1
|
Su Z, Liu B, Dai J, Han M, Lai JC, Wang S, Chen Y, Zhao Y, Zhang R, Ma H, Deng Y, Li Z. A simulated microgravity-oriented AIE probe-ECM hydrogel-integrated chip for cell culture and superoxide anion radical detection. Biosens Bioelectron 2024; 264:116656. [PMID: 39133993 DOI: 10.1016/j.bios.2024.116656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/17/2024] [Accepted: 08/08/2024] [Indexed: 09/01/2024]
Abstract
Human space activities have been continuously increasing. Astronauts experiencing spaceflight are faced with health problems caused by special space environments such as microgravity, and the investigation of cell injury is fundamental. The development of a platform capable of cell culture and injury detection is the prerequisite for the investigation. Constructing a platform suitable for special conditions in space life science research is the key issue. The ground-based investigation is an indispensable part of the research. Accordingly, a simulated microgravity (SMG)-oriented integrated chip platform capable of 3D cell culture and in situ visual detection of superoxide anion radical (O2•-) is developed. SMG can cause oxidative stress in human cells, and O2•- is one of the signaling molecules. Thus, a O2•--responsive aggregation-induced emission (AIE) probe is designed, which shows high selectivity and sensitivity to O2•-. Moreover, the probe exhibits abilities of long-term and wash-free staining to cells due to the AIE behavior, which is precious for space cell imaging. Meanwhile, a chip with a high-aspect-ratio chamber for adequate medium storage for the lack of the perfusion system during the SMG experiment and a cell culture chamber which can integrate the extracellular matrix (ECM) hydrogel for the bioinspired 3D cell culture is fabricated. In addition, a porous membrane is introduced between the chambers to prevent the hydrogel from separating during the SMG experiment. The afforded AIE probe-ECM hydrogel-integrated chip can achieve 3D culturing of U87-MG cells and in situ fluorescent detection of endogenous O2•- in the cells after long-term staining under SMG. The chip provides a powerful and potential platform for ground-based investigation in space life science and biomedical research.
Collapse
Affiliation(s)
- Zhaoqing Su
- Institute of Engineering Medicine, School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China; Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing, 100081, China
| | - Beiqin Liu
- Institute of Engineering Medicine, School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China; Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing, 100081, China
| | - Jing Dai
- Institute of Engineering Medicine, School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China; Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing, 100081, China
| | - Min Han
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, 250014, China
| | - Jian-Cheng Lai
- Tachin Technology Co., Ltd., Beijing, 100094, China; Beijing Institute of Collaborative Innovation, Beijing, 100094, China
| | - Shuyue Wang
- Institute of Engineering Medicine, School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China; Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing, 100081, China
| | - Yu Chen
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing, 100081, China; Aerospace Medical Center, Aerospace Center Hospital, Beijing, 100049, China
| | - Yimeng Zhao
- Institute of Engineering Medicine, School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China; Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing, 100081, China
| | - Ruoyao Zhang
- Institute of Engineering Medicine, School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China; Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing, 100081, China
| | - Hong Ma
- Institute of Engineering Medicine, School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China; Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing, 100081, China.
| | - Yulin Deng
- Institute of Engineering Medicine, School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China; Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing, 100081, China.
| | - Zhao Li
- Institute of Engineering Medicine, School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China; Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing, 100081, China.
| |
Collapse
|
2
|
Li K, Gu L, Cai H, Lu HC, Mackie K, Guo F. Human brain organoids for understanding substance use disorders. Drug Metab Pharmacokinet 2024:101036. [PMID: 39567282 DOI: 10.1016/j.dmpk.2024.101036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
Substance use disorders (SUDs) are complex mental health conditions involving a problematic pattern of substance use. Challenges remain in understanding its neural mechanisms, which are likely to lead to improved SUD treatments. Human brain organoids, brain-like 3D in vitro cultures derived from human stem cells, show unique potential in recapitulating the response of a developing human brain to substances. Here, we review the recent progress in understanding SUD using human brain organoid models focusing on neurodevelopmental perspectives. We first summarize the background of SUD in humans. Moreover, we introduce the development of various human brain organoid models and then discuss current progress and findings underlying the abuse of substances like nicotine, alcohol, and other addictive drugs using organoid models. Furthermore, we review efforts to develop organ chips and microphysiological systems to engineer better human brain organoids for advancing SUD studies. Lastly, we conclude by elaborating on the current challenges and future directions of SUD studies using human brain organoids.
Collapse
Affiliation(s)
- Kangle Li
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN, 47405, USA
| | - Longjun Gu
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN, 47405, USA
| | - Hongwei Cai
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN, 47405, USA
| | - Hui-Chen Lu
- Gill Center for Biomolecular Science, Department of Psychological and Brain Sciences, Indiana University Bloomington, IN, 47405, USA
| | - Ken Mackie
- Gill Center for Biomolecular Science, 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
|
3
|
Cui X, Li X, Zheng H, Su Y, Zhang S, Li M, Hao X, Zhang S, Hu Z, Xia Z, Shi C, Xu Y, Mao C. Human midbrain organoids: a powerful tool for advanced Parkinson's disease modeling and therapy exploration. NPJ Parkinsons Dis 2024; 10:189. [PMID: 39428415 PMCID: PMC11491477 DOI: 10.1038/s41531-024-00799-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 10/02/2024] [Indexed: 10/22/2024] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder marked by the loss of dopaminergic neurons in the substantia nigra. Despite progress, the pathogenesis remains unclear. Human midbrain organoids (hMLOs) have emerged as a promising model for studying PD, drug screening, and potential treatments. This review discusses the development of hMLOs, their application in PD research, and current challenges in organoid construction, highlighting possible optimization strategies.
Collapse
Affiliation(s)
- Xin Cui
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Xinwei Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Huimin Zheng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Yun Su
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Shuyu Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Neuro-Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mengjie Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Xiaoyan Hao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Shuo Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Zhengwei Hu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Zongping Xia
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Clinical Systems Biology Laboratories, Zhengzhou University, Zhengzhou, China
| | - Changhe Shi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China.
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China.
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, China.
| | - Chengyuan Mao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China.
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
4
|
Tian C, Cai H, Ao Z, Gu L, Li X, Niu VC, Bondesson M, Gu M, Mackie K, Guo F. Engineering human midbrain organoid microphysiological systems to model prenatal PFOS exposure. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 947:174478. [PMID: 38964381 PMCID: PMC11404128 DOI: 10.1016/j.scitotenv.2024.174478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/30/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024]
Abstract
Perfluorooctane sulfonate (PFOS), a class of synthetic chemicals detected in various environmental compartments, has been associated with dysfunctions of the human central nervous system (CNS). However, the underlying neurotoxicology of PFOS exposure is largely understudied due to the lack of relevant human models. Here, we report bioengineered human midbrain organoid microphysiological systems (hMO-MPSs) to recapitulate the response of a fetal human brain to multiple concurrent PFOS exposure conditions. Each hMO-MPS consists of an hMO on a fully 3D printed holder device with a perfusable organoid adhesion layer for enhancing air-liquid interface culturing. Leveraging the unique, simply-fabricated holder devices, hMO-MPSs are scalable, easy to use, and compatible with conventional well-plates, and allow easy transfer onto a multiple-electrode array (MEA) system for plug-and-play measurement of neural activity. Interestingly, the neural activity of hMO-MPSs initially increased and subsequently decreased by exposure to a concentration range of 0, 30, 100, to 300 μM of PFOS. Furthermore, PFOS exposure impaired neural development and promoted neuroinflammation in the engineered hMO-MPSs. Along with PFOS, our platform is broadly applicable for studies toxicology of various other environmental pollutants.
Collapse
Affiliation(s)
- Chunhui Tian
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN 47405, United States
| | - Hongwei Cai
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN 47405, United States
| | - Zheng Ao
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN 47405, United States
| | - Longjun Gu
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN 47405, United States
| | - Xiang Li
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN 47405, United States
| | - Vivian C Niu
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN 47405, United States; Bloomington High School South, Bloomington, IN 47401, United States
| | - Maria Bondesson
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN 47405, United States
| | - Mingxia Gu
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Pulmonary Biology, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, OH 45229, Cincinnati, United States; University of Cincinnati School of Medicine, OH 45229, Cincinnati, United States
| | - Ken Mackie
- Gill Center for Biomolecular Science, Department of Psychological and Brain Sciences, Indiana University Bloomington, IN 47405, United States
| | - Feng Guo
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN 47405, United States.
| |
Collapse
|
5
|
Serafini CE, Charles S, Casteleiro Costa P, Niu W, Cheng B, Wen Z, Lu H, Robles FE. Non-invasive label-free imaging analysis pipeline for in situ characterization of 3D brain organoids. Sci Rep 2024; 14:22331. [PMID: 39333572 PMCID: PMC11436713 DOI: 10.1038/s41598-024-72038-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 09/03/2024] [Indexed: 09/29/2024] Open
Abstract
Brain organoids provide a unique opportunity to model organ development in a system similar to human organogenesis in vivo. Brain organoids thus hold great promise for drug screening and disease modeling. Conventional approaches to organoid characterization predominantly rely on molecular analysis methods, which are expensive, time-consuming, labor-intensive, and involve the destruction of the valuable three-dimensional (3D) architecture of the organoids. This reliance on end-point assays makes it challenging to assess cellular and subcellular events occurring during organoid development in their 3D context. As a result, the long developmental processes are not monitored nor assessed. The ability to perform non-invasive assays is critical for longitudinally assessing features of organoid development during culture. In this paper, we demonstrate a label-free high-content imaging approach for observing changes in organoid morphology and structural changes occurring at the cellular and subcellular level. Enabled by microfluidic-based culture of 3D cell systems and a novel 3D quantitative phase imaging method, we demonstrate the ability to perform non-destructive high-resolution quantitative image analysis of the organoid. The highlighted results demonstrated in this paper provide a new approach to performing live, non-destructive monitoring of organoid systems during culture.
Collapse
Affiliation(s)
- Caroline E Serafini
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30318, USA
| | - Seleipiri Charles
- Georgia Institute of Technology, Interdisciplinary Program in Bioengineering, Atlanta, GA, 30332, USA
| | - Paloma Casteleiro Costa
- Georgia Institute of Technology, School of Electrical and Computer Engineering, Atlanta, GA, 30332, USA
| | - Weibo Niu
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia, 30322, USA
| | - Brian Cheng
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30318, USA
| | - Zhexing Wen
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia, 30322, USA
- Departments of Cell Biology and Neurology, Emory University School of Medicine, Atlanta, Georgia, 30322, USA
| | - Hang Lu
- Georgia Institute of Technology, Interdisciplinary Program in Bioengineering, Atlanta, GA, 30332, USA
- Georgia Institute of Technology, School of Chemical and Biomolecular Engineering, Atlanta, Georgia, 30332, USA
| | - Francisco E Robles
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30318, USA.
- Georgia Institute of Technology, Interdisciplinary Program in Bioengineering, Atlanta, GA, 30332, USA.
- Georgia Institute of Technology, School of Electrical and Computer Engineering, Atlanta, GA, 30332, USA.
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30318, USA.
| |
Collapse
|
6
|
Gu L, Cai H, Chen L, Gu M, Tchieu J, Guo F. Functional Neural Networks in Human Brain Organoids. BME FRONTIERS 2024; 5:0065. [PMID: 39314749 PMCID: PMC11418062 DOI: 10.34133/bmef.0065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/12/2024] [Accepted: 09/01/2024] [Indexed: 09/25/2024] Open
Abstract
Human brain organoids are 3-dimensional brain-like tissues derived from human pluripotent stem cells and hold promising potential for modeling neurological, psychiatric, and developmental disorders. While the molecular and cellular aspects of human brain organoids have been intensively studied, their functional properties such as organoid neural networks (ONNs) are largely understudied. Here, we summarize recent research advances in understanding, characterization, and application of functional ONNs in human brain organoids. We first discuss the formation of ONNs and follow up with characterization strategies including microelectrode array (MEA) technology and calcium imaging. Moreover, we highlight recent studies utilizing ONNs to investigate neurological diseases such as Rett syndrome and Alzheimer's disease. Finally, we provide our perspectives on the future challenges and opportunities for using ONNs in basic research and translational applications.
Collapse
Affiliation(s)
- Longjun Gu
- Department of Intelligent Systems Engineering,
Indiana University Bloomington, Bloomington, IN 47405, USA
| | - Hongwei Cai
- Department of Intelligent Systems Engineering,
Indiana University Bloomington, Bloomington, IN 47405, USA
| | - Lei Chen
- Department of Intelligent Systems Engineering,
Indiana University Bloomington, Bloomington, IN 47405, USA
| | - Mingxia Gu
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Pulmonary Biology, Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- University of Cincinnati School of Medicine, Cincinnati, OH 45229, USA
| | - Jason Tchieu
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Pulmonary Biology, Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- University of Cincinnati School of Medicine, Cincinnati, OH 45229, USA
| | - Feng Guo
- Department of Intelligent Systems Engineering,
Indiana University Bloomington, Bloomington, IN 47405, USA
| |
Collapse
|
7
|
Aili Y, Maimaitiming N, Wang Z, Wang Y. Brain organoids: A new tool for modelling of neurodevelopmental disorders. J Cell Mol Med 2024; 28:e18560. [PMID: 39258535 PMCID: PMC11388061 DOI: 10.1111/jcmm.18560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/07/2024] [Accepted: 07/09/2024] [Indexed: 09/12/2024] Open
Abstract
Neurodevelopmental disorders are mostly studied using mice as models. However, the mouse brain lacks similar cell types and structures as those of the human brain. In recent years, emergence of three-dimensional brain organoids derived from human embryonic stem cells or induced pluripotent stem cells allows for controlled monitoring and evaluation of early neurodevelopmental processes and has opened a window for studying various aspects of human brain development. However, such organoids lack original anatomical structure of the brain during maturation, and neurodevelopmental maturation processes that rely on unique cellular interactions and neural network connections are limited. Consequently, organoids are difficult to be used extensively and effectively while modelling later stages of human brain development and disease progression. To address this problem, several methods and technologies have emerged that aim to enhance the sophisticated regulation of brain organoids developmental processes through bioengineering approaches, which may alleviate some of the current limitations. This review discusses recent advances and application areas of human brain organoid culture methods, aiming to generalize optimization strategies for organoid systems, improve the ability to mimic human brain development, and enhance the application value of organoids.
Collapse
Affiliation(s)
- Yirizhati Aili
- Department of NeurosurgeryThe First Affiliated Hospital of Xinjiang Medical UniversityXinjiangPeople's Republic of China
- Key Laboratory of Precision Diagnosis and Clinical Transformation of Nervous System TumorsXinjiang Medical UniversityXinjiangPeople's Republic of China
| | | | - Zengliang Wang
- Department of NeurosurgeryThe First Affiliated Hospital of Xinjiang Medical UniversityXinjiangPeople's Republic of China
- Key Laboratory of Precision Diagnosis and Clinical Transformation of Nervous System TumorsXinjiang Medical UniversityXinjiangPeople's Republic of China
| | - Yongxin Wang
- Department of NeurosurgeryThe First Affiliated Hospital of Xinjiang Medical UniversityXinjiangPeople's Republic of China
- Key Laboratory of Precision Diagnosis and Clinical Transformation of Nervous System TumorsXinjiang Medical UniversityXinjiangPeople's Republic of China
| |
Collapse
|
8
|
Smirnova L, Hartung T. The Promise and Potential of Brain Organoids. Adv Healthc Mater 2024; 13:e2302745. [PMID: 38252094 DOI: 10.1002/adhm.202302745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 12/22/2023] [Indexed: 01/23/2024]
Abstract
Brain organoids are 3D in vitro culture systems derived from human pluripotent stem cells that self-organize to model features of the (developing) human brain. This review examines the techniques behind organoid generation, their current and potential applications, and future directions for the field. Brain organoids possess complex architecture containing various neural cell types, synapses, and myelination. They have been utilized for toxicology testing, disease modeling, infection studies, personalized medicine, and gene-environment interaction studies. An emerging concept termed Organoid Intelligence (OI) combines organoids with artificial intelligence systems to generate learning and memory, with the goals of modeling cognition and enabling biological computing applications. Brain organoids allow neuroscience studies not previously achievable with traditional techniques, and have the potential to transform disease modeling, drug development, and the understanding of human brain development and disorders. The aspirational vision of OI parallels the origins of artificial intelligence, and efforts are underway to map a roadmap toward its realization. In summary, brain organoids constitute a disruptive technology that is rapidly advancing and gaining traction across multiple disciplines.
Collapse
Affiliation(s)
- Lena Smirnova
- Center for Alternatives to Animal Testing (CAAT), Bloomberg School of Public Health and Whiting School of Engineering, Johns Hopkins University, 615 N Wolfe St, Baltimore, MD, 21205, USA
| | - Thomas Hartung
- Center for Alternatives to Animal Testing (CAAT), Bloomberg School of Public Health and Whiting School of Engineering, Johns Hopkins University, 615 N Wolfe St, Baltimore, MD, 21205, USA
- CAAT-Europe, University of Konstanz, Universitätsstr. 10, 78464, Konstanz, BW, Germany
| |
Collapse
|
9
|
Karatayev O, Collier AD, Targoff SR, Leibowitz SF. Neurological Disorders Induced by Drug Use: Effects of Adolescent and Embryonic Drug Exposure on Behavioral Neurodevelopment. Int J Mol Sci 2024; 25:8341. [PMID: 39125913 PMCID: PMC11313660 DOI: 10.3390/ijms25158341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/10/2024] [Accepted: 07/17/2024] [Indexed: 08/12/2024] Open
Abstract
Clinical studies demonstrate that the risk of developing neurological disorders is increased by overconsumption of the commonly used drugs, alcohol, nicotine and cannabis. These drug-induced neurological disorders, which include substance use disorder (SUD) and its co-occurring emotional conditions such as anxiety and depression, are observed not only in adults but also with drug use during adolescence and after prenatal exposure to these drugs, and they are accompanied by long-lasting disturbances in brain development. This report provides overviews of clinical and preclinical studies, which confirm these adverse effects in adolescents and the offspring prenatally exposed to the drugs and include a more in-depth description of specific neuronal systems, their neurocircuitry and molecular mechanisms, affected by drug exposure and of specific techniques used to determine if these effects in the brain are causally related to the behavioral disturbances. With analysis of further studies, this review then addresses four specific questions that are important for fully understanding the impact that drug use in young individuals can have on future pregnancies and their offspring. Evidence demonstrates that the adverse effects on their brain and behavior can occur: (1) at low doses with short periods of drug exposure during pregnancy; (2) after pre-conception drug use by both females and males; (3) in subsequent generations following the initial drug exposure; and (4) in a sex-dependent manner, with drug use producing a greater risk in females than males of developing SUDs with emotional conditions and female offspring after prenatal drug exposure responding more adversely than male offspring. With the recent rise in drug use by adolescents and pregnant women that has occurred in association with the legalization of cannabis and increased availability of vaping tools, these conclusions from the clinical and preclinical literature are particularly alarming and underscore the urgent need to educate young women and men about the possible harmful effects of early drug use and to seek novel therapeutic strategies that might help to limit drug use in young individuals.
Collapse
Affiliation(s)
| | | | | | - Sarah F. Leibowitz
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065, USA; (O.K.); (S.R.T.)
| |
Collapse
|
10
|
Yang Y, Cui J, Kong Y, Hou Y, Ma C. Organoids: new frontiers in tumor immune microenvironment research. Front Immunol 2024; 15:1422031. [PMID: 39136020 PMCID: PMC11317300 DOI: 10.3389/fimmu.2024.1422031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 07/15/2024] [Indexed: 08/15/2024] Open
Abstract
The tumor microenvironment (TME) contains cells that regulate medication response and cancer growth in a major way. Tumor immunology research has been rejuvenated and cancer treatment has been changed by immunotherapy, a rapidly developing therapeutic approach. The growth patterns of tumor cells in vivo and the heterogeneity, complexity, and individuality of tumors produced from patients are not reflected in traditional two-dimensional tumor cell profiles. On the other hand, an in vitro three-dimensional (3D) model called the organoid model is gaining popularity. It can replicate the physiological and pathological properties of the original tissues in vivo. Tumor cells are the source of immune organoids. The TME characteristics can be preserved while preserving the variety of tumors by cultivating epithelial tumor cells with various stromal and immunological components. In addition to having genetic and physical similarities to human diseases and the ability to partially reconstruct the complex structure of tumors, these models are now widely used in research fields including cancer, developmental biology, regenerative mechanisms, drug development, disease modeling, and organ transplantation. This study reviews the function of organoids in immunotherapy and the tumor immune milieu. We also discuss current developments and suggest translational uses of tumor organoids in immuno-oncology research, immunotherapy modeling, and precision medicine.
Collapse
Affiliation(s)
- Yujia Yang
- Department of Immunology, Key Laboratory of Immune Mechanism and Intervention on Serious Disease in Hebei Province, Hebei Medical University, Immunology Department of Hebei Medical University, Shijiazhuang, China
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jinlei Cui
- Department of Immunology, Key Laboratory of Immune Mechanism and Intervention on Serious Disease in Hebei Province, Hebei Medical University, Immunology Department of Hebei Medical University, Shijiazhuang, China
| | - Yajie Kong
- Department of Immunology, Key Laboratory of Immune Mechanism and Intervention on Serious Disease in Hebei Province, Hebei Medical University, Immunology Department of Hebei Medical University, Shijiazhuang, China
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yu Hou
- Department of Immunology, Key Laboratory of Immune Mechanism and Intervention on Serious Disease in Hebei Province, Hebei Medical University, Immunology Department of Hebei Medical University, Shijiazhuang, China
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Cuiqing Ma
- Department of Immunology, Key Laboratory of Immune Mechanism and Intervention on Serious Disease in Hebei Province, Hebei Medical University, Immunology Department of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
11
|
Li K, Gu L, Cai H, Lu HC, Mackie K, Guo F. Human brain organoids for understanding substance use disorders. Drug Metab Pharmacokinet 2024; 58:101031. [PMID: 39146603 DOI: 10.1016/j.dmpk.2024.101031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/23/2024] [Accepted: 07/26/2024] [Indexed: 08/17/2024]
Abstract
Substance use disorders (SUDs) are complex mental health conditions involving a problematic pattern of substance use. Challenges remain in understanding their neural mechanisms, which are likely to lead to improved SUD treatments. Human brain organoids, brain-like 3D in vitro cultures derived from human stem cells, show unique potential in recapitulating the response of a developing human brain to substances. Here, we review the recent progress in understanding SUDs using human brain organoid models focusing on neurodevelopmental perspectives. We first summarize the background of SUDs in humans. Moreover, we introduce the development of various human brain organoid models and then discuss current progress and findings underlying the abuse of substances like nicotine, alcohol, and other addictive drugs using organoid models. Furthermore, we review efforts to develop organ chips and microphysiological systems to engineer better human brain organoids for advancing SUD studies. Lastly, we conclude by elaborating on the current challenges and future directions of SUD studies using human brain organoids.
Collapse
Affiliation(s)
- Kangle Li
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN, 47405, United States
| | - Longjun Gu
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN, 47405, United States
| | - Hongwei Cai
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN, 47405, United States
| | - Hui-Chen Lu
- Gill Center for Biomolecular Science, Department of Psychological and Brain Sciences, Indiana University Bloomington, IN, 47405, United States
| | - Ken Mackie
- Gill Center for Biomolecular Science, Department of Psychological and Brain Sciences, Indiana University Bloomington, IN, 47405, United States
| | - Feng Guo
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN, 47405, United States.
| |
Collapse
|
12
|
Charles S, Jackson-Holmes E, Sun G, Zhou Y, Siciliano B, Niu W, Han H, Nikitina A, Kemp ML, Wen Z, Lu H. Non-Invasive Quality Control of Organoid Cultures Using Mesofluidic CSTR Bioreactors and High-Content Imaging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.19.604365. [PMID: 39091761 PMCID: PMC11291105 DOI: 10.1101/2024.07.19.604365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Human brain organoids produce anatomically relevant cellular structures and recapitulate key aspects of in vivo brain function, which holds great potential to model neurological diseases and screen therapeutics. However, the long growth time of 3D systems complicates the culturing of brain organoids and results in heterogeneity across samples hampering their applications. We developed an integrated platform to enable robust and long-term culturing of 3D brain organoids. We designed a mesofluidic bioreactor device based on a reaction-diffusion scaling theory, which achieves robust media exchange for sufficient nutrient delivery in long-term culture. We integrated this device with longitudinal tracking and machine learning-based classification tools to enable non-invasive quality control of live organoids. This integrated platform allows for sample pre-selection for downstream molecular analysis. Transcriptome analyses of organoids revealed that our mesofluidic bioreactor promoted organoid development while reducing cell death. Our platform thus offers a generalizable tool to establish reproducible culture standards for 3D cellular systems for a variety of applications beyond brain organoids.
Collapse
Affiliation(s)
- Seleipiri Charles
- Interdisciplinary Program in Bioengineering, Georgia Institute of Technology, 311 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
| | - Emily Jackson-Holmes
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, 311 Ferst Drive NW Atlanta, Georgia 30332, U.S.A
| | - Gongchen Sun
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, 311 Ferst Drive NW Atlanta, Georgia 30332, U.S.A
| | - Ying Zhou
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurology, Emory University School of Medicine, 615 Michael Street, Atlanta, Georgia 30322, U.S.A
| | - Benjamin Siciliano
- Graduate Program in Molecular and Systems Pharmacology, Laney Graduate School, Emory University, 615 Michael Street, Atlanta, GA, 30322, U.S.A
| | - Weibo Niu
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurology, Emory University School of Medicine, 615 Michael Street, Atlanta, Georgia 30322, U.S.A
| | - Haejun Han
- Interdisciplinary Program in Bioengineering, Georgia Institute of Technology, 311 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
- School of Biological Sciences, Georgia Institute of Technology, 310 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
| | - Arina Nikitina
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, 311 Ferst Drive NW Atlanta, Georgia 30332, U.S.A
| | - Melissa L Kemp
- Interdisciplinary Program in Bioengineering, Georgia Institute of Technology, 311 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
| | - Zhexing Wen
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurology, Emory University School of Medicine, 615 Michael Street, Atlanta, Georgia 30322, U.S.A
| | - Hang Lu
- Interdisciplinary Program in Bioengineering, Georgia Institute of Technology, 311 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, 311 Ferst Drive NW Atlanta, Georgia 30332, U.S.A
| |
Collapse
|
13
|
Chen H, Fang S, Zhu X, Liu H. Cancer-associated fibroblasts and prostate cancer stem cells: crosstalk mechanisms and implications for disease progression. Front Cell Dev Biol 2024; 12:1412337. [PMID: 39092186 PMCID: PMC11291335 DOI: 10.3389/fcell.2024.1412337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 07/05/2024] [Indexed: 08/04/2024] Open
Abstract
The functional heterogeneity and ecological niche of prostate cancer stem cells (PCSCs), which are major drivers of prostate cancer development and treatment resistance, have attracted considerable research attention. Cancer-associated fibroblasts (CAFs), which are crucial components of the tumor microenvironment (TME), substantially affect PCSC stemness. Additionally, CAFs promote PCSC growth and survival by releasing signaling molecules and modifying the surrounding environment. Conversely, PCSCs may affect the characteristics and behavior of CAFs by producing various molecules. This crosstalk mechanism is potentially crucial for prostate cancer progression and the development of treatment resistance. Using organoids to model the TME enables an in-depth study of CAF-PCSC interactions, providing a valuable preclinical tool to accurately evaluate potential target genes and design novel treatment strategies for prostate cancer. The objective of this review is to discuss the current research on the multilevel and multitarget regulatory mechanisms underlying CAF-PCSC interactions and crosstalk, aiming to inform therapeutic approaches that address challenges in prostate cancer treatment.
Collapse
Affiliation(s)
| | | | | | - Hao Liu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
14
|
Du C, Liu J, Liu S, Xiao P, Chen Z, Chen H, Huang W, Lei Y. Bone and Joint-on-Chip Platforms: Construction Strategies and Applications. SMALL METHODS 2024:e2400436. [PMID: 38763918 DOI: 10.1002/smtd.202400436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/28/2024] [Indexed: 05/21/2024]
Abstract
Organ-on-a-chip, also known as "tissue chip," is an advanced platform based on microfluidic systems for constructing miniature organ models in vitro. They can replicate the complex physiological and pathological responses of human organs. In recent years, the development of bone and joint-on-chip platforms aims to simulate the complex physiological and pathological processes occurring in human bones and joints, including cell-cell interactions, the interplay of various biochemical factors, the effects of mechanical stimuli, and the intricate connections between multiple organs. In the future, bone and joint-on-chip platforms will integrate the advantages of multiple disciplines, bringing more possibilities for exploring disease mechanisms, drug screening, and personalized medicine. This review explores the construction and application of Organ-on-a-chip technology in bone and joint disease research, proposes a modular construction concept, and discusses the new opportunities and future challenges in the construction and application of bone and joint-on-chip platforms.
Collapse
Affiliation(s)
- Chengcheng Du
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jiacheng Liu
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Senrui Liu
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Pengcheng Xiao
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhuolin Chen
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Hong Chen
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Wei Huang
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yiting Lei
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
15
|
Jin H, Xue Z, Liu J, Ma B, Yang J, Lei L. Advancing Organoid Engineering for Tissue Regeneration and Biofunctional Reconstruction. Biomater Res 2024; 28:0016. [PMID: 38628309 PMCID: PMC11018530 DOI: 10.34133/bmr.0016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 03/04/2024] [Indexed: 04/19/2024] Open
Abstract
Tissue damage and functional abnormalities in organs have become a considerable clinical challenge. Organoids are often applied as disease models and in drug discovery and screening. Indeed, several studies have shown that organoids are an important strategy for achieving tissue repair and biofunction reconstruction. In contrast to established stem cell therapies, organoids have high clinical relevance. However, conventional approaches have limited the application of organoids in clinical regenerative medicine. Engineered organoids might have the capacity to overcome these challenges. Bioengineering-a multidisciplinary field that applies engineering principles to biomedicine-has bridged the gap between engineering and medicine to promote human health. More specifically, bioengineering principles have been applied to organoids to accelerate their clinical translation. In this review, beginning with the basic concepts of organoids, we describe strategies for cultivating engineered organoids and discuss the multiple engineering modes to create conditions for breakthroughs in organoid research. Subsequently, studies on the application of engineered organoids in biofunction reconstruction and tissue repair are presented. Finally, we highlight the limitations and challenges hindering the utilization of engineered organoids in clinical applications. Future research will focus on cultivating engineered organoids using advanced bioengineering tools for personalized tissue repair and biofunction reconstruction.
Collapse
Affiliation(s)
- Hairong Jin
- Institute of Translational Medicine,
Zhejiang Shuren University, Hangzhou 310015, China
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
- Ningxia Medical University, Ningxia 750004, China
| | - Zengqi Xue
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Jinnv Liu
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Binbin Ma
- Department of Biology,
The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Jianfeng Yang
- Institute of Translational Medicine,
Zhejiang Shuren University, Hangzhou 310015, China
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Lanjie Lei
- Institute of Translational Medicine,
Zhejiang Shuren University, Hangzhou 310015, China
| |
Collapse
|
16
|
Amartumur S, Nguyen H, Huynh T, Kim TS, Woo RS, Oh E, Kim KK, Lee LP, Heo C. Neuropathogenesis-on-chips for neurodegenerative diseases. Nat Commun 2024; 15:2219. [PMID: 38472255 PMCID: PMC10933492 DOI: 10.1038/s41467-024-46554-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Developing diagnostics and treatments for neurodegenerative diseases (NDs) is challenging due to multifactorial pathogenesis that progresses gradually. Advanced in vitro systems that recapitulate patient-like pathophysiology are emerging as alternatives to conventional animal-based models. In this review, we explore the interconnected pathogenic features of different types of ND, discuss the general strategy to modelling NDs using a microfluidic chip, and introduce the organoid-on-a-chip as the next advanced relevant model. Lastly, we overview how these models are being applied in academic and industrial drug development. The integration of microfluidic chips, stem cells, and biotechnological devices promises to provide valuable insights for biomedical research and developing diagnostic and therapeutic solutions for NDs.
Collapse
Affiliation(s)
- Sarnai Amartumur
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Huong Nguyen
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Thuy Huynh
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Testaverde S Kim
- Center for Integrated Nanostructure Physics (CINAP), Institute for Basic Science (IBS), Suwon, 16419, Korea
| | - Ran-Sook Woo
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon, 34824, Korea
| | - Eungseok Oh
- Department of Neurology, Chungnam National University Hospital, Daejeon, 35015, Korea
| | - Kyeong Kyu Kim
- Department of Precision Medicine, Graduate School of Basic Medical Science (GSBMS), Institute for Anti-microbial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Korea
| | - Luke P Lee
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea.
- Harvard Medical School, Division of Engineering in Medicine and Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA.
- Department of Bioengineering, Department of Electrical Engineering and Computer Science, University of California, Berkeley, CA, 94720, USA.
| | - Chaejeong Heo
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea.
- Center for Integrated Nanostructure Physics (CINAP), Institute for Basic Science (IBS), Suwon, 16419, Korea.
| |
Collapse
|
17
|
Wu Z, Cai H, Tian C, Ao Z, Jiang L, Guo F. Exploiting Sound for Emerging Applications of Extracellular Vesicles. NANO RESEARCH 2024; 17:462-475. [PMID: 38712329 PMCID: PMC11073796 DOI: 10.1007/s12274-023-5840-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 05/08/2024]
Abstract
Extracellular vesicles are nano- to microscale, membrane-bound particles released by cells into extracellular space, and act as carriers of biomarkers and therapeutics, holding promising potential in translational medicine. However, the challenges remain in handling and detecting extracellular vesicles for disease diagnosis as well as exploring their therapeutic capability for disease treatment. Here, we review the recent engineering and technology advances by leveraging the power of sound waves to address the challenges in diagnostic and therapeutic applications of extracellular vesicles and biomimetic nanovesicles. We first introduce the fundamental principles of sound waves for understanding different acoustic-assisted extracellular vesicle technologies. We discuss the acoustic-assisted diagnostic methods including the purification, manipulation, biosensing, and bioimaging of extracellular vesicles. Then, we summarize the recent advances in acoustically enhanced therapeutics using extracellular vesicles and biomimetic nanovesicles. Finally, we provide perspectives into current challenges and future clinical applications of the promising extracellular vesicles and biomimetic nanovesicles powered by sound.
Collapse
Affiliation(s)
- Zhuhao Wu
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN 47405, United States
| | - Hongwei Cai
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN 47405, United States
| | - Chunhui Tian
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN 47405, United States
| | - Zheng Ao
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN 47405, United States
| | - Lei Jiang
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN 47405, United States
| | - Feng Guo
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN 47405, United States
| |
Collapse
|
18
|
Saglam-Metiner P, Yildirim E, Dincer C, Basak O, Yesil-Celiktas O. Humanized brain organoids-on-chip integrated with sensors for screening neuronal activity and neurotoxicity. Mikrochim Acta 2024; 191:71. [PMID: 38168828 DOI: 10.1007/s00604-023-06165-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/21/2023] [Indexed: 01/05/2024]
Abstract
The complex structure and function of the human central nervous system that develops from the neural tube made in vitro modeling quite challenging until the discovery of brain organoids. Human-induced pluripotent stem cells-derived brain organoids offer recapitulation of the features of early human neurodevelopment in vitro, including the generation, proliferation, and differentiation into mature neurons and micro-macroglial cells, as well as the complex interactions among these diverse cell types of the developing brain. Recent advancements in brain organoids, microfluidic systems, real-time sensing technologies, and their cutting-edge integrated use provide excellent models and tools for emulation of fundamental neurodevelopmental processes, the pathology of neurological disorders, personalized transplantation therapy, and high-throughput neurotoxicity testing by bridging the gap between two-dimensional models and the complex three-dimensional environment in vivo. In this review, we summarize how bioengineering approaches are applied to mitigate the limitations of brain organoids for biomedical and clinical research. We further provide an extensive overview and future perspectives of the humanized brain organoids-on-chip platforms with integrated sensors toward brain organoid intelligence and biocomputing studies. Such approaches might pave the way for increasing approvable clinical applications by solving their current limitations.
Collapse
Affiliation(s)
- Pelin Saglam-Metiner
- Department of Bioengineering, Faculty of Engineering, Ege University, Izmir, Turkey
- Department of Translational Neuroscience, Division of Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Ender Yildirim
- Department of Mechanical Engineering, Middle East Technical University, Ankara, Turkey
- ODTÜ MEMS Center, Ankara, Turkey
| | - Can Dincer
- Department of Microsystems Engineering (IMTEK), University of Freiburg, Freiburg, Germany
- FIT Freiburg Center for Interactive Materials and Bioinspired Technologies, University of Freiburg, Freiburg, Germany
| | - Onur Basak
- Department of Translational Neuroscience, Division of Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Ozlem Yesil-Celiktas
- Department of Bioengineering, Faculty of Engineering, Ege University, Izmir, Turkey.
| |
Collapse
|
19
|
Wang Z, Zhang Y, Li Z, Wang H, Li N, Deng Y. Microfluidic Brain-on-a-Chip: From Key Technology to System Integration and Application. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2304427. [PMID: 37653590 DOI: 10.1002/smll.202304427] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/02/2023] [Indexed: 09/02/2023]
Abstract
As an ideal in vitro model, brain-on-chip (BoC) is an important tool to comprehensively elucidate brain characteristics. However, the in vitro model for the definition scope of BoC has not been universally recognized. In this review, BoC is divided into brain cells-on-a- chip, brain slices-on-a-chip, and brain organoids-on-a-chip according to the type of culture on the chip. Although these three microfluidic BoCs are constructed in different ways, they all use microfluidic chips as carrier tools. This method can better meet the needs of maintaining high culture activity on a chip for a long time. Moreover, BoC has successfully integrated cell biology, the biological material platform technology of microenvironment on a chip, manufacturing technology, online detection technology on a chip, and so on, enabling the chip to present structural diversity and high compatibility to meet different experimental needs and expand the scope of applications. Here, the relevant core technologies, challenges, and future development trends of BoC are summarized.
Collapse
Affiliation(s)
- Zhaohe Wang
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China
| | - Yongqian Zhang
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China
| | - Zhe Li
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China
| | - Hao Wang
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China
| | - Nuomin Li
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China
| | - Yulin Deng
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China
| |
Collapse
|
20
|
Rezaei B, Giacomoni J, Nilsson F, Sozzi E, Fiorenzano A, Parmar M, Keller SS, Kajtez J. Modular 3D printed platform for fluidically connected human brain organoid culture. Biofabrication 2023; 16:015014. [PMID: 37956452 DOI: 10.1088/1758-5090/ad0c2c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 11/13/2023] [Indexed: 11/15/2023]
Abstract
Brain organoid technology has transformed both basic and applied biomedical research and paved the way for novel insights into developmental processes and disease states of the human brain. While the use of brain organoids has been rapidly growing in the past decade, the accompanying bioengineering and biofabrication solutions have remained scarce. As a result, most brain organoid protocols still rely on commercially available tools and culturing platforms that had previously been established for different purposes, thus entailing suboptimal culturing conditions and excessive use of plasticware. To address these issues, we developed a 3D printing pipeline for the fabrication of tailor-made culturing platforms for fluidically connected but spatially separated brain organoid array culture. This all-in-one platform allows all culturing steps-from cellular aggregation, spheroid growth, hydrogel embedding, and organoid maturation-to be performed in a single well plate without the need for organoid manipulation or transfer. Importantly, the approach relies on accessible materials and widely available 3D printing equipment. Furthermore, the developed design principles are modular and highly customizable. As such, we believe that the presented technology can be easily adapted by other research groups and fuel further development of culturing tools and platforms for brain organoids and other 3D cellular systems.
Collapse
Affiliation(s)
- Babak Rezaei
- National Centre for Nano Fabrication and Characterization (DTU Nanolab), Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Jessica Giacomoni
- Department of Experimental Medical Science, Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Fredrik Nilsson
- Department of Experimental Medical Science, Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Edoardo Sozzi
- Department of Experimental Medical Science, Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Alessandro Fiorenzano
- Department of Experimental Medical Science, Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Lund University, Lund, Sweden
- Stem Cell Fate Laboratory, Institute of Genetics and Biophysics"A.BuzzatiTraverso", CNR, Naples, Italy
| | - Malin Parmar
- Department of Experimental Medical Science, Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Stephan S Keller
- National Centre for Nano Fabrication and Characterization (DTU Nanolab), Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Janko Kajtez
- Department of Experimental Medical Science, Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Lund University, Lund, Sweden
| |
Collapse
|
21
|
Abdulla A, Chen S, Chen Z, Wang Y, Yan H, Chen R, Ahmad KZ, Liu K, Yan C, He J, Jiang L, Ding X. Three-dimensional microfluidics with dynamic fluidic perturbation promotes viability and uniformity of human cerebral organoids. Biosens Bioelectron 2023; 240:115635. [PMID: 37651948 DOI: 10.1016/j.bios.2023.115635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 08/14/2023] [Accepted: 08/23/2023] [Indexed: 09/02/2023]
Abstract
Human cerebral organoids (COs), generated from stem cells, are emerging animal alternatives for understanding brain development and neurodegeneration diseases. Long-term growth of COs is currently hindered by the limitation of efficient oxygen infiltration and continuous nutrient supply, leading to general inner hypoxia and cell death at the core region of the organoids. Here, we developed a three-dimensional (3D) microfluidic platform with dynamic fluidic perturbation and oxygen supply. We demonstrated COs cultured in the 3D microfluidic system grew continuously for over 50 days without cell death at the core region. Increased cell proliferation and enhanced cell differentiation were also observed and verified with immunofluorescence staining, proteomics and metabolomics. Time-lapse proteomics from 7 consecutive acquisitions between day 4 and day 30 identified 546 proteins differently expressed accompanying COs growth, which were mainly relevant to nervous system development, in utero embryonic development, brain development and neuron migration. Our 3D microfluidic platform provides potential utility for culturing high-homogeneous human organoids.
Collapse
Affiliation(s)
- Aynur Abdulla
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Shujin Chen
- Ministry of Education, Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhecong Chen
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yukun Wang
- School of Engineering and Design, Technical University of Munich, Munich, Germany
| | - Haoni Yan
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Rui Chen
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Khan Zara Ahmad
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Kun Liu
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chonghuai Yan
- Ministry of Education, Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jie He
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai, China.
| | - Lai Jiang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Xianting Ding
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
22
|
Sun S, Xue X, Fu J. Modeling development using microfluidics: bridging gaps to foster fundamental and translational research. Curr Opin Genet Dev 2023; 82:102097. [PMID: 37573835 PMCID: PMC11193336 DOI: 10.1016/j.gde.2023.102097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 07/01/2023] [Accepted: 07/16/2023] [Indexed: 08/15/2023]
Abstract
In vitro stem cell-derived embryo and organ models, termed embryoids and organoids, respectively, provide promising experimental tools to study physiological and pathological processes in mammalian development and organ formation. Most of current embryoid and organoid systems are developed using conventional three-dimensional cultures that lack controls of spatiotemporal extracellular signals. Microfluidics, an established technology for quantitative controls and quantifications of dynamic chemical and physical environments, has recently been utilized for developing next-generation embryoids and organoids in a controllable and reproducible manner. In this review, we summarize recent progress in constructing microfluidics-based embryoids and organoids. Development of these models demonstrates the successful applications of microfluidics in establishing morphogen gradients, accelerating medium transport, exerting mechanical forces, facilitating tissue coculture studies, and improving assay throughput, thus supporting using microfluidics for building next-generation embryoids and organoids for fundamental and translational research.
Collapse
Affiliation(s)
- Shiyu Sun
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Xufeng Xue
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
23
|
Tsai YC, Ozaki H, Morikawa A, Shiraiwa K, Pin AP, Salem AG, Phommahasay KA, Sugita BK, Vu CH, Mamoun Hammad S, Kamei KI, Watanabe M. Brain organoid-on-a-chip to create multiple domains in forebrain organoids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.18.558278. [PMID: 37781620 PMCID: PMC10541131 DOI: 10.1101/2023.09.18.558278] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Brain organoids are three-dimensionally reconstructed brain tissue derived from pluripotent stem cells in vitro. 3D tissue cultures have opened new avenues for exploring development and disease modeling. However, some physiological conditions, including signaling gradients in 3D cultures, have not yet been easily achieved. Here, we introduce Brain Organoid-on-a-Chip platforms that generate signaling gradients that in turn enable the induction of topographic forebrain organoids. This creates a more continuous spectrum of brain regions and provides a more complete mimic of the human brain for evaluating neurodevelopment and disease in unprecedented detail.
Collapse
|
24
|
Rabeling A, Goolam M. Cerebral organoids as an in vitro model to study autism spectrum disorders. Gene Ther 2023; 30:659-669. [PMID: 35790793 DOI: 10.1038/s41434-022-00356-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 06/01/2022] [Accepted: 06/23/2022] [Indexed: 11/09/2022]
Abstract
Autism spectrum disorders (ASDs) are a set of disorders characterised by social and communication deficits caused by numerous genetic lesions affecting brain development. Progress in ASD research has been hampered by the lack of appropriate models, as both 2D cell culture as well as animal models cannot fully recapitulate the developing human brain or the pathogenesis of ASD. Recently, cerebral organoids have been developed to provide a more accurate, 3D in vitro model of human brain development. Cerebral organoids have been shown to recapitulate the foetal brain gene expression profile, transcriptome, epigenome, as well as disease dynamics of both idiopathic and syndromic ASDs. They are thus an excellent tool to investigate development of foetal stage ASDs, as well as interventions that can reverse or rescue the altered phenotypes observed. In this review, we discuss the development of cerebral organoids, their recent applications in the study of both syndromic and idiopathic ASDs, their use as an ASD drug development platform, as well as limitations of their use in ASD research.
Collapse
Affiliation(s)
- Alexa Rabeling
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa
| | - Mubeen Goolam
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa.
- UCT Neuroscience Institute, Cape Town, South Africa.
| |
Collapse
|
25
|
Gimondi S, Ferreira H, Reis RL, Neves NM. Microfluidic Devices: A Tool for Nanoparticle Synthesis and Performance Evaluation. ACS NANO 2023; 17:14205-14228. [PMID: 37498731 PMCID: PMC10416572 DOI: 10.1021/acsnano.3c01117] [Citation(s) in RCA: 45] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 07/24/2023] [Indexed: 07/29/2023]
Abstract
The use of nanoparticles (NPs) in nanomedicine holds great promise for the treatment of diseases for which conventional therapies present serious limitations. Additionally, NPs can drastically improve early diagnosis and follow-up of many disorders. However, to harness their full capabilities, they must be precisely designed, produced, and tested in relevant models. Microfluidic systems can simulate dynamic fluid flows, gradients, specific microenvironments, and multiorgan complexes, providing an efficient and cost-effective approach for both NPs synthesis and screening. Microfluidic technologies allow for the synthesis of NPs under controlled conditions, enhancing batch-to-batch reproducibility. Moreover, due to the versatility of microfluidic devices, it is possible to generate and customize endless platforms for rapid and efficient in vitro and in vivo screening of NPs' performance. Indeed, microfluidic devices show great potential as advanced systems for small organism manipulation and immobilization. In this review, first we summarize the major microfluidic platforms that allow for controlled NPs synthesis. Next, we will discuss the most innovative microfluidic platforms that enable mimicking in vitro environments as well as give insights into organism-on-a-chip and their promising application for NPs screening. We conclude this review with a critical assessment of the current challenges and possible future directions of microfluidic systems in NPs synthesis and screening to impact the field of nanomedicine.
Collapse
Affiliation(s)
- Sara Gimondi
- 3B’s
Research Group, I3Bs − Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters
of the European Institute of Excellence on Tissue Engineering and
Regenerative Medicine, AvePark, Parque
de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B’s−PT
Government Associate Laboratory, 4805-017 Braga, Guimarães, Portugal
| | - Helena Ferreira
- 3B’s
Research Group, I3Bs − Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters
of the European Institute of Excellence on Tissue Engineering and
Regenerative Medicine, AvePark, Parque
de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B’s−PT
Government Associate Laboratory, 4805-017 Braga, Guimarães, Portugal
| | - Rui L. Reis
- 3B’s
Research Group, I3Bs − Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters
of the European Institute of Excellence on Tissue Engineering and
Regenerative Medicine, AvePark, Parque
de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B’s−PT
Government Associate Laboratory, 4805-017 Braga, Guimarães, Portugal
| | - Nuno M. Neves
- 3B’s
Research Group, I3Bs − Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters
of the European Institute of Excellence on Tissue Engineering and
Regenerative Medicine, AvePark, Parque
de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B’s−PT
Government Associate Laboratory, 4805-017 Braga, Guimarães, Portugal
| |
Collapse
|
26
|
Saorin G, Caligiuri I, Rizzolio F. Microfluidic organoids-on-a-chip: The future of human models. Semin Cell Dev Biol 2023; 144:41-54. [PMID: 36241560 DOI: 10.1016/j.semcdb.2022.10.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/06/2022] [Accepted: 10/06/2022] [Indexed: 11/06/2022]
Abstract
Microfluidics opened the possibility to model the physiological environment by controlling fluids flows, and therefore nutrients supply. It allows to integrate external stimuli such as electricals or mechanicals and in situ monitoring important parameters such as pH, oxygen and metabolite concentrations. Organoids are self-organized 3D organ-like clusters, which allow to closely model original organ functionalities. Applying microfluidics to organoids allows to generate powerful human models for studying organ development, diseases, and drug testing. In this review, after a brief introduction on microfluidics, organoids and organoids-on-a-chip are described by organs (brain, heart, gastrointestinal tract, liver, pancreas) highlighting the microfluidic approaches since this point of view was overlooked in previously published reviews. Indeed, the review aims to discuss from a different point of view, primary microfluidics, the available literature on organoids-on-a-chip, standing out from the published literature by focusing on each specific organ.
Collapse
Affiliation(s)
- Gloria Saorin
- Department of Molecular Sciences and Nanosystems, Ca' Foscari University of Venice, 30123 Venezia, Italy
| | - Isabella Caligiuri
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
| | - Flavio Rizzolio
- Department of Molecular Sciences and Nanosystems, Ca' Foscari University of Venice, 30123 Venezia, Italy; Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy.
| |
Collapse
|
27
|
Adlakha YK. Human 3D brain organoids: steering the demolecularization of brain and neurological diseases. Cell Death Discov 2023; 9:221. [PMID: 37400464 DOI: 10.1038/s41420-023-01523-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 06/19/2023] [Accepted: 06/22/2023] [Indexed: 07/05/2023] Open
Abstract
Understanding of human brain development, dysfunction and neurological diseases has remained limited and challenging due to inability to recapitulate human brain-specific features in animal models. Though the anatomy and physiology of the human brain has been understood in a remarkable way using post-mortem, pathological samples of human and animal models, however, modeling of human brain development and neurological diseases remains a challenge owing to distinct complexity of human brain. In this perspective, three-dimensional (3D) brain organoids have shown a beam of light. Tremendous growth in stem cell technologies has permitted the differentiation of pluripotent stem cells under 3D culture conditions into brain organoids, which recapitulate the unique features of human brain in many ways and also offer the detailed investigation of brain development, dysfunction and neurological diseases. Their translational value has also emerged and will benefit the society once the protocols for the upscaling of brain organoids are in place. Here, we summarize new advancements in methods for generation of more complex brain organoids including vascularized and mixed lineage tissue from PSCs. How synthetic biomaterials and microfluidic technology is boosting brain organoid development, has also been highlighted. We discuss the applications of brain organoids in studying preterm birth associated brain dysfunction; viral infections mediated neuroinflammation, neurodevelopmental and neurodegenerative diseases. We also highlight the translational value of brain organoids and current challenges that the field is experiencing.
Collapse
Affiliation(s)
- Yogita K Adlakha
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh, India.
- Maternal and Child Health Domain, Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, India.
| |
Collapse
|
28
|
Hetzel LA, Ali A, Corbo V, Hankemeier T. Microfluidics and Organoids, the Power Couple of Developmental Biology and Oncology Studies. Int J Mol Sci 2023; 24:10882. [PMID: 37446057 DOI: 10.3390/ijms241310882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Organoids are an advanced cell model that hold the key to unlocking a deeper understanding of in vivo cellular processes. This model can be used in understanding organ development, disease progression, and treatment efficacy. As the scientific world embraces the model, it must also establish the best practices for cultivating organoids and utilizing them to the greatest potential in assays. Microfluidic devices are emerging as a solution to overcome the challenges of organoids and adapt assays. Unfortunately, the various applications of organoids often depend on specific features in a device. In this review, we discuss the options and considerations for features and materials depending on the application and development of the organoid.
Collapse
Affiliation(s)
- Laura Ann Hetzel
- Leiden Academic Center for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| | - Ahmed Ali
- Leiden Academic Center for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| | - Vincenzo Corbo
- Department of Diagnostics and Public Health, ARC-Net Research Centre, University of Verona, 37134 Verona, Italy
| | - Thomas Hankemeier
- Leiden Academic Center for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| |
Collapse
|
29
|
Kilpatrick S, Irwin C, Singh KK. Human pluripotent stem cell (hPSC) and organoid models of autism: opportunities and limitations. Transl Psychiatry 2023; 13:217. [PMID: 37344450 PMCID: PMC10284884 DOI: 10.1038/s41398-023-02510-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 05/09/2023] [Accepted: 06/05/2023] [Indexed: 06/23/2023] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder caused by genetic or environmental perturbations during early development. Diagnoses are dependent on the identification of behavioral abnormalities that likely emerge well after the disorder is established, leaving critical developmental windows uncharacterized. This is further complicated by the incredible clinical and genetic heterogeneity of the disorder that is not captured in most mammalian models. In recent years, advancements in stem cell technology have created the opportunity to model ASD in a human context through the use of pluripotent stem cells (hPSCs), which can be used to generate 2D cellular models as well as 3D unguided- and region-specific neural organoids. These models produce profoundly intricate systems, capable of modeling the developing brain spatiotemporally to reproduce key developmental milestones throughout early development. When complemented with multi-omics, genome editing, and electrophysiology analysis, they can be used as a powerful tool to profile the neurobiological mechanisms underlying this complex disorder. In this review, we will explore the recent advancements in hPSC-based modeling, discuss present and future applications of the model to ASD research, and finally consider the limitations and future directions within the field to make this system more robust and broadly applicable.
Collapse
Affiliation(s)
- Savannah Kilpatrick
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Department of Biochemistry and Biomedical Science, McMaster University, Hamilton, ON, Canada
| | - Courtney Irwin
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Karun K Singh
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada.
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
30
|
Mulder LA, Depla JA, Sridhar A, Wolthers K, Pajkrt D, Vieira de Sá R. A beginner's guide on the use of brain organoids for neuroscientists: a systematic review. Stem Cell Res Ther 2023; 14:87. [PMID: 37061699 PMCID: PMC10105545 DOI: 10.1186/s13287-023-03302-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 03/27/2023] [Indexed: 04/17/2023] Open
Abstract
BACKGROUND The first human brain organoid protocol was presented in the beginning of the previous decade, and since then, the field witnessed the development of many new brain region-specific models, and subsequent protocol adaptations and modifications. The vast amount of data available on brain organoid technology may be overwhelming for scientists new to the field and consequently decrease its accessibility. Here, we aimed at providing a practical guide for new researchers in the field by systematically reviewing human brain organoid publications. METHODS Articles published between 2010 and 2020 were selected and categorised for brain organoid applications. Those describing neurodevelopmental studies or protocols for novel organoid models were further analysed for culture duration of the brain organoids, protocol comparisons of key aspects of organoid generation, and performed functional characterisation assays. We then summarised the approaches taken for different models and analysed the application of small molecules and growth factors used to achieve organoid regionalisation. Finally, we analysed articles for organoid cell type compositions, the reported time points per cell type, and for immunofluorescence markers used to characterise different cell types. RESULTS Calcium imaging and patch clamp analysis were the most frequently used neuronal activity assays in brain organoids. Neural activity was shown in all analysed models, yet network activity was age, model, and assay dependent. Induction of dorsal forebrain organoids was primarily achieved through combined (dual) SMAD and Wnt signalling inhibition. Ventral forebrain organoid induction was performed with dual SMAD and Wnt signalling inhibition, together with additional activation of the Shh pathway. Cerebral organoids and dorsal forebrain model presented the most cell types between days 35 and 60. At 84 days, dorsal forebrain organoids contain astrocytes and potentially oligodendrocytes. Immunofluorescence analysis showed cell type-specific application of non-exclusive markers for multiple cell types. CONCLUSIONS We provide an easily accessible overview of human brain organoid cultures, which may help those working with brain organoids to define their choice of model, culture time, functional assay, differentiation, and characterisation strategies.
Collapse
Affiliation(s)
- Lance A Mulder
- Department of Paediatric Infectious Diseases, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands.
- Department Medical Microbiology, OrganoVIR Labs, Amsterdam UMC Location University of Amsterdam, Amsterdam, the Netherlands.
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands.
| | - Josse A Depla
- Department of Paediatric Infectious Diseases, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Department Medical Microbiology, OrganoVIR Labs, Amsterdam UMC Location University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands
- uniQure Biopharma B.V., Amsterdam, The Netherlands
| | - Adithya Sridhar
- Department of Paediatric Infectious Diseases, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Department Medical Microbiology, OrganoVIR Labs, Amsterdam UMC Location University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands
| | - Katja Wolthers
- Department Medical Microbiology, OrganoVIR Labs, Amsterdam UMC Location University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands
| | - Dasja Pajkrt
- Department of Paediatric Infectious Diseases, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Department Medical Microbiology, OrganoVIR Labs, Amsterdam UMC Location University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands
| | - Renata Vieira de Sá
- Department Medical Microbiology, OrganoVIR Labs, Amsterdam UMC Location University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands
- uniQure Biopharma B.V., Amsterdam, The Netherlands
| |
Collapse
|
31
|
Cai H, Ao Z, Tian C, Wu Z, Kaurich C, Chen Z, Gu M, Hohmann AG, Mackie K, Guo F. Engineering human spinal microphysiological systems to model opioid-induced tolerance. Bioact Mater 2023; 22:482-490. [PMID: 36330161 PMCID: PMC9618681 DOI: 10.1016/j.bioactmat.2022.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/04/2022] [Accepted: 10/04/2022] [Indexed: 11/05/2022] Open
Abstract
pioids are commonly used for treating chronic pain. However, with continued use, they may induce tolerance and/or hyperalgesia, which limits therapeutic efficacy. The human mechanisms of opioid-induced tolerance and hyperalgesia are significantly understudied, in part, because current models cannot fully recapitulate human pathology. Here, we engineered novel human spinal microphysiological systems (MPSs) integrated with plug-and-play neural activity sensing for modeling human nociception and opioid-induced tolerance. Each spinal MPS consists of a flattened human spinal cord organoid derived from human stem cells and a 3D printed organoid holder device for plug-and-play neural activity measurement. We found that the flattened organoid design of MPSs not only reduces hypoxia and necrosis in the organoids, but also promotes their neuron maturation, neural activity, and functional development. We further demonstrated that prolonged opioid exposure resulted in neurochemical correlates of opioid tolerance and hyperalgesia, as measured by altered neural activity, and downregulation of μ-opioid receptor expression of human spinal MPSs. The MPSs are scalable, cost-effective, easy-to-use, and compatible with commonly-used well-plates, thus allowing plug-and-play measurements of neural activity. We believe the MPSs hold a promising translational potential for studying human pain etiology, screening new treatments, and validating novel therapeutics for human pain medicine.
Collapse
Affiliation(s)
- Hongwei Cai
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, 47405, United States
| | - Zheng Ao
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, 47405, United States
| | - Chunhui Tian
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, 47405, United States
| | - Zhuhao Wu
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, 47405, United States
| | - Connor Kaurich
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, 47405, United States
| | - Zi Chen
- Department of Surgery, Brigham and Women's Hospital/Harvard Medical School, Boston, MA, 02115, United States
| | - Mingxia Gu
- Division of Pulmonary Biology, Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, United States
- University of Cincinnati School of Medicine, Cincinnati, OH, 45229, United States
| | - Andrea G. Hohmann
- Gill Center for Biomolecular Science, and Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, United States
| | - Ken Mackie
- Gill Center for Biomolecular Science, and Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, United States
| | - Feng Guo
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, 47405, United States
| |
Collapse
|
32
|
Zhu Y, Zhang X, Sun L, Wang Y, Zhao Y. Engineering Human Brain Assembloids by Microfluidics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2210083. [PMID: 36634089 DOI: 10.1002/adma.202210083] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/17/2022] [Indexed: 06/17/2023]
Abstract
Brain assembloids offer a highly promising strategy to model human brain development and disease, and advance potential studies in regenerative medicine, therapeutic screening, and drug discovery, while it is challenging to produce uniform brain organoids and assemble them flexibly by conventional methods. Here, a multidisciplinary engineered strategy to generate human brain assembloids with desired patterning based on microfluidic technology is presented. By encapsulating human induced pluripotent stem cells in microcapsules via microfluidic electrospray, brain region-specific organoids are efficiently formed, which are then introduced into a microfluidic chip consisting of a bottom layer with a micropillar array and a movable upper layer with a complementary microhole array. These brain organoids can settle into microholes and fuse into brain assembloids. As varied organoid microcapsules with designed 1D sequences or 2D arrays can be assembled into the vertical microholes, large coding amounts of fused brain assembloids with desired patterning can be produced. It is found that brain assembloids composed of cortical, hippocampal, and thalamic organoids can grow and function well, characterized with active neural migration and interaction. These features indicate that the suggested flexible, scalable, and controlled microfluidic systems are remarkably potential in wide applications of brain assembloids in neurological and biomedical fields.
Collapse
Affiliation(s)
- Yujuan Zhu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| | - Xiaoxuan Zhang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Lingyu Sun
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Yu Wang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| |
Collapse
|
33
|
Spatio-temporal dynamics enhance cellular diversity, neuronal function and further maturation of human cerebral organoids. Commun Biol 2023; 6:173. [PMID: 36788328 PMCID: PMC9926461 DOI: 10.1038/s42003-023-04547-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 02/02/2023] [Indexed: 02/16/2023] Open
Abstract
The bioengineerined and whole matured human brain organoids stand as highly valuable three-dimensional in vitro brain-mimetic models to recapitulate in vivo brain development, neurodevelopmental and neurodegenerative diseases. Various instructive signals affecting multiple biological processes including morphogenesis, developmental stages, cell fate transitions, cell migration, stem cell function and immune responses have been employed for generation of physiologically functional cerebral organoids. However, the current approaches for maturation require improvement for highly harvestable and functional cerebral organoids with reduced batch-to-batch variabilities. Here, we demonstrate two different engineering approaches, the rotating cell culture system (RCCS) microgravity bioreactor and a newly designed microfluidic platform (µ-platform) to improve harvestability, reproducibility and the survival of high-quality cerebral organoids and compare with those of traditional spinner and shaker systems. RCCS and µ-platform organoids have reached ideal sizes, approximately 95% harvestability, prolonged culture time with Ki-67 + /CD31 + /β-catenin+ proliferative, adhesive and endothelial-like cells and exhibited enriched cellular diversity (abundant neural/glial/ endothelial cell population), structural brain morphogenesis, further functional neuronal identities (glutamate secreting glutamatergic, GABAergic and hippocampal neurons) and synaptogenesis (presynaptic-postsynaptic interaction) during whole human brain development. Both organoids expressed CD11b + /IBA1 + microglia and MBP + /OLIG2 + oligodendrocytes at high levels as of day 60. RCCS and µ-platform organoids showing high levels of physiological fidelity a high level of physiological fidelity can serve as functional preclinical models to test new therapeutic regimens for neurological diseases and benefit from multiplexing.
Collapse
|
34
|
Wu Z, Ao Z, Cai H, Li X, Chen B, Tu H, Wang Y, Lu RO, Gu M, Cheng L, Lu X, Guo F. Acoustofluidic assembly of primary tumor-derived organotypic cell clusters for rapid evaluation of cancer immunotherapy. J Nanobiotechnology 2023; 21:40. [PMID: 36739414 PMCID: PMC9899402 DOI: 10.1186/s12951-023-01786-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 01/15/2023] [Indexed: 02/06/2023] Open
Abstract
Cancer immunotherapy shows promising potential for treating breast cancer. While patients may have heterogeneous treatment responses for adjuvant therapy, it is challenging to predict an individual patient's response to cancer immunotherapy. Here, we report primary tumor-derived organotypic cell clusters (POCCs) for rapid and reliable evaluation of cancer immunotherapy. By using a label-free, contactless, and highly biocompatible acoustofluidic method, hundreds of cell clusters could be assembled from patient primary breast tumor dissociation within 2 min. Through the incorporation of time-lapse living cell imaging, the POCCs could faithfully recapitulate the cancer-immune interaction dynamics as well as their response to checkpoint inhibitors. Superior to current tumor organoids that usually take more than two weeks to develop, the POCCs can be established and used for evaluation of cancer immunotherapy within 12 h. The POCCs can preserve the cell components from the primary tumor due to the short culture time. Moreover, the POCCs can be assembled with uniform fabricate size and cell composition and served as an open platform for manipulating cell composition and ratio under controlled treatment conditions with a short turnaround time. Thus, we provide a new method to identify potentially immunogenic breast tumors and test immunotherapy, promoting personalized cancer therapy.
Collapse
Affiliation(s)
- Zhuhao Wu
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, 47405, USA
| | - 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
| | - Xiang Li
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, 47405, USA
| | - Bin Chen
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, 47405, USA
| | - Honglei Tu
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, 47405, USA
| | - Yijie Wang
- Computer Science Department, Indiana University, Bloomington, IN, 47408, USA
| | - Rongze Olivia Lu
- Department of Neurological Surgery, Brain Tumor Center, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, California, CA, 94143, USA
| | - Mingxia Gu
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Pulmonary Biology, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- University of Cincinnati School of Medicine, Cincinnati, OH, 45229, USA
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Brown University Warren Alpert Medical School, Lifespan Academic Medical Center, and the Legorreta Cancer Center at Brown University, Providence, RI, 02903, USA
| | - Xin Lu
- Department of Biological Sciences, Boler-Parseghian Center for Rare and Neglected Diseases, Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN, 46556, USA
- Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Feng Guo
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, 47405, USA.
- Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
35
|
Jeong E, Choi S, Cho SW. Recent Advances in Brain Organoid Technology for Human Brain Research. ACS APPLIED MATERIALS & INTERFACES 2023; 15:200-219. [PMID: 36468535 DOI: 10.1021/acsami.2c17467] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Brain organoids are self-assembled three-dimensional aggregates with brain-like cell types and structures and have emerged as new model systems that can be used to investigate human neurodevelopment and neurological disorders. However, brain organoids are not as mature and functional as real human brains due to limitations of the culture system with insufficient developmental patterning signals and a lack of components that are important for brain development and function, such as the non-neural population and vasculature. In addition, establishing the desired brain-like environment and monitoring the complex neural networks and physiological functions of the brain organoids remain challenging. The current protocols to generate brain organoids also have problems with heterogeneity and batch variation due to spontaneous self-organization of brain organoids into complex architectures of the brain. To address these limitations of current brain organoid technologies, various engineering platforms, such as extracellular matrices, fluidic devices, three-dimensional bioprinting, bioreactors, polymeric scaffolds, microelectrodes, and biochemical sensors, have been employed to improve neuronal development and maturation, reduce structural heterogeneity, and facilitate functional analysis and monitoring. In this review, we provide an overview of the latest engineering techniques that overcome these limitations in the production and application of brain organoids.
Collapse
Affiliation(s)
- Eunseon Jeong
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Suah Choi
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
36
|
Pal A, Kaswan K, Barman SR, Lin YZ, Chung JH, Sharma MK, Liu KL, Chen BH, Wu CC, Lee S, Choi D, Lin ZH. Microfluidic nanodevices for drug sensing and screening applications. Biosens Bioelectron 2023; 219:114783. [PMID: 36257116 PMCID: PMC9533638 DOI: 10.1016/j.bios.2022.114783] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 09/18/2022] [Accepted: 10/01/2022] [Indexed: 11/03/2022]
Abstract
The outbreak of pandemics (e.g., severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2 in 2019), influenza A viruses (H1N1 in 2009), etc.), and worldwide spike in the aging population have created unprecedented urgency for developing new drugs to improve disease treatment. As a result, extensive efforts have been made to design novel techniques for efficient drug monitoring and screening, which form the backbone of drug development. Compared to traditional techniques, microfluidics-based platforms have emerged as promising alternatives for high-throughput drug screening due to their inherent miniaturization characteristics, low sample consumption, integration, and compatibility with diverse analytical strategies. Moreover, the microfluidic-based models utilizing human cells to produce in-vitro biomimetics of the human body pave new ways to predict more accurate drug effects in humans. This review provides a comprehensive summary of different microfluidics-based drug sensing and screening strategies and briefly discusses their advantages. Most importantly, an in-depth outlook of the commonly used detection techniques integrated with microfluidic chips for highly sensitive drug screening is provided. Then, the influence of critical parameters such as sensing materials and microfluidic platform geometries on screening performance is summarized. This review also outlines the recent applications of microfluidic approaches for screening therapeutic and illicit drugs. Moreover, the current challenges and the future perspective of this research field is elaborately highlighted, which we believe will contribute immensely towards significant achievements in all aspects of drug development.
Collapse
Affiliation(s)
- Arnab Pal
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan; International Intercollegiate PhD Program, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Kuldeep Kaswan
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan; International Intercollegiate PhD Program, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Snigdha Roy Barman
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan; International Intercollegiate PhD Program, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Yu-Zih Lin
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Jun-Hsuan Chung
- Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Manish Kumar Sharma
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Kuei-Lin Liu
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Bo-Huan Chen
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan; International Intercollegiate PhD Program, National Tsing Hua University, Hsinchu, 30013, Taiwan; Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, 333, Taiwan
| | - Chih-Cheng Wu
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan; Center of Quality Management, National Taiwan University Hospital, Hsinchu Branch, Hsinchu, 30059, Taiwan; College of Medicine, National Taiwan University, Taipei, 10051, Taiwan; Institute of Cellular and System Medicine, National Health Research Institute, Zhunan, 35053, Taiwan
| | - Sangmin Lee
- School of Mechanical Engineering, Chung-Ang University, Seoul, 06974, South Korea.
| | - Dongwhi Choi
- Department of Mechanical Engineering (Integrated Engineering Program), Kyung Hee University, Gyeonggi, 17104, South Korea.
| | - Zong-Hong Lin
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan; International Intercollegiate PhD Program, National Tsing Hua University, Hsinchu, 30013, Taiwan; Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan; Department of Chemistry, National Tsing Hua University, Hsinchu, 30013, Taiwan; Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, 30013, Taiwan; Department of Mechanical Engineering (Integrated Engineering Program), Kyung Hee University, Gyeonggi, 17104, South Korea.
| |
Collapse
|
37
|
Tian T, Liu J, Zhu H. Organ Chips and Visualization of Biological Systems. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1199:155-183. [PMID: 37460731 DOI: 10.1007/978-981-32-9902-3_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Organ-on-a-chip (OOC) is an emerging frontier cross-cutting science and technology developed in the past 10 years. It was first proposed by the Wyss Institute for Biologically Inspired Engineering of Harvard Medical School. It consists of a transparent flexible polymer the size of a computer memory stick, with hollow microfluidic channels lined with living human cells. Researchers used bionics methods to simulate the microenvironment of human cells on microfluidic chips, so as to realize the basic physiological functions of corresponding tissues and organs in vitro. Transparent chip materials can perform real-time visualization and high-resolution analysis of various human life processes in a way that is impossible in animal models, so as to better reproduce the microenvironment of human tissue and simulate biological systems in vitro to observe drug metabolism and other life processes. It provides innovative research systems and system solutions for in vitro bionics of biological systems. It also has gradually become a new tool for disease mechanism research and new drug development. In this chapter, we will take the current research mature single-organ-on-a-chip and multi-organ human-on-a-chip as examples; give an overview of the research background and underlying technologies in this field, especially the application of in vitro bionic models in visualized medicine; and look forward to the foreseeable future development prospects after the integration of organ-on-chip and organoid technology.
Collapse
Affiliation(s)
- Tian Tian
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China.
| | - Jun Liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - He Zhu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| |
Collapse
|
38
|
Dixon TA, Muotri AR. Advancing preclinical models of psychiatric disorders with human brain organoid cultures. Mol Psychiatry 2023; 28:83-95. [PMID: 35948659 PMCID: PMC9812789 DOI: 10.1038/s41380-022-01708-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/05/2022] [Accepted: 07/07/2022] [Indexed: 01/11/2023]
Abstract
Psychiatric disorders are often distinguished from neurological disorders in that the former do not have characteristic lesions or findings from cerebrospinal fluid, electroencephalograms (EEGs), or brain imaging, and furthermore do not have commonly recognized convergent mechanisms. Psychiatric disorders commonly involve clinical diagnosis of phenotypic behavioral disturbances of mood and psychosis, often with a poorly understood contribution of environmental factors. As such, psychiatric disease has been challenging to model preclinically for mechanistic understanding and pharmaceutical development. This review compares commonly used animal paradigms of preclinical testing with evolving techniques of induced pluripotent cell culture with a focus on emerging three-dimensional models. Advances in complexity of 3D cultures, recapitulating electrical activity in utero, and disease modeling of psychosis, mood, and environmentally induced disorders are reviewed. Insights from these rapidly expanding technologies are discussed as they pertain to the utility of human organoid and other models in finding novel research directions, validating pharmaceutical action, and recapitulating human disease.
Collapse
Affiliation(s)
- Thomas Anthony Dixon
- grid.266100.30000 0001 2107 4242Department of Psychiatry, University of California San Diego, La Jolla, CA 92093 USA
| | - Alysson R. Muotri
- grid.266100.30000 0001 2107 4242Department of Pediatrics and Department of Cellular & Molecular Medicine, University of California San Diego, School of Medicine, Center for Academic Research and Training in Anthropogeny (CARTA), Kavli Institute for Brain and Mind, Archealization Center (ArchC), La Jolla, CA 92037 USA
| |
Collapse
|
39
|
Di Cristo L, Sabella S. Cell Cultures at the Air-Liquid Interface and Their Application in Cancer Research. Methods Mol Biol 2023; 2645:41-64. [PMID: 37202611 DOI: 10.1007/978-1-0716-3056-3_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Air-liquid interface (ALI) cell cultures are considered a valid tool for the replacement of animals in biomedical research. By mimicking crucial features of the human in vivo epithelial barriers (e.g., lung, intestine, and skin), ALI cell cultures enable proper structural architectures and differentiated functions of normal and diseased tissue barriers. Thereby, ALI models realistically resemble tissue conditions and provide in vivo-like responses. Since their implementation, they are routinely used in several applications, from toxicity testing to cancer research, receiving an appreciable level of acceptance (in some cases a regulatory acceptance) as attractive testing alternatives to animals. In this chapter, an overview of the ALI cell cultures will be presented together with their application in cancer cell culture, highlighting the potential advantages and disadvantages of the model.
Collapse
Affiliation(s)
- Luisana Di Cristo
- D3 PharmaChemistry, Nanoregulatory Group, Italian Institute of Technology, Genoa, Italy.
| | - Stefania Sabella
- D3 PharmaChemistry, Nanoregulatory Group, Italian Institute of Technology, Genoa, Italy
| |
Collapse
|
40
|
Unagolla JM, Jayasuriya AC. Recent advances in organoid engineering: A comprehensive review. APPLIED MATERIALS TODAY 2022; 29:101582. [PMID: 38264423 PMCID: PMC10804911 DOI: 10.1016/j.apmt.2022.101582] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
Organoid, a 3D structure derived from various cell sources including progenitor and differentiated cells that self-organize through cell-cell and cell-matrix interactions to recapitulate the tissue/organ-specific architecture and function in vitro. The advancement of stem cell culture and the development of hydrogel-based extracellular matrices (ECM) have made it possible to derive self-assembled 3D tissue constructs like organoids. The ability to mimic the actual physiological conditions is the main advantage of organoids, reducing the excessive use of animal models and variability between animal models and humans. However, the complex microenvironment and complex cellular structure of organoids cannot be easily developed only using traditional cell biology. Therefore, several bioengineering approaches, including microfluidics, bioreactors, 3D bioprinting, and organoids-on-a-chip techniques, are extensively used to generate more physiologically relevant organoids. In this review, apart from organoid formation and self-assembly basics, the available bioengineering technologies are extensively discussed as solutions for traditional cell biology-oriented problems in organoid cultures. Also, the natural and synthetic hydrogel systems used in organoid cultures are discussed when necessary to highlight the significance of the stem cell microenvironment. The selected organoid models and their therapeutic applications in drug discovery and disease modeling are also presented.
Collapse
Affiliation(s)
- Janitha M. Unagolla
- Biomedical Engineering Program, Department of Bioengineering, College of Engineering, The University of Toledo, Toledo OH, United States
| | - Ambalangodage C. Jayasuriya
- Biomedical Engineering Program, Department of Bioengineering, College of Engineering, The University of Toledo, Toledo OH, United States
- Department of Orthopaedic Surgery, College of Medicine and Life Sciences, The University of Toledo, 3000 Arlington Avenue, Toledo, OH 43614, United States
| |
Collapse
|
41
|
Song J, Bang S, Choi N, Kim HN. Brain organoid-on-a-chip: A next-generation human brain avatar for recapitulating human brain physiology and pathology. BIOMICROFLUIDICS 2022; 16:061301. [PMID: 36438549 PMCID: PMC9691285 DOI: 10.1063/5.0121476] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 11/01/2022] [Indexed: 06/16/2023]
Abstract
Neurodegenerative diseases and neurodevelopmental disorders have become increasingly prevalent; however, the development of new pharmaceuticals to treat these diseases has lagged. Animal models have been extensively utilized to identify underlying mechanisms and to validate drug efficacies, but they possess inherent limitations including genetic heterogeneity with humans. To overcome these limitations, human cell-based in vitro brain models including brain-on-a-chip and brain organoids have been developed. Each technique has distinct advantages and disadvantages in terms of the mimicry of structure and microenvironment, but each technique could not fully mimic the structure and functional aspects of the brain tissue. Recently, a brain organoid-on-a-chip (BOoC) platform has emerged, which merges brain-on-a-chip and brain organoids. BOoC can potentially reflect the detailed structure of the brain tissue, vascular structure, and circulation of fluid. Hence, we summarize recent advances in BOoC as a human brain avatar and discuss future perspectives. BOoC platform can pave the way for mechanistic studies and the development of pharmaceuticals to treat brain diseases in future.
Collapse
Affiliation(s)
- Jiyoung Song
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Seokyoung Bang
- Department of Medical Biotechnology, Dongguk University, Goyang 10326, Republic of Korea
| | - Nakwon Choi
- Authors to whom correspondence should be addressed:; ; and
| | - Hong Nam Kim
- Authors to whom correspondence should be addressed:; ; and
| |
Collapse
|
42
|
Huang Y, Guo L, Cao C, Ma R, Huang Y, Zhong K, Gao H, Huang Y, Bu Q. Silver nanoparticles exposure induces developmental neurotoxicity in hiPSC-derived cerebral organoids. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 845:157047. [PMID: 35780879 DOI: 10.1016/j.scitotenv.2022.157047] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/16/2022] [Accepted: 06/25/2022] [Indexed: 06/15/2023]
Abstract
Silver nanoparticles (AgNPs) are used in various research fields. Although the neurotoxicity of AgNPs has been explored in animal models and 2D cell-culture models, including human stem cells, these models cannot accurately mimic the development of the human brain. Therefore, the potential mechanisms of AgNPs-induced developmental neurotoxicity in humans are still largely unclear. In this study, cerebral organoids derived from induced pluripotent stem cells were treated with 0.1 μg/mL or 0.5 μg/mL AgNPs for 7 days. At the low concentration (0.1 μg/mL), AgNPs increased the cell proliferation and inhibited the neural apoptosis in the organoids, but impaired the cilium assembly and elongation, which may perturb the cell cycle and induce abnormal cerebral-organoid growth. Conversely, at the high concentration (0.5 μg/mL), AgNPs significantly inhibited cell proliferation and induced apoptosis in cerebral organoids. High-concentration AgNPs reduced the expression and co-localization of the cytoskeleton proteins F-actin, myosin, and tubulin, thereby perturbing neurite growth. In conclusion, AgNPs exposure induces developmental neurotoxic effects in cerebral organoids and is thus a potential congenital risk factor.
Collapse
Affiliation(s)
- Yan Huang
- West China School of Public Health and West China Fourth Hospital, 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
| | - Rui Ma
- Department of Food Science and Technology, College of Biomass and Engineering and Healthy Food Evaluation Research Center, Sichuan University, Chengdu 610065, 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
| | - Kai Zhong
- Department of Food Science and Technology, College of Biomass and Engineering and Healthy Food Evaluation Research Center, Sichuan University, Chengdu 610065, 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
| | - Yina Huang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, 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
|
43
|
Castiglione H, Vigneron PA, Baquerre C, Yates F, Rontard J, Honegger T. Human Brain Organoids-on-Chip: Advances, Challenges, and Perspectives for Preclinical Applications. Pharmaceutics 2022; 14:2301. [PMID: 36365119 PMCID: PMC9699341 DOI: 10.3390/pharmaceutics14112301] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/19/2022] [Accepted: 10/21/2022] [Indexed: 09/26/2023] Open
Abstract
There is an urgent need for predictive in vitro models to improve disease modeling and drug target identification and validation, especially for neurological disorders. Cerebral organoids, as alternative methods to in vivo studies, appear now as powerful tools to decipher complex biological processes thanks to their ability to recapitulate many features of the human brain. Combining these innovative models with microfluidic technologies, referred to as brain organoids-on-chips, allows us to model the microenvironment of several neuronal cell types in 3D. Thus, this platform opens new avenues to create a relevant in vitro approach for preclinical applications in neuroscience. The transfer to the pharmaceutical industry in drug discovery stages and the adoption of this approach by the scientific community requires the proposition of innovative microphysiological systems allowing the generation of reproducible cerebral organoids of high quality in terms of structural and functional maturation, and compatibility with automation processes and high-throughput screening. In this review, we will focus on the promising advantages of cerebral organoids for disease modeling and how their combination with microfluidic systems can enhance the reproducibility and quality of these in vitro models. Then, we will finish by explaining why brain organoids-on-chips could be considered promising platforms for pharmacological applications.
Collapse
Affiliation(s)
- Héloïse Castiglione
- NETRI, 69007 Lyon, France
- Sup’Biotech/CEA-IBFJ-SEPIA, Bâtiment 60, 18 Route du Panorama, 94260 Fontenay-aux-Roses, France
| | - Pierre-Antoine Vigneron
- Sup’Biotech/CEA-IBFJ-SEPIA, Bâtiment 60, 18 Route du Panorama, 94260 Fontenay-aux-Roses, France
- Sup’Biotech, Ecole D’ingénieurs, 66 Rue Guy Môquet, 94800 Villejuif, France
| | | | - Frank Yates
- Sup’Biotech/CEA-IBFJ-SEPIA, Bâtiment 60, 18 Route du Panorama, 94260 Fontenay-aux-Roses, France
- Sup’Biotech, Ecole D’ingénieurs, 66 Rue Guy Môquet, 94800 Villejuif, France
| | | | | |
Collapse
|
44
|
Sozzi E, Kajtez J, Bruzelius A, Wesseler MF, Nilsson F, Birtele M, Larsen NB, Ottosson DR, Storm P, Parmar M, Fiorenzano A. Silk scaffolding drives self-assembly of functional and mature human brain organoids. Front Cell Dev Biol 2022; 10:1023279. [PMID: 36313550 PMCID: PMC9614032 DOI: 10.3389/fcell.2022.1023279] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 09/28/2022] [Indexed: 11/13/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) are intrinsically able to self-organize into cerebral organoids that mimic features of developing human brain tissue. These three-dimensional structures provide a unique opportunity to generate cytoarchitecture and cell-cell interactions reminiscent of human brain complexity in a dish. However, current in vitro brain organoid methodologies often result in intra-organoid variability, limiting their use in recapitulating later developmental stages as well as in disease modeling and drug discovery. In addition, cell stress and hypoxia resulting from long-term culture lead to incomplete maturation and cell death within the inner core. Here, we used a recombinant silk microfiber network as a scaffold to drive hPSCs to self-arrange into engineered cerebral organoids. Silk scaffolding promoted neuroectoderm formation and reduced heterogeneity of cellular organization within individual organoids. Bulk and single cell transcriptomics confirmed that silk cerebral organoids display more homogeneous and functionally mature neuronal properties than organoids grown in the absence of silk scaffold. Furthermore, oxygen sensing analysis showed that silk scaffolds create more favorable growth and differentiation conditions by facilitating the delivery of oxygen and nutrients. The silk scaffolding strategy appears to reduce intra-organoid variability and enhances self-organization into functionally mature human brain organoids.
Collapse
Affiliation(s)
- Edoardo Sozzi
- Department of Experimental Medical Science, Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Janko Kajtez
- Department of Experimental Medical Science, Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Andreas Bruzelius
- Department of Experimental Medical Science, Regenerative Neurophysiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Milan Finn Wesseler
- Department of Health Technology (DTU Health Tech), Technical University of Denmark, Kongens Lyngby, Denmark
| | - Fredrik Nilsson
- Department of Experimental Medical Science, Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Marcella Birtele
- Department of Experimental Medical Science, Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Niels B. Larsen
- Department of Health Technology (DTU Health Tech), Technical University of Denmark, Kongens Lyngby, Denmark
| | - Daniella Rylander Ottosson
- Department of Experimental Medical Science, Regenerative Neurophysiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Petter Storm
- Department of Experimental Medical Science, Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Malin Parmar
- Department of Experimental Medical Science, Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Alessandro Fiorenzano
- Department of Experimental Medical Science, Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Lund University, Lund, Sweden
- *Correspondence: Alessandro Fiorenzano,
| |
Collapse
|
45
|
Tran HN, Gautam V. Micro/nano devices for integration with human brain organoids. Biosens Bioelectron 2022; 218:114750. [DOI: 10.1016/j.bios.2022.114750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 09/17/2022] [Accepted: 09/22/2022] [Indexed: 11/02/2022]
|
46
|
Tran HN, Gautam V. Micro- and nanodevices for integration with human brain organoids. Biosens Bioelectron 2022:114734. [PMID: 36990931 DOI: 10.1016/j.bios.2022.114734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/18/2022] [Accepted: 09/14/2022] [Indexed: 12/01/2022]
Affiliation(s)
- Hao Nguyen Tran
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Victoria, 3010, Australia
| | - Vini Gautam
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Victoria, 3010, Australia.
| |
Collapse
|
47
|
Hu X, Zheng J, Hu Q, Liang L, Yang D, Cheng Y, Li SS, Chen LJ, Yang Y. Smart acoustic 3D cell construct assembly with high-resolution. Biofabrication 2022; 14. [PMID: 35764072 DOI: 10.1088/1758-5090/ac7c90] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 06/22/2022] [Indexed: 11/12/2022]
Abstract
Precise and flexible three-dimensional (3D) cell construct assembly using external forces or fields can produce micro-scale cellular architectures with intercellular connections, which is an important prerequisite to reproducing the structures and functions of biological systems. Currently, it is also a substantial challenge in the bioengineering field. Here, we propose a smart acoustic 3D cell assembly strategy that utilizes a 3D printed module and hydrogel sheets. Digitally controlled six wave beams offer a high degree of freedom (including wave vector combination, frequency, phase, and amplitude) that enables versatile biomimetic micro cellular patterns in hydrogel sheets. Further, replaceable frames can be used to fix the acoustic-built micro-scale cellular structures in these sheets, enabling user-defined hierarchical or heterogeneous constructs through layer-by-layer assembly. This strategy can be employed to construct vasculature with different diameters and lengths, composed of human umbilical vein endothelial cells and smooth muscle cells. These constructs can also induce controllable vascular network formation. Overall, the findings of this work extend the capabilities of acoustic cell assembly into 3D space, offering advantages including innovative, flexible, and precise patterning, and displaying great potential for the manufacture of various artificial tissue structures that duplicate in vivo functions.
Collapse
Affiliation(s)
- Xuejia Hu
- School of Electronic Science and Engineering, Xiamen University, Xiamen University, No. 422 Siming south road, Xiamen, Fujian, 361005, CHINA
| | - Jingjing Zheng
- School of physics and engineering, Wuhan University, luojia mountain street, Wuhan, Wuhan, Hubei, 430072, CHINA
| | - Qinghao Hu
- School of physics and engineering, Wuhan University, luojia street, Wuhan, Wuhan, Hubei, 430072, CHINA
| | - Li Liang
- School of Physics and Electronic Technology, Anhui Normal University, No. 189 of jiuhua south road, Wuhu, Wuhu, Anhui, 241000, CHINA
| | - Dongyong Yang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, No. 238, Jiefang road, Wuhan, Hubei, 430060, CHINA
| | - Yanxiang Cheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, No. 238, Jiefang road, Wuhan, Hubei, 430060, CHINA
| | - Sen-Sen Li
- School of Electronic Science and Engineering, Xiamen University, Xiamen University, No. 422 Siming south road, Xiamen, Fujian, 361005, CHINA
| | - Lu-Jian Chen
- School of Electronic Science and Engineering, Xiamen University, Xiamen University, No. 422 Siming south road, Xiamen, Fujian, 361005, CHINA
| | - Yi Yang
- School of physics and engineering, Wuhan University, luojia street, Wuhan, Wuhan, Hubei, 430072, CHINA
| |
Collapse
|
48
|
Li M, Gong J, Gao L, Zou T, Kang J, Xu H. Advanced human developmental toxicity and teratogenicity assessment using human organoid models. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 235:113429. [PMID: 35325609 DOI: 10.1016/j.ecoenv.2022.113429] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/12/2022] [Accepted: 03/16/2022] [Indexed: 06/14/2023]
Abstract
Tremendous progress has been made in the field of toxicology leading to the advance of developmental toxicity assessment. Conventional animal models and in vitro two-dimensional models cannot accurately describe toxic effects and predict actual in vivo responses due to obvious inter-species differences between humans and animals, as well as the lack of a physiologically relevant tissue microenvironment. Human embryonic stem cell (hESC)- and induced pluripotent stem cell (iPSC)-derived three-dimensional organoids are ideal complex and multicellular organotypic models, which are indispensable in recapitulating morphogenesis, cellular interactions, and molecular processes of early human organ development. Recently, human organoids have been used for drug discovery, chemical toxicity and safety in vitro assessment. This review discusses the recent advances in the use of human organoid models, (i.e., brain, retinal, cardiac, liver, kidney, lung, and intestinal organoid models) for developmental toxicity and teratogenicity assessment of distinct tissues/organs following exposure to pharmaceutical compounds, heavy metals, persistent organic pollutants, nanomaterials, and ambient air pollutants. Combining next-generation organoid models with innovative engineering technologies generates novel and powerful tools for developmental toxicity and teratogenicity assessment, and the rapid progress in this field is expected to continue.
Collapse
Affiliation(s)
- Minghui Li
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Jing Gong
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Lixiong Gao
- Department of Ophthalmology, Third Medical Center of PLA General Hospital, Beijing 100039, China
| | - Ting Zou
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Jiahui Kang
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Haiwei Xu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China.
| |
Collapse
|
49
|
Chrishtop V, Nikonorova V, Gutsalova A, Rumyantseva T, Dukhinova M, Salmina А. Systematic comparison of basic animal models of cerebral hypoperfusion. Tissue Cell 2022; 75:101715. [DOI: 10.1016/j.tice.2021.101715] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 12/12/2021] [Accepted: 12/14/2021] [Indexed: 02/07/2023]
|
50
|
Stankovic IN, Colak D. Prenatal Drugs and Their Effects on the Developing Brain: Insights From Three-Dimensional Human Organoids. Front Neurosci 2022; 16:848648. [PMID: 35401083 PMCID: PMC8990163 DOI: 10.3389/fnins.2022.848648] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 02/01/2022] [Indexed: 11/13/2022] Open
Abstract
Decades of research have unequivocally demonstrated that fetal exposure to both recreational and prescription drugs in utero negatively impacts the developing brain. More recently, the application of cutting-edge techniques in neurodevelopmental research has attempted to identify how the fetal brain responds to specific environmental stimuli. Meanwhile, human fetal brain studies still encounter ethical considerations and technical limitations in tissue collection. Human-induced pluripotent stem cell (iPSC)-derived brain organoid technology has emerged as a powerful alternative to examine fetal neurobiology. In fact, human 3D organoid tissues recapitulate cerebral development during the first trimester of pregnancy. In this review, we aim to provide a comprehensive summary of fetal brain metabolic studies related to drug abuse in animal and human models. Additionally, we will discuss the current challenges and prospects of using brain organoids for large-scale metabolomics. Incorporating cutting-edge techniques in human brain organoids may lead to uncovering novel molecular and cellular mechanisms of neurodevelopment, direct novel therapeutic approaches, and raise new exciting questions.
Collapse
Affiliation(s)
- Isidora N. Stankovic
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, Cornell University, New York, NY, United States
- *Correspondence: Isidora N. Stankovic,
| | - Dilek Colak
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, Cornell University, New York, NY, United States
- Gale & Ira Drukier Institute for Children’s Health, Weill Cornell Medicine, Cornell University, New York, NY, United States
- Dilek Colak,
| |
Collapse
|