101
|
Namchaiw P, Bunreangsri P, Eiamcharoen P, Eiamboonsert S, P. Poo-arporn R. An in vitro workflow of neuron-laden agarose-laminin hydrogel for studying small molecule-induced amyloidogenic condition. PLoS One 2022; 17:e0273458. [PMID: 36026506 PMCID: PMC9416999 DOI: 10.1371/journal.pone.0273458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 08/08/2022] [Indexed: 11/18/2022] Open
Abstract
In vitro studies have been popularly used to determine the cellular and molecular mechanisms for many decades. However, the traditional two-dimension (2D) cell culture which grows cells on a flat surface does not fully recapitulate the pathological phenotypes. Alternatively, the three-dimension (3D) cell culture provides cell-cell and cell-ECM interaction that better mimics tissue-like structure. Thus, it has gained increasing attention recently. Yet, the expenses, time-consuming, and complications of cellular and biomolecular analysis are still major limitations of 3D culture. Herein, we describe a cost-effective and simplified workflow of the 3D neuronal cell-laden agarose-laminin preparation and the isolation of cells, RNAs, and proteins from the scaffold. To study the effects of the amyloidogenic condition in neurons, we utilized a neuron-like cell line, SH-SY5Y, and induced the amyloidogenic condition by using an amyloid forty-two inducer (Aftin-4). The effectiveness of RNAs, proteins and cells isolation from 3D scaffold enables us to investigate the cellular and molecular mechanisms underlying amyloidogenic cascade in neuronal cells. The results show that SH-SY5Y cultured in agarose-laminin scaffold differentiated to a mature TUJ1-expressing neuron cell on day 7. Furthermore, the gene expression profile from the Aftin-4-induced amyloidogenic condition revealed the expression of relevant gene-encoding proteins in the amyloidogenic pathway, including APP, BACE1, PS1, and PS2. This platform could induce the amyloid-beta 42 secretion and entrap secreted proteins in the scaffold. The induction of amyloidogenic conditions in a 3D culture facilitates the interaction between secreted amyloid-beta and neurons, which makes it resembles the pathological environment in Alzheimer’s brain. Together, this workflow is applicable for studying the cellular and molecular analysis of amyloid-induced neuronal toxicity, such as those occurred in Alzheimer’s disease progression. Importantly, our method is cost-effective, reproducible, and easy to manipulate.
Collapse
Affiliation(s)
- Poommaree Namchaiw
- Biological Engineering Program, Faculty of Engineering, King Mongkut’s University of Technology Thonburi, Thung Kru, Bangkok, Thailand
- Neuroscience Center for Research and Innovation, Learning Institute, King Mongkut’s University of Technology Thonburi, Thung Kru, Bangkok, Thailand
- * E-mail:
| | - Patapon Bunreangsri
- Neuroscience Center for Research and Innovation, Learning Institute, King Mongkut’s University of Technology Thonburi, Thung Kru, Bangkok, Thailand
| | - Piyaporn Eiamcharoen
- Department of Pathology, Faculty of Veterinary Medicine, Kasetsart University, Thung Kru, Bangkok, Thailand
- Veterinary Medical Teaching Hospital, University of California Davis, Davis, California, United States of America
| | - Salita Eiamboonsert
- Biological Engineering Program, Faculty of Engineering, King Mongkut’s University of Technology Thonburi, Thung Kru, Bangkok, Thailand
- Media Technology, King Mongkut’s University of Technology Thonburi, Thung Kru, Bangkok, Thailand
| | - Rungtiva P. Poo-arporn
- Biological Engineering Program, Faculty of Engineering, King Mongkut’s University of Technology Thonburi, Thung Kru, Bangkok, Thailand
| |
Collapse
|
102
|
Human Brain Organoid: A Versatile Tool for Modeling Neurodegeneration Diseases and for Drug Screening. Stem Cells Int 2022; 2022:2150680. [PMID: 36061149 PMCID: PMC9436613 DOI: 10.1155/2022/2150680] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 05/28/2022] [Accepted: 06/18/2022] [Indexed: 11/17/2022] Open
Abstract
Clinical trials serve as the fundamental prerequisite for clinical therapy of human disease, which is primarily based on biomedical studies in animal models. Undoubtedly, animal models have made a significant contribution to gaining insight into the developmental and pathophysiological understanding of human diseases. However, none of the existing animal models could efficiently simulate the development of human organs and systems due to a lack of spatial information; the discrepancy in genetic, anatomic, and physiological basis between animals and humans limits detailed investigation. Therefore, the translational efficiency of the research outcomes in clinical applications was significantly weakened, especially for some complex, chronic, and intractable diseases. For example, the clinical trials for human fragile X syndrome (FXS) solely based on animal models have failed such as mGluR5 antagonists. To mimic the development of human organs more faithfully and efficiently translate in vitro biomedical studies to clinical trials, extensive attention to organoids derived from stem cells contributes to a deeper understanding of this research. The organoids are a miniaturized version of an organ generated in vitro, partially recapitulating key features of human organ development. As such, the organoids open a novel avenue for in vitro models of human disease, advantageous over the existing animal models. The invention of organoids has brought an innovative breakthrough in regeneration medicine. The organoid-derived human tissues or organs could potentially function as invaluable platforms for biomedical studies, pathological investigation of human diseases, and drug screening. Importantly, the study of regeneration medicine and the development of therapeutic strategies for human diseases could be conducted in a dish, facilitating in vitro analysis and experimentation. Thus far, the pilot breakthrough has been made in the generation of numerous types of organoids representing different human organs. Most of these human organoids have been employed for in vitro biomedical study and drug screening. However, the efficiency and quality of the organoids in recapitulating the development of human organs have been hindered by engineering and conceptual challenges. The efficiency and quality of the organoids are essential for downstream applications. In this article, we highlight the application in the modeling of human neurodegenerative diseases (NDDs) such as FXS, Alzheimer's disease (AD), Parkinson's disease (PD), and autistic spectrum disorders (ASD), and organoid-based drug screening. Additionally, challenges and weaknesses especially for limits of the brain organoid models in modeling late onset NDDs such as AD and PD., and future perspectives regarding human brain organoids are addressed.
Collapse
|
103
|
Glasauer SMK, Goderie SK, Rauch JN, Guzman E, Audouard M, Bertucci T, Joy S, Rommelfanger E, Luna G, Keane-Rivera E, Lotz S, Borden S, Armando AM, Quehenberger O, Temple S, Kosik KS. Human tau mutations in cerebral organoids induce a progressive dyshomeostasis of cholesterol. Stem Cell Reports 2022; 17:2127-2140. [PMID: 35985329 PMCID: PMC9481908 DOI: 10.1016/j.stemcr.2022.07.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 11/26/2022] Open
Abstract
Mutations in the MAPT gene that encodes tau lead to frontotemporal dementia (FTD) with pathology evident in both cerebral neurons and glia. Human cerebral organoids (hCOs) from individuals harboring pathogenic tau mutations can reveal the earliest downstream effects on molecular pathways within a developmental context, generating interacting neurons and glia. We found that in hCOs carrying the V337M and R406W tau mutations, the cholesterol biosynthesis pathway in astrocytes was the top upregulated gene set compared with isogenic controls by single-cell RNA sequencing (scRNA-seq). The 15 upregulated genes included HMGCR, ACAT2, STARD4, LDLR, and SREBF2. This result was confirmed in a homozygous R406W mutant cell line by immunostaining and sterol measurements. Cholesterol abundance in the brain is tightly regulated by efflux and cholesterol biosynthetic enzyme levels in astrocytes, and dysregulation can cause aberrant phosphorylation of tau. Our findings suggest that cholesterol dyshomeostasis is an early event in the etiology of neurodegeneration caused by tau mutations. Cerebral organoid models of tauopathy caused by MAPT mutations Upregulated cholesterol and fatty acid biosynthesis genes in MAPT mutant astrocytes Elevation of cholesterol and its precursors in MAPT mutant cerebral organoids
Collapse
Affiliation(s)
- Stella M K Glasauer
- Neuroscience Research Institute and Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | | | - Jennifer N Rauch
- Neuroscience Research Institute and Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - Elmer Guzman
- Neuroscience Research Institute and Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - Morgane Audouard
- Neuroscience Research Institute and Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | | | - Shona Joy
- Neural Stem Cell Institute, Rensselaer, NY 12144, USA
| | - Emma Rommelfanger
- Neuroscience Research Institute and Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - Gabriel Luna
- Neuroscience Research Institute and Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - Erica Keane-Rivera
- Neuroscience Research Institute and Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - Steven Lotz
- Neural Stem Cell Institute, Rensselaer, NY 12144, USA
| | - Susan Borden
- Neural Stem Cell Institute, Rensselaer, NY 12144, USA
| | - Aaron M Armando
- Department of Pharmacology, University of California, San Diego, San Diego, CA 92093, USA
| | - Oswald Quehenberger
- Department of Pharmacology, University of California, San Diego, San Diego, CA 92093, USA
| | - Sally Temple
- Neural Stem Cell Institute, Rensselaer, NY 12144, USA.
| | - Kenneth S Kosik
- Neuroscience Research Institute and Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106, USA.
| |
Collapse
|
104
|
Liu C, Fu Z, Wu S, Wang X, Zhang S, Chu C, Hong Y, Wu W, Chen S, Jiang Y, Wu Y, Song Y, Liu Y, Guo X. Mitochondrial HSF1 triggers mitochondrial dysfunction and neurodegeneration in Huntington's disease. EMBO Mol Med 2022; 14:e15851. [PMID: 35670111 PMCID: PMC9260212 DOI: 10.15252/emmm.202215851] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 05/10/2022] [Accepted: 05/10/2022] [Indexed: 12/18/2022] Open
Affiliation(s)
- Chunyue Liu
- State Key Laboratory of Reproductive Medicine Key Laboratory of Human Functional Genomics of Jiangsu Province Department of Neurobiology Interdisciplinary InnoCenter for Organoids School of Basic Medical Sciences Nanjing Medical University Nanjing China
- State Key Laboratory of Reproductive Medicine Interdisciplinary InnoCenter for Organoids Institute for Stem Cell and Neural Regeneration School of Pharmacy Nanjing Medical University Nanjing China
| | - Zixing Fu
- State Key Laboratory of Reproductive Medicine Key Laboratory of Human Functional Genomics of Jiangsu Province Department of Neurobiology Interdisciplinary InnoCenter for Organoids School of Basic Medical Sciences Nanjing Medical University Nanjing China
| | - Shanshan Wu
- State Key Laboratory of Reproductive Medicine Interdisciplinary InnoCenter for Organoids Institute for Stem Cell and Neural Regeneration School of Pharmacy Nanjing Medical University Nanjing China
| | - Xiaosong Wang
- State Key Laboratory of Reproductive Medicine Key Laboratory of Human Functional Genomics of Jiangsu Province Department of Neurobiology Interdisciplinary InnoCenter for Organoids School of Basic Medical Sciences Nanjing Medical University Nanjing China
| | - Shengrong Zhang
- State Key Laboratory of Reproductive Medicine Key Laboratory of Human Functional Genomics of Jiangsu Province Department of Neurobiology Interdisciplinary InnoCenter for Organoids School of Basic Medical Sciences Nanjing Medical University Nanjing China
| | - Chu Chu
- State Key Laboratory of Reproductive Medicine Interdisciplinary InnoCenter for Organoids Institute for Stem Cell and Neural Regeneration School of Pharmacy Nanjing Medical University Nanjing China
| | - Yuan Hong
- State Key Laboratory of Reproductive Medicine Interdisciplinary InnoCenter for Organoids Institute for Stem Cell and Neural Regeneration School of Pharmacy Nanjing Medical University Nanjing China
| | - Wenbo Wu
- State Key Laboratory of Reproductive Medicine Key Laboratory of Human Functional Genomics of Jiangsu Province Department of Neurobiology Interdisciplinary InnoCenter for Organoids School of Basic Medical Sciences Nanjing Medical University Nanjing China
| | - Shengqi Chen
- State Key Laboratory of Reproductive Medicine Key Laboratory of Human Functional Genomics of Jiangsu Province Department of Neurobiology Interdisciplinary InnoCenter for Organoids School of Basic Medical Sciences Nanjing Medical University Nanjing China
| | - Yueqing Jiang
- State Key Laboratory of Reproductive Medicine Key Laboratory of Human Functional Genomics of Jiangsu Province Department of Neurobiology Interdisciplinary InnoCenter for Organoids School of Basic Medical Sciences Nanjing Medical University Nanjing China
| | - Yang Wu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics Key Laboratory of Magnetic Resonance in Biological Systems Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology Chinese Academy of Sciences Wuhan China
| | - Yongbo Song
- Department of Pharmacology Shenyang Pharmaceutical University Shenyang China
| | - Yan Liu
- State Key Laboratory of Reproductive Medicine Interdisciplinary InnoCenter for Organoids Institute for Stem Cell and Neural Regeneration School of Pharmacy Nanjing Medical University Nanjing China
| | - Xing Guo
- State Key Laboratory of Reproductive Medicine Key Laboratory of Human Functional Genomics of Jiangsu Province Department of Neurobiology Interdisciplinary InnoCenter for Organoids School of Basic Medical Sciences Nanjing Medical University Nanjing China
- Department of Endocrinology Sir Run Run Hospital Nanjing Medical University Nanjing Jiangsu China
| |
Collapse
|
105
|
Qiao H, Zhao W, Guo M, Zhu L, Chen T, Wang J, Xu X, Zhang Z, Wu Y, Chen P. Cerebral Organoids for Modeling of HSV-1-Induced-Amyloid β Associated Neuropathology and Phenotypic Rescue. Int J Mol Sci 2022; 23:ijms23115981. [PMID: 35682661 PMCID: PMC9181143 DOI: 10.3390/ijms23115981] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/20/2022] [Accepted: 05/23/2022] [Indexed: 11/16/2022] Open
Abstract
Herpes simplex virus type I (HSV-1) infection is a potential risk factor involved in the Amyloid β (Aβ) associated neuropathology. However, further understanding of the neuropathological effects of the HSV-1 infection is hampered by the limitations of existing infection models due to the distinct differences between human brains and other mammalians’ brains. Here we generated cerebral organoid models derived from pluripotent stem cells to investigate the HSV-induced Aβ associated neuropathology and the role of antiviral drugs in the phenotypic rescue. Our results identified that the HSV-1-infected cerebral organoids recapitulated Aβ associated neuropathology including the multicellular Aβ deposition, dysregulated endogenous AD mediators, reactive gliosis, neuroinflammation, and neural loss, indicating that cerebral organoids offer an opportunity for modeling the interaction of HSV-1 with the complex phenotypes across the genetic, cellular, and tissue levels of the human Alzheimer’s disease (AD). Furthermore, we identified that two antiviral drugs, namely Ribavirin (RBV) and Valacyclovir (VCV), inhibited HSV-1 replication and rescued the neuropathological phenotypes associated with AD in the HSV-1-infected cerebral organoids, implying their therapeutic potential to slow down the progression of AD. Our study provides a high-fidelity human-relevant in-vitro HSV-1 infection model to reconstitute the multiscale neuropathological features associated with AD and discover therapeutic drug candidates relevant to the AD viral hypothesis.
Collapse
Affiliation(s)
- Haowen Qiao
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China; (H.Q.); (W.Z.); (L.Z.); (T.C.); (J.W.); (X.X.)
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China;
- State Key Laboratory of Virology, Wuhan University, Wuhan 430071, China
| | - Wen Zhao
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China; (H.Q.); (W.Z.); (L.Z.); (T.C.); (J.W.); (X.X.)
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China;
| | - Moujian Guo
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China;
- State Key Laboratory of Virology, Wuhan University, Wuhan 430071, China
- Institute of Medical Virology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China
| | - Lili Zhu
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China; (H.Q.); (W.Z.); (L.Z.); (T.C.); (J.W.); (X.X.)
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China;
| | - Tao Chen
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China; (H.Q.); (W.Z.); (L.Z.); (T.C.); (J.W.); (X.X.)
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China;
| | - Jibo Wang
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China; (H.Q.); (W.Z.); (L.Z.); (T.C.); (J.W.); (X.X.)
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China;
| | - Xiaodong Xu
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China; (H.Q.); (W.Z.); (L.Z.); (T.C.); (J.W.); (X.X.)
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China;
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430050, China;
| | - Ying Wu
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China;
- State Key Laboratory of Virology, Wuhan University, Wuhan 430071, China
- Institute of Medical Virology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China
- Correspondence: (Y.W.); (P.C.)
| | - Pu Chen
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China; (H.Q.); (W.Z.); (L.Z.); (T.C.); (J.W.); (X.X.)
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China;
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430050, China;
- Correspondence: (Y.W.); (P.C.)
| |
Collapse
|
106
|
Reducing PDK1/Akt Activity: An Effective Therapeutic Target in the Treatment of Alzheimer's Disease. Cells 2022; 11:cells11111735. [PMID: 35681431 PMCID: PMC9179555 DOI: 10.3390/cells11111735] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/16/2022] [Accepted: 05/24/2022] [Indexed: 12/11/2022] Open
Abstract
Alzheimer’s disease (AD) is a common age-related neurodegenerative disease that leads to memory loss and cognitive function damage due to intracerebral neurofibrillary tangles (NFTs) and amyloid-β (Aβ) protein deposition. The phosphoinositide-dependent protein kinase (PDK1)/protein kinase B (Akt) signaling pathway plays a significant role in neuronal differentiation, synaptic plasticity, neuronal survival, and neurotransmission via the axon–dendrite axis. The phosphorylation of PDK1 and Akt rises in the brain, resulting in phosphorylation of the TNF-α-converting enzyme (TACE) at its cytoplasmic tail (the C-terminal end), changing its internalization as well as its trafficking. The current review aimed to explain the mechanisms of the PDK1/Akt/TACE signaling axis that exerts its modulatory effect on AD physiopathology. We provide an overview of the neuropathological features, genetics, Aβ aggregation, Tau protein hyperphosphorylation, neuroinflammation, and aging in the AD brain. Additionally, we summarized the phosphoinositide 3-kinase (PI3K)/PDK1/Akt pathway-related features and its molecular mechanism that is dependent on TACE in the pathogenesis of AD. This study reviewed the relationship between the PDK1/Akt signaling pathway and AD, and discussed the role of PDK1/Akt in resisting neuronal toxicity by suppressing TACE expression in the cell membrane. This work also provides a perspective for developing new therapeutics targeting PDK1/Akt and TACE for the treatment of AD.
Collapse
|
107
|
Ding L, Xiao Z, Gong X, Peng Y. Knowledge graphs of ethical concerns of cerebral organoids. Cell Prolif 2022; 55:e13239. [PMID: 35582763 PMCID: PMC9357362 DOI: 10.1111/cpr.13239] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/11/2022] [Accepted: 04/13/2022] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVES The rapid development of cerebral organoid technology and the gradual maturity of cerebral organoids highlight the necessity of foresighted research on relevant ethical concerns. We employed knowledge graphs and conducted statistical analysis with CiteSpace for a comprehensive analysis of the status quo of the research on the ethical concerns of cerebral organoids from a bibliometric perspective. MATERIALS AND METHODS We performed a statistical analysis of published papers on cerebral organoid ethics, keyword co-occurrence graph, literature co-citation and knowledge clustering graph to examine the status of the ethics research, internal relationship between technological development and ethical research, and ethical concerns of the academia. Finally, we used a keyword time zone graph and related statistics to analyze and predict the trends and popular topics of future cerebral organoids ethics research. RESULTS We demonstrated that although the ethical concerns of cerebral organoids have long been discussed, it was not until 2017 that the ethical issues began to receive more attention, when cerebral organoids were gradually mimicking the human brain more closely and increasingly being combined with chimera research. The recent key ethical concerns are primarily divided into three categories: concerns that are common in life sciences, specific to cerebral organoids, and present in cross-fields. These increasing ethical concerns are inherently related to the continual development of technology. The analysis pointed out that future research should focus on the ethical concerns of consciousness that are unique to cerebral organoids, ethical concerns of cross-fields, and construction and improvement of legislative and regulatory systems. CONCLUSIONS Although research on cerebral organoids can benefit the biomedicine field, the relevant ethical concerns are significant and have received increasing attention, which are inherently related to the continual development of technology. Future studies in ethics regarding cerebral organoid research should focus on the ethical concerns of consciousness, and cross-fields, as well as the improvement of regulatory systems.
Collapse
Affiliation(s)
- Lulu Ding
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Zhenyu Xiao
- School of Life Science, Beijing Institute of Technology, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Xia Gong
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yaojin Peng
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
108
|
Scuto M, Modafferi S, Rampulla F, Zimbone V, Tomasello M, Spano’ S, Ontario M, Palmeri A, Trovato Salinaro A, Siracusa R, Di Paola R, Cuzzocrea S, Calabrese E, Wenzel U, Calabrese V. Redox modulation of stress resilience by Crocus Sativus L. for potential neuroprotective and anti-neuroinflammatory applications in brain disorders: From molecular basis to therapy. Mech Ageing Dev 2022; 205:111686. [DOI: 10.1016/j.mad.2022.111686] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/18/2022] [Accepted: 05/18/2022] [Indexed: 12/13/2022]
|
109
|
Hasan MF, Trushina E. Advances in Recapitulating Alzheimer's Disease Phenotypes Using Human Induced Pluripotent Stem Cell-Based In Vitro Models. Brain Sci 2022; 12:552. [PMID: 35624938 PMCID: PMC9138647 DOI: 10.3390/brainsci12050552] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/24/2022] [Accepted: 04/24/2022] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is an incurable neurodegenerative disorder and the leading cause of death among older individuals. Available treatment strategies only temporarily mitigate symptoms without modifying disease progression. Recent studies revealed the multifaceted neurobiology of AD and shifted the target of drug development. Established animal models of AD are mostly tailored to yield a subset of disease phenotypes, which do not recapitulate the complexity of sporadic late-onset AD, the most common form of the disease. The use of human induced pluripotent stem cells (HiPSCs) offers unique opportunities to fill these gaps. Emerging technology allows the development of disease models that recapitulate a brain-like microenvironment using patient-derived cells. These models retain the individual's unraveled genetic background, yielding clinically relevant disease phenotypes and enabling cost-effective, high-throughput studies for drug discovery. Here, we review the development of various HiPSC-based models to study AD mechanisms and their application in drug discovery.
Collapse
Affiliation(s)
- Md Fayad Hasan
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA;
| | - Eugenia Trushina
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
110
|
Han ZZ, Kang SG, Arce L, Westaway D. Prion-like strain effects in tauopathies. Cell Tissue Res 2022; 392:179-199. [PMID: 35460367 PMCID: PMC9034081 DOI: 10.1007/s00441-022-03620-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 03/25/2022] [Indexed: 12/30/2022]
Abstract
Tau is a microtubule-associated protein that plays crucial roles in physiology and pathophysiology. In the realm of dementia, tau protein misfolding is associated with a wide spectrum of clinicopathologically diverse neurodegenerative diseases, collectively known as tauopathies. As proposed by the tau strain hypothesis, the intrinsic heterogeneity of tauopathies may be explained by the existence of structurally distinct tau conformers, “strains”. Tau strains can differ in their associated clinical features, neuropathological profiles, and biochemical signatures. Although prior research into infectious prion proteins offers valuable lessons for studying how a protein-only pathogen can encompass strain diversity, the underlying mechanism by which tau subtypes are generated remains poorly understood. Here we summarize recent advances in understanding different tau conformers through in vivo and in vitro experimental paradigms, and the implications of heterogeneity of pathological tau species for drug development.
Collapse
Affiliation(s)
- Zhuang Zhuang Han
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2M8, Canada.,Department of Medicine, University of Alberta, Edmonton, AB, Canada.,Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Sang-Gyun Kang
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2M8, Canada.,Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Luis Arce
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2M8, Canada.,Department of Medicine, University of Alberta, Edmonton, AB, Canada.,Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - David Westaway
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2M8, Canada. .,Department of Medicine, University of Alberta, Edmonton, AB, Canada. .,Department of Biochemistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
111
|
Martinelli I, Tayebati SK, Tomassoni D, Nittari G, Roy P, Amenta F. Brain and Retinal Organoids for Disease Modeling: The Importance of In Vitro Blood–Brain and Retinal Barriers Studies. Cells 2022; 11:cells11071120. [PMID: 35406683 PMCID: PMC8997725 DOI: 10.3390/cells11071120] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/16/2022] [Accepted: 03/22/2022] [Indexed: 11/16/2022] Open
Abstract
Brain and retinal organoids are functional and dynamic in vitro three-dimensional (3D) structures derived from pluripotent stem cells that spontaneously organize themselves to their in vivo counterparts. Here, we review the main literature data of how these organoids have been developed through different protocols and how they have been technically analyzed. Moreover, this paper reviews recent advances in using organoids to model neurological and retinal diseases, considering their potential for translational applications but also pointing out their limitations. Since the blood–brain barrier (BBB) and blood–retinal barrier (BRB) are understood to play a fundamental role respectively in brain and eye functions, both in health and in disease, we provide an overview of the progress in the development techniques of in vitro models as reliable and predictive screening tools for BBB and BRB-penetrating compounds. Furthermore, we propose potential future directions for brain and retinal organoids, in which dedicated biobanks will represent a novel tool for neuroscience and ophthalmology research.
Collapse
Affiliation(s)
- Ilenia Martinelli
- School of Medicinal and Health Products Sciences, University of Camerino, 62032 Camerino, Italy; (S.K.T.); (G.N.); (F.A.)
- Correspondence:
| | - Seyed Khosrow Tayebati
- School of Medicinal and Health Products Sciences, University of Camerino, 62032 Camerino, Italy; (S.K.T.); (G.N.); (F.A.)
| | - Daniele Tomassoni
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy; (D.T.); (P.R.)
| | - Giulio Nittari
- School of Medicinal and Health Products Sciences, University of Camerino, 62032 Camerino, Italy; (S.K.T.); (G.N.); (F.A.)
| | - Proshanta Roy
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy; (D.T.); (P.R.)
| | - Francesco Amenta
- School of Medicinal and Health Products Sciences, University of Camerino, 62032 Camerino, Italy; (S.K.T.); (G.N.); (F.A.)
| |
Collapse
|
112
|
Cell models for Down syndrome-Alzheimer’s disease research. Neuronal Signal 2022; 6:NS20210054. [PMID: 35449591 PMCID: PMC8996251 DOI: 10.1042/ns20210054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 03/07/2022] [Accepted: 03/21/2022] [Indexed: 11/29/2022] Open
Abstract
Down syndrome (DS) is the most common chromosomal abnormality and leads to intellectual disability, increased risk of cardiac defects, and an altered immune response. Individuals with DS have an extra full or partial copy of chromosome 21 (trisomy 21) and are more likely to develop early-onset Alzheimer’s disease (AD) than the general population. Changes in expression of human chromosome 21 (Hsa21)-encoded genes, such as amyloid precursor protein (APP), play an important role in the pathogenesis of AD in DS (DS-AD). However, the mechanisms of DS-AD remain poorly understood. To date, several mouse models with an extra copy of genes syntenic to Hsa21 have been developed to characterise DS-AD-related phenotypes. Nonetheless, due to genetic and physiological differences between mouse and human, mouse models cannot faithfully recapitulate all features of DS-AD. Cells differentiated from human-induced pluripotent stem cells (iPSCs), isolated from individuals with genetic diseases, can be used to model disease-related cellular and molecular pathologies, including DS. In this review, we will discuss the limitations of mouse models of DS and how these can be addressed using recent advancements in modelling DS using human iPSCs and iPSC-mouse chimeras, and potential applications of iPSCs in preclinical studies for DS-AD.
Collapse
|
113
|
Susaimanickam PJ, Kiral FR, Park IH. Region Specific Brain Organoids to Study Neurodevelopmental Disorders. Int J Stem Cells 2022; 15:26-40. [PMID: 35220290 PMCID: PMC8889336 DOI: 10.15283/ijsc22006] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 01/17/2022] [Indexed: 12/03/2022] Open
Abstract
Region specific brain organoids are brain organoids derived by patterning protocols using extrinsic signals as opposed to cerebral organoids obtained by self-patterning. The main focus of this review is to discuss various region-specific brain organoids developed so far and their application in modeling neurodevelopmental disease. We first discuss the principles of neural axis formation by series of growth factors, such as SHH, WNT, BMP signalings, that are critical to generate various region-specific brain organoids. Then we discuss various neurodevelopmental disorders modeled so far with these region-specific brain organoids, and findings made on mechanism and treatment options for neurodevelopmental disorders (NDD).
Collapse
Affiliation(s)
- Praveen Joseph Susaimanickam
- Department of Genetics, Yale Stem Cell Center, Yale Child Study Center, Yale School of Medicine, New Haven, CT, USA
| | - Ferdi Ridvan Kiral
- Department of Genetics, Yale Stem Cell Center, Yale Child Study Center, Yale School of Medicine, New Haven, CT, USA
| | - In-Hyun Park
- Department of Genetics, Yale Stem Cell Center, Yale Child Study Center, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
114
|
Cuanalo-Contreras K, Benkmann D. Towards More Human and Humane Testing: The Role of the Device Supplier Industry. Altern Lab Anim 2022; 50:62-70. [PMID: 35184611 DOI: 10.1177/02611929211073132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In vivo testing has been the gold standard for preclinical drug development and toxicology. However, animal-based methods often lack human relevance and have a low predictability rate, not to mention the enormous ethical and financial concerns associated with their use. For instance, according to the US Congressional Budget Office (cbo.gov), it takes an average of 10.5 years to take a compound from the preclinical phase to the market, with a cost of US$1-US$2 billion. 90% of drugs that are tested in animals and enter clinical trials fail due to lack of safety and efficacy - this fact questions the significance of in vivo testing. Cells in culture can recapitulate certain aspects of physiology and disease, as well as indicate drug responses and toxicity. Thus, they represent a sophisticated human-relevant and humane alternative. With advances in the in vitro field, it is anticipated that confidence will be gained towards a move away from traditional in vivo models. Specialised supplier industries have been a driving force in the transition to non-animal research, by translating new approach methodologies into scalable products that have been adopted by the regulatory and testing industries - but we are still at the beginning. In this article, we introduce the perspective of the device supplier industry on the current challenges and opportunities surrounding the adoption of new in vitro methods, with the goal of promoting effective co-operation with scientists and other stakeholders. In addition, we highlight some examples of where non-animal approaches have been used in regulatory submissions, as well as listing some educational and training resources that can help when selecting the most appropriate assay.
Collapse
|
115
|
Barak M, Fedorova V, Pospisilova V, Raska J, Vochyanova S, Sedmik J, Hribkova H, Klimova H, Vanova T, Bohaciakova D. Human iPSC-Derived Neural Models for Studying Alzheimer's Disease: from Neural Stem Cells to Cerebral Organoids. Stem Cell Rev Rep 2022; 18:792-820. [PMID: 35107767 PMCID: PMC8930932 DOI: 10.1007/s12015-021-10254-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2021] [Indexed: 12/05/2022]
Abstract
During the past two decades, induced pluripotent stem cells (iPSCs) have been widely used to study mechanisms of human neural development, disease modeling, and drug discovery in vitro. Especially in the field of Alzheimer’s disease (AD), where this treatment is lacking, tremendous effort has been put into the investigation of molecular mechanisms behind this disease using induced pluripotent stem cell-based models. Numerous of these studies have found either novel regulatory mechanisms that could be exploited to develop relevant drugs for AD treatment or have already tested small molecules on in vitro cultures, directly demonstrating their effect on amelioration of AD-associated pathology. This review thus summarizes currently used differentiation strategies of induced pluripotent stem cells towards neuronal and glial cell types and cerebral organoids and their utilization in modeling AD and potential drug discovery.
Collapse
Affiliation(s)
- Martin Barak
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic
| | - Veronika Fedorova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic
| | - Veronika Pospisilova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic
| | - Jan Raska
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic
| | - Simona Vochyanova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic
| | - Jiri Sedmik
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic
- International Clinical Research Center, St. Anne's Faculty Hospital Brno, Brno, Czech Republic
| | - Hana Hribkova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic
| | - Hana Klimova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic
| | - Tereza Vanova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic
- International Clinical Research Center, St. Anne's Faculty Hospital Brno, Brno, Czech Republic
| | - Dasa Bohaciakova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic.
- International Clinical Research Center, St. Anne's Faculty Hospital Brno, Brno, Czech Republic.
| |
Collapse
|
116
|
Hou PS, Kuo HC. Central nervous system organoids for modeling neurodegenerative diseases. IUBMB Life 2022; 74:812-825. [PMID: 35102668 DOI: 10.1002/iub.2595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/18/2021] [Accepted: 12/02/2021] [Indexed: 11/07/2022]
Abstract
Recent advances in induced pluripotent stem cell (iPSC) technology have allowed researchers to generate neurodegenerative disease-specific iPSCs and use the cells to derive a variety of relevant cell populations for laboratory modeling and drug testing. Nevertheless, these efforts have faced challenges related to immaturity and lack of complex developmental niches in the derived cell populations, limiting the utility of these in vitro models of neurodegenerative disease. Such limitations may be overcome by using human iPSC technology to generate three-dimensional (3D) brain organoids, which better recapitulate in vivo tissue architecture than traditional neuronal cultures to provide more complex and representative disease models and drug testing systems. In this review, we focus on the application of pluripotent stem cell-derived central nervous system (CNS) organoids to model neurodegenerative diseases. We first summarize recent progress in generating and characterizing various CNS organoids from pluripotent stem cells. We then review the application of CNS organoids for modeling several different human neurodegenerative diseases. We also describe several novel pathological mechanisms and drugs that were studied using patient iPSC-derived CNS organoids. Finally, we discuss remaining challenges and emerging opportunities for the use of 3D brain organoids for in vitro modeling of CNS development and neurodegeneration.
Collapse
Affiliation(s)
- Pei-Shan Hou
- Institute of Anatomy and Cell Biology, National Yang-Ming Chiao-Tung University, Taipei, Taiwan.,Brain Research Center, National Yang-Ming Chiao-Tung University, Taipei, Taiwan
| | - Hung-Chih Kuo
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan.,Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
117
|
Chyr J, Gong H, Zhou X. DOTA: Deep Learning Optimal Transport Approach to Advance Drug Repositioning for Alzheimer's Disease. Biomolecules 2022; 12:196. [PMID: 35204697 PMCID: PMC8961573 DOI: 10.3390/biom12020196] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/16/2022] [Accepted: 01/22/2022] [Indexed: 02/04/2023] Open
Abstract
Alzheimer's disease (AD) is the leading cause of age-related dementia, affecting over 5 million people in the United States and incurring a substantial global healthcare cost. Unfortunately, current treatments are only palliative and do not cure AD. There is an urgent need to develop novel anti-AD therapies; however, drug discovery is a time-consuming, expensive, and high-risk process. Drug repositioning, on the other hand, is an attractive approach to identify drugs for AD treatment. Thus, we developed a novel deep learning method called DOTA (Drug repositioning approach using Optimal Transport for Alzheimer's disease) to repurpose effective FDA-approved drugs for AD. Specifically, DOTA consists of two major autoencoders: (1) a multi-modal autoencoder to integrate heterogeneous drug information and (2) a Wasserstein variational autoencoder to identify effective AD drugs. Using our approach, we predict that antipsychotic drugs with circadian effects, such as quetiapine, aripiprazole, risperidone, suvorexant, brexpiprazole, olanzapine, and trazadone, will have efficacious effects in AD patients. These drugs target important brain receptors involved in memory, learning, and cognition, including serotonin 5-HT2A, dopamine D2, and orexin receptors. In summary, DOTA repositions promising drugs that target important biological pathways and are predicted to improve patient cognition, circadian rhythms, and AD pathogenesis.
Collapse
Affiliation(s)
- Jacqueline Chyr
- Center for Computational Systems Medicine, School of Biomedical Informatics, University of Texas Health Science Center, Houston, TX 77030, USA;
| | - Haoran Gong
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China;
| | - Xiaobo Zhou
- Center for Computational Systems Medicine, School of Biomedical Informatics, University of Texas Health Science Center, Houston, TX 77030, USA;
| |
Collapse
|
118
|
In Vitro Methodologies to Study the Role of Advanced Glycation End Products (AGEs) in Neurodegeneration. Nutrients 2022; 14:nu14020363. [PMID: 35057544 PMCID: PMC8777776 DOI: 10.3390/nu14020363] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 02/07/2023] Open
Abstract
Advanced glycation end products (AGEs) can be present in food or be endogenously produced in biological systems. Their formation has been associated with chronic neurodegenerative diseases such as Alzheimer’s disease, Parkinson’s disease, multiple sclerosis, and amyotrophic lateral sclerosis. The implication of AGEs in neurodegeneration is related to their ability to bind to AGE-specific receptors and the ability of their precursors to induce the so-called “dicarbonyl stress”, resulting in cross-linking and protein damage. However, the mode of action underlying their role in neurodegeneration remains unclear. While some research has been carried out in observational clinical studies, further in vitro studies may help elucidate these underlying modes of action. This review presents and discusses in vitro methodologies used in research on the potential role of AGEs in neuroinflammation and neurodegeneration. The overview reveals the main concepts linking AGEs to neurodegeneration, the current findings, and the available and advisable in vitro models to study their role. Moreover, the major questions regarding the role of AGEs in neurodegenerative diseases and the challenges and discrepancies in the research field are discussed.
Collapse
|
119
|
Borges AC, Broersen K, Leandro P, Fernandes TG. Engineering Organoids for in vitro Modeling of Phenylketonuria. Front Mol Neurosci 2022; 14:787242. [PMID: 35082602 PMCID: PMC8784555 DOI: 10.3389/fnmol.2021.787242] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/29/2021] [Indexed: 12/15/2022] Open
Abstract
Phenylketonuria is a recessive genetic disorder of amino-acid metabolism, where impaired phenylalanine hydroxylase function leads to the accumulation of neurotoxic phenylalanine levels in the brain. Severe cognitive and neuronal impairment are observed in untreated/late-diagnosed patients, and even early treated ones are not safe from life-long sequelae. Despite the wealth of knowledge acquired from available disease models, the chronic effect of Phenylketonuria in the brain is still poorly understood and the consequences to the aging brain remain an open question. Thus, there is the need for better predictive models, able to recapitulate specific mechanisms of this disease. Human induced pluripotent stem cells (hiPSCs), with their ability to differentiate and self-organize in multiple tissues, might provide a new exciting in vitro platform to model specific PKU-derived neuronal impairment. In this review, we gather what is known about the impact of phenylalanine in the brain of patients and highlight where hiPSC-derived organoids could contribute to the understanding of this disease.
Collapse
Affiliation(s)
- Alice C. Borges
- Department of Bioengineering and iBB – Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory i4HB – Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Kerensa Broersen
- Department of Applied Stem Cell Technologies, Faculty of Science and Technology, Technical Medical Centre, University of Twente, Enschede, Netherlands
| | - Paula Leandro
- Faculty of Pharmacy, iMed.ULisboa - Research Institute for Medicines, Universidade de Lisboa, Lisbon, Portugal
| | - Tiago G. Fernandes
- Department of Bioengineering and iBB – Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory i4HB – Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- *Correspondence: Tiago G. Fernandes,
| |
Collapse
|
120
|
Fanizza F, Campanile M, Forloni G, Giordano C, Albani D. Induced pluripotent stem cell-based organ-on-a-chip as personalized drug screening tools: A focus on neurodegenerative disorders. J Tissue Eng 2022; 13:20417314221095339. [PMID: 35570845 PMCID: PMC9092580 DOI: 10.1177/20417314221095339] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 04/04/2022] [Indexed: 01/15/2023] Open
Abstract
The Organ-on-a-Chip (OoC) technology shows great potential to revolutionize the drugs development pipeline by mimicking the physiological environment and functions of human organs. The translational value of OoC is further enhanced when combined with patient-specific induced pluripotent stem cells (iPSCs) to develop more realistic disease models, paving the way for the development of a new generation of patient-on-a-chip devices. iPSCs differentiation capacity leads to invaluable improvements in personalized medicine. Moreover, the connection of single-OoC into multi-OoC or body-on-a-chip allows to investigate drug pharmacodynamic and pharmacokinetics through the study of multi-organs cross-talks. The need of a breakthrough thanks to this technology is particularly relevant within the field of neurodegenerative diseases, where the number of patients is increasing and the successful rate in drug discovery is worryingly low. In this review we discuss current iPSC-based OoC as drug screening models and their implication in development of new therapies for neurodegenerative disorders.
Collapse
Affiliation(s)
- Francesca Fanizza
- Department of Chemistry, Materials and
Chemical Engineering “Giulio Natta,” Politecnico di Milano, Milan, Italy
| | - Marzia Campanile
- Department of Chemistry, Materials and
Chemical Engineering “Giulio Natta,” Politecnico di Milano, Milan, Italy
| | - Gianluigi Forloni
- Department of Neuroscience, Istituto di
Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Carmen Giordano
- Department of Chemistry, Materials and
Chemical Engineering “Giulio Natta,” Politecnico di Milano, Milan, Italy
| | - Diego Albani
- Department of Neuroscience, Istituto di
Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| |
Collapse
|
121
|
Allen GE, Dhanda AS, Julian LM. Emerging Methods in Modeling Brain Development and Disease with Human Pluripotent Stem Cells. Methods Mol Biol 2022; 2515:319-342. [PMID: 35776361 DOI: 10.1007/978-1-0716-2409-8_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The Nobel Prize-winning discovery that human somatic cells can be readily reprogrammed into pluripotent cells has revolutionized our potential to understand the human brain. The rapid technological progression of this field has made it possible to easily obtain human neural cells and even intact tissues, offering invaluable resources to model human brain development. In this chapter, we present a brief history of hPSC-based approaches to study brain development and then, provide new insights into neurological diseases, focusing on those driven by aberrant cell death. Furthermore, we will shed light on the latest technologies and highlight the methods that researchers can use to employ established hPSC approaches in their research. Our intention is to demonstrate that hPSC-based modeling is a technical approach accessible to all researchers who seek a deeper understanding of the human brain.
Collapse
Affiliation(s)
- George E Allen
- Department of Biological Sciences; Centre for Cell Biology, Development, and Disease, Faculty of Science, Simon Fraser University, Burnaby, BC, Canada
| | - Aaron S Dhanda
- Department of Biological Sciences; Centre for Cell Biology, Development, and Disease, Faculty of Science, Simon Fraser University, Burnaby, BC, Canada
| | - Lisa M Julian
- Department of Biological Sciences; Centre for Cell Biology, Development, and Disease, Faculty of Science, Simon Fraser University, Burnaby, BC, Canada.
| |
Collapse
|
122
|
Cordella F, Brighi C, Soloperto A, Di Angelantonio S. Stem cell-based 3D brain organoids for mimicking, investigating, and challenging Alzheimer's diseases. Neural Regen Res 2022; 17:330-332. [PMID: 34269204 PMCID: PMC8463991 DOI: 10.4103/1673-5374.317976] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Affiliation(s)
- Federica Cordella
- Center for Life Nanoscience, Istituto Italiano di Tecnologia; Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Carlo Brighi
- Center for Life Nanoscience, Istituto Italiano di Tecnologia; Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | | | - Silvia Di Angelantonio
- Center for Life Nanoscience, Istituto Italiano di Tecnologia; Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| |
Collapse
|
123
|
Kumar D, Hassan MI. Neurodegenerative brain models vs. cell replacement or restoration therapy: A review on promises and pitfalls. Biochem Biophys Res Commun 2021; 585:124-131. [PMID: 34801932 DOI: 10.1016/j.bbrc.2021.11.040] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 11/11/2021] [Indexed: 01/17/2023]
Abstract
Disease models have proven useful tools for gaining deeper mechanistic insights into neurodegenerative diseases. In this context, stem cell technology is effective, especially induced pluripotent stem cell (iPSC)-derived brain organoids and cell replacement/restoration which can be used for personalized medicine, allowing physicians to test the efficacy of drugs in vitro before delivering them to patients, enabling more precise and personalized treatment. Nonetheless, it offers the potential to minimize (or even eliminate) the use of animals, provides important clues for disease processes, and accelerates therapeutic strategies. Perhaps in the not-too-distant future, organoid models of the human brain will be able to link blood-brain barrier cultures with other liver cultures, simulating blood flow across organs and as a method of testing medicines, giving crucial pharmacokinetics and pharmacodynamics data. Simultaneously, stem cell interventions for cell replacements or restoration therapy would enable us to realize efficacious and realistic therapeutic options for Neurodegenerative diseases.
Collapse
Affiliation(s)
- Dhiraj Kumar
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India.
| |
Collapse
|
124
|
Ramirez S, Mukherjee A, Sepulveda SE, Gherardelli C, Becerra-Calixto A, Bravo-Vasquez N, Soto C. Protocol for controlled cortical impact in human cerebral organoids to model traumatic brain injury. STAR Protoc 2021; 2:100987. [PMID: 34927096 PMCID: PMC8649394 DOI: 10.1016/j.xpro.2021.100987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Modeling traumatic brain injury (TBI) has been a challenge. Rodent and cellular models have provided relevant contributions despite their limitations. Here, we present a protocol for a TBI model based on the controlled cortical impact (CCI) performed on human cerebral organoids (COs), self-assembled 3D cultures that recapitulate features of the human brain. Here, we generate COs from iPSCs obtained from reprogrammed fibroblasts. For complete details on the use and execution of this protocol, please refer to Ramirez et al. (2021). Steps for generating human cerebral organoids (COs)from iPSCs Protocol to adapt controlled cortical impact system for use with human COs Enables the study of traumatic brain injury in human COs
Collapse
Affiliation(s)
- Santiago Ramirez
- Mitchell Center for Alzheimer’s Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School, University of Texas Health Science at Houston, Houston, TX 77030, USA
| | - Abhisek Mukherjee
- Mitchell Center for Alzheimer’s Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School, University of Texas Health Science at Houston, Houston, TX 77030, USA
| | - Sofia E. Sepulveda
- Mitchell Center for Alzheimer’s Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School, University of Texas Health Science at Houston, Houston, TX 77030, USA
| | - Camila Gherardelli
- Mitchell Center for Alzheimer’s Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School, University of Texas Health Science at Houston, Houston, TX 77030, USA
| | - Andrea Becerra-Calixto
- Mitchell Center for Alzheimer’s Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School, University of Texas Health Science at Houston, Houston, TX 77030, USA
| | - Nicolas Bravo-Vasquez
- Mitchell Center for Alzheimer’s Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School, University of Texas Health Science at Houston, Houston, TX 77030, USA
| | - Claudio Soto
- Mitchell Center for Alzheimer’s Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School, University of Texas Health Science at Houston, Houston, TX 77030, USA
- Corresponding author
| |
Collapse
|
125
|
Sharma NS, Karan A, Lee D, Yan Z, Xie J. Advances in Modeling Alzheimer's Disease In Vitro. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202100097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- Navatha Shree Sharma
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program University of Nebraska Medical Center Omaha NE 68198 USA
| | - Anik Karan
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program University of Nebraska Medical Center Omaha NE 68198 USA
| | - Donghee Lee
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program University of Nebraska Medical Center Omaha NE 68198 USA
| | - Zheng Yan
- Department of Mechanical & Aerospace Engineering and Department of Biomedical Biological and Chemical Engineering University of Missouri Columbia MO 65211 USA
| | - Jingwei Xie
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program University of Nebraska Medical Center Omaha NE 68198 USA
- Department of Mechanical and Materials Engineering College of Engineering University of Nebraska Lincoln Lincoln NE 68588 USA
| |
Collapse
|
126
|
Supakul S, Okano H, Maeda S. Utilization of Human Induced Pluripotent Stem Cells-Derived In vitro Models for the Future Study of Sex Differences in Alzheimer's Disease. Front Aging Neurosci 2021; 13:768948. [PMID: 34803659 PMCID: PMC8599796 DOI: 10.3389/fnagi.2021.768948] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/15/2021] [Indexed: 01/15/2023] Open
Abstract
Alzheimer’s disease (AD) is an aging-dependent neurodegenerative disease that impairs cognitive function. Although the main pathologies of AD are the aggregation of amyloid-beta (Aβ) and phosphorylated Tau protein, the mechanisms that lead to these pathologies and their effects are believed to be heterogeneous among patients. Many epidemiological studies have suggested that sex is involved in disease prevalence and progression. The reduction of sex hormones contributes to the pathogenesis of AD, especially in females, suggesting that the supplementation of sex hormones could be a therapeutic intervention for AD. However, interventional studies have revealed that hormone therapy is beneficial under limited conditions in certain populations with specific administration methods. Thus, this suggests the importance of identifying crucial factors that determine hormonal effects in patients with AD. Based on these factors, it is necessary to decide which patients will receive the intervention before starting it. However, the long observational period and many uncontrollable environmental factors in clinical trials made it difficult to identify such factors, except for the APOE ε4 allele. Induced pluripotent stem cells (iPSCs) derived from patients can differentiate into neurons and recapitulate some aspects of AD pathogenesis. This in vitro model allows us to control non-cell autonomous factors, including the amount of Aβ aggregates and sex hormones. Hence, iPSCs provide opportunities to investigate sex-dependent pathogenesis and predict a suitable population for clinical trials of hormone treatment.
Collapse
Affiliation(s)
- Sopak Supakul
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Sumihiro Maeda
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
127
|
Bubnys A, Tsai LH. Harnessing cerebral organoids for Alzheimer's disease research. Curr Opin Neurobiol 2021; 72:120-130. [PMID: 34818608 DOI: 10.1016/j.conb.2021.10.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/20/2021] [Accepted: 10/19/2021] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder affecting the aging population. Despite many studies, there remains an urgent need to identify the root causes of AD, together with potential treatments. Cerebral organoid technology has made it possible to model human neurophysiology and disease with increasing accuracy in patient-derived tissue cultures. Here, we review the most recent advances in modeling AD in organoids and other engineered three-dimensional cell culture systems. Early studies demonstrated that familial AD patient-derived organoids robustly develop disease pathology. Ongoing work has expanded this focus to investigate the genetic and environmental causes of late-onset sporadic AD and harness organoids for high-throughput drug screens. Future organoid models will need to incorporate additional cell types and tissues implicated in disease pathogenesis, including microglia and vasculature. We anticipate the continuation of this rapid progress in developing cerebral organoid technology toward facilitating our understanding of and informing treatment strategies for AD.
Collapse
|
128
|
Steinberg DJ, Aqeilan RI. WWOX-Related Neurodevelopmental Disorders: Models and Future Perspectives. Cells 2021; 10:cells10113082. [PMID: 34831305 PMCID: PMC8623516 DOI: 10.3390/cells10113082] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/28/2021] [Accepted: 11/03/2021] [Indexed: 12/12/2022] Open
Abstract
The WW domain-containing oxidoreductase (WWOX) gene was originally discovered as a putative tumor suppressor spanning the common fragile site FRA16D, but as time has progressed the extent of its pleiotropic function has become apparent. At present, WWOX is a major source of interest in the context of neurological disorders, and more specifically developmental and epileptic encephalopathies (DEEs). This review article aims to introduce the many model systems used through the years to study its function and roles in neuropathies. Similarities and fundamental differences between rodent and human models are discussed. Finally, future perspectives and promising research avenues are suggested.
Collapse
|
129
|
Walczak PA, Perez-Esteban P, Bassett DC, Hill EJ. Modelling the central nervous system: tissue engineering of the cellular microenvironment. Emerg Top Life Sci 2021; 5:507-517. [PMID: 34524411 PMCID: PMC8589431 DOI: 10.1042/etls20210245] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/16/2021] [Accepted: 08/27/2021] [Indexed: 12/30/2022]
Abstract
With the increasing prevalence of neurodegenerative diseases, improved models of the central nervous system (CNS) will improve our understanding of neurophysiology and pathogenesis, whilst enabling exploration of novel therapeutics. Studies of brain physiology have largely been carried out using in vivo models, ex vivo brain slices or primary cell culture from rodents. Whilst these models have provided great insight into complex interactions between brain cell types, key differences remain between human and rodent brains, such as degree of cortical complexity. Unfortunately, comparative models of human brain tissue are lacking. The development of induced Pluripotent Stem Cells (iPSCs) has accelerated advancement within the field of in vitro tissue modelling. However, despite generating accurate cellular representations of cortical development and disease, two-dimensional (2D) iPSC-derived cultures lack an entire dimension of environmental information on structure, migration, polarity, neuronal circuitry and spatiotemporal organisation of cells. As such, researchers look to tissue engineering in order to develop advanced biomaterials and culture systems capable of providing necessary cues for guiding cell fates, to construct in vitro model systems with increased biological relevance. This review highlights experimental methods for engineering of in vitro culture systems to recapitulate the complexity of the CNS with consideration given to previously unexploited biophysical cues within the cellular microenvironment.
Collapse
Affiliation(s)
- Paige A. Walczak
- College of Health and Life Sciences, School of Biosciences, Aston University, Birmingham, U.K
| | - Patricia Perez-Esteban
- College of Health and Life Sciences, School of Biosciences, Aston University, Birmingham, U.K
| | - David C. Bassett
- Healthcare Technologies Institute, School of Chemical Engineering, University of Birmingham, Birmingham, U.K
| | - Eric James Hill
- College of Health and Life Sciences, School of Biosciences, Aston University, Birmingham, U.K
| |
Collapse
|
130
|
Foliaki ST, Schwarz B, Groveman BR, Walters RO, Ferreira NC, Orrù CD, Smith A, Wood A, Schmit OM, Freitag P, Yuan J, Zou W, Bosio CM, Carroll JA, Haigh CL. Neuronal excitatory-to-inhibitory balance is altered in cerebral organoid models of genetic neurological diseases. Mol Brain 2021; 14:156. [PMID: 34635127 PMCID: PMC8507222 DOI: 10.1186/s13041-021-00864-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 09/29/2021] [Indexed: 12/31/2022] Open
Abstract
The neuro-physiological properties of individuals with genetic pre-disposition to neurological disorders are largely unknown. Here we aimed to explore these properties using cerebral organoids (COs) derived from fibroblasts of individuals with confirmed genetic mutations including PRNPE200K, trisomy 21 (T21), and LRRK2G2019S, which are associated with Creutzfeldt Jakob disease, Down Syndrome, and Parkinson's disease. We utilized no known disease/healthy COs (HC) as normal function controls. At 3-4 and 6-10 months post-differentiation, COs with mutations showed no evidence of disease-related pathology. Electrophysiology assessment showed that all COs exhibited mature neuronal firing at 6-10 months old. At this age, we observed significant changes in the electrophysiology of the COs with disease-associated mutations (dCOs) as compared with the HC, including reduced neuronal network communication, slowing neuronal oscillations, and increased coupling of delta and theta phases to the amplitudes of gamma oscillations. Such changes were linked with the detection of hypersynchronous events like spike-and-wave discharges. These dysfunctions were associated with altered production and release of neurotransmitters, compromised activity of excitatory ionotropic receptors including receptors of kainate, AMPA, and NMDA, and changed levels and function of excitatory glutamatergic synapses and inhibitory GABAergic synapses. Neuronal properties that modulate GABAergic inhibition including the activity of Na-K-Cl cotransport 1 (NKCC1) in Cl- homeostasis and the levels of synaptic and extra-synaptic localization of GABA receptors (GABARs) were altered in the T21 COs only. The neurosteroid allopregnanolone, a positive modulator of GABARs, was downregulated in all the dCOs. Treatment with this neurosteroid significantly improved the neuronal communication in the dCOs, possibly through improving the GABAergic inhibition. Overall, without the manifestation of any disease-related pathology, the genetic mutations PRNPE200K, T21, and LRRK2G2019S significantly altered the neuronal network communication in dCOs by disrupting the excitatory-to-inhibitory balance.
Collapse
Affiliation(s)
- Simote T Foliaki
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Benjamin Schwarz
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Bradley R Groveman
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Ryan O Walters
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Natalia C Ferreira
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Christina D Orrù
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Anna Smith
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Aleksandar Wood
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Olivia M Schmit
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Phoebe Freitag
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Jue Yuan
- Departments of Pathology and Neurology, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Wenquan Zou
- Departments of Pathology and Neurology, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Catharine M Bosio
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - James A Carroll
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Cathryn L Haigh
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA.
| |
Collapse
|
131
|
Ramirez S, Mukherjee A, Sepulveda S, Becerra-Calixto A, Bravo-Vasquez N, Gherardelli C, Chavez M, Soto C. Modeling Traumatic Brain Injury in Human Cerebral Organoids. Cells 2021; 10:2683. [PMID: 34685663 PMCID: PMC8534257 DOI: 10.3390/cells10102683] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/30/2021] [Accepted: 10/01/2021] [Indexed: 01/22/2023] Open
Abstract
Traumatic brain injury (TBI) is a head injury that disrupts the normal brain structure and function. TBI has been extensively studied using various in vitro and in vivo models. Most of the studies have been done with rodent models, which may respond differently to TBI than human nerve cells. Taking advantage of the recent development of cerebral organoids (COs) derived from human induced pluripotent stem cells (iPSCs), which resemble the architecture of specific human brain regions, here, we adapted the controlled cortical impact (CCI) model to induce TBI in human COs as a novel in vitro platform. To adapt the CCI procedure into COs, we have developed a phantom brain matrix, matching the mechanical characteristics of the brain, altogether with an empty mouse skull as a platform to allow the use of the stereotactic CCI equipment on COs. After the CCI procedure, COs were histologically prepared to evaluate neurons and astrocyte populations using the microtubule-associated protein 2 (MAP2) and the glial fibrillary acidic protein (GFAP). Moreover, a marker of metabolic response, the neuron-specific enolase (NSE), and cellular death using cleaved caspase 3 were also analyzed. Our results show that human COs recapitulate the primary pathological changes of TBI, including metabolic alterations related to neuronal damage, neuronal loss, and astrogliosis. This novel approach using human COs to model TBI in vitro holds great potential and opens new alternatives for understanding brain abnormalities produced by TBI, and for the development and testing of new therapeutic approaches.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Claudio Soto
- Mitchell Center for Alzheimer’s Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School, University of Texas Health Science at Houston, Houston, TX 77030, USA; (S.R.); (A.M.); (S.S.); (A.B.-C.); (N.B.-V.); (C.G.); (M.C.)
| |
Collapse
|
132
|
Kang Y, Zhou Y, Li Y, Han Y, Xu J, Niu W, Li Z, Liu S, Feng H, Huang W, Duan R, Xu T, Raj N, Zhang F, Dou J, Xu C, Wu H, Bassell GJ, Warren ST, Allen EG, Jin P, Wen Z. A human forebrain organoid model of fragile X syndrome exhibits altered neurogenesis and highlights new treatment strategies. Nat Neurosci 2021; 24:1377-1391. [PMID: 34413513 PMCID: PMC8484073 DOI: 10.1038/s41593-021-00913-6] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 07/15/2021] [Indexed: 02/07/2023]
Abstract
Fragile X syndrome (FXS) is caused by the loss of fragile X mental retardation protein (FMRP), an RNA-binding protein that can regulate the translation of specific mRNAs. In this study, we developed an FXS human forebrain organoid model and observed that the loss of FMRP led to dysregulated neurogenesis, neuronal maturation and neuronal excitability. Bulk and single-cell gene expression analyses of FXS forebrain organoids revealed that the loss of FMRP altered gene expression in a cell-type-specific manner. The developmental deficits in FXS forebrain organoids could be rescued by inhibiting the phosphoinositide 3-kinase pathway but not the metabotropic glutamate pathway disrupted in the FXS mouse model. We identified a large number of human-specific mRNAs bound by FMRP. One of these human-specific FMRP targets, CHD2, contributed to the altered gene expression in FXS organoids. Collectively, our study revealed molecular, cellular and electrophysiological abnormalities associated with the loss of FMRP during human brain development.
Collapse
Affiliation(s)
- Yunhee Kang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA;,Department of Biostatistics and Bioinformatics, Emory University School of Public Health, Atlanta, GA 30322, USA
| | - Ying Zhou
- Department of Psychiatry and Behavioral Scieces, Emory University School of Medicine, Atlanta, GA 30322, USA;,Department of Biostatistics and Bioinformatics, Emory University School of Public Health, Atlanta, GA 30322, USA
| | - Yujing Li
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA;,Department of Biostatistics and Bioinformatics, Emory University School of Public Health, Atlanta, GA 30322, USA
| | - Yanfei Han
- Department of Psychiatry and Behavioral Scieces, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jie Xu
- The Graduate Program in Genetics and Molecular Biology, Emory University, GA 30322, USA
| | - Weibo Niu
- Department of Psychiatry and Behavioral Scieces, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ziyi Li
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Shiying Liu
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, OH 44106, USA
| | - Hao Feng
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, OH 44106, USA
| | - Wen Huang
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410078, China
| | - Ranhui Duan
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410078, China
| | - Tianmin Xu
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Nisha Raj
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Feiran Zhang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Juan Dou
- Department of Psychiatry and Behavioral Scieces, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Chongchong Xu
- Department of Psychiatry and Behavioral Scieces, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Hao Wu
- Department of Biostatistics and Bioinformatics, Emory University School of Public Health, Atlanta, GA 30322, USA
| | - Gary J Bassell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Stephen T Warren
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Emily G Allen
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Peng Jin
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA;,To whom correspondence should be addressed: (P.J.) and (Z.W.)
| | - Zhexing Wen
- Department of Psychiatry and Behavioral Scieces, Emory University School of Medicine, Atlanta, GA 30322, USA;,Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA;,To whom correspondence should be addressed: (P.J.) and (Z.W.)
| |
Collapse
|
133
|
Alić I, Goh PA, Murray A, Portelius E, Gkanatsiou E, Gough G, Mok KY, Koschut D, Brunmeir R, Yeap YJ, O'Brien NL, Groet J, Shao X, Havlicek S, Dunn NR, Kvartsberg H, Brinkmalm G, Hithersay R, Startin C, Hamburg S, Phillips M, Pervushin K, Turmaine M, Wallon D, Rovelet-Lecrux A, Soininen H, Volpi E, Martin JE, Foo JN, Becker DL, Rostagno A, Ghiso J, Krsnik Ž, Šimić G, Kostović I, Mitrečić D, Francis PT, Blennow K, Strydom A, Hardy J, Zetterberg H, Nižetić D. Patient-specific Alzheimer-like pathology in trisomy 21 cerebral organoids reveals BACE2 as a gene dose-sensitive AD suppressor in human brain. Mol Psychiatry 2021; 26:5766-5788. [PMID: 32647257 PMCID: PMC8190957 DOI: 10.1038/s41380-020-0806-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 05/18/2020] [Accepted: 05/29/2020] [Indexed: 11/15/2022]
Abstract
A population of more than six million people worldwide at high risk of Alzheimer's disease (AD) are those with Down Syndrome (DS, caused by trisomy 21 (T21)), 70% of whom develop dementia during lifetime, caused by an extra copy of β-amyloid-(Aβ)-precursor-protein gene. We report AD-like pathology in cerebral organoids grown in vitro from non-invasively sampled strands of hair from 71% of DS donors. The pathology consisted of extracellular diffuse and fibrillar Aβ deposits, hyperphosphorylated/pathologically conformed Tau, and premature neuronal loss. Presence/absence of AD-like pathology was donor-specific (reproducible between individual organoids/iPSC lines/experiments). Pathology could be triggered in pathology-negative T21 organoids by CRISPR/Cas9-mediated elimination of the third copy of chromosome 21 gene BACE2, but prevented by combined chemical β and γ-secretase inhibition. We found that T21 organoids secrete increased proportions of Aβ-preventing (Aβ1-19) and Aβ-degradation products (Aβ1-20 and Aβ1-34). We show these profiles mirror in cerebrospinal fluid of people with DS. We demonstrate that this protective mechanism is mediated by BACE2-trisomy and cross-inhibited by clinically trialled BACE1 inhibitors. Combined, our data prove the physiological role of BACE2 as a dose-sensitive AD-suppressor gene, potentially explaining the dementia delay in ~30% of people with DS. We also show that DS cerebral organoids could be explored as pre-morbid AD-risk population detector and a system for hypothesis-free drug screens as well as identification of natural suppressor genes for neurodegenerative diseases.
Collapse
Grants
- MR/S011277/1 Medical Research Council
- MR/L501542/1 Medical Research Council
- G-0907 Parkinson's UK
- MR/N026004/1 Medical Research Council
- MR/R024901/1 Medical Research Council
- Wellcome Trust
- 217199 Wellcome Trust
- G0901254 Medical Research Council
- MR/T002581/1 Medical Research Council
- RF1 AG059695 NIA NIH HHS
- G0701075 Medical Research Council
- 098330 Wellcome Trust
- William Harvey Academy Fellowship, co-funded by the People Programme (Marie Curie Actions) of the European Union’s Seventh Framework Programme (FP7/2007-2013) under REA grant agreement n° 608765
- Fondation pour la Recherche Médicale (Foundation for Medical Research in France)
- National Research Foundation Singapore (National Research Foundation-Prime Minister’s office, Republic of Singapore)
- BrightFocus Foundation (BrightFocus)
- Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- Svenska Forskningsrådet Formas (Swedish Research Council Formas)
- KB holds the Torsten Söderberg Professorship in Medicine at the Royal Swedish Academy of Sciences, and is supported by the Swedish Alzheimer Foundation (#AF-742881), Hjärnfonden, Sweden (#FO2017-0243), and the Swedish State Support for Clinical Research (#ALFGBG-715986).
- Wellcome Trust (Wellcome)
- JH received funding from the Dementia Research Institute, an anonymous foundation and the Dolby foundation
- HZ is a Wallenberg Academy Fellow supported by grants from the Swedish Research Council, the European Research Council, Swedish State Support for Clinical Research (ALFGBG-720931) the UK Dementia Research Institute at UCL
Collapse
Affiliation(s)
- Ivan Alić
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
- The Blizard Institute, Barts & The London School of Medicine, Queen Mary University of London, London, E1 2AT, UK
- Department of Anatomy, Histology and Embryology, Faculty of Veterinary Medicine, University of Zagreb, 10000, Zagreb, Croatia
| | - Pollyanna A Goh
- The Blizard Institute, Barts & The London School of Medicine, Queen Mary University of London, London, E1 2AT, UK
- LonDownS Consortium, London, UK
| | - Aoife Murray
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
| | - Erik Portelius
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, S-405 30, Sweden
| | - Eleni Gkanatsiou
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, S-405 30, Sweden
| | - Gillian Gough
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
| | - Kin Y Mok
- LonDownS Consortium, London, UK
- Dementia Research Institute & Reta Lila Weston Institute, Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - David Koschut
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
| | - Reinhard Brunmeir
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
| | - Yee Jie Yeap
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
| | - Niamh L O'Brien
- The Blizard Institute, Barts & The London School of Medicine, Queen Mary University of London, London, E1 2AT, UK
- LonDownS Consortium, London, UK
| | - Jürgen Groet
- The Blizard Institute, Barts & The London School of Medicine, Queen Mary University of London, London, E1 2AT, UK
- LonDownS Consortium, London, UK
| | - Xiaowei Shao
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
| | - Steven Havlicek
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, 138672, Singapore
| | - N Ray Dunn
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Singapore, 138648, Singapore
| | - Hlin Kvartsberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, S-405 30, Sweden
| | - Gunnar Brinkmalm
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, S-405 30, Sweden
| | - Rosalyn Hithersay
- LonDownS Consortium, London, UK
- Division of Psychiatry, University College London, London, WC1E 6BT, UK
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE5 8AF, UK
| | - Carla Startin
- LonDownS Consortium, London, UK
- Division of Psychiatry, University College London, London, WC1E 6BT, UK
| | - Sarah Hamburg
- LonDownS Consortium, London, UK
- Division of Psychiatry, University College London, London, WC1E 6BT, UK
| | - Margaret Phillips
- School of Biological Sciences, Nanyang Technological University, Singapore, 639798, Singapore
| | - Konstantin Pervushin
- School of Biological Sciences, Nanyang Technological University, Singapore, 639798, Singapore
| | - Mark Turmaine
- Division of Biosciences, University College London, Gower Street, London, WC1E 6BT, UK
| | - David Wallon
- Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, Department of Neurology and CNR-MAJ, F 76000, Normandy Center for Genomic and Personalized Medicine, Rouen, France
| | - Anne Rovelet-Lecrux
- Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, Department of Neurology and CNR-MAJ, F 76000, Normandy Center for Genomic and Personalized Medicine, Rouen, France
| | - Hilkka Soininen
- University of Eastern Finland, Institute of Clinical Medicine/Neurology, Kuopio, FI-70211, Finland
| | - Emanuela Volpi
- School of Life Sciences, University of Westminster, London, W1W 6UW, UK
| | - Joanne E Martin
- The Blizard Institute, Barts & The London School of Medicine, Queen Mary University of London, London, E1 2AT, UK
| | - Jia Nee Foo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, 138672, Singapore
| | - David L Becker
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
| | - Agueda Rostagno
- Department of Pathology & Department of Psychiatry, New York University School of Medicine, New York, NY, 10016, USA
| | - Jorge Ghiso
- Department of Pathology & Department of Psychiatry, New York University School of Medicine, New York, NY, 10016, USA
| | - Željka Krsnik
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, 10000, Zagreb, Croatia
| | - Goran Šimić
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, 10000, Zagreb, Croatia
| | - Ivica Kostović
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, 10000, Zagreb, Croatia
| | - Dinko Mitrečić
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, 10000, Zagreb, Croatia
| | - Paul T Francis
- Wolfson Centre for Age-Related Diseases, King's College London, London, SE1 1UL, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, S-405 30, Sweden
| | - Andre Strydom
- LonDownS Consortium, London, UK
- Division of Psychiatry, University College London, London, WC1E 6BT, UK
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE5 8AF, UK
| | - John Hardy
- LonDownS Consortium, London, UK
- Dementia Research Institute & Reta Lila Weston Institute, Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, S-405 30, Sweden
- Dementia Research Institute & Reta Lila Weston Institute, Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Dean Nižetić
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore.
- The Blizard Institute, Barts & The London School of Medicine, Queen Mary University of London, London, E1 2AT, UK.
- LonDownS Consortium, London, UK.
| |
Collapse
|
134
|
Torrens-Mas M, Perelló-Reus C, Navas-Enamorado C, Ibargüen-González L, Sanchez-Polo A, Segura-Sampedro JJ, Masmiquel L, Barcelo C, Gonzalez-Freire M. Organoids: An Emerging Tool to Study Aging Signature across Human Tissues. Modeling Aging with Patient-Derived Organoids. Int J Mol Sci 2021; 22:10547. [PMID: 34638891 PMCID: PMC8508868 DOI: 10.3390/ijms221910547] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 09/24/2021] [Accepted: 09/25/2021] [Indexed: 12/12/2022] Open
Abstract
The biology of aging is focused on the identification of novel pathways that regulate the underlying processes of aging to develop interventions aimed at delaying the onset and progression of chronic diseases to extend lifespan. However, the research on the aging field has been conducted mainly in animal models, yeast, Caenorhabditis elegans, and cell cultures. Thus, it is unclear to what extent this knowledge is transferable to humans since they might not reflect the complexity of aging in people. An organoid culture is an in vitro 3D cell-culture technology that reproduces the physiological and cellular composition of the tissues and/or organs. This technology is being used in the cancer field to predict the response of a patient-derived tumor to a certain drug or treatment serving as patient stratification and drug-guidance approaches. Modeling aging with patient-derived organoids has a tremendous potential as a preclinical model tool to discover new biomarkers of aging, to predict adverse outcomes during aging, and to design personalized approaches for the prevention and treatment of aging-related diseases and geriatric syndromes. This could represent a novel approach to study chronological and/or biological aging, paving the way to personalized interventions targeting the biology of aging.
Collapse
Affiliation(s)
- Margalida Torrens-Mas
- Vascular and Metabolic Pathologies Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain; (M.T.-M.); (C.N.-E.); (A.S.-P.); (L.M.)
- Translational Research in Aging and Longevity (TRIAL) Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain
| | - Catalina Perelló-Reus
- Translational Pancreatic Cancer Oncogenesis Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain; (C.P.-R.); (L.I.-G.)
| | - Cayetano Navas-Enamorado
- Vascular and Metabolic Pathologies Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain; (M.T.-M.); (C.N.-E.); (A.S.-P.); (L.M.)
- Translational Research in Aging and Longevity (TRIAL) Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain
| | - Lesly Ibargüen-González
- Translational Pancreatic Cancer Oncogenesis Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain; (C.P.-R.); (L.I.-G.)
| | - Andres Sanchez-Polo
- Vascular and Metabolic Pathologies Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain; (M.T.-M.); (C.N.-E.); (A.S.-P.); (L.M.)
- Translational Research in Aging and Longevity (TRIAL) Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain
| | - Juan Jose Segura-Sampedro
- Translational Research in Aging and Longevity (TRIAL) Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain
- General & Digestive Surgery Department, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain;
- School of Medicine, University of the Balearic Islands, 07120 Palma de Mallorca, Spain
| | - Luis Masmiquel
- Vascular and Metabolic Pathologies Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain; (M.T.-M.); (C.N.-E.); (A.S.-P.); (L.M.)
| | - Carles Barcelo
- Translational Pancreatic Cancer Oncogenesis Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain; (C.P.-R.); (L.I.-G.)
| | - Marta Gonzalez-Freire
- Vascular and Metabolic Pathologies Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain; (M.T.-M.); (C.N.-E.); (A.S.-P.); (L.M.)
- Translational Research in Aging and Longevity (TRIAL) Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain
| |
Collapse
|
135
|
Xiong Y, Lim CS. Understanding the Modulatory Effects of Cannabidiol on Alzheimer's Disease. Brain Sci 2021; 11:brainsci11091211. [PMID: 34573232 PMCID: PMC8472755 DOI: 10.3390/brainsci11091211] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/03/2021] [Accepted: 09/04/2021] [Indexed: 11/23/2022] Open
Abstract
Alzheimer’s disease (AD), the most common neurodegenerative disease, is characterized by progressive cognitive impairment. The deposition of amyloid beta (Aβ) and hyperphosphorylated tau is considered the hallmark of AD pathology. Many therapeutic approaches such as Food and Drug Administration-approved cholinesterase inhibitors and N–methyl–D–aspartate receptor antagonists have been used to intervene in AD pathology. However, current therapies only provide limited symptomatic relief and are ineffective in preventing AD progression. Cannabidiol (CBD), a phytocannabinoid devoid of psychoactive responses, provides neuroprotective effects through both cannabinoid and noncannabinoid receptors. Recent studies using an AD mouse model have suggested that CBD can reverse cognitive deficits along with Aβ-induced neuroinflammatory, oxidative responses, and neuronal death. Furthermore, CBD can reduce the accumulation of Aβ and hyperphosphorylation of tau, suggesting the possibility of delaying AD progression. Particularly, the noncannabinoid receptor, peroxisome proliferator-activated receptor gamma, has been suggested to be involved in multiple functions of CBD. Therefore, understanding the underlying mechanisms of CBD is necessary for intervening in AD pathology in depth and for the translation of preclinical studies into clinical settings. In this review, we summarize recent studies on the effect of CBD in AD and suggest problems to be overcome for the therapeutic use of CBD.
Collapse
Affiliation(s)
- Yinyi Xiong
- Department of Pharmacology, Wonkwang University School of Medicine, Iksan 54538, Korea;
- Department of Rehabilitation, Affiliated Hospital of Jiujiang University, Jiujiang 332000, China
| | - Chae-Seok Lim
- Department of Pharmacology, Wonkwang University School of Medicine, Iksan 54538, Korea;
- Correspondence: ; Tel.: +82-63-850-6765; Fax: +82-63-850-7262
| |
Collapse
|
136
|
Xu J, Wen Z. Brain Organoids: Studying Human Brain Development and Diseases in a Dish. Stem Cells Int 2021; 2021:5902824. [PMID: 34539790 PMCID: PMC8448601 DOI: 10.1155/2021/5902824] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 08/19/2021] [Indexed: 12/31/2022] Open
Abstract
With the rapid development of stem cell technology, the advent of three-dimensional (3D) cultured brain organoids has opened a new avenue for studying human neurodevelopment and neurological disorders. Brain organoids are stem-cell-derived 3D suspension cultures that self-assemble into an organized structure with cell types and cytoarchitectures recapitulating the developing brain. In recent years, brain organoids have been utilized in various aspects, ranging from basic biology studies, to disease modeling, and high-throughput screening of pharmaceutical compounds. In this review, we overview the establishment and development of brain organoid technology, its recent progress, and translational applications, as well as existing limitations and future directions.
Collapse
Affiliation(s)
- Jie Xu
- The Graduate Program in Genetics and Molecular Biology, Laney Graduate School, Emory University, GA 30322, USA
| | - Zhexing Wen
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
137
|
Chen X, Sun G, Tian E, Zhang M, Davtyan H, Beach TG, Reiman EM, Blurton‐Jones M, Holtzman DM, Shi Y. Modeling Sporadic Alzheimer's Disease in Human Brain Organoids under Serum Exposure. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2101462. [PMID: 34337898 PMCID: PMC8456220 DOI: 10.1002/advs.202101462] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/29/2021] [Indexed: 05/30/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease with no cure. Huge efforts have been made to develop anti-AD drugs in the past decades. However, all drug development programs for disease-modifying therapies have failed. Possible reasons for the high failure rate include incomplete understanding of complex pathophysiology of AD, especially sporadic AD (sAD), and species difference between humans and animal models used in preclinical studies. In this study, sAD is modeled using human induced pluripotent stem cell (hiPSC)-derived 3D brain organoids. Because the blood-brain barrier (BBB) leakage is a well-known risk factor for AD, brain organoids are exposed to human serum to mimic the serum exposure consequence of BBB breakdown in AD patient brains. The serum-exposed brain organoids are able to recapitulate AD-like pathologies, including increased amyloid beta (Aβ) aggregates and phosphorylated microtubule-associated tau protein (p-Tau) level, synaptic loss, and impaired neural network. Serum exposure increases Aβ and p-Tau levels through inducing beta-secretase 1 (BACE) and glycogen synthase kinase-3 alpha / beta (GSK3α/β) levels, respectively. In addition, single-cell transcriptomic analysis of brain organoids reveals that serum exposure reduced synaptic function in both neurons and astrocytes and induced immune response in astrocytes. The human brain organoid-based sAD model established in this study can provide a powerful platform for both mechanistic study and therapeutic development in the future.
Collapse
Affiliation(s)
- Xianwei Chen
- Division of Stem Cell Biology ResearchDepartment of Developmental and Stem Cell BiologyBeckman Research Institute of City of Hope1500 E. Duarte RdDuarteCA91010USA
| | - Guoqiang Sun
- Division of Stem Cell Biology ResearchDepartment of Developmental and Stem Cell BiologyBeckman Research Institute of City of Hope1500 E. Duarte RdDuarteCA91010USA
| | - E Tian
- Division of Stem Cell Biology ResearchDepartment of Developmental and Stem Cell BiologyBeckman Research Institute of City of Hope1500 E. Duarte RdDuarteCA91010USA
| | - Mingzi Zhang
- Division of Stem Cell Biology ResearchDepartment of Developmental and Stem Cell BiologyBeckman Research Institute of City of Hope1500 E. Duarte RdDuarteCA91010USA
| | - Hayk Davtyan
- Institute for Memory Impairments & Neurological Disorders and Sue & Bill Gross Stem Cell Research CenterUniversity of California IrvineIrvineCA92697USA
| | - Thomas G. Beach
- Banner Sun Health Research Institute105015 West Santa Fe DriveSun CityAZ85351USA
| | - Eric M. Reiman
- Banner Alzheimer Institute901 East Willetta StreetPhoenixAZ95006USA
| | - Mathew Blurton‐Jones
- Department of Neurobiology & BehaviorInstitute for Memory Impairments & Neurological Disorders and Sue & Bill Gross Stem Cell Research CenterUniversity of California IrvineIrvineCA92697USA
| | - David M. Holtzman
- Department of NeurologyHope Center for Neurological DisordersKnight Alzheimer's Disease Research CenterWashington University in St. LouisSt. LouisMO63110USA
| | - Yanhong Shi
- Division of Stem Cell Biology ResearchDepartment of Developmental and Stem Cell BiologyBeckman Research Institute of City of Hope1500 E. Duarte RdDuarteCA91010USA
| |
Collapse
|
138
|
Toshikawa H, Ikenaka A, Li L, Nishinaka-Arai Y, Niwa A, Ashida A, Kazuki Y, Nakahata T, Tamai H, Russell DW, Saito MK. N-Acetylcysteine prevents amyloid-β secretion in neurons derived from human pluripotent stem cells with trisomy 21. Sci Rep 2021; 11:17377. [PMID: 34462463 PMCID: PMC8405674 DOI: 10.1038/s41598-021-96697-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 08/10/2021] [Indexed: 11/09/2022] Open
Abstract
Down syndrome (DS) is caused by the trisomy of chromosome 21. Among the many disabilities found in individuals with DS is an increased risk of early-onset Alzheimer's disease (AD). Although higher oxidative stress and an upregulation of amyloid β (Aβ) peptides from an extra copy of the APP gene are attributed to the AD susceptibility, the relationship between the two factors is unclear. To address this issue, we established an in vitro cellular model using neurons differentiated from DS patient-derived induced pluripotent stem cells (iPSCs) and isogenic euploid iPSCs. Neurons differentiated from DS patient-derived iPSCs secreted more Aβ compared to those differentiated from the euploid iPSCs. Treatment of the neurons with an antioxidant, N-acetylcysteine, significantly suppressed the Aβ secretion. These findings suggest that oxidative stress has an important role in controlling the Aβ level in neurons differentiated from DS patient-derived iPSCs and that N-acetylcysteine can be a potential therapeutic option to ameliorate the Aβ secretion.
Collapse
Affiliation(s)
- Hiromitsu Toshikawa
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan.,Osaka Medical and Pharmaceutical University, Takatsuki, 5690801, Japan.,Social Welfare Organization "SAISEIKAI" Imperial Gift Foundation Inc., Saiseikai Suita Hospital, Suita, 5640013, Japan
| | - Akihiro Ikenaka
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Li Li
- Division of Hematology, School of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Yoko Nishinaka-Arai
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan.,Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, 6068507, Japan
| | - Akira Niwa
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Akira Ashida
- Osaka Medical and Pharmaceutical University, Takatsuki, 5690801, Japan
| | - Yasuhiro Kazuki
- Chromosome Engineering Research Center, Tottori University, Tottori, Japan.,Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine, Tottori University, Tottori, Japan
| | - Tatsutoshi Nakahata
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hiroshi Tamai
- Osaka Medical and Pharmaceutical University, Takatsuki, 5690801, Japan.,Institute for Developmental Brain Research, Osaka Medical and Pharmaceutical University, Takatsuki, 5690801, Japan
| | - David W Russell
- Division of Hematology, School of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Megumu K Saito
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan.
| |
Collapse
|
139
|
Bowles KR, Silva MC, Whitney K, Bertucci T, Berlind JE, Lai JD, Garza JC, Boles NC, Mahali S, Strang KH, Marsh JA, Chen C, Pugh DA, Liu Y, Gordon RE, Goderie SK, Chowdhury R, Lotz S, Lane K, Crary JF, Haggarty SJ, Karch CM, Ichida JK, Goate AM, Temple S. ELAVL4, splicing, and glutamatergic dysfunction precede neuron loss in MAPT mutation cerebral organoids. Cell 2021; 184:4547-4563.e17. [PMID: 34314701 PMCID: PMC8635409 DOI: 10.1016/j.cell.2021.07.003] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 05/06/2021] [Accepted: 06/30/2021] [Indexed: 12/21/2022]
Abstract
Frontotemporal dementia (FTD) because of MAPT mutation causes pathological accumulation of tau and glutamatergic cortical neuronal death by unknown mechanisms. We used human induced pluripotent stem cell (iPSC)-derived cerebral organoids expressing tau-V337M and isogenic corrected controls to discover early alterations because of the mutation that precede neurodegeneration. At 2 months, mutant organoids show upregulated expression of MAPT, glutamatergic signaling pathways, and regulators, including the RNA-binding protein ELAVL4, and increased stress granules. Over the following 4 months, mutant organoids accumulate splicing changes, disruption of autophagy function, and build-up of tau and P-tau-S396. By 6 months, tau-V337M organoids show specific loss of glutamatergic neurons as seen in individuals with FTD. Mutant neurons are susceptible to glutamate toxicity, which can be rescued pharmacologically by the PIKFYVE kinase inhibitor apilimod. Our results demonstrate a sequence of events that precede neurodegeneration, revealing molecular pathways associated with glutamate signaling as potential targets for therapeutic intervention in FTD.
Collapse
Affiliation(s)
- Kathryn R Bowles
- Ronald M. Loeb Center for Alzheimer's Disease, Friedman Brain Institute, Departments of Genetics and Genomic Sciences, Neuroscience, and Neurology, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY 10029, USA
| | - M Catarina Silva
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Departments of Neurology and Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Kristen Whitney
- Ronald M. Loeb Center for Alzheimer's Disease, Friedman Brain Institute, Departments of Genetics and Genomic Sciences, Neuroscience, and Neurology, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY 10029, USA; Department of Pathology, Neuropathology Brain Bank and Research Core, ISMMS, New York, NY 10029, USA
| | | | - Joshua E Berlind
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Jesse D Lai
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Amgen Research, One Amgen Center Dr., Thousand Oaks, CA 91320, USA
| | - Jacob C Garza
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Departments of Neurology and Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | | | - Sidhartha Mahali
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Kevin H Strang
- Ronald M. Loeb Center for Alzheimer's Disease, Friedman Brain Institute, Departments of Genetics and Genomic Sciences, Neuroscience, and Neurology, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY 10029, USA; Department of Pathology, Neuropathology Brain Bank and Research Core, ISMMS, New York, NY 10029, USA
| | - Jacob A Marsh
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Cynthia Chen
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Derian A Pugh
- Ronald M. Loeb Center for Alzheimer's Disease, Friedman Brain Institute, Departments of Genetics and Genomic Sciences, Neuroscience, and Neurology, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY 10029, USA
| | - Yiyuan Liu
- Ronald M. Loeb Center for Alzheimer's Disease, Friedman Brain Institute, Departments of Genetics and Genomic Sciences, Neuroscience, and Neurology, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY 10029, USA
| | - Ronald E Gordon
- Department of Pathology, Neuropathology Brain Bank and Research Core, ISMMS, New York, NY 10029, USA
| | | | | | - Steven Lotz
- Neural Stem Cell Institute, Rensselaer, NY 12144, USA
| | - Keith Lane
- Neural Stem Cell Institute, Rensselaer, NY 12144, USA
| | - John F Crary
- Department of Pathology, Neuropathology Brain Bank and Research Core, ISMMS, New York, NY 10029, USA
| | - Stephen J Haggarty
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Departments of Neurology and Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Celeste M Karch
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Justin K Ichida
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Alison M Goate
- Ronald M. Loeb Center for Alzheimer's Disease, Friedman Brain Institute, Departments of Genetics and Genomic Sciences, Neuroscience, and Neurology, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY 10029, USA.
| | - Sally Temple
- Neural Stem Cell Institute, Rensselaer, NY 12144, USA.
| |
Collapse
|
140
|
Vignon A, Salvador-Prince L, Lehmann S, Perrier V, Torrent J. Deconstructing Alzheimer's Disease: How to Bridge the Gap between Experimental Models and the Human Pathology? Int J Mol Sci 2021; 22:8769. [PMID: 34445475 PMCID: PMC8395727 DOI: 10.3390/ijms22168769] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/05/2021] [Accepted: 08/06/2021] [Indexed: 02/07/2023] Open
Abstract
Discovered more than a century ago, Alzheimer's disease (AD) is not only still present in our societies but has also become the most common dementia, with 50 million people worldwide affected by the disease. This number is expected to double in the next generation, and no cure is currently available to slow down or stop the disease progression. Recently, some advances were made due to the approval of the aducanumab treatment by the American Food and Drug Administration. The etiology of this human-specific disease remains poorly understood, and the mechanisms of its development have not been completely clarified. Several hypotheses concerning the molecular mechanisms of AD have been proposed, but the existing studies focus primarily on the two main markers of the disease: the amyloid β peptides, whose aggregation in the brain generates amyloid plaques, and the abnormally phosphorylated tau proteins, which are responsible for neurofibrillary tangles. These protein aggregates induce neuroinflammation and neurodegeneration, which, in turn, lead to cognitive and behavioral deficits. The challenge is, therefore, to create models that best reproduce this pathology. This review aims at gathering the different existing AD models developed in vitro, in cellulo, and in vivo. Many models have already been set up, but it is necessary to identify the most relevant ones for our investigations. The purpose of the review is to help researchers to identify the most pertinent disease models, from the most often used to the most recently generated and from simple to complex, explaining their specificities and giving concrete examples.
Collapse
Affiliation(s)
- Anaïs Vignon
- INM, University of Montpellier, INSERM, 34095 Montpellier, France; (A.V.); (L.S.-P.)
| | - Lucie Salvador-Prince
- INM, University of Montpellier, INSERM, 34095 Montpellier, France; (A.V.); (L.S.-P.)
| | - Sylvain Lehmann
- INM, University of Montpellier, INSERM, CHU Montpellier, 34095 Montpellier, France;
| | - Véronique Perrier
- INM, University of Montpellier, INSERM, CNRS, 34095 Montpellier, France
| | - Joan Torrent
- INM, University of Montpellier, INSERM, 34095 Montpellier, France; (A.V.); (L.S.-P.)
| |
Collapse
|
141
|
Porciúncula LO, Goto-Silva L, Ledur PF, Rehen SK. The Age of Brain Organoids: Tailoring Cell Identity and Functionality for Normal Brain Development and Disease Modeling. Front Neurosci 2021. [DOI: 10.3389/fnins.2021.674563
expr 918028134 + 817050540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Over the past years, brain development has been investigated in rodent models, which were particularly relevant to establish the role of specific genes in this process. However, the cytoarchitectonic features, which determine neuronal network formation complexity, are unique to humans. This implies that the developmental program of the human brain and neurological disorders can only partly be reproduced in rodents. Advancement in the study of the human brain surged with cultures of human brain tissue in the lab, generated from induced pluripotent cells reprogrammed from human somatic tissue. These cultures, termed brain organoids, offer an invaluable model for the study of the human brain. Brain organoids reproduce the cytoarchitecture of the cortex and can develop multiple brain regions and cell types. Integration of functional activity of neural cells within brain organoids with genetic, cellular, and morphological data in a comprehensive model for human development and disease is key to advance in the field. Because the functional activity of neural cells within brain organoids relies on cell repertoire and time in culture, here, we review data supporting the gradual formation of complex neural networks in light of cell maturity within brain organoids. In this context, we discuss how the technology behind brain organoids brought advances in understanding neurodevelopmental, pathogen-induced, and neurodegenerative diseases.
Collapse
|
142
|
Porciúncula LO, Goto-Silva L, Ledur PF, Rehen SK. The Age of Brain Organoids: Tailoring Cell Identity and Functionality for Normal Brain Development and Disease Modeling. Front Neurosci 2021; 15:674563. [PMID: 34483818 PMCID: PMC8414411 DOI: 10.3389/fnins.2021.674563] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 07/19/2021] [Indexed: 12/15/2022] Open
Abstract
Over the past years, brain development has been investigated in rodent models, which were particularly relevant to establish the role of specific genes in this process. However, the cytoarchitectonic features, which determine neuronal network formation complexity, are unique to humans. This implies that the developmental program of the human brain and neurological disorders can only partly be reproduced in rodents. Advancement in the study of the human brain surged with cultures of human brain tissue in the lab, generated from induced pluripotent cells reprogrammed from human somatic tissue. These cultures, termed brain organoids, offer an invaluable model for the study of the human brain. Brain organoids reproduce the cytoarchitecture of the cortex and can develop multiple brain regions and cell types. Integration of functional activity of neural cells within brain organoids with genetic, cellular, and morphological data in a comprehensive model for human development and disease is key to advance in the field. Because the functional activity of neural cells within brain organoids relies on cell repertoire and time in culture, here, we review data supporting the gradual formation of complex neural networks in light of cell maturity within brain organoids. In this context, we discuss how the technology behind brain organoids brought advances in understanding neurodevelopmental, pathogen-induced, and neurodegenerative diseases.
Collapse
Affiliation(s)
- Lisiane O. Porciúncula
- Department of Biochemistry, Program of Biological Sciences - Biochemistry, Institute of Health and Basic Sciences, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Livia Goto-Silva
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | - Pitia F. Ledur
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | - Stevens K. Rehen
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
- Department of Genetics, Institute of Biology, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| |
Collapse
|
143
|
Barth M, Bacioglu M, Schwarz N, Novotny R, Brandes J, Welzer M, Mazzitelli S, Häsler LM, Schweighauser M, Wuttke TV, Kronenberg-Versteeg D, Fog K, Ambjørn M, Alik A, Melki R, Kahle PJ, Shimshek DR, Koch H, Jucker M, Tanriöver G. Microglial inclusions and neurofilament light chain release follow neuronal α-synuclein lesions in long-term brain slice cultures. Mol Neurodegener 2021; 16:54. [PMID: 34380535 PMCID: PMC8356412 DOI: 10.1186/s13024-021-00471-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 07/06/2021] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Proteopathic brain lesions are a hallmark of many age-related neurodegenerative diseases including synucleinopathies and develop at least a decade before the onset of clinical symptoms. Thus, understanding of the initiation and propagation of such lesions is key for developing therapeutics to delay or halt disease progression. METHODS Alpha-synuclein (αS) inclusions were induced in long-term murine and human slice cultures by seeded aggregation. An αS seed-recognizing human antibody was tested for blocking seeding and/or spreading of the αS lesions. Release of neurofilament light chain (NfL) into the culture medium was assessed. RESULTS To study initial stages of α-synucleinopathies, we induced αS inclusions in murine hippocampal slice cultures by seeded aggregation. Induction of αS inclusions in neurons was apparent as early as 1week post-seeding, followed by the occurrence of microglial inclusions in vicinity of the neuronal lesions at 2-3 weeks. The amount of αS inclusions was dependent on the type of αS seed and on the culture's genetic background (wildtype vs A53T-αS genotype). Formation of αS inclusions could be monitored by neurofilament light chain protein release into the culture medium, a fluid biomarker of neurodegeneration commonly used in clinical settings. Local microinjection of αS seeds resulted in spreading of αS inclusions to neuronally connected hippocampal subregions, and seeding and spreading could be inhibited by an αS seed-recognizing human antibody. We then applied parameters of the murine cultures to surgical resection-derived adult human long-term neocortical slice cultures from 22 to 61-year-old donors. Similarly, in these human slice cultures, proof-of-principle induction of αS lesions was achieved at 1week post-seeding in combination with viral A53T-αS expressions. CONCLUSION The successful translation of these brain cultures from mouse to human with the first reported induction of human αS lesions in a true adult human brain environment underlines the potential of this model to study proteopathic lesions in intact mouse and now even aged human brain environments.
Collapse
Affiliation(s)
- Melanie Barth
- DZNE, German Center for Neurodegenerative Diseases, 72076 Tuebingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
- Graduate Training Center of Neuroscience, University of Tuebingen, 72076 Tuebingen, Germany
| | - Mehtap Bacioglu
- DZNE, German Center for Neurodegenerative Diseases, 72076 Tuebingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
- Graduate Training Center of Neuroscience, University of Tuebingen, 72076 Tuebingen, Germany
| | - Niklas Schwarz
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
| | - Renata Novotny
- DZNE, German Center for Neurodegenerative Diseases, 72076 Tuebingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
| | - Janine Brandes
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
| | - Marc Welzer
- DZNE, German Center for Neurodegenerative Diseases, 72076 Tuebingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
- Graduate Training Center of Neuroscience, University of Tuebingen, 72076 Tuebingen, Germany
| | - Sonia Mazzitelli
- DZNE, German Center for Neurodegenerative Diseases, 72076 Tuebingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
| | - Lisa M. Häsler
- DZNE, German Center for Neurodegenerative Diseases, 72076 Tuebingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
| | - Manuel Schweighauser
- DZNE, German Center for Neurodegenerative Diseases, 72076 Tuebingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
| | - Thomas V. Wuttke
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
- Department of Neurosurgery, University of Tuebingen, 72076 Tuebingen, Germany
| | - Deborah Kronenberg-Versteeg
- DZNE, German Center for Neurodegenerative Diseases, 72076 Tuebingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
| | - Karina Fog
- Division of Neuroscience, H. Lundbeck A/S, 2500 Valby, Denmark
| | - Malene Ambjørn
- Division of Neuroscience, H. Lundbeck A/S, 2500 Valby, Denmark
| | - Ania Alik
- MIRCen, CEA and Laboratory of Neurodegenerative Diseases, CNRS, Institut François Jacob, 92265 Fontenay-aux-Roses, France
| | - Ronald Melki
- MIRCen, CEA and Laboratory of Neurodegenerative Diseases, CNRS, Institut François Jacob, 92265 Fontenay-aux-Roses, France
| | - Philipp J. Kahle
- DZNE, German Center for Neurodegenerative Diseases, 72076 Tuebingen, Germany
- Laboratory of Functional Neurogenetics, Department of Neurodegeneration, Hertie-Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
| | - Derya R. Shimshek
- Neuroscience Research, Novartis Institutes for BioMedical Research, CH-4056 Basel, Switzerland
| | - Henner Koch
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
- Department of Epileptology, Neurology, RWTH Aachen University, Aachen, Germany
| | - Mathias Jucker
- DZNE, German Center for Neurodegenerative Diseases, 72076 Tuebingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
| | - Gaye Tanriöver
- DZNE, German Center for Neurodegenerative Diseases, 72076 Tuebingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
| |
Collapse
|
144
|
Bi FC, Yang XH, Cheng XY, Deng WB, Guo XL, Yang H, Wang Y, Li J, Yao Y. Optimization of cerebral organoids: a more qualified model for Alzheimer's disease research. Transl Neurodegener 2021; 10:27. [PMID: 34372927 PMCID: PMC8349709 DOI: 10.1186/s40035-021-00252-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 07/17/2021] [Indexed: 12/18/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that currently cannot be cured by any drug or intervention, due to its complicated pathogenesis. Current animal and cellular models of AD are unable to meet research needs for AD. However, recent three-dimensional (3D) cerebral organoid models derived from human stem cells have provided a new tool to study molecular mechanisms and pharmaceutical developments of AD. In this review, we discuss the advantages and key limitations of the AD cerebral organoid system in comparison to the commonly used AD models, and propose possible solutions, in order to improve their application in AD research. Ethical concerns associated with human cerebral organoids are also discussed. We also summarize future directions of studies that will improve the cerebral organoid system to better model the pathological events observed in AD brains.
Collapse
Affiliation(s)
- Feng-Chen Bi
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
- Key Laboratory of Traditional Chinese Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, 750004, China
| | - Xin-He Yang
- School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
| | - Xiao-Yu Cheng
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital, Soochow University, Suzhou, 215004, China
| | - Wen-Bin Deng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510275, China
| | - Xiao-Li Guo
- School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
| | - Hui Yang
- Research Center of Medical Science and Technology, Ningxia Medical University, Yinchuan, 750004, China
| | - Yin Wang
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China.
| | - Juan Li
- Key Laboratory of Traditional Chinese Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, 750004, China.
- School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China.
| | - Yao Yao
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China.
| |
Collapse
|
145
|
Heydari Z, Moeinvaziri F, Agarwal T, Pooyan P, Shpichka A, Maiti TK, Timashev P, Baharvand H, Vosough M. Organoids: a novel modality in disease modeling. Biodes Manuf 2021; 4:689-716. [PMID: 34395032 PMCID: PMC8349706 DOI: 10.1007/s42242-021-00150-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 06/12/2021] [Indexed: 12/17/2022]
Abstract
Limitations of monolayer culture conditions have motivated scientists to explore new models that can recapitulate the architecture and function of human organs more accurately. Recent advances in the improvement of protocols have resulted in establishing three-dimensional (3D) organ-like architectures called ‘organoids’ that can display the characteristics of their corresponding real organs, including morphological features, functional activities, and personalized responses to specific pathogens. We discuss different organoid-based 3D models herein, which are classified based on their original germinal layer. Studies of organoids simulating the complexity of real tissues could provide novel platforms and opportunities for generating practical knowledge along with preclinical studies, including drug screening, toxicology, and molecular pathophysiology of diseases. This paper also outlines the key challenges, advantages, and prospects of current organoid systems.
Collapse
Affiliation(s)
- Zahra Heydari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 14155-4364 Iran
- Department of Developmental Biology, University of Science and Culture, Tehran, 14155-4364 Iran
| | - Farideh Moeinvaziri
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 14155-4364 Iran
- Department of Developmental Biology, University of Science and Culture, Tehran, 14155-4364 Iran
| | - Tarun Agarwal
- Department of Biotechnology, Indian Institute of Technology, Kharagpur, West Bengal 721302 India
| | - Paria Pooyan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 14155-4364 Iran
| | - Anastasia Shpichka
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, 19991 Moscow, Russia
- Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
- Chemistry Department, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Tapas K. Maiti
- Department of Biotechnology, Indian Institute of Technology, Kharagpur, West Bengal 721302 India
| | - Peter Timashev
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, 19991 Moscow, Russia
- Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
- Chemistry Department, Lomonosov Moscow State University, 119991 Moscow, Russia
- Department of Polymers and Composites, N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 14155-4364 Iran
- Department of Developmental Biology, University of Science and Culture, Tehran, 14155-4364 Iran
| | - Massoud Vosough
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 14155-4364 Iran
- Department of Regenerative Medicine, Cell Science Research Centre, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 14155-4364 Iran
| |
Collapse
|
146
|
Jalink P, Caiazzo M. Brain Organoids: Filling the Need for a Human Model of Neurological Disorder. BIOLOGY 2021; 10:740. [PMID: 34439972 PMCID: PMC8389592 DOI: 10.3390/biology10080740] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 07/25/2021] [Accepted: 07/26/2021] [Indexed: 02/06/2023]
Abstract
Neurological disorders are among the leading causes of death worldwide, accounting for almost all onsets of dementia in the elderly, and are known to negatively affect motor ability, mental and cognitive performance, as well as overall wellbeing and happiness. Currently, most neurological disorders go untreated due to a lack of viable treatment options. The reason for this lack of options is s poor understanding of the disorders, primarily due to research models that do not translate well into the human in vivo system. Current models for researching neurological disorders, neurodevelopment, and drug interactions in the central nervous system include in vitro monolayer cell cultures, and in vivo animal models. These models have shortcomings when it comes to translating research about disorder pathology, development, and treatment to humans. Brain organoids are three-dimensional (3D) cultures of stem cell-derived neural cells that mimic the development of the in vivo human brain with high degrees of accuracy. Researchers have started developing these miniature brains to model neurodevelopment, and neuropathology. Brain organoids have been used to model a wide range of neurological disorders, including the complex and poorly understood neurodevelopmental and neurodegenerative disorders. In this review, we discuss the brain organoid technology, placing special focus on the different brain organoid models that have been developed, discussing their strengths, weaknesses, and uses in neurological disease modeling.
Collapse
Affiliation(s)
- Philip Jalink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Universiteitsweg 99, CG 3584 Utrecht, The Netherlands;
| | - Massimiliano Caiazzo
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Universiteitsweg 99, CG 3584 Utrecht, The Netherlands;
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy
| |
Collapse
|
147
|
Agboola OS, Hu X, Shan Z, Wu Y, Lei L. Brain organoid: a 3D technology for investigating cellular composition and interactions in human neurological development and disease models in vitro. Stem Cell Res Ther 2021; 12:430. [PMID: 34332630 PMCID: PMC8325286 DOI: 10.1186/s13287-021-02369-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 05/03/2021] [Indexed: 01/01/2023] Open
Abstract
Abstract The study of human brain physiology, including cellular interactions in normal and disease conditions, has been a challenge due to its complexity and unavailability. Induced pluripotent stem cell (iPSC) study is indispensable in the study of the pathophysiology of neurological disorders. Nevertheless, monolayer systems lack the cytoarchitecture necessary for cellular interactions and neurological disease modeling. Brain organoids generated from human pluripotent stem cells supply an ideal environment to model both cellular interactions and pathophysiology of the human brain. This review article discusses the composition and interactions among neural lineage and non-central nervous system cell types in brain organoids, current studies, and future perspectives in brain organoid research. Ultimately, the promise of brain organoids is to unveil previously inaccessible features of neurobiology that emerge from complex cellular interactions and to improve our mechanistic understanding of neural development and diseases. Graphical abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02369-8.
Collapse
Affiliation(s)
- Oluwafemi Solomon Agboola
- Department of Histology and Embryology, Basic Medical Science College, Harbin Medical University, 194 Xuefu Rd, Nangang District, Heilongjiang Province, Harbin, 150081, People's Republic of China
| | - Xinglin Hu
- Department of Histology and Embryology, Basic Medical Science College, Harbin Medical University, 194 Xuefu Rd, Nangang District, Heilongjiang Province, Harbin, 150081, People's Republic of China
| | - Zhiyan Shan
- Department of Histology and Embryology, Basic Medical Science College, Harbin Medical University, 194 Xuefu Rd, Nangang District, Heilongjiang Province, Harbin, 150081, People's Republic of China
| | - Yanshuang Wu
- Department of Histology and Embryology, Basic Medical Science College, Harbin Medical University, 194 Xuefu Rd, Nangang District, Heilongjiang Province, Harbin, 150081, People's Republic of China.
| | - Lei Lei
- Department of Histology and Embryology, Basic Medical Science College, Harbin Medical University, 194 Xuefu Rd, Nangang District, Heilongjiang Province, Harbin, 150081, People's Republic of China. .,Key Laboratory of Preservative of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China.
| |
Collapse
|
148
|
Trudler D, Ghatak S, Lipton SA. Emerging hiPSC Models for Drug Discovery in Neurodegenerative Diseases. Int J Mol Sci 2021; 22:8196. [PMID: 34360966 PMCID: PMC8347370 DOI: 10.3390/ijms22158196] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 12/12/2022] Open
Abstract
Neurodegenerative diseases affect millions of people worldwide and are characterized by the chronic and progressive deterioration of neural function. Neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD), represent a huge social and economic burden due to increasing prevalence in our aging society, severity of symptoms, and lack of effective disease-modifying therapies. This lack of effective treatments is partly due to a lack of reliable models. Modeling neurodegenerative diseases is difficult because of poor access to human samples (restricted in general to postmortem tissue) and limited knowledge of disease mechanisms in a human context. Animal models play an instrumental role in understanding these diseases but fail to comprehensively represent the full extent of disease due to critical differences between humans and other mammals. The advent of human-induced pluripotent stem cell (hiPSC) technology presents an advantageous system that complements animal models of neurodegenerative diseases. Coupled with advances in gene-editing technologies, hiPSC-derived neural cells from patients and healthy donors now allow disease modeling using human samples that can be used for drug discovery.
Collapse
Affiliation(s)
- Dorit Trudler
- Neurodegeneration New Medicines Center and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; (D.T.); (S.G.)
| | - Swagata Ghatak
- Neurodegeneration New Medicines Center and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; (D.T.); (S.G.)
| | - Stuart A. Lipton
- Neurodegeneration New Medicines Center and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; (D.T.); (S.G.)
- Department of Neurosciences, University of California San Diego School of Medicine, La Jolla, CA 92093, USA
| |
Collapse
|
149
|
Groveman BR, Smith A, Williams K, Haigh CL. Cerebral organoids as a new model for prion disease. PLoS Pathog 2021; 17:e1009747. [PMID: 34288977 PMCID: PMC8294539 DOI: 10.1371/journal.ppat.1009747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Bradley R. Groveman
- Prion Cell Biology Unit, Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Anna Smith
- Prion Cell Biology Unit, Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Katie Williams
- Prion Cell Biology Unit, Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Cathryn L. Haigh
- Prion Cell Biology Unit, Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
- * E-mail:
| |
Collapse
|
150
|
Abstract
Induced pluripotent stem cell (iPSC) technology holds promise for modeling neurodegenerative diseases. Traditional approaches for disease modeling using animal and cellular models require knowledge of disease mutations. However, many patients with neurodegenerative diseases do not have a known genetic cause. iPSCs offer a way to generate patient-specific models and study pathways of dysfunction in an in vitro setting in order to understand the causes and subtypes of neurodegeneration. Furthermore, iPSC-based models can be used to search for candidate therapeutics using high-throughput screening. Here we review how iPSC-based models are currently being used to further our understanding of neurodegenerative diseases, as well as discuss their challenges and future directions.
Collapse
Affiliation(s)
- Jonathan Li
- Computational and Systems Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA;
| | - Ernest Fraenkel
- Computational and Systems Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA; .,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| |
Collapse
|