401
|
Hopkins HK, Traverse EM, Barr KL. Methodologies for Generating Brain Organoids to Model Viral Pathogenesis in the CNS. Pathogens 2021; 10:1510. [PMID: 34832665 PMCID: PMC8625030 DOI: 10.3390/pathogens10111510] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 12/22/2022] Open
Abstract
(1) Background: The human brain is of interest in viral research because it is often the target of viruses. Neurological infections can result in consequences in the CNS, which can result in death or lifelong sequelae. Organoids modeling the CNS are notable because they are derived from stem cells that differentiate into specific brain cells such as neural progenitors, neurons, astrocytes, and glial cells. Numerous protocols have been developed for the generation of CNS organoids, and our goal was to describe the various CNS organoid models available for viral pathogenesis research to serve as a guide to determine which protocol might be appropriate based on research goal, timeframe, and budget. (2) Methods: Articles for this review were found in Pubmed, Scopus and EMBASE. The search terms used were "brain + organoid" and "CNS + organoid" (3) Results: There are two main methods for organoid generation, and the length of time for organoid generation varied from 28 days to over 2 months. The costs for generating a population of organoids ranged from USD 1000 to 5000. (4) Conclusions: There are numerous methods for generating organoids representing multiple regions of the brain, with several types of modifications for fine-tuning the model to a researcher's specifications. Organoid models of the CNS can serve as a platform for characterization and mechanistic studies that can reduce or eliminate the use of animals, especially for viruses that only cause disease in the human CNS.
Collapse
Affiliation(s)
| | | | - Kelli L. Barr
- Center for Global Health and Infectious Disease Research, University of South Florida, Tampa, FL 33612, USA; (H.K.H.); (E.M.T.)
| |
Collapse
|
402
|
Matsui T, Shinozawa T. Human Organoids for Predictive Toxicology Research and Drug Development. Front Genet 2021; 12:767621. [PMID: 34790228 PMCID: PMC8591288 DOI: 10.3389/fgene.2021.767621] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/11/2021] [Indexed: 12/11/2022] Open
Abstract
Organoids are three-dimensional structures fabricated in vitro from pluripotent stem cells or adult tissue stem cells via a process of self-organization that results in the formation of organ-specific cell types. Human organoids are expected to mimic complex microenvironments and many of the in vivo physiological functions of relevant tissues, thus filling the translational gap between animals and humans and increasing our understanding of the mechanisms underlying disease and developmental processes. In the last decade, organoid research has attracted increasing attention in areas such as disease modeling, drug development, regenerative medicine, toxicology research, and personalized medicine. In particular, in the field of toxicology, where there are various traditional models, human organoids are expected to blaze a new path in future research by overcoming the current limitations, such as those related to differences in drug responses among species. Here, we discuss the potential usefulness, limitations, and future prospects of human liver, heart, kidney, gut, and brain organoids from the viewpoints of predictive toxicology research and drug development, providing cutting edge information on their fabrication methods and functional characteristics.
Collapse
Affiliation(s)
- Toshikatsu Matsui
- Drug Safety Research and Evaluation, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Tadahiro Shinozawa
- Drug Safety Research and Evaluation, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| |
Collapse
|
403
|
Kelley KW, Pașca SP. Human brain organogenesis: Toward a cellular understanding of development and disease. Cell 2021; 185:42-61. [PMID: 34774127 DOI: 10.1016/j.cell.2021.10.003] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/24/2021] [Accepted: 10/01/2021] [Indexed: 02/06/2023]
Abstract
The construction of the human nervous system is a distinctly complex although highly regulated process. Human tissue inaccessibility has impeded a molecular understanding of the developmental specializations from which our unique cognitive capacities arise. A confluence of recent technological advances in genomics and stem cell-based tissue modeling is laying the foundation for a new understanding of human neural development and dysfunction in neuropsychiatric disease. Here, we review recent progress on uncovering the cellular and molecular principles of human brain organogenesis in vivo as well as using organoids and assembloids in vitro to model features of human evolution and disease.
Collapse
Affiliation(s)
- Kevin W Kelley
- Department of Psychiatry and Behavioral Sciences, Stanford University, CA, USA; Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute, Stanford, CA, USA
| | - Sergiu P Pașca
- Department of Psychiatry and Behavioral Sciences, Stanford University, CA, USA; Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute, Stanford, CA, USA.
| |
Collapse
|
404
|
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: 0.8] [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
|
405
|
Blanchard JW, Victor MB, Tsai LH. Dissecting the complexities of Alzheimer disease with in vitro models of the human brain. Nat Rev Neurol 2021; 18:25-39. [PMID: 34750588 DOI: 10.1038/s41582-021-00578-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/05/2021] [Indexed: 12/12/2022]
Abstract
Alzheimer disease (AD) is the most prevalent type of dementia. It is marked by severe memory loss and cognitive decline, and currently has limited effective treatment options. Although individuals with AD have common neuropathological hallmarks, emerging data suggest that the disease has a complex polygenic aetiology, and more than 25 genetic loci have been linked to an elevated risk of AD and dementia. Nevertheless, our ability to decipher the cellular and molecular mechanisms that underlie genetic susceptibility to AD, and its progression and severity, remains limited. Here, we discuss ongoing efforts to leverage genomic data from patients using cellular reprogramming technologies to recapitulate complex brain systems and build in vitro discovery platforms. Much attention has already been given to methodologies to derive major brain cell types from pluripotent stem cells. We therefore focus on technologies that combine multiple cell types to recreate anatomical and physiological properties of human brain tissue in vitro. We discuss the advances in the field for modelling four domains that have come into view as key contributors to the pathogenesis of AD: the blood-brain barrier, myelination, neuroinflammation and neuronal circuits. We also highlight opportunities for the field to further interrogate the complex genetic and environmental factors of AD using in vitro models.
Collapse
Affiliation(s)
- Joel W Blanchard
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Neuroscience, Black Family Stem Cell Institute, Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
| | - Matheus B Victor
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Li-Huei Tsai
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA. .,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA. .,Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
406
|
McTague A, Rossignoli G, Ferrini A, Barral S, Kurian MA. Genome Editing in iPSC-Based Neural Systems: From Disease Models to Future Therapeutic Strategies. Front Genome Ed 2021; 3:630600. [PMID: 34713254 PMCID: PMC8525405 DOI: 10.3389/fgeed.2021.630600] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 02/19/2021] [Indexed: 12/14/2022] Open
Abstract
Therapeutic advances for neurological disorders are challenging due to limited accessibility of the human central nervous system and incomplete understanding of disease mechanisms. Many neurological diseases lack precision treatments, leading to significant disease burden and poor outcome for affected patients. Induced pluripotent stem cell (iPSC) technology provides human neuronal cells that facilitate disease modeling and development of therapies. The use of genome editing, in particular CRISPR-Cas9 technology, has extended the potential of iPSCs, generating new models for a number of disorders, including Alzheimers and Parkinson Disease. Editing of iPSCs, in particular with CRISPR-Cas9, allows generation of isogenic pairs, which differ only in the disease-causing mutation and share the same genetic background, for assessment of phenotypic differences and downstream effects. Moreover, genome-wide CRISPR screens allow high-throughput interrogation for genetic modifiers in neuronal phenotypes, leading to discovery of novel pathways, and identification of new therapeutic targets. CRISPR-Cas9 has now evolved beyond altering gene expression. Indeed, fusion of a defective Cas9 (dCas9) nuclease with transcriptional repressors or activation domains allows down-regulation or activation of gene expression (CRISPR interference, CRISPRi; CRISPR activation, CRISPRa). These new tools will improve disease modeling and facilitate CRISPR and cell-based therapies, as seen for epilepsy and Duchenne muscular dystrophy. Genome engineering holds huge promise for the future understanding and treatment of neurological disorders, but there are numerous barriers to overcome. The synergy of iPSC-based model systems and gene editing will play a vital role in the route to precision medicine and the clinical translation of genome editing-based therapies.
Collapse
Affiliation(s)
- Amy McTague
- Developmental Neurosciences, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom.,Department of Neurology, Great Ormond Street Hospital, London, United Kingdom
| | - Giada Rossignoli
- Developmental Neurosciences, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Arianna Ferrini
- Developmental Neurosciences, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Serena Barral
- Developmental Neurosciences, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Manju A Kurian
- Developmental Neurosciences, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom.,Department of Neurology, Great Ormond Street Hospital, London, United Kingdom
| |
Collapse
|
407
|
Shin N, Kim Y, Ko J, Choi SW, Hyung S, Lee SE, Park S, Song J, Jeon NL, Kang KS. Vascularization of iNSC spheroid in a 3D spheroid-on-a-chip platform enhances neural maturation. Biotechnol Bioeng 2021; 119:566-574. [PMID: 34716703 PMCID: PMC9298365 DOI: 10.1002/bit.27978] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/26/2021] [Accepted: 10/26/2021] [Indexed: 11/12/2022]
Abstract
In vitro platforms for studying the human brain have been developed, and brain organoids derived from stem cells have been studied. However, current organoid models lack three-dimensional (3D) vascular networks, limiting organoid proliferation, differentiation, and apoptosis. In this study, we created a 3D model of vascularized spheroid cells using an injection-molded microfluidic chip. We cocultured spheroids derived from induced neural stem cells (iNSCs) with perfusable blood vessels. Gene expression analysis and immunostaining revealed that the vascular network greatly enhanced spheroid differentiation and reduced apoptosis. This platform can be used to further study the functional and structural interactions between blood vessels and neural spheroids, and ultimately to simulate brain development and disease.
Collapse
Affiliation(s)
- Nari Shin
- Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Youngtaek Kim
- Department of Mechanical Engineering, Seoul National University, Seoul, South Korea
| | - Jihoon Ko
- Department of Mechanical Engineering, Seoul National University, Seoul, South Korea
| | - Soon Won Choi
- Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Sujin Hyung
- Department of Mechanical Engineering, Seoul National University, Seoul, South Korea
| | - Seung-Eun Lee
- Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Seunghyuk Park
- Department of Mechanical Engineering, Seoul National University, Seoul, South Korea
| | - Jiyoung Song
- Department of Mechanical Engineering, Seoul National University, Seoul, South Korea
| | - Noo Li Jeon
- Department of Mechanical Engineering, Seoul National University, Seoul, South Korea.,Institute of Bioengineering, Seoul National University, Seoul, South Korea.,Institute of Advanced Machinery and Design, Seoul National University, Seoul, South Korea
| | - Kyung-Sun Kang
- Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| |
Collapse
|
408
|
Non-Animal Models in Experimental Subarachnoid Hemorrhage Research: Potentials and the Dilemma of the Translation from Bench to Bedside. Transl Stroke Res 2021; 13:218-221. [PMID: 34714498 PMCID: PMC8918456 DOI: 10.1007/s12975-021-00950-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 09/24/2021] [Accepted: 09/27/2021] [Indexed: 11/08/2022]
|
409
|
Lavazza A. 'Consciousnessoids': clues and insights from human cerebral organoids for the study of consciousness. Neurosci Conscious 2021; 7:niab029. [PMID: 34729213 PMCID: PMC8557395 DOI: 10.1093/nc/niab029] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 08/02/2021] [Accepted: 08/17/2021] [Indexed: 12/21/2022] Open
Abstract
Human cerebral organoids (HCOs) are an in vitro three-dimensional model of early neural development, aimed at modelling and understanding brain development and neurological disorders. In just a few years, there has been a rapid and considerable progress in the attempt to create a brain model capable of showcasing the structure and functions of the human brain. There are still strong limitations to address, including the absence of vascularization that makes it difficult to feed the central layers of organoids. Nevertheless, some important features of the nervous system have recently been observed: HCOs manifest electrical activity, are sensitive to light stimulation and are able to connect to a spinal cord by sending impulses that make a muscle contract. Recent data show that cortical organoid network development at 10 months resembles some preterm babies' electroencephalography (EEG) patterns. In the light of the fast pace of research in this field, one might consider the hypothesis that HCOs might become a living laboratory for studying the emergence of consciousness and investigating its mechanisms and neural correlates. HCOs could be also a benchmark for different neuroscientific theories of consciousness. In this paper, I propose some potential lines of research and offer some clues and insights so as to use HCOs in trying to unveil some puzzles concerning our conscious states. Finally, I consider some relevant ethical issues regarding this specific experimentation on HCOs and conclude that some of them could require strict regulation in this field.
Collapse
Affiliation(s)
- Andrea Lavazza
- Centro Universitario Internazionale, Via Garbasso, 42, Arezzo 52100, Italy
- University of Pavia, Department of Brain and Behavioural Sciences, Piazza Botta, 11, Pavia 27100, Italy
| |
Collapse
|
410
|
Singh K, Hotchkiss KM, Patel KK, Wilkinson DS, Mohan AA, Cook SL, Sampson JH. Enhancing T Cell Chemotaxis and Infiltration in Glioblastoma. Cancers (Basel) 2021; 13:5367. [PMID: 34771532 PMCID: PMC8582389 DOI: 10.3390/cancers13215367] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma is an immunologically 'cold' tumor, which are characterized by absent or minimal numbers of tumor-infiltrating lymphocytes (TILs). For those tumors that have been invaded by lymphocytes, they are profoundly exhausted and ineffective. While many immunotherapy approaches seek to reinvigorate immune cells at the tumor, this requires TILs to be present. Therefore, to unleash the full potential of immunotherapy in glioblastoma, the trafficking of lymphocytes to the tumor is highly desirable. However, the process of T cell recruitment into the central nervous system (CNS) is tightly regulated. Naïve T cells may undergo an initial licensing process to enter the migratory phenotype necessary to enter the CNS. T cells then must express appropriate integrins and selectin ligands to interact with transmembrane proteins at the blood-brain barrier (BBB). Finally, they must interact with antigen-presenting cells and undergo further licensing to enter the parenchyma. These T cells must then navigate the tumor microenvironment, which is rich in immunosuppressive factors. Altered tumoral metabolism also interferes with T cell motility. In this review, we will describe these processes and their mediators, along with potential therapeutic approaches to enhance trafficking. We also discuss safety considerations for such approaches as well as potential counteragents.
Collapse
Affiliation(s)
- Kirit Singh
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC 27710, USA; (K.M.H.); (K.K.P.); (D.S.W.); (A.A.M.); (S.L.C.)
| | | | | | | | | | | | - John H. Sampson
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC 27710, USA; (K.M.H.); (K.K.P.); (D.S.W.); (A.A.M.); (S.L.C.)
| |
Collapse
|
411
|
A Multi-Modal Toolkit for Studying Neutrophils in Cancer and Beyond. Cancers (Basel) 2021; 13:cancers13215331. [PMID: 34771495 PMCID: PMC8582456 DOI: 10.3390/cancers13215331] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/18/2021] [Accepted: 10/21/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Neutrophils are critical immune cells in host defense and maintenance of tissue homeostasis. Studying the complex and diverse functions of these innate immune cells requires a comprehensive toolkit of experimental techniques to elucidate the function and regulation of neutrophils in health and disease. In this review, we discuss key methodologies and their applications in neutrophil research, including in vivo imaging, ex vivo functional assays, and high dimensional single-cell technologies, and how they can be integrated into a multi-modal approach to study neutrophil function in cancer and other diseases. Abstract As key effector cells of the innate immune response, neutrophils are rapidly deployed to sites of inflammation where they deliver a payload of potent effector mechanisms that are essential for host defense against pathogens as well as tissue homeostasis. In addition, neutrophils are central contributors to the pathogenesis of a vast spectrum of inflammatory, degenerative, and neoplastic diseases. As our understanding of neutrophils in health and disease continually expands, so too does our appreciation of their complex and dynamic nature in vivo; from development, maturation, and trafficking to cellular heterogeneity and functional plasticity. Therefore, contemporary neutrophil research relies on multiple complementary methodologies to perform integrated analysis of neutrophil phenotypic heterogeneity, organ- and stimulus-specific trafficking mechanisms, as well as tailored effector functions in vivo. This review discusses established and emerging technologies used to study neutrophils, with a focus on in vivo imaging in animal models, as well as next-generation ex vivo model systems to study mechanisms of neutrophil function. Furthermore, we discuss how high-dimensional single-cell analysis technologies are driving a renaissance in neutrophil biology by redefining our understanding of neutrophil development, heterogeneity, and functional plasticity. Finally, we discuss innovative applications and emerging opportunities to integrate these high-dimensional, multi-modal techniques to deepen our understanding of neutrophils in cancer research and beyond.
Collapse
|
412
|
Chang CY, Ting HC, Liu CA, Su HL, Chiou TW, Harn HJ, Lin SZ, Ho TJ. Differentiation of Human Pluripotent Stem Cells Into Specific Neural Lineages. Cell Transplant 2021; 30:9636897211017829. [PMID: 34665040 PMCID: PMC8529300 DOI: 10.1177/09636897211017829] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) are sources of several somatic cell
types for human developmental studies, in vitro disease modeling, and
cell transplantation therapy. Improving strategies of derivation of
high-purity specific neural and glial lineages from hPSCs is critical
for application to the study and therapy of the nervous system. Here,
we will focus on the principles behind establishment of neuron and
glia differentiation methods according to developmental studies. We
will also highlight the limitations and challenges associated with the
differentiation of several “difficult” neural lineages and delay in
neuronal maturation and functional integration. To overcome these
challenges, we will introduce strategies and novel technologies aimed
at improving the differentiation of various neural lineages to expand
the application potential of hPSCs to the study of the nervous
system.
Collapse
Affiliation(s)
- Chia-Yu Chang
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,Department of Medical Research, Hualien Tzu Chi Hospital, Hualien, Taiwan.,Neuroscience Center, Hualien Tzu Chi Hospital, Hualien, Taiwan
| | - Hsiao-Chien Ting
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Ching-Ann Liu
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,Department of Medical Research, Hualien Tzu Chi Hospital, Hualien, Taiwan.,Neuroscience Center, Hualien Tzu Chi Hospital, Hualien, Taiwan
| | - Hong-Lin Su
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Tzyy-Wen Chiou
- Department of Life Science, National Dong Hwa University, Hualien, Taiwan
| | - Horng-Jyh Harn
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,Department of Pathology, Hualien Tzu Chi Hospital and Tzu Chi University, Hualien, Taiwan
| | - Shinn-Zong Lin
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,Department of Neurosurgery, Hualien Tzu Chi Hospital, Hualien, Taiwan
| | - Tsung-Jung Ho
- Department of Chinese Medicine, Hualien Tzu Chi Hospital, Hualien, Taiwan.,Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Hualien, Taiwan.,School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
413
|
Dailamy A, Parekh U, Katrekar D, Kumar A, McDonald D, Moreno A, Bagheri P, Ng TN, Mali P. Programmatic introduction of parenchymal cell types into blood vessel organoids. Stem Cell Reports 2021; 16:2432-2441. [PMID: 34559998 PMCID: PMC8515093 DOI: 10.1016/j.stemcr.2021.08.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 08/22/2021] [Accepted: 08/23/2021] [Indexed: 10/25/2022] Open
Abstract
Pluripotent stem cell-derived organoids have transformed our ability to recreate complex three-dimensional models of human tissue. However, the directed differentiation methods used to create them do not afford the ability to introduce cross-germ-layer cell types. Here, we present a bottom-up engineering approach to building vascularized human tissue by combining genetic reprogramming with chemically directed organoid differentiation. As a proof of concept, we created neuro-vascular and myo-vascular organoids via transcription factor overexpression in vascular organoids. We comprehensively characterized neuro-vascular organoids in terms of marker gene expression and composition, and demonstrated that the organoids maintain neural and vascular function for at least 45 days in culture. Finally, we demonstrated chronic electrical stimulation of myo-vascular organoid aggregates as a potential path toward engineering mature and large-scale vascularized skeletal muscle tissue from organoids. Our approach offers a roadmap to build diverse vascularized tissues of any type derived entirely from pluripotent stem cells.
Collapse
Affiliation(s)
- Amir Dailamy
- Department of Bioengineering, University of California San Diego, CA 92093, USA
| | - Udit Parekh
- Department of Electrical and Computer Engineering, University of California San Diego, CA 92093, USA
| | - Dhruva Katrekar
- Department of Bioengineering, University of California San Diego, CA 92093, USA
| | - Aditya Kumar
- Department of Bioengineering, University of California San Diego, CA 92093, USA
| | - Daniella McDonald
- Biomedical Sciences Graduate Program, University of California San Diego, CA 92093, USA
| | - Ana Moreno
- Department of Bioengineering, University of California San Diego, CA 92093, USA
| | - Pegah Bagheri
- Department of Bioengineering, University of California San Diego, CA 92093, USA
| | - Tse Nga Ng
- Department of Electrical and Computer Engineering, University of California San Diego, CA 92093, USA
| | - Prashant Mali
- Department of Bioengineering, University of California San Diego, CA 92093, USA.
| |
Collapse
|
414
|
Xu X, Li L, Luo L, Shu L, Si X, Chen Z, Xia W, Huang J, Liu Y, Shao A, Ke Y. Opportunities and challenges of glioma organoids. Cell Commun Signal 2021; 19:102. [PMID: 34635112 PMCID: PMC8504127 DOI: 10.1186/s12964-021-00777-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 08/15/2021] [Indexed: 12/15/2022] Open
Abstract
Glioma is the most common primary brain tumor and its prognosis is poor. Despite surgical removal, glioma is still prone to recurrence because it grows rapidly in the brain, is resistant to chemotherapy, and is highly aggressive. Therefore, there is an urgent need for a platform to study the cell dynamics of gliomas in order to discover the characteristics of the disease and develop more effective treatments. Although 2D cell models and animal models in previous studies have provided great help for our research, they also have many defects. Recently, scientific researchers have constructed a 3D structure called Organoids, which is similar to the structure of human tissues and organs. Organoids can perfectly compensate for the shortcomings of previous glioma models and are currently the most suitable research platform for glioma research. Therefore, we review the three methods currently used to establish glioma organoids. And introduced how they play a role in the diagnosis and treatment of glioma. Finally, we also summarized the current bottlenecks and difficulties encountered by glioma organoids, and the current efforts to solve these difficulties. Video Abstract
Collapse
Affiliation(s)
- Xiangdong Xu
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory On Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Lingfei Li
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Linting Luo
- Department of Neurology, Liwan Central Hospital of GuangZhou, Guangzhou, People's Republic of China
| | - Lingling Shu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China.,Department of Hematological Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China.,Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Xiaoli Si
- Department of Neurology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Zhenzhen Chen
- Department of Hematology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Wenqing Xia
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Jinyu Huang
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Yang Liu
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory On Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, People's Republic of China.
| | - Anwen Shao
- Department of Neurosurgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, People's Republic of China.
| | - Yiquan Ke
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory On Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, People's Republic of China.
| |
Collapse
|
415
|
Fernandes S, Klein D, Marchetto MC. Unraveling Human Brain Development and Evolution Using Organoid Models. Front Cell Dev Biol 2021; 9:737429. [PMID: 34692694 PMCID: PMC8529117 DOI: 10.3389/fcell.2021.737429] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 09/14/2021] [Indexed: 12/17/2022] Open
Abstract
Brain organoids are proving to be physiologically relevant models for studying human brain development in terms of temporal transcriptional signature recapitulation, dynamic cytoarchitectural development, and functional electrophysiological maturation. Several studies have employed brain organoid technologies to elucidate human-specific processes of brain development, gene expression, and cellular maturation by comparing human-derived brain organoids to those of non-human primates (NHPs). Brain organoids have been established from a variety of NHP pluripotent stem cell (PSC) lines and many protocols are now available for generating brain organoids capable of reproducibly representing specific brain region identities. Innumerous combinations of brain region specific organoids derived from different human and NHP PSCs, with CRISPR-Cas9 gene editing techniques and strategies to promote advanced stages of maturation, will successfully establish complex brain model systems for the accurate representation and elucidation of human brain development. Identified human-specific processes of brain development are likely vulnerable to dysregulation and could result in the identification of therapeutic targets or disease prevention strategies. Here, we discuss the potential of brain organoids to successfully model human-specific processes of brain development and explore current strategies for pinpointing these differences.
Collapse
Affiliation(s)
- Sarah Fernandes
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, United States
- Department of Biological Sciences, University of California, San Diego, San Diego, CA, United States
| | - Davis Klein
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, United States
- Department of Chemistry and Biochemistry, San Diego State University, San Diego, CA, United States
| | - Maria C. Marchetto
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, United States
- Department of Anthropology, Center for Academic Research and Training in Anthropogeny (CARTA), University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
416
|
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: 34] [Impact Index Per Article: 8.5] [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
|
417
|
Dellaquila A, Le Bao C, Letourneur D, Simon‐Yarza T. In Vitro Strategies to Vascularize 3D Physiologically Relevant Models. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100798. [PMID: 34351702 PMCID: PMC8498873 DOI: 10.1002/advs.202100798] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/23/2021] [Indexed: 05/04/2023]
Abstract
Vascularization of 3D models represents a major challenge of tissue engineering and a key prerequisite for their clinical and industrial application. The use of prevascularized models built from dedicated materials could solve some of the actual limitations, such as suboptimal integration of the bioconstructs within the host tissue, and would provide more in vivo-like perfusable tissue and organ-specific platforms. In the last decade, the fabrication of vascularized physiologically relevant 3D constructs has been attempted by numerous tissue engineering strategies, which are classified here in microfluidic technology, 3D coculture models, namely, spheroids and organoids, and biofabrication. In this review, the recent advancements in prevascularization techniques and the increasing use of natural and synthetic materials to build physiological organ-specific models are discussed. Current drawbacks of each technology, future perspectives, and translation of vascularized tissue constructs toward clinics, pharmaceutical field, and industry are also presented. By combining complementary strategies, these models are envisioned to be successfully used for regenerative medicine and drug development in a near future.
Collapse
Affiliation(s)
- Alessandra Dellaquila
- Université de ParisINSERM U1148X Bichat HospitalParisF‐75018France
- Elvesys Microfluidics Innovation CenterParis75011France
- Biomolecular PhotonicsDepartment of PhysicsUniversity of BielefeldBielefeld33615Germany
| | - Chau Le Bao
- Université de ParisINSERM U1148X Bichat HospitalParisF‐75018France
- Université Sorbonne Paris NordGalilée InstituteVilletaneuseF‐93430France
| | | | | |
Collapse
|
418
|
Guy B, Zhang JS, Duncan LH, Johnston RJ. Human neural organoids: Models for developmental neurobiology and disease. Dev Biol 2021; 478:102-121. [PMID: 34181916 PMCID: PMC8364509 DOI: 10.1016/j.ydbio.2021.06.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/08/2021] [Accepted: 06/24/2021] [Indexed: 12/25/2022]
Abstract
Human organoids stand at the forefront of basic and translational research, providing experimentally tractable systems to study human development and disease. These stem cell-derived, in vitro cultures can generate a multitude of tissue and organ types, including distinct brain regions and sensory systems. Neural organoid systems have provided fundamental insights into molecular mechanisms governing cell fate specification and neural circuit assembly and serve as promising tools for drug discovery and understanding disease pathogenesis. In this review, we discuss several human neural organoid systems, how they are generated, advances in 3D imaging and bioengineering, and the impact of organoid studies on our understanding of the human nervous system.
Collapse
Affiliation(s)
- Brian Guy
- Department of Biology, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD, 21218, USA
| | - Jingliang Simon Zhang
- Department of Biology, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD, 21218, USA
| | - Leighton H Duncan
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Robert J Johnston
- Department of Biology, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD, 21218, USA.
| |
Collapse
|
419
|
Prasad M, Kumar R, Buragohain L, Kumari A, Ghosh M. Organoid Technology: A Reliable Developmental Biology Tool for Organ-Specific Nanotoxicity Evaluation. Front Cell Dev Biol 2021; 9:696668. [PMID: 34631696 PMCID: PMC8495170 DOI: 10.3389/fcell.2021.696668] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 08/13/2021] [Indexed: 12/14/2022] Open
Abstract
Engineered nanomaterials are bestowed with certain inherent physicochemical properties unlike their parent materials, rendering them suitable for the multifaceted needs of state-of-the-art biomedical, and pharmaceutical applications. The log-phase development of nano-science along with improved "bench to beside" conversion carries an enhanced probability of human exposure with numerous nanoparticles. Thus, toxicity assessment of these novel nanoscale materials holds a key to ensuring the safety aspects or else the global biome will certainly face a debacle. The toxicity may span from health hazards due to direct exposure to indirect means through food chain contamination or environmental pollution, even causing genotoxicity. Multiple ways of nanotoxicity evaluation include several in vitro and in vivo methods, with in vitro methods occupying the bulk of the "experimental space." The underlying reason may be multiple, but ethical constraints in in vivo animal experiments are a significant one. Two-dimensional (2D) monoculture is undoubtedly the most exploited in vitro method providing advantages in terms of cost-effectiveness, high throughput, and reproducibility. However, it often fails to mimic a tissue or organ which possesses a defined three-dimensional structure (3D) along with intercellular communication machinery. Instead, microtissues such as spheroids or organoids having a precise 3D architecture and proximate in vivo tissue-like behavior can provide a more realistic evaluation than 2D monocultures. Recent developments in microfluidics and bioreactor-based organoid synthesis have eased the difficulties to prosper nano-toxicological analysis in organoid models surpassing the obstacle of ethical issues. The present review will enlighten applications of organoids in nanotoxicological evaluation, their advantages, and prospects toward securing commonplace nano-interventions.
Collapse
Affiliation(s)
- Minakshi Prasad
- Department of Animal Biotechnology, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, India
| | - Rajesh Kumar
- Department of Veterinary Physiology and Biochemistry, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, India
| | - Lukumoni Buragohain
- Department of Animal Biotechnology, College of Veterinary Science, Assam Agricultural University, Guwahati, India
| | | | - Mayukh Ghosh
- Department of Veterinary Physiology and Biochemistry, RGSC, Banaras Hindu University, Varanasi, India
| |
Collapse
|
420
|
Retinal Organoid Technology: Where Are We Now? Int J Mol Sci 2021; 22:ijms221910244. [PMID: 34638582 PMCID: PMC8549701 DOI: 10.3390/ijms221910244] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/17/2021] [Accepted: 09/20/2021] [Indexed: 12/25/2022] Open
Abstract
It is difficult to regenerate mammalian retinal cells once the adult retina is damaged, and current clinical approaches to retinal damages are very limited. The introduction of the retinal organoid technique empowers researchers to study the molecular mechanisms controlling retinal development, explore the pathogenesis of retinal diseases, develop novel treatment options, and pursue cell/tissue transplantation under a certain genetic background. Here, we revisit the historical background of retinal organoid technology, categorize current methods of organoid induction, and outline the obstacles and potential solutions to next-generation retinal organoids. Meanwhile, we recapitulate recent research progress in cell/tissue transplantation to treat retinal diseases, and discuss the pros and cons of transplanting single-cell suspension versus retinal organoid sheet for cell therapies.
Collapse
|
421
|
What can biofabrication do for space and what can space do for biofabrication? Trends Biotechnol 2021; 40:398-411. [PMID: 34544616 DOI: 10.1016/j.tibtech.2021.08.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/19/2021] [Accepted: 08/23/2021] [Indexed: 01/08/2023]
Abstract
Biofabrication in space is one of the novel promising and prospective research directions in the rapidly emerging field of space STEM. There are several advantages of biofabrication in space. Under microgravity, it is possible to engineer constructs using more fluidic channels and thus more biocompatible bioinks. Microgravity enables biofabrication of tissue and organ constructs of more complex geometries, thus facilitating novel scaffold-, label-, and nozzle-free technologies based on multi-levitation principles. However, when exposed to microgravity and cosmic radiation, biofabricated tissues could be used to study pathophysiological phenomena that will be useful on Earth and for deep space manned missions. Here, we provide leading concepts about the potential mutual benefits of the application of biofabrication technologies in space.
Collapse
|
422
|
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: 8] [Impact Index Per Article: 2.0] [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
|
423
|
Novel Approaches Used to Examine and Control Neurogenesis in Parkinson's Disease. Int J Mol Sci 2021; 22:ijms22179608. [PMID: 34502516 PMCID: PMC8431772 DOI: 10.3390/ijms22179608] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/31/2021] [Accepted: 09/02/2021] [Indexed: 12/16/2022] Open
Abstract
Neurogenesis is a key mechanism of brain development and plasticity, which is impaired in chronic neurodegeneration, including Parkinson’s disease. The accumulation of aberrant α-synuclein is one of the features of PD. Being secreted, this protein produces a prominent neurotoxic effect, alters synaptic plasticity, deregulates intercellular communication, and supports the development of neuroinflammation, thereby providing propagation of pathological events leading to the establishment of a PD-specific phenotype. Multidirectional and ambiguous effects of α-synuclein on adult neurogenesis suggest that impaired neurogenesis should be considered as a target for the prevention of cell loss and restoration of neurological functions. Thus, stimulation of endogenous neurogenesis or cell-replacement therapy with stem cell-derived differentiated neurons raises new hopes for the development of effective and safe technologies for treating PD neurodegeneration. Given the rapid development of optogenetics, it is not surprising that this method has already been repeatedly tested in manipulating neurogenesis in vivo and in vitro via targeting stem or progenitor cells. However, niche astrocytes could also serve as promising candidates for controlling neuronal differentiation and improving the functional integration of newly formed neurons within the brain tissue. In this review, we mainly focus on current approaches to assess neurogenesis and prospects in the application of optogenetic protocols to restore the neurogenesis in Parkinson’s disease.
Collapse
|
424
|
Ryan AR, England AR, Chaney CP, Cowdin MA, Hiltabidle M, Daniel E, Gupta AK, Oxburgh L, Carroll TJ, Cleaver O. Vascular deficiencies in renal organoids and ex vivo kidney organogenesis. Dev Biol 2021; 477:98-116. [PMID: 34000274 PMCID: PMC8382085 DOI: 10.1016/j.ydbio.2021.04.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 04/24/2021] [Accepted: 04/27/2021] [Indexed: 12/18/2022]
Abstract
Chronic kidney disease (CKD) and end stage renal disease (ESRD) are increasingly frequent and devastating conditions that have driven a surge in the need for kidney transplantation. A stark shortage of organs has fueled interest in generating viable replacement tissues ex vivo for transplantation. One promising approach has been self-organizing organoids, which mimic developmental processes and yield multicellular, organ-specific tissues. However, a recognized roadblock to this approach is that many organoid cell types fail to acquire full maturity and function. Here, we comprehensively assess the vasculature in two distinct kidney organoid models as well as in explanted embryonic kidneys. Using a variety of methods, we show that while organoids can develop a wide range of kidney cell types, as previously shown, endothelial cells (ECs) initially arise but then rapidly regress over time in culture. Vasculature of cultured embryonic kidneys exhibit similar regression. By contrast, engraftment of kidney organoids under the kidney capsule results in the formation of a stable, perfused vasculature that integrates into the organoid. This work demonstrates that kidney organoids offer a promising model system to define the complexities of vascular-nephron interactions, but the establishment and maintenance of a vascular network present unique challenges when grown ex vivo.
Collapse
Affiliation(s)
- Anne R Ryan
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Alicia R England
- Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Christopher P Chaney
- Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Mitzy A Cowdin
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Max Hiltabidle
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Edward Daniel
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | | | - Thomas J Carroll
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ondine Cleaver
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
425
|
Maoz BM. Brain-on-a-Chip: Characterizing the next generation of advanced in vitro platforms for modeling the central nervous system. APL Bioeng 2021; 5:030902. [PMID: 34368601 PMCID: PMC8325567 DOI: 10.1063/5.0055812] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 07/19/2021] [Indexed: 02/07/2023] Open
Abstract
The complexity of the human brain creates significant, almost insurmountable challenges for neurological drug development. Advanced in vitro platforms are increasingly enabling researchers to overcome these challenges, by mimicking key features of the brain's composition and functionality. Many of these platforms are called "Brains-on-a-Chip"-a term that was originally used to refer to microfluidics-based systems containing miniature engineered tissues, but that has since expanded to describe a vast range of in vitro central nervous system (CNS) modeling approaches. This Perspective seeks to refine the definition of a Brain-on-a-Chip for the next generation of in vitro platforms, identifying criteria that determine which systems should qualify. These criteria reflect the extent to which a given platform overcomes the challenges unique to in vitro CNS modeling (e.g., recapitulation of the brain's microenvironment; inclusion of critical subunits, such as the blood-brain barrier) and thereby provides meaningful added value over conventional cell culture systems. The paper further outlines practical considerations for the development and implementation of Brain-on-a-Chip platforms and concludes with a vision for where these technologies may be heading.
Collapse
Affiliation(s)
- Ben M. Maoz
- Author to whom correspondence should be addressed:
| |
Collapse
|
426
|
Liu S, Striebel J, Pasquini G, Ng AHM, Khoshakhlagh P, Church GM, Busskamp V. Neuronal Cell-type Engineering by Transcriptional Activation. Front Genome Ed 2021; 3:715697. [PMID: 34713262 PMCID: PMC8525383 DOI: 10.3389/fgeed.2021.715697] [Citation(s) in RCA: 4] [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: 05/27/2021] [Accepted: 08/12/2021] [Indexed: 12/26/2022] Open
Abstract
Gene activation with the CRISPR-Cas system has great implications in studying gene function, controlling cellular behavior, and modulating disease progression. In this review, we survey recent studies on targeted gene activation and multiplexed screening for inducing neuronal differentiation using CRISPR-Cas transcriptional activation (CRISPRa) and open reading frame (ORF) expression. Critical technical parameters of CRISPRa and ORF-based strategies for neuronal programming are presented and discussed. In addition, recent progress on in vivo applications of CRISPRa to the nervous system are highlighted. Overall, CRISPRa represents a valuable addition to the experimental toolbox for neuronal cell-type programming.
Collapse
Affiliation(s)
- Songlei Liu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, United States
| | - Johannes Striebel
- Department of Ophthalmology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Giovanni Pasquini
- Department of Ophthalmology, Medical Faculty, University of Bonn, Bonn, Germany
| | | | | | - George M. Church
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, United States
- GC Therapeutics, Inc, Cambridge, MA, United States
| | - Volker Busskamp
- Department of Ophthalmology, Medical Faculty, University of Bonn, Bonn, Germany
| |
Collapse
|
427
|
Wang YF, Liu C, Xu PF. Deciphering and reconstitution of positional information in the human brain development. ACTA ACUST UNITED AC 2021; 10:29. [PMID: 34467458 PMCID: PMC8408296 DOI: 10.1186/s13619-021-00091-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 08/02/2021] [Indexed: 12/29/2022]
Abstract
Organoid has become a novel in vitro model to research human development and relevant disorders in recent years. With many improvements on the culture protocols, current brain organoids could self-organize into a complicated three-dimensional organization that mimics most of the features of the real human brain at the molecular, cellular, and further physiological level. However, lacking positional information, an important characteristic conveyed by gradients of signaling molecules called morphogens, leads to the deficiency of spatiotemporally regulated cell arrangements and cell–cell interactions in the brain organoid development. In this review, we will overview the role of morphogen both in the vertebrate neural development in vivo as well as the brain organoid culture in vitro, the strategies to apply morphogen concentration gradients in the organoid system and future perspectives of the brain organoid technology.
Collapse
Affiliation(s)
- Yi-Fan Wang
- Women's Hospital, and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Institute of Zhejiang University and University of Edinburgh, Jiaxing, Zhejiang, China.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Dr, Singapore, 117599, Singapore
| | - Cong Liu
- Women's Hospital, and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Peng-Fei Xu
- Women's Hospital, and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
428
|
Sun L, Hui L. Progress in human liver organoids. J Mol Cell Biol 2021; 12:607-617. [PMID: 32236564 PMCID: PMC7683012 DOI: 10.1093/jmcb/mjaa013] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 12/30/2019] [Accepted: 03/30/2020] [Indexed: 12/24/2022] Open
Abstract
Understanding the development, regeneration, and disorders of the liver is the major goal in liver biology. Current mechanistic knowledge of human livers has been largely derived from mouse models and cell lines, which fall short in recapitulating the features of human liver cells or the structures and functions of human livers. Organoids as an in vitro system hold the promise to generate organ-like tissues in a dish. Recent advances in human liver organoids also facilitate the understanding of the biology and diseases in this complex organ. Here we review the progress in human liver organoids, mainly focusing on the methods to generate liver organoids, their applications, and possible future directions.
Collapse
Affiliation(s)
- Lulu Sun
- State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Lijian Hui
- State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.,Bio-Research Innovation Center, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Suzhou 215121, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
429
|
Haase FD, Coorey B, Riley L, Cantrill LC, Tam PPL, Gold WA. Pre-clinical Investigation of Rett Syndrome Using Human Stem Cell-Based Disease Models. Front Neurosci 2021; 15:698812. [PMID: 34512241 PMCID: PMC8423999 DOI: 10.3389/fnins.2021.698812] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 07/19/2021] [Indexed: 01/02/2023] Open
Abstract
Rett syndrome (RTT) is an X-linked neurodevelopmental disorder, mostly caused by mutations in MECP2. The disorder mainly affects girls and it is associated with severe cognitive and physical disabilities. Modeling RTT in neural and glial cell cultures and brain organoids derived from patient- or mutation-specific human induced pluripotent stem cells (iPSCs) has advanced our understanding of the pathogenesis of RTT, such as disease-causing mechanisms, disease progression, and cellular and molecular pathology enabling the identification of actionable therapeutic targets. Brain organoid models that recapitulate much of the tissue architecture and the complexity of cell types in the developing brain, offer further unprecedented opportunity for elucidating human neural development, without resorting to conventional animal models and the limited resource of human neural tissues. This review focuses on the new knowledge of RTT that has been gleaned from the iPSC-based models as well as limitations of the models and strategies to refine organoid technology in the quest for clinically relevant disease models for RTT and the broader spectrum of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Florencia D. Haase
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Kids Neuroscience Centre, Kids Research, Children’s Hospital at Westmead, Westmead, NSW, Australia
- Molecular Neurobiology Research Laboratory, Kids Research, Children’s Hospital at Westmead, and Children’s Medical Research Institute, Westmead, NSW, Australia
| | - Bronte Coorey
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Kids Neuroscience Centre, Kids Research, Children’s Hospital at Westmead, Westmead, NSW, Australia
- Molecular Neurobiology Research Laboratory, Kids Research, Children’s Hospital at Westmead, and Children’s Medical Research Institute, Westmead, NSW, Australia
| | - Lisa Riley
- Rare Diseases Functional Genomics Laboratory, Kids Research, Children’s Hospital at Westmead, and Children’s Medical Research Institute, Westmead, NSW, Australia
| | - Laurence C. Cantrill
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Kids Research, Children’s Hospital at Westmead, Westmead, NSW, Australia
| | - Patrick P. L. Tam
- Embryology Research Unit, Children’s Medical Research Institute, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Wendy A. Gold
- Kids Neuroscience Centre, Kids Research, Children’s Hospital at Westmead, Westmead, NSW, Australia
- Molecular Neurobiology Research Laboratory, Kids Research, Children’s Hospital at Westmead, and Children’s Medical Research Institute, Westmead, NSW, Australia
- Rare Diseases Functional Genomics Laboratory, Kids Research, Children’s Hospital at Westmead, and Children’s Medical Research Institute, Westmead, NSW, Australia
| |
Collapse
|
430
|
Iwata R. Temporal differences of neurodevelopment processes between species. Neurosci Res 2021; 177:8-15. [PMID: 34419562 DOI: 10.1016/j.neures.2021.08.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 07/08/2021] [Accepted: 08/17/2021] [Indexed: 02/06/2023]
Abstract
The ontogeny programs are highly conserved across all vertebrates, although there are significant temporal variations in interspecies developmental processes. Changing the timing and rate of developmental processes could affect subsequent organogenesis profoundly and may also have been critical factors in evolutionary diversity. However, despite their potential importance, the cellular and molecular mechanisms that control interspecies differences in developmental timescale remain unclear. This review highlights recent advances in the experimental models to compare interspecies differences in neurodevelopmental processes, neurogenesis, and neuronal maturation and discusses the possible mechanisms that could generate species-specific timescales.
Collapse
Affiliation(s)
- Ryohei Iwata
- VIB KU Leuven Center for Brain & Disease Research, KU Leuven Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium; Université Libre de Bruxelles (ULB), Institut de Recherches en Biologie Humaine et Moléculaire (IRIBHM), ULB Neuroscience Institute (UNI), 1070 Brussels, Belgium.
| |
Collapse
|
431
|
Günther C, Rothhammer V, Karow M, Neurath M, Winner B. The Gut-Brain Axis in Inflammatory Bowel Disease-Current and Future Perspectives. Int J Mol Sci 2021; 22:ijms22168870. [PMID: 34445575 PMCID: PMC8396333 DOI: 10.3390/ijms22168870] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/11/2021] [Accepted: 08/13/2021] [Indexed: 12/13/2022] Open
Abstract
The gut–brain axis is a bidirectional communication system driven by neural, hormonal, metabolic, immunological, and microbial signals. Signaling events from the gut can modulate brain function and recent evidence suggests that the gut–brain axis may play a pivotal role in linking gastrointestinal and neurological diseases. Accordingly, accumulating evidence has suggested a link between inflammatory bowel diseases (IBDs) and neurodegenerative, as well as neuroinflammatory diseases. In this context, clinical, epidemiological and experimental data have demonstrated that IBD predisposes a person to pathologies of the central nervous system (CNS). Likewise, a number of neurological disorders are associated with changes in the intestinal environment, which are indicative for disease-mediated gut–brain inter-organ communication. Although this axis was identified more than 20 years ago, the sequence of events and underlying molecular mechanisms are poorly defined. The emergence of precision medicine has uncovered the need to take into account non-intestinal symptoms in the context of IBD that could offer the opportunity to tailor therapies to individual patients. The aim of this review is to highlight recent findings supporting the clinical and biological link between the gut and brain, as well as its clinical significance for IBD as well as neurodegeneration and neuroinflammation. Finally, we focus on novel human-specific preclinical models that will help uncover disease mechanisms to better understand and modulate the function of this complex system.
Collapse
Affiliation(s)
- Claudia Günther
- Department of Medicine 1, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany;
- Correspondence: (C.G.); (B.W.); Tel.: +49-(0)9131-85-45240 (C.G.); +49-(0)9131-85-39301 (B.W.)
| | - Veit Rothhammer
- Department of Neurology, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - Marisa Karow
- Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - Markus Neurath
- Department of Medicine 1, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - Beate Winner
- Department of Stem Cell Biology, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany
- Correspondence: (C.G.); (B.W.); Tel.: +49-(0)9131-85-45240 (C.G.); +49-(0)9131-85-39301 (B.W.)
| |
Collapse
|
432
|
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: 24] [Impact Index Per Article: 6.0] [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
|
433
|
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: 19] [Impact Index Per Article: 4.8] [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
|
434
|
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: 42] [Impact Index Per Article: 10.5] [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
|
435
|
The Renin-Angiotensin System in the Tumor Microenvironment of Glioblastoma. Cancers (Basel) 2021; 13:cancers13164004. [PMID: 34439159 PMCID: PMC8392691 DOI: 10.3390/cancers13164004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 08/03/2021] [Accepted: 08/06/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Glioblastoma (GB) is the most aggressive brain cancer in humans. Patient survival outcomes have remained dismal despite intensive research over the past 50 years, with a median overall survival of only 14.6 months. We highlight the critical role of the renin–angiotensin system (RAS) on GB cancer stem cells and the tumor microenvironment which, in turn, influences cancer stem cells in driving tumorigenesis and treatment resistance. We present recent developments and underscore the need for further research into the GB tumor microenvironment. We discuss the novel therapeutic targeting of the RAS using existing commonly available medications and utilizing model systems to further this critical investigation. Abstract Glioblastoma (GB) is an aggressive primary brain tumor. Despite intensive research over the past 50 years, little advance has been made to improve the poor outcome, with an overall median survival of 14.6 months following standard treatment. Local recurrence is inevitable due to the quiescent cancer stem cells (CSCs) in GB that co-express stemness-associated markers and components of the renin–angiotensin system (RAS). The dynamic and heterogeneous tumor microenvironment (TME) plays a fundamental role in tumor development, progression, invasiveness, and therapy resistance. There is increasing evidence showing the critical role of the RAS in the TME influencing CSCs via its upstream and downstream pathways. Drugs that alter the hallmarks of cancer by modulating the RAS present a potential new therapeutic alternative or adjunct to conventional treatment of GB. Cerebral and GB organoids may offer a cost-effective method for evaluating the efficacy of RAS-modulating drugs on GB. We review the nexus between the GB TME, CSC niche, and the RAS, and propose re-purposed RAS-modulating drugs as a potential therapeutic alternative or adjunct to current standard therapy for GB.
Collapse
|
436
|
Cho AN, Jin Y, An Y, Kim J, Choi YS, Lee JS, Kim J, Choi WY, Koo DJ, Yu W, Chang GE, Kim DY, Jo SH, Kim J, Kim SY, Kim YG, Kim JY, Choi N, Cheong E, Kim YJ, Je HS, Kang HC, Cho SW. Microfluidic device with brain extracellular matrix promotes structural and functional maturation of human brain organoids. Nat Commun 2021; 12:4730. [PMID: 34354063 PMCID: PMC8342542 DOI: 10.1038/s41467-021-24775-5] [Citation(s) in RCA: 193] [Impact Index Per Article: 48.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 07/06/2021] [Indexed: 11/10/2022] Open
Abstract
Brain organoids derived from human pluripotent stem cells provide a highly valuable in vitro model to recapitulate human brain development and neurological diseases. However, the current systems for brain organoid culture require further improvement for the reliable production of high-quality organoids. Here, we demonstrate two engineering elements to improve human brain organoid culture, (1) a human brain extracellular matrix to provide brain-specific cues and (2) a microfluidic device with periodic flow to improve the survival and reduce the variability of organoids. A three-dimensional culture modified with brain extracellular matrix significantly enhanced neurogenesis in developing brain organoids from human induced pluripotent stem cells. Cortical layer development, volumetric augmentation, and electrophysiological function of human brain organoids were further improved in a reproducible manner by dynamic culture in microfluidic chamber devices. Our engineering concept of reconstituting brain-mimetic microenvironments facilitates the development of a reliable culture platform for brain organoids, enabling effective modeling and drug development for human brain diseases.
Collapse
Affiliation(s)
- Ann-Na Cho
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Yoonhee Jin
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Yeonjoo An
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Jin Kim
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Yi Sun Choi
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Jung Seung Lee
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Junghoon Kim
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Won-Young Choi
- Department of Biochemistry, Yonsei University, Seoul, Republic of Korea
| | - Dong-Jun Koo
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea
| | - Weonjin Yu
- Signature Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Gyeong-Eon Chang
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Dong-Yoon Kim
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea
| | - Sung-Hyun Jo
- Department of Chemical Engineering, Soongsil University, Seoul, Republic of Korea
| | - Jihun Kim
- Division of Pediatric Neurology, Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sung-Yon Kim
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea
| | - Yun-Gon Kim
- Department of Chemical Engineering, Soongsil University, Seoul, Republic of Korea
| | - Ju Young Kim
- Department of Advanced Materials Engineering, Kangwon National University, Samcheok, Republic of Korea
| | - Nakwon Choi
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Eunji Cheong
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Young-Joon Kim
- Department of Biochemistry, Yonsei University, Seoul, Republic of Korea
| | - Hyunsoo Shawn Je
- Signature Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Hoon-Chul Kang
- Division of Pediatric Neurology, Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea.
- Center for Nanomedicine, Institute for Basic science (IBS), Seoul, Republic of Korea.
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
437
|
Current tools to interrogate microglial biology. Neuron 2021; 109:2805-2819. [PMID: 34390649 DOI: 10.1016/j.neuron.2021.07.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 06/23/2021] [Accepted: 07/06/2021] [Indexed: 12/26/2022]
Abstract
Microglial cells perform a plethora of functions in the central nervous system (CNS), involving them in brain development, maintenance of homeostasis in adulthood, and CNS diseases. Significant technical advancements have prompted the development of novel systems adapted to analyze microglia with increasing specificity and intricacy. The advent of single-cell technologies combined with targeted mouse models has been decisive in deciphering microglia heterogeneity and dissecting microglial functions. However sophisticated these tools have become, clear limitations remain. Understanding their pitfalls and advantages ensures their correct application. Therefore, we provide a guide to the cutting-edge methods currently available to dissect microglial biology.
Collapse
|
438
|
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: 15] [Impact Index Per Article: 3.8] [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
|
439
|
Wu Z, Cai H, Ao Z, Xu J, Heaps S, Guo F. Microfluidic Printing of Tunable Hollow Microfibers for Vascular Tissue Engineering. ADVANCED MATERIALS TECHNOLOGIES 2021; 6:2000683. [PMID: 34458563 PMCID: PMC8386518 DOI: 10.1002/admt.202000683] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Indexed: 05/28/2023]
Abstract
Bioprinting of vascular tissues holds great potential in tissue engineering and regenerative medicine. However, challenges remain in fabricating biocompatible and versatile scaffolds for the rapid engineering of vascular tissues and vascularized organs. Here, we report novel bioink-enabled microfluidic printing of tunable hollow microfibers for the rapid formation of blood vessels. By compositing biomaterials including sodium alginate, gelatin methacrylate (GelMA), and glycidyl-methacrylate silk fibroin (SilkMA), we prepared a novel composite bioink with excellent printability and biocompatibility. This composite bioink can be printed into hollow microfibers with tunable dimensions using a microfluidic co-axial printing. After seeding human umbilical vein endothelial cells (HUVEC) into the hollow chambers via a microfluidic prefusion device, these cells can adhere to, grow, proliferate, and then cover the internal surface of the printed hollow scaffolds to form vessel-like tissue structures within three days. By combining the unique composite bioink, microfluidic printing of vascular scaffolds, and microfluidic cell seeding and culturing, our strategy can fabricate vascular-like tissue structures with high viability and tunable dimension within three days. The presented method may engineer in vitro vasculatures for the broad applications in basic research and translational medicine including in vitro disease models, tissue microcirculation, and tissue transplantation.
Collapse
|
440
|
Chen KG, Park K, Spence JR. Studying SARS-CoV-2 infectivity and therapeutic responses with complex organoids. Nat Cell Biol 2021; 23:822-833. [PMID: 34341531 PMCID: PMC8355201 DOI: 10.1038/s41556-021-00721-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 06/22/2021] [Indexed: 12/15/2022]
Abstract
Clinical management of patients with severe complications of COVID-19 has been hindered by a lack of effective drugs and a failure to capture the extensive heterogeneity of the disease with conventional methods. Here we review the emerging roles of complex organoids in the study of SARS-CoV-2 infection, modelling of COVID-19 disease pathology and in drug, antibody and vaccine development. We discuss opportunities for COVID-19 research and remaining challenges in the application of organoids.
Collapse
Affiliation(s)
- Kevin G Chen
- NIH Stem Cell Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| | - Kyeyoon Park
- NIH Stem Cell Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Jason R Spence
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
441
|
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: 26] [Impact Index Per Article: 6.5] [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
|
442
|
Wörsdörfer P, Ergün S. The Impact of Oxygen Availability and Multilineage Communication on Organoid Maturation. Antioxid Redox Signal 2021; 35:217-233. [PMID: 33334234 DOI: 10.1089/ars.2020.8195] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Significance: An optimal supply with oxygen is of high importance during embryogenesis and a prerequisite for proper organ development. Different tissues require varying amounts of oxygen, and even within single organs, different phases of development go alongside with either physiological hypoxia or the need for sufficient oxygen supply. Recent Advances: Human induced pluripotent stem cell-derived organoid models are state of the art cell culture platforms for the investigation of developmental processes, disease modeling, and drug testing. Organoids modeling the development of multiple tissues were developed within the past years. Critical Issues: Until now, optimization of oxygen supply and its role during organoid growth, differentiation, and maturation have only rarely been addressed. Recent publications indicate that hypoxia-induced processes play an important role in three-dimensional tissue cultures, triggering multilineage communication between mesenchymal cells, the endothelium, as well as organotypic cells. Later in culture, a sufficient supply with oxygen is of high importance to allow larger organoid sizes. Moreover, cellular stress is reduced and tissue maturation is improved. Therefore, a functional blood vessel network is required. Future Directions: In this review, we will briefly summarize aspects of the role of oxygen during embryonic development and organogenesis, present an update on novel organoid models with a special focus on organoid vascularization, and discuss the importance of complex organoids involving parenchymal cells, mesenchymal cells, inflammatory cells, and functional blood vessels for the generation of mature and fully functional tissues in vitro. Antioxid. Redox Signal. 35, 217-233.
Collapse
Affiliation(s)
- Philipp Wörsdörfer
- Institute of Anatomy and Cell Biology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Süleyman Ergün
- Institute of Anatomy and Cell Biology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| |
Collapse
|
443
|
Wu YC, Sonninen TM, Peltonen S, Koistinaho J, Lehtonen Š. Blood-Brain Barrier and Neurodegenerative Diseases-Modeling with iPSC-Derived Brain Cells. Int J Mol Sci 2021; 22:7710. [PMID: 34299328 PMCID: PMC8307585 DOI: 10.3390/ijms22147710] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/11/2021] [Accepted: 07/12/2021] [Indexed: 12/12/2022] Open
Abstract
The blood-brain barrier (BBB) regulates the delivery of oxygen and important nutrients to the brain through active and passive transport and prevents neurotoxins from entering the brain. It also has a clearance function and removes carbon dioxide and toxic metabolites from the central nervous system (CNS). Several drugs are unable to cross the BBB and enter the CNS, adding complexity to drug screens targeting brain disorders. A well-functioning BBB is essential for maintaining healthy brain tissue, and a malfunction of the BBB, linked to its permeability, results in toxins and immune cells entering the CNS. This impairment is associated with a variety of neurological diseases, including Alzheimer's disease and Parkinson's disease. Here, we summarize current knowledge about the BBB in neurodegenerative diseases. Furthermore, we focus on recent progress of using human-induced pluripotent stem cell (iPSC)-derived models to study the BBB. We review the potential of novel stem cell-based platforms in modeling the BBB and address advances and key challenges of using stem cell technology in modeling the human BBB. Finally, we highlight future directions in this area.
Collapse
Affiliation(s)
- Ying-Chieh Wu
- Neuroscience Center, University of Helsinki, 00014 Helsinki, Finland; (Y.-C.W.); (T.-M.S.); (S.P.); (J.K.)
| | - Tuuli-Maria Sonninen
- Neuroscience Center, University of Helsinki, 00014 Helsinki, Finland; (Y.-C.W.); (T.-M.S.); (S.P.); (J.K.)
| | - Sanni Peltonen
- Neuroscience Center, University of Helsinki, 00014 Helsinki, Finland; (Y.-C.W.); (T.-M.S.); (S.P.); (J.K.)
| | - Jari Koistinaho
- Neuroscience Center, University of Helsinki, 00014 Helsinki, Finland; (Y.-C.W.); (T.-M.S.); (S.P.); (J.K.)
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| | - Šárka Lehtonen
- Neuroscience Center, University of Helsinki, 00014 Helsinki, Finland; (Y.-C.W.); (T.-M.S.); (S.P.); (J.K.)
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| |
Collapse
|
444
|
Huang Y, Dai Y, Li M, Guo L, Cao C, Huang Y, Ma R, Qiu S, Su X, Zhong K, Huang Y, Gao H, Bu Q. Exposure to cadmium induces neuroinflammation and impairs ciliogenesis in hESC-derived 3D cerebral organoids. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 797:149043. [PMID: 34303983 DOI: 10.1016/j.scitotenv.2021.149043] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/06/2021] [Accepted: 07/10/2021] [Indexed: 02/08/2023]
Abstract
Cadmium (Cd) is an environmental heavy metal toxicant with central nervous system toxicity and has a greater negative impact on fetal neurodevelopment. However, the causative mechanisms for the neurodevelopmental toxicity of Cd have remained unclear. The human cerebral organoids can better mimic the three-dimensional structure of the early fetal nerve tissue, which can be used to study the developmental neurotoxicity under the condition of maternal exposure to Cd. Our study identified that Cd exposure specifically induced apoptosis in neurons and inhibited the proliferation of neural progenitor cells, but neural differentiation was not significantly affected in cerebral organoids. Cd exposure also elicited overexpression of GFAP, a marker of astrocytes and resulted in IL-6 release. This study revealed that mineral absorption was significantly disturbed with metallothioneins expression up-regulation. Moreover, we found Cd exposure inhibited cilium-related gene expression and reduced ciliary length with increasing dose. In conclusion, our study has shown that Cd exposure regulated neural cell proliferation and death, induced neuroinflammation, enhanced metal ion absorption, and impaired ciliogenesis, which hinder the normal development of the fetal brain.
Collapse
Affiliation(s)
- Yan Huang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Yanping Dai
- National Chengdu Center for Safety Evaluation of Drugs, State Key Lab of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Min Li
- National Chengdu Center for Safety Evaluation of Drugs, State Key Lab of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Lulu Guo
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Chulin Cao
- College of Life Sciences, Northwest A&F University, Yangling 712100, China
| | - Yuting Huang
- Department of Food Science and Technology, College of Biomass and Engineering and Healthy Food Evaluation Research Center, Sichuan University, Chengdu 610065, China
| | - Rui Ma
- Department of Food Science and Technology, College of Biomass and Engineering and Healthy Food Evaluation Research Center, Sichuan University, Chengdu 610065, China
| | - Shengyue Qiu
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaoyi Su
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Kai Zhong
- Department of Food Science and Technology, College of Biomass and Engineering and Healthy Food Evaluation Research Center, Sichuan University, Chengdu 610065, China
| | - Yina Huang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Hong Gao
- Department of Food Science and Technology, College of Biomass and Engineering and Healthy Food Evaluation Research Center, Sichuan University, Chengdu 610065, China
| | - Qian Bu
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China; National Chengdu Center for Safety Evaluation of Drugs, State Key Lab of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
445
|
Azar J, Bahmad HF, Daher D, Moubarak MM, Hadadeh O, Monzer A, Al Bitar S, Jamal M, Al-Sayegh M, Abou-Kheir W. The Use of Stem Cell-Derived Organoids in Disease Modeling: An Update. Int J Mol Sci 2021; 22:7667. [PMID: 34299287 PMCID: PMC8303386 DOI: 10.3390/ijms22147667] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/17/2021] [Accepted: 06/18/2021] [Indexed: 02/06/2023] Open
Abstract
Organoids represent one of the most important advancements in the field of stem cells during the past decade. They are three-dimensional in vitro culturing models that originate from self-organizing stem cells and can mimic the in vivo structural and functional specificities of body organs. Organoids have been established from multiple adult tissues as well as pluripotent stem cells and have recently become a powerful tool for studying development and diseases in vitro, drug screening, and host-microbe interaction. The use of stem cells-that have self-renewal capacity to proliferate and differentiate into specialized cell types-for organoids culturing represents a major advancement in biomedical research. Indeed, this new technology has a great potential to be used in a multitude of fields, including cancer research, hereditary and infectious diseases. Nevertheless, organoid culturing is still rife with many challenges, not limited to being costly and time consuming, having variable rates of efficiency in generation and maintenance, genetic stability, and clinical applications. In this review, we aim to provide a synopsis of pluripotent stem cell-derived organoids and their use for disease modeling and other clinical applications.
Collapse
Affiliation(s)
- Joseph Azar
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2260, Lebanon; (J.A.); (H.F.B.); (D.D.); (M.M.M.); (O.H.); (A.M.); (S.A.B.)
| | - Hisham F. Bahmad
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2260, Lebanon; (J.A.); (H.F.B.); (D.D.); (M.M.M.); (O.H.); (A.M.); (S.A.B.)
- Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL 33140, USA
| | - Darine Daher
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2260, Lebanon; (J.A.); (H.F.B.); (D.D.); (M.M.M.); (O.H.); (A.M.); (S.A.B.)
| | - Maya M. Moubarak
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2260, Lebanon; (J.A.); (H.F.B.); (D.D.); (M.M.M.); (O.H.); (A.M.); (S.A.B.)
| | - Ola Hadadeh
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2260, Lebanon; (J.A.); (H.F.B.); (D.D.); (M.M.M.); (O.H.); (A.M.); (S.A.B.)
| | - Alissar Monzer
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2260, Lebanon; (J.A.); (H.F.B.); (D.D.); (M.M.M.); (O.H.); (A.M.); (S.A.B.)
| | - Samar Al Bitar
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2260, Lebanon; (J.A.); (H.F.B.); (D.D.); (M.M.M.); (O.H.); (A.M.); (S.A.B.)
| | - Mohamed Jamal
- Hamdan Bin Mohammed College of Dental Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai 66566, United Arab Emirates
| | - Mohamed Al-Sayegh
- Biology Division, New York University Abu Dhabi, Abu Dhabi 2460, United Arab Emirates
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2260, Lebanon; (J.A.); (H.F.B.); (D.D.); (M.M.M.); (O.H.); (A.M.); (S.A.B.)
| |
Collapse
|
446
|
Li H, Gao L, Du J, Ma T, Ye Z, Li Z. To Better Generate Organoids, What Can We Learn From Teratomas? Front Cell Dev Biol 2021; 9:700482. [PMID: 34336851 PMCID: PMC8324104 DOI: 10.3389/fcell.2021.700482] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/21/2021] [Indexed: 12/12/2022] Open
Abstract
The genomic profile of animal models is not completely matched with the genomic profile of humans, and 2D cultures do not represent the cellular heterogeneity and tissue architecture found in tissues of their origin. Derived from 3D culture systems, organoids establish a crucial bridge between 2D cell cultures and in vivo animal models. Organoids have wide and promising applications in developmental research, disease modeling, drug screening, precision therapy, and regenerative medicine. However, current organoids represent only single or partial components of a tissue, which lack blood vessels, native microenvironment, communication with near tissues, and a continuous dorsal-ventral axis within 3D culture systems. Although efforts have been made to solve these problems, unfortunately, there is no ideal method. Teratoma, which has been frequently studied in pathological conditions, was recently discovered as a new in vivo model for developmental studies. In contrast to organoids, teratomas have vascularized 3D structures and regions of complex tissue-like organization. Studies have demonstrated that teratomas can be used to mimic multilineage human development, enrich specific somatic progenitor/stem cells, and even generate brain organoids. These results provide unique opportunities to promote our understanding of the vascularization and maturation of organoids. In this review, we first summarize the basic characteristics, applications, and limitations of both organoids and teratomas and further discuss the possibility that in vivo teratoma systems can be used to promote the vascularization and maturation of organoids within an in vitro 3D culture system.
Collapse
Affiliation(s)
- Hongyu Li
- Department of Ophthalmology, The Chinese People's Liberation Army General Hospital, Beijing, China
| | - Lixiong Gao
- Department of Ophthalmology, The Chinese People's Liberation Army General Hospital, Beijing, China
| | - Jinlin Du
- Department of Ophthalmology, The Chinese People's Liberation Army General Hospital, Beijing, China
| | - Tianju Ma
- Department of Ophthalmology, The Chinese People's Liberation Army General Hospital, Beijing, China
| | - Zi Ye
- Department of Ophthalmology, The Chinese People's Liberation Army General Hospital, Beijing, China
| | - Zhaohui Li
- Department of Ophthalmology, The Chinese People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
447
|
Jung O, Song MJ, Ferrer M. Operationalizing the Use of Biofabricated Tissue Models as Preclinical Screening Platforms for Drug Discovery and Development. SLAS DISCOVERY 2021; 26:1164-1176. [PMID: 34269079 DOI: 10.1177/24725552211030903] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
A wide range of complex in vitro models (CIVMs) are being developed for scientific research and preclinical drug efficacy and safety testing. The hope is that these CIVMs will mimic human physiology and pathology and predict clinical responses more accurately than the current cellular models. The integration of these CIVMs into the drug discovery and development pipeline requires rigorous scientific validation, including cellular, morphological, and functional characterization; benchmarking of clinical biomarkers; and operationalization as robust and reproducible screening platforms. It will be critical to establish the degree of physiological complexity that is needed in each CIVM to accurately reproduce native-like homeostasis and disease phenotypes, as well as clinical pharmacological responses. Choosing which CIVM to use at each stage of the drug discovery and development pipeline will be driven by a fit-for-purpose approach, based on the specific disease pathomechanism to model and screening throughput needed. Among the different CIVMs, biofabricated tissue equivalents are emerging as robust and versatile cellular assay platforms. Biofabrication technologies, including bioprinting approaches with hydrogels and biomaterials, have enabled the production of tissues with a range of physiological complexity and controlled spatial arrangements in multiwell plate platforms, which make them amenable for medium-throughput screening. However, operationalization of such 3D biofabricated models using existing automation screening platforms comes with a unique set of challenges. These challenges will be discussed in this perspective, including examples and thoughts coming from a laboratory dedicated to designing and developing assays for automated screening.
Collapse
Affiliation(s)
- Olive Jung
- 3D Tissue Bioprinting Laboratory (3DTBL), Division of Pre-clinical Innovation (DPI), National Center for Advancing Translational Sciences (NCATS), NIH, Rockville, MD, USA.,Biomedical Ultrasonics, Biotherapy and Biopharmaceuticals Laboratory, Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Min Jae Song
- 3D Tissue Bioprinting Laboratory (3DTBL), Division of Pre-clinical Innovation (DPI), National Center for Advancing Translational Sciences (NCATS), NIH, Rockville, MD, USA
| | - Marc Ferrer
- 3D Tissue Bioprinting Laboratory (3DTBL), Division of Pre-clinical Innovation (DPI), National Center for Advancing Translational Sciences (NCATS), NIH, Rockville, MD, USA
| |
Collapse
|
448
|
Cerebral Organoids-Challenges to Establish a Brain Prototype. Cells 2021; 10:cells10071790. [PMID: 34359959 PMCID: PMC8306666 DOI: 10.3390/cells10071790] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/06/2021] [Accepted: 07/13/2021] [Indexed: 12/21/2022] Open
Abstract
The new cellular models based on neural cells differentiated from induced pluripotent stem cells have greatly enhanced our understanding of human nervous system development. Highly efficient protocols for the differentiation of iPSCs into different types of neural cells have allowed the creation of 2D models of many neurodegenerative diseases and nervous system development. However, the 2D culture of neurons is an imperfect model of the 3D brain tissue architecture represented by many functionally active cell types. The development of protocols for the differentiation of iPSCs into 3D cerebral organoids made it possible to establish a cellular model closest to native human brain tissue. Cerebral organoids are equally suitable for modeling various CNS pathologies, testing pharmacologically active substances, and utilization in regenerative medicine. Meanwhile, this technology is still at the initial stage of development.
Collapse
|
449
|
Ao Z, Cai H, Wu Z, Song S, Karahan H, Kim B, Lu HC, Kim J, Mackie K, Guo F. Tubular human brain organoids to model microglia-mediated neuroinflammation. LAB ON A CHIP 2021; 21:2751-2762. [PMID: 34021557 PMCID: PMC8493632 DOI: 10.1039/d1lc00030f] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Human brain organoids, 3D brain tissue cultures derived from human pluripotent stem cells, hold promising potential in modeling neuroinflammation for a variety of neurological diseases. However, challenges remain in generating standardized human brain organoids that can recapitulate key physiological features of a human brain. Here, we present tubular organoid-on-a-chip devices to generate better organoids and model neuroinflammation. By employing 3D printed hollow mesh scaffolds, our device can be easily incorporated into multiwell-plates for reliable, scalable, and reproducible generation of tubular organoids. By introducing rocking flows through the tubular device channel, our device can perfuse nutrients and oxygen to minimize organoid necrosis and hypoxia, and incorporate immune cells into organoids to model neuro-immune interactions. Compared with conventional protocols, our method increased neural progenitor proliferation and reduced heterogeneity of human brain organoids. As a proof-of-concept application, we applied this method to model the microglia-mediated neuroinflammation after exposure to an opioid receptor agonist. We found isogenic microglia were activated after exposure to an opioid receptor agonist (DAMGO) and transformed back to the homeostatic status with further treatment by a cannabinoid receptor 2 (CB2) agonist (LY2828360). Importantly, the activated microglia in tubular organoids had stronger cytokine responses compared to those in 2D microglial cultures. Our tubular organoid device is simple, versatile, inexpensive, easy-to-use, and compatible with multiwell-plates, so it can be widely used in common research and clinical laboratory settings. This technology can be broadly used for basic and translational applications in inflammatory diseases including substance use disorders, neural diseases, autoimmune disorders, and infectious diseases.
Collapse
Affiliation(s)
- Zheng Ao
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN 47405, USA.
| | - Hongwei Cai
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN 47405, USA.
| | - Zhuhao Wu
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN 47405, USA.
| | - Sunghwa Song
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN 47405, USA.
| | - Hande Karahan
- Stark Neurosciences Research Institute, and Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Byungwook Kim
- Stark Neurosciences Research Institute, and Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Hui-Chen Lu
- Gill Center for Biomolecular Science, and Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN 47405, USA
| | - Jungsu Kim
- Stark Neurosciences Research Institute, and Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Ken Mackie
- Gill Center for Biomolecular Science, and Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN 47405, USA
| | - Feng Guo
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN 47405, USA.
| |
Collapse
|
450
|
Abstract
The blood-brain barrier (BBB) is one of the most selective endothelial barriers. An understanding of its cellular, morphological, and biological properties in health and disease is necessary to develop therapeutics that can be transported from blood to brain. In vivo models have provided some insight into these features and transport mechanisms adopted at the brain, yet they have failed as a robust platform for the translation of results into clinical outcomes. In this article, we provide a general overview of major BBB features and describe various models that have been designed to replicate this barrier and neurological pathologies linked with the BBB. We propose several key parameters and design characteristics that can be employed to engineer physiologically relevant models of the blood-brain interface and highlight the need for a consensus in the measurement of fundamental properties of this barrier.
Collapse
Affiliation(s)
- Cynthia Hajal
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Baptiste Le Roi
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Roger D Kamm
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Ben M Maoz
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| |
Collapse
|