture the precise patient genome, in the cell type of interest, with physiological expression levels of the gene(s) of interest. Induced pluripotent stem cell (iPSC) technology, together with advances in 2D and 3D neuronal differentiation, offers a unique opportunity to overcome this challenge and generate a limitless supply of human neurons for in vitro studies. iPSC-neuron models have been widely employed to model AD and we discuss in this review the progress that has been made to date using patient-derived neurons to recapitulate key aspects of AD pathology and how these models have contributed to a deeper understanding of AD molecular mechanisms, as well as addressing the key challenges posed by using this technology and what progress is being made to overcome these. Finally, we highlight future directions for the use of iPSC-neurons in AD research and highlight the potential value of this technology to neurodegenerative research in the coming years.
Collapse
Affiliation(s)
- Charles Arber
- Department of Molecular Neuroscience, UCL Institute of Neurology, 1 Wakefield Street, London, WC1N 1PJ, UK
| | - Christopher Lovejoy
- Department of Molecular Neuroscience, UCL Institute of Neurology, 1 Wakefield Street, London, WC1N 1PJ, UK
| | - Selina Wray
- Department of Molecular Neuroscience, UCL Institute of Neurology, 1 Wakefield Street, London, WC1N 1PJ, UK.
| |
Collapse
|
972
|
Lemke KA, Aghayee A, Ashton RS. Deriving, regenerating, and engineering CNS tissues using human pluripotent stem cells. Curr Opin Biotechnol 2017; 47:36-42. [PMID: 28605638 DOI: 10.1016/j.copbio.2017.05.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 05/08/2017] [Indexed: 01/08/2023]
Abstract
Progress in deriving a spectrum of central nervous system cell phenotypes from human pluripotent stem cells has spurred significant advances in in vitro modeling and development of regenerative therapies for neurological disorders. While the clinical impact of these advances is still being evaluated, their integration with advanced tissue engineering methodologies and therapeutic approaches that induce neural circuit plasticity, respectively, remain underexplored frontiers.
Collapse
Affiliation(s)
- Kristen A Lemke
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53706, United States
| | - Alireza Aghayee
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53706, United States; Department of Material Science and Engineering, University of Wisconsin-Madison, Madison, WI 53715, United States
| | - Randolph S Ashton
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53706, United States; Department of Material Science and Engineering, University of Wisconsin-Madison, Madison, WI 53715, United States; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53715, United States.
| |
Collapse
|
973
|
Pistollato F, Canovas-Jorda D, Zagoura D, Price A. Protocol for the Differentiation of Human Induced Pluripotent Stem Cells into Mixed Cultures of Neurons and Glia for Neurotoxicity Testing. J Vis Exp 2017. [PMID: 28654077 PMCID: PMC5608344 DOI: 10.3791/55702] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Human pluripotent stem cells can differentiate into various cell types that can be applied to human-based in vitro toxicity assays. One major advantage is that the reprogramming of somatic cells to produce human induced pluripotent stem cells (hiPSCs) avoids the ethical and legislative issues related to the use of human embryonic stem cells (hESCs). HiPSCs can be expanded and efficiently differentiated into different types of neuronal and glial cells, serving as test systems for toxicity testing and, in particular, for the assessment of different pathways involved in neurotoxicity. This work describes a protocol for the differentiation of hiPSCs into mixed cultures of neuronal and glial cells. The signaling pathways that are regulated and/or activated by neuronal differentiation are defined. This information is critical to the application of the cell model to the new toxicity testing paradigm, in which chemicals are assessed based on their ability to perturb biological pathways. As a proof of concept, rotenone, an inhibitor of mitochondrial respiratory complex I, was used to assess the activation of the Nrf2 signaling pathway, a key regulator of the antioxidant-response-element-(ARE)-driven cellular defense mechanism against oxidative stress.
Collapse
Affiliation(s)
- Francesca Pistollato
- Directorate F - Health, Consumers and Reference Materials, Joint Research Centre
| | - David Canovas-Jorda
- Directorate F - Health, Consumers and Reference Materials, Joint Research Centre
| | - Dimitra Zagoura
- Directorate F - Health, Consumers and Reference Materials, Joint Research Centre
| | - Anna Price
- Directorate F - Health, Consumers and Reference Materials, Joint Research Centre;
| |
Collapse
|
974
|
Poon A, Zhang Y, Chandrasekaran A, Phanthong P, Schmid B, Nielsen TT, Freude KK. Modeling neurodegenerative diseases with patient-derived induced pluripotent cells: Possibilities and challenges. N Biotechnol 2017; 39:190-198. [PMID: 28579476 DOI: 10.1016/j.nbt.2017.05.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 04/21/2017] [Accepted: 05/29/2017] [Indexed: 02/06/2023]
Abstract
The rising prevalence of progressive neurodegenerative diseases coupled with increasing longevity poses an economic burden at individual and societal levels. There is currently no effective cure for the majority of neurodegenerative diseases and disease-affected tissues from patients have been difficult to obtain for research and drug discovery in pre-clinical settings. While the use of animal models has contributed invaluable mechanistic insights and potential therapeutic targets, the translational value of animal models could be further enhanced when combined with in vitro models derived from patient-specific induced pluripotent stem cells (iPSCs) and isogenic controls generated using CRISPR-Cas9 mediated genome editing. The iPSCs are self-renewable and capable of being differentiated into the cell types affected by the diseases. These in vitro models based on patient-derived iPSCs provide the opportunity to model disease development, uncover novel mechanisms and test potential therapeutics. Here we review findings from iPSC-based modeling of selected neurodegenerative diseases, including Alzheimer's disease, frontotemporal dementia and spinocerebellar ataxia. Furthermore, we discuss the possibilities of generating three-dimensional (3D) models using the iPSCs-derived cells and compare their advantages and disadvantages to conventional two-dimensional (2D) models.
Collapse
Affiliation(s)
- Anna Poon
- Faculty of Health and Medical Sciences, University of Copenhagen, Gronnegaardsvej 7, 1870 Frederiksberg C, Denmark
| | - Yu Zhang
- Faculty of Health and Medical Sciences, University of Copenhagen, Gronnegaardsvej 7, 1870 Frederiksberg C, Denmark
| | | | - Phetcharat Phanthong
- Stem Cell Research Group, Institute of Molecular Biosciences and Deparment of Anatomy, Faculty of Science, Bangkok, Mahidol University, 10400, Thailand
| | | | - Troels T Nielsen
- Danish Dementia Research Centre, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100 Copenhagen Ø, Denmark
| | - Kristine K Freude
- Faculty of Health and Medical Sciences, University of Copenhagen, Gronnegaardsvej 7, 1870 Frederiksberg C, Denmark.
| |
Collapse
|
975
|
Akhtar AA, Sances S, Barrett R, Breunig JJ. Organoid and Organ-On-A-Chip Systems: New Paradigms for Modeling Neurological and Gastrointestinal Disease. CURRENT STEM CELL REPORTS 2017; 3:98-111. [PMID: 28983454 PMCID: PMC5624725 DOI: 10.1007/s40778-017-0080-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW The modeling of biological processes in vitro provides an important tool to better understand mechanisms of development and disease, allowing for the rapid testing of therapeutics. However, a critical constraint in traditional monolayer culture systems is the absence of the multicellularity, spatial organization, and overall microenvironment present in vivo. This limitation has resulted in numerous therapeutics showing efficacy in vitro, but failing in patient trials. In this review, we discuss several organoid and "organ-on-a-chip" systems with particular regard to the modeling of neurological diseases and gastrointestinal disorders. RECENT FINDINGS Recently, the in vitro generation of multicellular organ-like structures, coined organoids, has allowed the modeling of human development, tissue architecture, and disease with human-specific pathophysiology. Additionally, microfluidic "organ-on-a-chip" technologies add another level of physiological mimicry by allowing biological mediums to be shuttled through 3D cultures. SUMMARY Organoids and organ-chips are rapidly evolving in vitro platforms which hold great promise for the modeling of development and disease.
Collapse
Affiliation(s)
- Aslam Abbasi Akhtar
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Samuel Sances
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Robert Barrett
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Joshua J. Breunig
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
| |
Collapse
|
976
|
Genetic evidence for role of integration of fast and slow neurotransmission in schizophrenia. Mol Psychiatry 2017; 22:792-801. [PMID: 28348379 PMCID: PMC5495879 DOI: 10.1038/mp.2017.33] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 01/05/2017] [Accepted: 01/17/2017] [Indexed: 12/12/2022]
Abstract
The most recent genome-wide association studies (GWAS) of schizophrenia (SCZ) identified hundreds of risk variants potentially implicated in the disease. Further, novel statistical methodology designed for polygenic architecture revealed more potential risk variants. This can provide a link between individual genetic factors and the mechanistic underpinnings of SCZ. Intriguingly, a large number of genes coding for ionotropic and metabotropic receptors for various neurotransmitters-glutamate, γ-aminobutyric acid (GABA), dopamine, serotonin, acetylcholine and opioids-and numerous ion channels were associated with SCZ. Here, we review these findings from the standpoint of classical neurobiological knowledge of neuronal synaptic transmission and regulation of electrical excitability. We show that a substantial proportion of the identified genes are involved in intracellular cascades known to integrate 'slow' (G-protein-coupled receptors) and 'fast' (ionotropic receptors) neurotransmission converging on the protein DARPP-32. Inspection of the Human Brain Transcriptome Project database confirms that that these genes are indeed expressed in the brain, with the expression profile following specific developmental trajectories, underscoring their relevance to brain organization and function. These findings extend the existing pathophysiology hypothesis by suggesting a unifying role of dysregulation in neuronal excitability and synaptic integration in SCZ. This emergent model supports the concept of SCZ as an 'associative' disorder-a breakdown in the communication across different slow and fast neurotransmitter systems through intracellular signaling pathways-and may unify a number of currently competing hypotheses of SCZ pathophysiology.
Collapse
|
977
|
Hartley BJ, Brennand KJ. Neural organoids for disease phenotyping, drug screening and developmental biology studies. Neurochem Int 2017; 106:85-93. [PMID: 27744003 PMCID: PMC5389930 DOI: 10.1016/j.neuint.2016.10.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 10/04/2016] [Accepted: 10/10/2016] [Indexed: 02/01/2023]
Abstract
Human induced pluripotent stem cells (hiPSCs) can theoretically yield limitless supplies of cells fated to any cell type that comprise the human organism, making them a new tool by which to potentially overcome caveats in current biomedical research. In vitro derivation of central nervous system (CNS) cell types has the potential to provide material for drug discovery and validation, safety and toxicity assays, cell replacement therapy and the elucidation of previously unknown disease mechanisms. However, current two-dimensional (2D) CNS differentiation protocols do not faithfully recapitulate the spatial organization of heterogeneous tissue, nor the cell-cell interactions, cell-extracellular matrix interactions, or specific physiological functions generated within complex tissue such as the brain. In an effort to overcome 2D protocol limitations, there have been advancements in deriving highly complicated 3D neural organoid structures. Herein we provide a synopsis of the derivation and application of neural organoids and discuss recent advancements and remaining challenges on the full potential of this novel technological platform.
Collapse
Affiliation(s)
- Brigham J Hartley
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, United States
| | - Kristen J Brennand
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, United States; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY, 10029, United States.
| |
Collapse
|
978
|
Lancaster MA, Corsini NS, Wolfinger S, Gustafson EH, Phillips AW, Burkard TR, Otani T, Livesey FJ, Knoblich JA. Guided self-organization and cortical plate formation in human brain organoids. Nat Biotechnol 2017; 35:659-666. [PMID: 28562594 DOI: 10.1038/nbt.3906] [Citation(s) in RCA: 534] [Impact Index Per Article: 66.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Accepted: 05/18/2017] [Indexed: 01/13/2023]
Abstract
Three-dimensional cell culture models have either relied on the self-organizing properties of mammalian cells or used bioengineered constructs to arrange cells in an organ-like configuration. While self-organizing organoids excel at recapitulating early developmental events, bioengineered constructs reproducibly generate desired tissue architectures. Here, we combine these two approaches to reproducibly generate human forebrain tissue while maintaining its self-organizing capacity. We use poly(lactide-co-glycolide) copolymer (PLGA) fiber microfilaments as a floating scaffold to generate elongated embryoid bodies. Microfilament-engineered cerebral organoids (enCORs) display enhanced neuroectoderm formation and improved cortical development. Furthermore, reconstitution of the basement membrane leads to characteristic cortical tissue architecture, including formation of a polarized cortical plate and radial units. Thus, enCORs model the distinctive radial organization of the cerebral cortex and allow for the study of neuronal migration. Our data demonstrate that combining 3D cell culture with bioengineering can increase reproducibility and improve tissue architecture.
Collapse
Affiliation(s)
- Madeline A Lancaster
- IMBA-Institute of Molecular Biotechnology of the Austrian Academy of Science, Vienna, Austria.,MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Nina S Corsini
- IMBA-Institute of Molecular Biotechnology of the Austrian Academy of Science, Vienna, Austria
| | - Simone Wolfinger
- IMBA-Institute of Molecular Biotechnology of the Austrian Academy of Science, Vienna, Austria
| | - E Hilary Gustafson
- IMBA-Institute of Molecular Biotechnology of the Austrian Academy of Science, Vienna, Austria
| | - Alex W Phillips
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Thomas R Burkard
- IMBA-Institute of Molecular Biotechnology of the Austrian Academy of Science, Vienna, Austria.,IMP-Institute of Molecular Pathology, Vienna, Austria
| | - Tomoki Otani
- Gurdon Institute and Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge, UK
| | - Frederick J Livesey
- Gurdon Institute and Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge, UK
| | - Juergen A Knoblich
- IMBA-Institute of Molecular Biotechnology of the Austrian Academy of Science, Vienna, Austria
| |
Collapse
|
979
|
Powell SK, Gregory J, Akbarian S, Brennand KJ. Application of CRISPR/Cas9 to the study of brain development and neuropsychiatric disease. Mol Cell Neurosci 2017; 82:157-166. [PMID: 28549865 DOI: 10.1016/j.mcn.2017.05.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 05/22/2017] [Indexed: 12/18/2022] Open
Abstract
CRISPR/Cas9 technology has transformed our ability to manipulate the genome and epigenome, from efficient genomic editing to targeted localization of effectors to specific loci. Through the manipulation of DNA- and histone-modifying enzyme activities, activation or repression of gene expression, and targeting of transcriptional regulators, the role of gene-regulatory and epigenetic pathways in basic biology and disease processes can be directly queried. Here, we discuss emerging CRISPR-based methodologies, with specific consideration of neurobiological applications of human induced pluripotent stem cell (hiPSC)-based models.
Collapse
Affiliation(s)
- S K Powell
- Medical Scientist Training Program, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - J Gregory
- Instructional Technology Group, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - S Akbarian
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - K J Brennand
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States.
| |
Collapse
|
980
|
Renner M, Lancaster MA, Bian S, Choi H, Ku T, Peer A, Chung K, Knoblich JA. Self-organized developmental patterning and differentiation in cerebral organoids. EMBO J 2017; 36:1316-1329. [PMID: 28283582 PMCID: PMC5430225 DOI: 10.15252/embj.201694700] [Citation(s) in RCA: 250] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 02/02/2017] [Accepted: 02/09/2017] [Indexed: 12/11/2022] Open
Abstract
Cerebral organoids recapitulate human brain development at a considerable level of detail, even in the absence of externally added signaling factors. The patterning events driving this self-organization are currently unknown. Here, we examine the developmental and differentiative capacity of cerebral organoids. Focusing on forebrain regions, we demonstrate the presence of a variety of discrete ventral and dorsal regions. Clearing and subsequent 3D reconstruction of entire organoids reveal that many of these regions are interconnected, suggesting that the entire range of dorso-ventral identities can be generated within continuous neuroepithelia. Consistent with this, we demonstrate the presence of forebrain organizing centers that express secreted growth factors, which may be involved in dorso-ventral patterning within organoids. Furthermore, we demonstrate the timed generation of neurons with mature morphologies, as well as the subsequent generation of astrocytes and oligodendrocytes. Our work provides the methodology and quality criteria for phenotypic analysis of brain organoids and shows that the spatial and temporal patterning events governing human brain development can be recapitulated in vitro.
Collapse
Affiliation(s)
- Magdalena Renner
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Vienna, Austria
| | - Madeline A Lancaster
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Vienna, Austria
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Shan Bian
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Vienna, Austria
| | - Heejin Choi
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Taeyun Ku
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Angela Peer
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Vienna, Austria
| | - Kwanghun Chung
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (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
- Broad Institute of Harvard University and MIT, Cambridge, MA, USA
| | - Juergen A Knoblich
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Vienna, Austria
| |
Collapse
|
981
|
Fused cerebral organoids model interactions between brain regions. Nat Methods 2017; 14:743-751. [PMID: 28504681 PMCID: PMC5540177 DOI: 10.1038/nmeth.4304] [Citation(s) in RCA: 523] [Impact Index Per Article: 65.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 05/04/2017] [Indexed: 02/08/2023]
Abstract
Human brain development involves complex interactions between different areas, including long distance neuronal migration or formation of major axonal tracts. 3D cerebral organoids allow the growth of diverse brain regions in vitro, but the random arrangement of regional identities limits the reliable analysis of complex phenotypes. Here, we describe a co-culture method combining various brain regions of choice within one organoid tissue. By fusing organoids specified toward dorsal and ventral forebrain, we generate a dorsal-ventral axis. Using fluorescent reporters, we demonstrate robust directional GABAergic interneuron migration from ventral into dorsal forebrain. We describe methodology for time-lapse imaging of human interneuron migration that is inhibited by the CXCR4 antagonist AMD3100. Our results demonstrate that cerebral organoid fusion cultures can model complex interactions between different brain regions. Combined with reprogramming technology, fusions offer the possibility to analyze complex neurodevelopmental defects using cells from neurological disease patients, and to test potential therapeutic compounds.
Collapse
|
982
|
Hříbková H, Zelinková J, Sun YM. Progress in human pluripotent stem cell-based modeling systems for neurological diseases. NEUROGENESIS 2017. [DOI: 10.1080/23262133.2017.1324258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Hana Hříbková
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jana Zelinková
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Yuh-Man Sun
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| |
Collapse
|
983
|
Haziza S, Mohan N, Loe-Mie Y, Lepagnol-Bestel AM, Massou S, Adam MP, Le XL, Viard J, Plancon C, Daudin R, Koebel P, Dorard E, Rose C, Hsieh FJ, Wu CC, Potier B, Herault Y, Sala C, Corvin A, Allinquant B, Chang HC, Treussart F, Simonneau M. Fluorescent nanodiamond tracking reveals intraneuronal transport abnormalities induced by brain-disease-related genetic risk factors. NATURE NANOTECHNOLOGY 2017; 12:322-328. [PMID: 27893730 DOI: 10.1038/nnano.2016.260] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 10/24/2016] [Indexed: 05/24/2023]
Abstract
Brain diseases such as autism and Alzheimer's disease (each inflicting >1% of the world population) involve a large network of genes displaying subtle changes in their expression. Abnormalities in intraneuronal transport have been linked to genetic risk factors found in patients, suggesting the relevance of measuring this key biological process. However, current techniques are not sensitive enough to detect minor abnormalities. Here we report a sensitive method to measure the changes in intraneuronal transport induced by brain-disease-related genetic risk factors using fluorescent nanodiamonds (FNDs). We show that the high brightness, photostability and absence of cytotoxicity allow FNDs to be tracked inside the branches of dissociated neurons with a spatial resolution of 12 nm and a temporal resolution of 50 ms. As proof of principle, we applied the FND tracking assay on two transgenic mouse lines that mimic the slight changes in protein concentration (∼30%) found in the brains of patients. In both cases, we show that the FND assay is sufficiently sensitive to detect these changes.
Collapse
Affiliation(s)
- Simon Haziza
- Laboratoire Aimé Cotton, CNRS, Université Paris-Sud, ENS Cachan, Université Paris-Saclay, 91405 Orsay, France
- Centre de Psychiatrie et Neurosciences, INSERM U894, Université Paris-Descartes, 75014 Paris, France
| | - Nitin Mohan
- Laboratoire Aimé Cotton, CNRS, Université Paris-Sud, ENS Cachan, Université Paris-Saclay, 91405 Orsay, France
| | - Yann Loe-Mie
- Centre de Psychiatrie et Neurosciences, INSERM U894, Université Paris-Descartes, 75014 Paris, France
| | | | - Sophie Massou
- Laboratoire Aimé Cotton, CNRS, Université Paris-Sud, ENS Cachan, Université Paris-Saclay, 91405 Orsay, France
| | - Marie-Pierre Adam
- Laboratoire Aimé Cotton, CNRS, Université Paris-Sud, ENS Cachan, Université Paris-Saclay, 91405 Orsay, France
| | - Xuan Loc Le
- Laboratoire Aimé Cotton, CNRS, Université Paris-Sud, ENS Cachan, Université Paris-Saclay, 91405 Orsay, France
| | - Julia Viard
- Centre de Psychiatrie et Neurosciences, INSERM U894, Université Paris-Descartes, 75014 Paris, France
| | - Christine Plancon
- Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Institut de Génomique, Centre National de Génotypage, 91057 Evry, France
| | - Rachel Daudin
- Centre de Psychiatrie et Neurosciences, INSERM U894, Université Paris-Descartes, 75014 Paris, France
| | - Pascale Koebel
- Institut de génétique et de biologie moléculaire et cellulaire, CNRS UMR 7104, INSERM U 964, Université de Strasbourg, 67400 Illkirch-Graffenstaden, France
| | - Emilie Dorard
- Centre de Psychiatrie et Neurosciences, INSERM U894, Université Paris-Descartes, 75014 Paris, France
| | - Christiane Rose
- Centre de Psychiatrie et Neurosciences, INSERM U894, Université Paris-Descartes, 75014 Paris, France
| | - Feng-Jen Hsieh
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei 106, Taiwan
| | - Chih-Che Wu
- Department of Applied Chemistry, National Chi Nan University, Puli, Nantou Hsien 545, Taiwan
| | - Brigitte Potier
- Centre de Psychiatrie et Neurosciences, INSERM U894, Université Paris-Descartes, 75014 Paris, France
| | - Yann Herault
- Institut de génétique et de biologie moléculaire et cellulaire, CNRS UMR 7104, INSERM U 964, Université de Strasbourg, 67400 Illkirch-Graffenstaden, France
| | - Carlo Sala
- Neuroscience Institute, CNR, 20129 Milano, Italy
| | - Aiden Corvin
- Department of Psychiatry, Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland
| | - Bernadette Allinquant
- Centre de Psychiatrie et Neurosciences, INSERM U894, Université Paris-Descartes, 75014 Paris, France
| | - Huan-Cheng Chang
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei 106, Taiwan
| | - François Treussart
- Laboratoire Aimé Cotton, CNRS, Université Paris-Sud, ENS Cachan, Université Paris-Saclay, 91405 Orsay, France
| | - Michel Simonneau
- Laboratoire Aimé Cotton, CNRS, Université Paris-Sud, ENS Cachan, Université Paris-Saclay, 91405 Orsay, France
- Centre de Psychiatrie et Neurosciences, INSERM U894, Université Paris-Descartes, 75014 Paris, France
- Department of Biology, ENS Cachan, Université Paris-Saclay, 94235 Cachan, France
| |
Collapse
|
984
|
Murphy AR, Laslett A, O'Brien CM, Cameron NR. Scaffolds for 3D in vitro culture of neural lineage cells. Acta Biomater 2017; 54:1-20. [PMID: 28259835 DOI: 10.1016/j.actbio.2017.02.046] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 02/28/2017] [Accepted: 02/28/2017] [Indexed: 12/22/2022]
Abstract
Understanding how neurodegenerative disorders develop is not only a key challenge for researchers but also for the wider society, given the rapidly aging populations in developed countries. Advances in this field require new tools with which to recreate neural tissue in vitro and produce realistic disease models. This in turn requires robust and reliable systems for performing 3D in vitro culture of neural lineage cells. This review provides a state of the art update on three-dimensional culture systems for in vitro development of neural tissue, employing a wide range of scaffold types including hydrogels, solid porous polymers, fibrous materials and decellularised tissues as well as microfluidic devices and lab-on-a-chip systems. To provide some context with in vivo development of the central nervous system (CNS), we also provide a brief overview of the neural stem cell niche, neural development and neural differentiation in vitro. We conclude with a discussion of future directions for this exciting and important field of biomaterials research. STATEMENT OF SIGNIFICANCE Neurodegenerative diseases, including dementia, Parkinson's and Alzheimer's diseases and motor neuron diseases, are a major societal challenge for aging populations. Understanding these conditions and developing therapies against them will require the development of new physical models of healthy and diseased neural tissue. Cellular models resembling neural tissue can be cultured in the laboratory with the help of 3D scaffolds - materials that allow the organization of neural cells into tissue-like structures. This review presents recent work on the development of different types of scaffolds for the 3D culture of neural lineage cells and the generation of functioning neural-like tissue. These in vitro culture systems are enabling the development of new approaches for modelling and tackling diseases of the brain and CNS.
Collapse
Affiliation(s)
- Ashley R Murphy
- Department of Materials Science and Engineering, Monash University, 22 Alliance Lane, Clayton, VIC 3800, Australia
| | - Andrew Laslett
- CSIRO Manufacturing, Bag 10, Clayton South MDC, VIC 3168, Australia; Australian Regenerative Medicine Institute, Science, Technology, Research and Innovation Precinct (STRIP), Monash University, Clayton Campus, Wellington Road, Clayton, VIC 3800, Australia
| | - Carmel M O'Brien
- CSIRO Manufacturing, Bag 10, Clayton South MDC, VIC 3168, Australia; Australian Regenerative Medicine Institute, Science, Technology, Research and Innovation Precinct (STRIP), Monash University, Clayton Campus, Wellington Road, Clayton, VIC 3800, Australia
| | - Neil R Cameron
- Department of Materials Science and Engineering, Monash University, 22 Alliance Lane, Clayton, VIC 3800, Australia.
| |
Collapse
|
985
|
Gitler AD, Dhillon P, Shorter J. Neurodegenerative disease: models, mechanisms, and a new hope. Dis Model Mech 2017; 10:499-502. [PMID: 28468935 PMCID: PMC5451177 DOI: 10.1242/dmm.030205] [Citation(s) in RCA: 437] [Impact Index Per Article: 54.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Neurodegeneration is a feature of many debilitating, incurable diseases that are rapidly rising in prevalence, such as Parkinson's disease. There is an urgent need to develop new and more effective therapeutic strategies to combat these devastating diseases. Models - from cell-based systems, to unicellular organisms, to complex animals - have proven to be a useful tool to help the research community shed light on the mechanisms underlying neurodegenerative diseases, and these advances have now begun to provide promising therapeutic avenues. In this themed issue of Disease Models & Mechanisms, a special collection of articles focused on neurodegenerative diseases is introduced. The collection includes original research articles that provide new insights into the complex pathophysiology of such diseases, revealing candidate biomarkers or therapeutic targets. Some of the articles describe a new disease model that enables deeper exploration of key mechanisms. We also present a series of reviews that highlight some of the recent translational advances made in studies of neurodegenerative diseases. In this Editorial, we summarize the articles featured in this collection, emphasizing the impact that model-based studies have made in this exciting area of research.
Collapse
Affiliation(s)
- Aaron D Gitler
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94404, USA
| | - Paraminder Dhillon
- Reviews Editor, Disease Models & Mechanisms, The Company of Biologists, Cambridge CB24 9LF, UK
| | - James Shorter
- Department of Biochemistry & Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
986
|
Ochocinska MJ, Zlokovic BV, Searson PC, Crowder AT, Kraig RP, Ljubimova JY, Mainprize TG, Banks WA, Warren RQ, Kindzelski A, Timmer W, Liu CH. NIH workshop report on the trans-agency blood-brain interface workshop 2016: exploring key challenges and opportunities associated with the blood, brain and their interface. Fluids Barriers CNS 2017; 14:12. [PMID: 28457227 PMCID: PMC5410699 DOI: 10.1186/s12987-017-0061-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 04/08/2017] [Indexed: 01/01/2023] Open
Abstract
A trans-agency workshop on the blood–brain interface (BBI), sponsored by the National Heart, Lung and Blood Institute, the National Cancer Institute and the Combat Casualty Care Research Program at the Department of Defense, was conducted in Bethesda MD on June 7–8, 2016. The workshop was structured into four sessions: (1) blood sciences; (2) exosome therapeutics; (3) next generation in vitro blood–brain barrier (BBB) models; and (4) BBB delivery and targeting. The first day of the workshop focused on the physiology of the blood and neuro-vascular unit, blood or biofluid-based molecular markers, extracellular vesicles associated with brain injury, and how these entities can be employed to better evaluate injury states and/or deliver therapeutics. The second day of the workshop focused on technical advances in in vitro models, BBB manipulations and nanoparticle-based drug carrier designs, with the goal of improving drug delivery to the central nervous system. The presentations and discussions underscored the role of the BBI in brain injury, as well as the role of the BBB as both a limiting factor and a potential conduit for drug delivery to the brain. At the conclusion of the meeting, the participants discussed challenges and opportunities confronting BBI translational researchers. In particular, the participants recommended using BBI translational research to stimulate advances in diagnostics, as well as targeted delivery approaches for detection and therapy of both brain injury and disease.
Collapse
Affiliation(s)
- Margaret J Ochocinska
- National Heart, Lung, and Blood Institute, National Institutes of Health, 6701 Rockledge Dr., Room 9149, Bethesda, MD, 20892-7950, USA.
| | | | | | | | | | | | | | | | - Ronald Q Warren
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Andrei Kindzelski
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - William Timmer
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Christina H Liu
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
987
|
Assembly of functionally integrated human forebrain spheroids. Nature 2017; 545:54-59. [PMID: 28445465 PMCID: PMC5805137 DOI: 10.1038/nature22330] [Citation(s) in RCA: 874] [Impact Index Per Article: 109.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 04/04/2017] [Indexed: 12/12/2022]
Abstract
The development of the nervous system involves a coordinated succession
of events including the migration of GABAergic neurons from ventral to dorsal
forebrain and their integration into cortical circuits. However, these
interregional interactions have not yet been modelled with human cells. Here, we
generate from human pluripotent cells three-dimensional spheroids resembling
either the dorsal or ventral forebrain and containing cortical glutamatergic or
GABAergic neurons. These subdomain-specific forebrain spheroids can be assembled
to recapitulate the saltatory migration of interneurons similar to migration in
fetal forebrain. Using this system, we find that in Timothy syndrome– a
neurodevelopmental disorder that is caused by mutations in the CaV1.2
calcium channel, interneurons display abnormal migratory saltations. We also
show that after migration, interneurons functionally integrate with
glutamatergic neurons to form a microphysiological system. We anticipate that
this approach will be useful for studying development and disease, and for
deriving spheroids that resemble other brain regions to assemble circuits
in vitro.
Collapse
|
988
|
Abstract
The mechanisms by which human stem cells self‐organise into brain‐like tissues in 3D organoid culture are poorly understood. In this issue of The EMBO Journal, Renner and Lancaster et al demonstrate that in the absence of external stimuli, human cerebral organoids develop large forebrain structures that display specific landmarks of spatial and temporal patterning, including signalling centres producing known morphogens. The generation of cerebral organoids is therefore likely to reflect normal brain development.
Collapse
|
989
|
Wu F, Su RQ, Lai YC, Wang X. Engineering of a synthetic quadrastable gene network to approach Waddington landscape and cell fate determination. eLife 2017; 6. [PMID: 28397688 PMCID: PMC5388541 DOI: 10.7554/elife.23702] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Accepted: 03/10/2017] [Indexed: 11/13/2022] Open
Abstract
The process of cell fate determination has been depicted intuitively as cells travelling and resting on a rugged landscape, which has been probed by various theoretical studies. However, few studies have experimentally demonstrated how underlying gene regulatory networks shape the landscape and hence orchestrate cellular decision-making in the presence of both signal and noise. Here we tested different topologies and verified a synthetic gene circuit with mutual inhibition and auto-activations to be quadrastable, which enables direct study of quadruple cell fate determination on an engineered landscape. We show that cells indeed gravitate towards local minima and signal inductions dictate cell fates through modulating the shape of the multistable landscape. Experiments, guided by model predictions, reveal that sequential inductions generate distinct cell fates by changing landscape in sequence and hence navigating cells to different final states. This work provides a synthetic biology framework to approach cell fate determination and suggests a landscape-based explanation of fixed induction sequences for targeted differentiation. DOI:http://dx.doi.org/10.7554/eLife.23702.001 Cells in animals use a process called differentiation to specialize into specific cell types such as skin cells and liver cells. Proteins called transcription factors drive particular steps in differentiation by controlling the activity of specific genes. Many transcription factors interact with each other to form complex networks that regulate gene activity to determine the fate of a cell and control the whole differentiation process. Some individual gene networks can program cells to become any one of several different cell fates, a feature known as multistability. In the 1950s, a scientist called Conrad Waddington proposed the concept of an “epigenetic landscape” to describe how the fate of a cell is decided as an animal develops. The cell, depicted as a ball, rolls down a rugged landscape and has the option of taking several different routes. Each route will eventually lead to a distinct cell fate. As the ball moves down the hill, the choice of routes and final destinations becomes more limited. Theoretical approaches have been used to understand how gene regulatory networks shape the epigenetic landscape of an animal. However, few studies have experimentally tested the findings of the theoretical approaches and it is not clear how environmental inputs help to determine which path a cell will take. Although bacteria cells do not generally specialize into particular cell types, bacteria cells can use multistability in transcription factor networks to switch between different behaviors or “states” in response to cues from the environment. Wu et al. used a bacterium called E. coli as a model to investigate how a gene network called MINPA from mammals, which is involved in differentiation and is believed to show multistability, can guide cells to adopt different states. The work combined experimental and mathematical approaches to design, construct and test an artificial version of the MINPA gene network in E. coli. The experiments showed that MINPA could direct the cells to adopt four different stable states in which the cells produced fluorescent proteins of different colors. With the help of mathematical modeling, Wu et al. charted how the landscape of cell states changed when external chemical cues were applied. Exposing the cells to several cues in particular orders guided the cells to different final states. The findings of Wu et al. shed new light on how the fate of a cell is determined and provide a theoretical framework for understanding the complex networks that control cell differentiation. This could help develop new ways of directing cell fate that could ultimately be used to generate cells to treat human diseases. DOI:http://dx.doi.org/10.7554/eLife.23702.002
Collapse
Affiliation(s)
- Fuqing Wu
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, United States
| | - Ri-Qi Su
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, United States.,School of Electrical, Computer and Energy Engineering, Arizona State University, Tempe, United States
| | - Ying-Cheng Lai
- School of Electrical, Computer and Energy Engineering, Arizona State University, Tempe, United States.,Institute for Complex Systems and Mathematical Biology, King's College, University of Aberdeen, Aberdeen, United Kingdom.,Department of Physics, Arizona State University, Tempe, United States
| | - Xiao Wang
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, United States
| |
Collapse
|
990
|
Cooper DJ, Zunino G, Bixby JL, Lemmon VP. Phenotypic screening with primary neurons to identify drug targets for regeneration and degeneration. Mol Cell Neurosci 2017; 80:161-169. [PMID: 27444126 PMCID: PMC5243932 DOI: 10.1016/j.mcn.2016.07.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 07/04/2016] [Accepted: 07/16/2016] [Indexed: 12/13/2022] Open
Abstract
High-throughput, target-based screening techniques have been utilized extensively for drug discovery in the past several decades. However, the need for more predictive in vitro models of in vivo disease states has generated a shift in strategy towards phenotype-based screens. Phenotype based screens are particularly valuable in studying complex conditions such as CNS injury and degenerative disease, as many factors can contribute to a specific cellular response. In this review, we will discuss different screening frameworks and their relative utility in examining mechanisms of neurodegeneration and axon regrowth, particularly in cell-based in vitro disease models.
Collapse
Affiliation(s)
- Daniel J. Cooper
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami, 1400 NW 12th Ave, Miami, FL 33136, USA
| | - Giulia Zunino
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami, 1400 NW 12th Ave, Miami, FL 33136, USA
| | - John L. Bixby
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami, 1400 NW 12th Ave, Miami, FL 33136, USA
- Center for Computational Science, University of Miami, 1400 NW 12th Ave, Miami, FL 33136, USA
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, 1400 NW 12th Ave, Miami, FL 33136, USA
| | - Vance P. Lemmon
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami, 1400 NW 12th Ave, Miami, FL 33136, USA
- Center for Computational Science, University of Miami, 1400 NW 12th Ave, Miami, FL 33136, USA
| |
Collapse
|
991
|
Watmuff B, Liu B, Karmacharya R. Stem cell-derived neurons in the development of targeted treatment for schizophrenia and bipolar disorder. Pharmacogenomics 2017; 18:471-479. [PMID: 28346060 DOI: 10.2217/pgs-2016-0187] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The recent advent of induced pluripotent stem cells has enabled the study of patient-specific and disease-related neurons in vitro and has facilitated new directions of inquiry into disease mechanisms. With these approaches, we now have the possibility of correlating ex vivo cellular phenotypes with individual patient response to treatment and/or side effects, which makes targeted treatments for schizophrenia and bipolar disorder a distinct prospect in the coming years. Here, we briefly review the current state of stem cell-based models and explore studies that are providing new insights into the disease biology of schizophrenia and bipolar disorder, which are laying the foundations for the development of novel targeted therapies.
Collapse
Affiliation(s)
- Bradley Watmuff
- Center for Experimental Drugs & Diagnostics, Psychiatric & Neurodevelopmental Genetics Unit, Center for Human Genetic Research, Harvard Medical School & Massachusetts General Hospital, Boston, MA 02114, USA.,Chemical Biology Program, Broad Institute of Harvard & MIT, Cambridge, MA 02142, USA
| | - Bangyan Liu
- Center for Experimental Drugs & Diagnostics, Psychiatric & Neurodevelopmental Genetics Unit, Center for Human Genetic Research, Harvard Medical School & Massachusetts General Hospital, Boston, MA 02114, USA
| | - Rakesh Karmacharya
- Center for Experimental Drugs & Diagnostics, Psychiatric & Neurodevelopmental Genetics Unit, Center for Human Genetic Research, Harvard Medical School & Massachusetts General Hospital, Boston, MA 02114, USA.,Chemical Biology Program, Broad Institute of Harvard & MIT, Cambridge, MA 02142, USA.,Schizophrenia & Bipolar Disorder Program, McLean Hospital, Belmont, MA 02478, USA
| |
Collapse
|
992
|
Dezonne RS, Sartore RC, Nascimento JM, Saia-Cereda VM, Romão LF, Alves-Leon SV, de Souza JM, Martins-de-Souza D, Rehen SK, Gomes FCA. Derivation of Functional Human Astrocytes from Cerebral Organoids. Sci Rep 2017; 7:45091. [PMID: 28345587 PMCID: PMC5366860 DOI: 10.1038/srep45091] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 02/15/2017] [Indexed: 12/17/2022] Open
Abstract
Astrocytes play a critical role in the development and homeostasis of the central nervous system (CNS). Astrocyte dysfunction results in several neurological and degenerative diseases. However, a major challenge to our understanding of astrocyte physiology and pathology is the restriction of studies to animal models, human post-mortem brain tissues, or samples obtained from invasive surgical procedures. Here, we report a protocol to generate human functional astrocytes from cerebral organoids derived from human pluripotent stem cells. The cellular isolation of cerebral organoids yielded cells that were morphologically and functionally like astrocytes. Immunolabelling and proteomic assays revealed that human organoid-derived astrocytes express the main astrocytic molecular markers, including glutamate transporters, specific enzymes and cytoskeletal proteins. We found that organoid-derived astrocytes strongly supported neuronal survival and neurite outgrowth and responded to ATP through transient calcium wave elevations, which are hallmarks of astrocyte physiology. Additionally, these astrocytes presented similar functional pathways to those isolated from adult human cortex by surgical procedures. This is the first study to provide proteomic and functional analyses of astrocytes isolated from human cerebral organoids. The isolation of these astrocytes holds great potential for the investigation of developmental and evolutionary features of the human brain and provides a useful approach to drug screening and neurodegenerative disease modelling.
Collapse
Affiliation(s)
- Rômulo Sperduto Dezonne
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ., Brasil
| | - Rafaela Costa Sartore
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ., Brasil.,Instituto D'Or de Pesquisa e Ensino (IDOR), Rio de Janeiro, RJ, Brasil
| | - Juliana Minardi Nascimento
- Instituto D'Or de Pesquisa e Ensino (IDOR), Rio de Janeiro, RJ, Brasil.,Instituto de Biologia, Universidade Estadual de Campinas, Campinas, SP, Brasil
| | | | - Luciana Ferreira Romão
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ., Brasil.,Universidade Federal do Rio de Janeiro,Campus Xerém, RJ, Brasil
| | - Soniza Vieira Alves-Leon
- Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - Jorge Marcondes de Souza
- Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | | | - Stevens Kastrup Rehen
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ., Brasil.,Instituto D'Or de Pesquisa e Ensino (IDOR), Rio de Janeiro, RJ, Brasil
| | | |
Collapse
|
993
|
Abstract
Here, we report a 3D cerebellar differentiation protocol with quick startup method, defined medium and no special materials or handling requirements. Three fibroblast growth factors (FGF2, 4 and 8) were used for cerebellar patterning and smoothened agonist (SAG) for granule cell development. After 35 days, differentiation products exhibited similar structures and neuronal markers reported in prior ‘organoid’ and ‘spheroid’ protocols. This included cells positive for KIRREL2 (a marker of early cerebellar neuroepithelium) and ZIC1 (a marker for granule cells). Follow-up tests indicated that addition of FGFs, if helpful, was not required to generate observed structures and cell types. This suggests that intrinsic production of patterning factors by aggregates themselves may be adequate for region-specific 3D modeling. This protocol may be used as a quick, easy and cost-efficient method for 3D culture, whether to research development of the early cerebellar neuroepithelium, a base to generate mature cortical structures, or to optimize minimal-factor protocols for other brain regions. Summary: We present a simplified method to generate early neuroepithelium of the cerebellar cortex in a 3D culture using defined medium and minimal addition of growth factors.
Collapse
Affiliation(s)
- Dwayne B Holmes
- Department of Pediatrics/Child Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam 1081 HV, The Netherlands
| | - Vivi M Heine
- Department of Pediatrics/Child Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam 1081 HV, The Netherlands .,Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University, Amsterdam 1081 HV, The Netherlands
| |
Collapse
|
994
|
|
995
|
Nondestructive nanostraw intracellular sampling for longitudinal cell monitoring. Proc Natl Acad Sci U S A 2017; 114:E1866-E1874. [PMID: 28223521 DOI: 10.1073/pnas.1615375114] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Here, we report a method for time-resolved, longitudinal extraction and quantitative measurement of intracellular proteins and mRNA from a variety of cell types. Cytosolic contents were repeatedly sampled from the same cell or population of cells for more than 5 d through a cell-culture substrate, incorporating hollow 150-nm-diameter nanostraws (NS) within a defined sampling region. Once extracted, the cellular contents were analyzed with conventional methods, including fluorescence, enzymatic assays (ELISA), and quantitative real-time PCR. This process was nondestructive with >95% cell viability after sampling, enabling long-term analysis. It is important to note that the measured quantities from the cell extract were found to constitute a statistically significant representation of the actual contents within the cells. Of 48 mRNA sequences analyzed from a population of cardiomyocytes derived from human induced pluripotent stem cells (hiPSC-CMs), 41 were accurately quantified. The NS platform samples from a select subpopulation of cells within a larger culture, allowing native cell-to-cell contact and communication even during vigorous activity such as cardiomyocyte beating. This platform was applied both to cell lines and to primary cells, including CHO cells, hiPSC-CMs, and human astrocytes derived in 3D cortical spheroids. By tracking the same cell or group of cells over time, this method offers an avenue to understand dynamic cell behavior, including processes such as induced pluripotency and differentiation.
Collapse
|
996
|
Nie J, Hashino E. Organoid technologies meet genome engineering. EMBO Rep 2017; 18:367-376. [PMID: 28202491 DOI: 10.15252/embr.201643732] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 01/13/2017] [Accepted: 01/18/2017] [Indexed: 01/08/2023] Open
Abstract
Three-dimensional (3D) stem cell differentiation cultures recently emerged as a novel model system for investigating human embryonic development and disease progression in vitro, complementing existing animal and two-dimensional (2D) cell culture models. Organoids, the 3D self-organizing structures derived from pluripotent or somatic stem cells, can recapitulate many aspects of structural organization and functionality of their in vivo organ counterparts, thus holding great promise for biomedical research and translational applications. Importantly, faithful recapitulation of disease and development processes relies on the ability to modify the genomic contents in organoid cells. The revolutionary genome engineering technologies, CRISPR/Cas9 in particular, enable investigators to generate various reporter cell lines for prompt validation of specific cell lineages as well as to introduce disease-associated mutations for disease modeling. In this review, we provide historical overviews, and discuss technical considerations, and potential future applications of genome engineering in 3D organoid models.
Collapse
Affiliation(s)
- Jing Nie
- Department of Otolaryngology - Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Eri Hashino
- Department of Otolaryngology - Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, USA .,Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
997
|
Wen Z. Modeling neurodevelopmental and psychiatric diseases with human iPSCs. J Neurosci Res 2017; 95:1097-1109. [PMID: 28186671 DOI: 10.1002/jnr.24031] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 12/27/2016] [Accepted: 01/10/2017] [Indexed: 12/29/2022]
Abstract
Neurodevelopmental and psychiatric disorders, including autism spectrum disorder and schizophrenia, are complex and heterogeneous disorders that affect a large portion of the world's population. While the causes are still poorly understood, currently available treatments are limited; the development of rational therapeutics based on an understanding of the etiology and pathogenesis of the disease is imperative. The breakthrough technology of deriving induced pluripotent stem cells (iPSCs), reprogrammed from somatic cells of healthy subjects or patients, offers an unprecedented opportunity to recapitulate both normal and pathological development of human tissue, thereby opening up a new avenue for disease modeling and drug development in a more genetically tractable and disease-relevant system. Here, I review the recent progress in the use of human iPSCs for modeling neurodevelopmental and psychiatric disorders and developing novel therapeutic strategies, and discuss challenges in this rapidly moving field. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Zhexing Wen
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurology, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
998
|
Chailangkarn T, Muotri AR. Modeling Williams syndrome with induced pluripotent stem cells. NEUROGENESIS (AUSTIN, TEX.) 2017; 4:e1283187. [PMID: 28229087 PMCID: PMC5305168 DOI: 10.1080/23262133.2017.1283187] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 12/01/2016] [Accepted: 01/10/2017] [Indexed: 12/20/2022]
Abstract
The development of induced pluripotent stem cells (iPSCs) like never before has opened novel opportunity to study diseases in relevant cell types. In our recent study, Williams syndrome (WS), a rare genetic neurodevelopmental disorder, that is caused by hemizygous deletion of 25-28 genes on chromosome 7, is of interest because of its unique cognitive and social profiles. Little is known about haploinsufficiency effect of those deleted genes on molecular and cellular phenotypes at the neural level due to the lack of relevant human cellular model. Using the cellular reprogramming approach, we reported that WS iPSC-derived neural progenitor cells (NPCs) has increased apoptosis and therefore increased doubling time, which could be rescued by complementation of frizzled 9, one of the genes typically deleted in WS. Moreover, WS iPSC-derived CTIP2-positive pyramidal neurons exhibit morphologic alterations including longer total dendrites and increasing dendritic spine number. In addition, WS iPSC-derived neurons show an increase in calcium transient frequency and synchronized activity likely due to increased number of dendritic spines and synapses. Our work integrated cross-level data from genetics to behavior of WS individuals and revealed altered cellular phenotypes in WS human NPCs and neurons that could be validated in other model systems such as magnetic resonance imaging (MRI) in live subjects and postmortem brain tissues.
Collapse
Affiliation(s)
- Thanathom Chailangkarn
- University of California San Diego, School of Medicine, Department of Pediatrics/Rady Children's Hospital San Diego, Department of Cellular & Molecular Medicine, Stem Cell Program, Center for Academic Research and Training in Anthropogeny (CARTA), La Jolla, CA, USA
- National Center for Genetic Engineering and Biotechnology (BIOTEC), Virology and Cell Technology Laboratory, Pathum Thani, Thailand
| | - Alysson R. Muotri
- University of California San Diego, School of Medicine, Department of Pediatrics/Rady Children's Hospital San Diego, Department of Cellular & Molecular Medicine, Stem Cell Program, Center for Academic Research and Training in Anthropogeny (CARTA), La Jolla, CA, USA
| |
Collapse
|
999
|
Giandomenico SL, Lancaster MA. Probing human brain evolution and development in organoids. Curr Opin Cell Biol 2017; 44:36-43. [DOI: 10.1016/j.ceb.2017.01.001] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Revised: 12/23/2016] [Accepted: 01/10/2017] [Indexed: 02/06/2023]
|
1000
|
CYP3A5 Mediates Effects of Cocaine on Human Neocorticogenesis: Studies using an In Vitro 3D Self-Organized hPSC Model with a Single Cortex-Like Unit. Neuropsychopharmacology 2017; 42:774-784. [PMID: 27534267 PMCID: PMC5240177 DOI: 10.1038/npp.2016.156] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 05/26/2016] [Accepted: 08/10/2016] [Indexed: 12/31/2022]
Abstract
Because of unavoidable confounding variables in the direct study of human subjects, it has been difficult to unravel the effects of prenatal cocaine exposure on the human fetal brain, as well as the cellular and biochemical mechanisms involved. Here, we propose a novel approach using a human pluripotent stem cell (hPSC)-based 3D neocortical organoid model. This model retains essential features of human neocortical development by encompassing a single self-organized neocortical structure, without including an animal-derived gelatinous matrix. We reported previously that prenatal cocaine exposure to rats during the most active period of neural progenitor proliferation induces cytoarchitectural changes in the embryonic neocortex. We also identified a role of CYP450 and consequent oxidative ER stress signaling in these effects. However, because of differences between humans and rodents in neocorticogenesis and brain CYP metabolism, translation of the research findings from the rodent model to human brain development is uncertain. Using hPSC 3D neocortical organoids, we demonstrate that the effects of cocaine are mediated through CYP3A5-induced generation of reactive oxygen species, inhibition of neocortical progenitor cell proliferation, induction of premature neuronal differentiation, and interruption of neural tissue development. Furthermore, knockdown of CYP3A5 reversed these cocaine-induced pathological phenotypes, suggesting CYP3A5 as a therapeutic target to mitigate the deleterious neurodevelopmental effects of prenatal cocaine exposure in humans. Moreover, 3D organoid methodology provides an innovative platform for identifying adverse effects of abused psychostimulants and pharmaceutical agents, and can be adapted for use in neurodevelopmental disorders with genetic etiologies.
Collapse
|