1
|
Di Stefano J, Di Marco F, Cicalini I, FitzGerald U, Pieragostino D, Verhoye M, Ponsaerts P, Van Breedam E. Generation, interrogation, and future applications of microglia-containing brain organoids. Neural Regen Res 2025; 20:3448-3460. [PMID: 39665813 PMCID: PMC11974650 DOI: 10.4103/nrr.nrr-d-24-00921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/29/2024] [Accepted: 11/05/2024] [Indexed: 12/13/2024] Open
Abstract
Brain organoids encompass a large collection of in vitro stem cell-derived 3D culture systems that aim to recapitulate multiple aspects of in vivo brain development and function. First, this review provides a brief introduction to the current state-of-the-art for neuro-ectoderm brain organoid development, emphasizing their biggest advantages in comparison with classical two-dimensional cell cultures and animal models. However, despite their usefulness for developmental studies, a major limitation for most brain organoid models is the absence of contributing cell types from endodermal and mesodermal origin. As such, current research is highly investing towards the incorporation of a functional vasculature and the microglial immune component. In this review, we will specifically focus on the development of immune-competent brain organoids. By summarizing the different approaches applied to incorporate microglia, it is highlighted that immune-competent brain organoids are not only important for studying neuronal network formation, but also offer a clear future as a new tool to study inflammatory responses in vitro in 3D in a brain-like environment. Therefore, our main focus here is to provide a comprehensive overview of assays to measure microglial phenotype and function within brain organoids, with an outlook on how these findings could better understand neuronal network development or restoration, as well as the influence of physical stress on microglia-containing brain organoids. Finally, we would like to stress that even though the development of immune-competent brain organoids has largely evolved over the past decade, their full potential as a pre-clinical tool to study novel therapeutic approaches to halt or reduce inflammation-mediated neurodegeneration still needs to be explored and validated.
Collapse
Affiliation(s)
- Julia Di Stefano
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
- Bio-Imaging Lab, University of Antwerp, Wilrijk, Belgium
| | - Federica Di Marco
- Center for Advanced Studies and Technology (CAST), G. d’Annunzio University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, Chieti, Italy
| | - Ilaria Cicalini
- Center for Advanced Studies and Technology (CAST), G. d’Annunzio University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, Chieti, Italy
| | - Una FitzGerald
- CÚRAM, Center for Research in Medical Devices, Biomedical Engineering, University of Galway, Ireland
- Galway Neuroscience Center, University of Galway, Ireland
| | - Damiana Pieragostino
- Center for Advanced Studies and Technology (CAST), G. d’Annunzio University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, Chieti, Italy
| | - Marleen Verhoye
- Bio-Imaging Lab, University of Antwerp, Wilrijk, Belgium
- μNEURO Research Center of Excellence, University of Antwerp, Wilrijk, Belgium
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
| | - Elise Van Breedam
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
2
|
He M, Lin Y, Zhang X, Wang S, Yang X, Cui F, Sheng X. 6-Chloronicotinic Acid Induces Toxicity in Mouse Neural Stem Cells via the C3ar1 Signaling. J Appl Toxicol 2025; 45:783-794. [PMID: 40205829 DOI: 10.1002/jat.4746] [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: 10/16/2024] [Revised: 11/23/2024] [Accepted: 12/03/2024] [Indexed: 04/11/2025]
Abstract
Neural stem cells (NSCs) are essential for brain development due to their ability to proliferate and differentiate into various neural cell types. Neonicotinoid insecticides (NNIs), which have replaced traditional pesticides, are now widely used and frequently detected in environmental and biological samples. Prenatal exposure to NNIs has been associated with an increased risk of neurodevelopmental disorders in offspring, yet the causal relationship and the underpinning mechanism remain to be clarified. As one of the primary metabolites of chloropyridinyl neonicotinoids, 6-chloronicotinic acid (6-ClNA) has been identified as a potential neurotoxin, though its effects on NSCs have not been fully explored. Here, we demonstrate that 6-ClNA exposure significantly disrupted NSC proliferation and differentiation in vitro. Transcriptomic analyses revealed that 6-ClNA altered the expression of pathways related to proliferation, apoptosis, and inflammation, with notable activation of the C3ar1/C1qa signaling axis. Genetic ablation of C3ar1 using siRNA markedly restored NSC proliferation and neurosphere formation, as well as reduced apoptosis, suggesting a central role of C3ar1/C1qa in mediating 6-ClNA's neurotoxic effects. These findings imply that early-life exposure to NNIs may affect the fitness and function of NSCs, wherein the C3ar1 pathway plays an indispensable role.
Collapse
Affiliation(s)
- Min He
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yahang Lin
- Department of Neurology, Wuhan Fourth Hospital/Pu'ai Hospital, Wuhan, China
| | - Xiaojing Zhang
- School of Life and Health Sciences, Hainan Province Key Laboratory of One Health, Collaborative Innovation Center of One Health, Hainan University, Haikou, China
| | - Siyi Wang
- Department of Neurology, Wuhan Fourth Hospital/Pu'ai Hospital, Wuhan, China
| | - Xinyu Yang
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fengzhen Cui
- Dongguan Key Laboratory of Environmental Medicine, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan, China
| | - Xia Sheng
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Life and Health Sciences, Hainan Province Key Laboratory of One Health, Collaborative Innovation Center of One Health, Hainan University, Haikou, China
| |
Collapse
|
3
|
Zhao Y, Wang T, Liu J, Wang Z, Lu Y. Emerging brain organoids: 3D models to decipher, identify and revolutionize brain. Bioact Mater 2025; 47:378-402. [PMID: 40026825 PMCID: PMC11869974 DOI: 10.1016/j.bioactmat.2025.01.025] [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: 10/23/2024] [Revised: 01/20/2025] [Accepted: 01/20/2025] [Indexed: 03/05/2025] Open
Abstract
Brain organoids are an emerging in vitro 3D brain model that is integrated from pluripotent stem cells. This model mimics the human brain's developmental process and disease-related phenotypes to a certain extent while advancing the development of human brain-based biological intelligence. However, many limitations of brain organoid culture (e.g., lacking a functional vascular system, etc.) prevent in vitro-cultured organoids from truly replicating the human brain in terms of cell type and structure. To improve brain organoids' scalability, efficiency, and stability, this paper discusses important contributions of material biology and microprocessing technology in solving the related limitations of brain organoids and applying the latest imaging technology to make real-time imaging of brain organoids possible. In addition, the related applications of brain organoids, especially the development of organoid intelligence combined with artificial intelligence, are analyzed, which will help accelerate the rational design and guidance of brain organoids.
Collapse
Affiliation(s)
- Yuli Zhao
- College of Life Sciences, Shenyang Normal University, Shenyang, 110034, Liaoning, China
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
| | - Ting Wang
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
| | - Jiajun Liu
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
- Tianjin Industrial Microbiology Key Laboratory, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Ze Wang
- College of Life Sciences, Shenyang Normal University, Shenyang, 110034, Liaoning, China
| | - Yuan Lu
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
4
|
Tao B, Li X, Hao M, Tian T, Li Y, Li X, Yang C, Li Q, Feng Q, Zhou H, Zhao Y, Wang D, Liu W. Organoid-Guided Precision Medicine: From Bench to Bedside. MedComm (Beijing) 2025; 6:e70195. [PMID: 40321594 PMCID: PMC12046123 DOI: 10.1002/mco2.70195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 03/16/2025] [Accepted: 03/18/2025] [Indexed: 05/08/2025] Open
Abstract
Organoid technology, as an emerging field within biotechnology, has demonstrated transformative potential in advancing precision medicine. This review systematically outlines the translational trajectory of organoids from bench to bedside, emphasizing their construction methodologies, key regulatory factors, and multifaceted applications in personalized healthcare. By recapitulating physiological architectures and disease phenotypes through three-dimensional culture systems, organoids leverage natural and synthetic scaffolds, stem cell sources, and spatiotemporal cytokine regulation to model tissue-specific microenvironments. Diverse organoid types-including skin, intestinal, lung, and tumor organoids-have facilitated breakthroughs in modeling tissue development, drug efficacy and toxicity screening, disease pathogenesis studies, and patient-tailored diagnostics. For instance, patient-derived tumor organoids preserve tumor heterogeneity and genomic profiles, serving as predictive platforms for individualized chemotherapy responses. In precision medicine, organoid-guided multiomics analyses identify actionable biomarkers and resistance mechanisms, while clustered regularly interspaced short palindromic repeats-based functional screens optimize therapeutic targeting. Despite preclinical successes, challenges persist in standardization, vascularization, and ethical considerations. Future integration of artificial intelligence, microfluidics, and spatial transcriptomics will enhance organoid scalability, reproducibility, and clinical relevance. By bridging molecular insights with patient-specific therapies, organoids are poised to revolutionize precision medicine, offering dynamic platforms for drug development, regenerative strategies, and individualized treatment paradigms.
Collapse
Affiliation(s)
- Boqiang Tao
- Department of Oral and Maxillofacial SurgeryHospital of StomatologyJilin UniversityChangchunChina
| | - Xiaolan Li
- Laboratory of Allergy and Precision MedicineChengdu Institute of Respiratory Healththe Third People's Hospital of ChengduAffiliated Hospital of Southwest Jiaotong UniversityChengduChina
| | - Ming Hao
- Department of Oral and Maxillofacial SurgeryHospital of StomatologyJilin UniversityChangchunChina
| | - Tian Tian
- Laboratory Animal CenterCollege of Animal ScienceJilin UniversityChangchunChina
| | - Yuyang Li
- Department of Oral and Maxillofacial SurgeryHospital of StomatologyJilin UniversityChangchunChina
| | - Xiang Li
- Department of Oral and Maxillofacial SurgeryHospital of StomatologyJilin UniversityChangchunChina
| | - Chun Yang
- College of Basic MedicineBeihua UniversityJilinChina
| | - Qirong Li
- Laboratory Animal CenterCollege of Animal ScienceJilin UniversityChangchunChina
| | - Qiang Feng
- Laboratory Animal CenterCollege of Animal ScienceJilin UniversityChangchunChina
| | - Hengzong Zhou
- Laboratory Animal CenterCollege of Animal ScienceJilin UniversityChangchunChina
| | - Yicheng Zhao
- Laboratory Animal CenterCollege of Animal ScienceJilin UniversityChangchunChina
| | - Dongxu Wang
- Laboratory Animal CenterCollege of Animal ScienceJilin UniversityChangchunChina
- Zhichuang Gene Editing Animal Model Research CenterWenzhou Institute of TechnologyWenzhouChina
| | - Weiwei Liu
- Department of Oral and Maxillofacial SurgeryHospital of StomatologyJilin UniversityChangchunChina
| |
Collapse
|
5
|
Karam M, Ortega-Gascó A, Tornero D. Emerging Insights into Brain Inflammation: Stem-Cell-Based Approaches for Regenerative Medicine. Int J Mol Sci 2025; 26:3275. [PMID: 40244116 PMCID: PMC11989304 DOI: 10.3390/ijms26073275] [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: 02/28/2025] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/18/2025] Open
Abstract
Neuroinflammation is a complex immune response triggered by brain injury or pathological stimuli, and is highly exacerbated in neurodegenerative diseases. It plays a dual role in the central nervous system, promoting repair in acute stages while aggravating disease progression by contributing to neuronal loss, synaptic dysfunction, and glial dysregulation in chronic phases. Inflammatory responses are mainly orchestrated by microglia and infiltrated monocytes, which, when dysregulated, not only harm existing neurons, but also impair the survival and differentiation of neural stem and progenitor cells in the affected brain regions. Modulating neuroinflammation is crucial for harnessing its protective functions while minimizing its detrimental effects. Current therapeutic strategies focus on fine-tuning inflammatory responses through pharmacological agents, bioactive molecules, and stem cell-based therapies. These approaches aim to restore immune homeostasis, support neuroprotection, and promote regeneration in various neurological disorders. However, animal models sometimes fail to reproduce human-specific inflammatory responses in the brain. In this context, stem-cell-derived models provide a powerful tool to study neuroinflammatory mechanisms in a patient-specific and physiologically relevant context. These models facilitate high-throughput screening, personalized medicine, and the development of targeted therapies while addressing the limitations of traditional animal models, paving the way for more targeted and effective treatments.
Collapse
Affiliation(s)
- Marie Karam
- Laboratory of Neural Stem Cells and Brain Damage, Department of Biomedical Sciences, Institute of Neurosciences, University of Barcelona, 08036 Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Alba Ortega-Gascó
- Laboratory of Neural Stem Cells and Brain Damage, Department of Biomedical Sciences, Institute of Neurosciences, University of Barcelona, 08036 Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Daniel Tornero
- Laboratory of Neural Stem Cells and Brain Damage, Department of Biomedical Sciences, Institute of Neurosciences, University of Barcelona, 08036 Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28029 Madrid, Spain
| |
Collapse
|
6
|
Wu H, Feng E, Yin H, Zhang Y, Chen G, Zhu B, Yue X, Zhang H, Liu Q, Xiong L. Biomaterials for neuroengineering: applications and challenges. Regen Biomater 2025; 12:rbae137. [PMID: 40007617 PMCID: PMC11855295 DOI: 10.1093/rb/rbae137] [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: 09/07/2024] [Revised: 10/19/2024] [Accepted: 11/03/2024] [Indexed: 02/27/2025] Open
Abstract
Neurological injuries and diseases are a leading cause of disability worldwide, underscoring the urgent need for effective therapies. Neural regaining and enhancement therapies are seen as the most promising strategies for restoring neural function, offering hope for individuals affected by these conditions. Despite their promise, the path from animal research to clinical application is fraught with challenges. Neuroengineering, particularly through the use of biomaterials, has emerged as a key field that is paving the way for innovative solutions to these challenges. It seeks to understand and treat neurological disorders, unravel the nature of consciousness, and explore the mechanisms of memory and the brain's relationship with behavior, offering solutions for neural tissue engineering, neural interfaces and targeted drug delivery systems. These biomaterials, including both natural and synthetic types, are designed to replicate the cellular environment of the brain, thereby facilitating neural repair. This review aims to provide a comprehensive overview for biomaterials in neuroengineering, highlighting their application in neural functional regaining and enhancement across both basic research and clinical practice. It covers recent developments in biomaterial-based products, including 2D to 3D bioprinted scaffolds for cell and organoid culture, brain-on-a-chip systems, biomimetic electrodes and brain-computer interfaces. It also explores artificial synapses and neural networks, discussing their applications in modeling neural microenvironments for repair and regeneration, neural modulation and manipulation and the integration of traditional Chinese medicine. This review serves as a comprehensive guide to the role of biomaterials in advancing neuroengineering solutions, providing insights into the ongoing efforts to bridge the gap between innovation and clinical application.
Collapse
Affiliation(s)
- Huanghui Wu
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Enduo Feng
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Huanxin Yin
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Yuxin Zhang
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Guozhong Chen
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Beier Zhu
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Xuezheng Yue
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Haiguang Zhang
- Rapid Manufacturing Engineering Center, School of Mechatronical Engineering and Automation, Shanghai University, Shanghai 200444, China
- Shanghai Key Laboratory of Intelligent Manufacturing and Robotics, Shanghai University, Shanghai 200072, China
| | - Qiong Liu
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200434, China
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China
| | - Lize Xiong
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| |
Collapse
|
7
|
Neziri S, Köseoğlu AE, Deniz Köseoğlu G, Özgültekin B, Özgentürk NÖ. Animal models in neuroscience with alternative approaches: Evolutionary, biomedical, and ethical perspectives. Animal Model Exp Med 2024; 7:868-880. [PMID: 39375824 DOI: 10.1002/ame2.12487] [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: 05/27/2024] [Accepted: 08/07/2024] [Indexed: 10/09/2024] Open
Abstract
Animal models have been a crucial tool in neuroscience research for decades, providing insights into the biomedical and evolutionary mechanisms of the nervous system, disease, and behavior. However, their use has raised concerns on several ethical, clinical, and scientific considerations. The welfare of animals and the 3R principles (replacement, reduction, refinement) are the focus of the ethical concerns, targeting the importance of reducing the stress and suffering of these models. Several laws and guidelines are applied and developed to protect animal rights during experimenting. Concurrently, in the clinic and biomedical fields, discussions on the relevance of animal model findings on human organisms have increased. Latest data suggest that in a considerable amount of time the animal model results are not translatable in humans, costing time and money. Alternative methods, such as in vitro (cell culture, microscopy, organoids, and micro physiological systems) techniques and in silico (computational) modeling, have emerged as potential replacements for animal models, providing more accurate data in a minimized cost. By adopting alternative methods and promoting ethical considerations in research practices, we can achieve the 3R goals while upholding our responsibility to both humans and other animals. Our goal is to present a thorough review of animal models used in neuroscience from the biomedical, evolutionary, and ethical perspectives. The novelty of this research lies in integrating diverse points of views to provide an understanding of the advantages and disadvantages of animal models in neuroscience and in discussing potential alternative methods.
Collapse
Affiliation(s)
- Sabina Neziri
- Department of Molecular Biology and Genetics, Faculty of Art and Science, Yıldız Technical University, Istanbul, Turkey
| | | | | | - Buminhan Özgültekin
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Acıbadem University, Istanbul, Turkey
| | - Nehir Özdemir Özgentürk
- Department of Molecular Biology and Genetics, Faculty of Art and Science, Yıldız Technical University, Istanbul, Turkey
| |
Collapse
|
8
|
Li M, Yuan Y, Hou Z, Hao S, Jin L, Wang B. Human brain organoid: trends, evolution, and remaining challenges. Neural Regen Res 2024; 19:2387-2399. [PMID: 38526275 PMCID: PMC11090441 DOI: 10.4103/1673-5374.390972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/26/2023] [Accepted: 10/28/2023] [Indexed: 03/26/2024] Open
Abstract
Advanced brain organoids provide promising platforms for deciphering the cellular and molecular processes of human neural development and diseases. Although various studies and reviews have described developments and advancements in brain organoids, few studies have comprehensively summarized and analyzed the global trends in this area of neuroscience. To identify and further facilitate the development of cerebral organoids, we utilized bibliometrics and visualization methods to analyze the global trends and evolution of brain organoids in the last 10 years. First, annual publications, countries/regions, organizations, journals, authors, co-citations, and keywords relating to brain organoids were identified. The hotspots in this field were also systematically identified. Subsequently, current applications for brain organoids in neuroscience, including human neural development, neural disorders, infectious diseases, regenerative medicine, drug discovery, and toxicity assessment studies, are comprehensively discussed. Towards that end, several considerations regarding the current challenges in brain organoid research and future strategies to advance neuroscience will be presented to further promote their application in neurological research.
Collapse
Affiliation(s)
- Minghui Li
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yuhan Yuan
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Zongkun Hou
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Shilei Hao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Liang Jin
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Bochu Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| |
Collapse
|
9
|
Meng L, Zhou B, Liu H, Chen Y, Yuan R, Chen Z, Luo S, Chen H. Advancing toxicity studies of per- and poly-fluoroalkyl substances (pfass) through machine learning: Models, mechanisms, and future directions. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 946:174201. [PMID: 38936709 DOI: 10.1016/j.scitotenv.2024.174201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 06/17/2024] [Accepted: 06/20/2024] [Indexed: 06/29/2024]
Abstract
Perfluorinated and perfluoroalkyl substances (PFASs), encompassing a vast array of isomeric chemicals, are recognized as typical emerging contaminants with direct or potential impacts on human health and the ecological environment. With the complex and elusive toxicological profiles of PFASs, machine learning (ML) has been increasingly employed in their toxicity studies due to its proficiency in prediction and data analytics. This integration is poised to become a predominant trend in environmental toxicology, propelled by the swift advancements in computational technology. This review diligently examines the literature to encapsulate the varied objectives of employing ML in the toxicity studies of PFASs: (1) Utilizing ML to establish Quantitative Structure-Activity Relationship (QSAR) models for PFASs with diverse toxicity endpoints, facilitating the targeted toxicity prediction of unidentified PFASs; (2) Investigating and substantiating the Adverse Outcome Pathway (AOP) through the synergy of ML and traditional toxicological methods, with this refining the toxicity assessment framework for PFASs; (3) Dissecting and elucidating the features of established ML models to advance Open Research into the toxicity of PFASs, with a primary focus on determinants and mechanisms. The discourse extends to an in-depth examination of ML studies, segregating findings based on their distinct application trajectories. Given that ML represents a nascent paradigm within PFASs research, this review delineates the collective challenges encountered in the ML-mediated study of PFAS toxicity and proffers strategic guidance for ensuing investigations.
Collapse
Affiliation(s)
- Lingxuan Meng
- Beijing Key Laboratory of Resource-oriented Treatment of Industrial Pollutants, School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Beihai Zhou
- Beijing Key Laboratory of Resource-oriented Treatment of Industrial Pollutants, School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Haijun Liu
- School of Resources and Environment, Anqing Normal University, Anqing, China.
| | - Yuefang Chen
- Beijing Key Laboratory of Resource-oriented Treatment of Industrial Pollutants, School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China.
| | - Rongfang Yuan
- Beijing Key Laboratory of Resource-oriented Treatment of Industrial Pollutants, School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Zhongbing Chen
- Faculty of Environmental Sciences, Czech University of Life Sciences Prague, Kamýcká 129, 16500 Praha-Suchdol, Czech Republic.
| | - Shuai Luo
- Beijing Key Laboratory of Resource-oriented Treatment of Industrial Pollutants, School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Huilun Chen
- Beijing Key Laboratory of Resource-oriented Treatment of Industrial Pollutants, School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China.
| |
Collapse
|
10
|
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
|
11
|
Kang S, Chen EC, Cifuentes H, Co JY, Cole G, Graham J, Hsia R, Kiyota T, Klein JA, Kroll KT, Nieves Lopez LM, Norona LM, Peiris H, Potla R, Romero-Lopez M, Roth JG, Tseng M, Fullerton AM, Homan KA. Complex in vitromodels positioned for impact to drug testing in pharma: a review. Biofabrication 2024; 16:042006. [PMID: 39189069 DOI: 10.1088/1758-5090/ad6933] [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: 12/22/2023] [Accepted: 07/30/2024] [Indexed: 08/28/2024]
Abstract
Recent years have seen the creation and popularization of various complexin vitromodels (CIVMs), such as organoids and organs-on-chip, as a technology with the potential to reduce animal usage in pharma while also enhancing our ability to create safe and efficacious drugs for patients. Public awareness of CIVMs has increased, in part, due to the recent passage of the FDA Modernization Act 2.0. This visibility is expected to spur deeper investment in and adoption of such models. Thus, end-users and model developers alike require a framework to both understand the readiness of current models to enter the drug development process, and to assess upcoming models for the same. This review presents such a framework for model selection based on comparative -omics data (which we term model-omics), and metrics for qualification of specific test assays that a model may support that we term context-of-use (COU) assays. We surveyed existing healthy tissue models and assays for ten drug development-critical organs of the body, and provide evaluations of readiness and suggestions for improving model-omics and COU assays for each. In whole, this review comes from a pharma perspective, and seeks to provide an evaluation of where CIVMs are poised for maximum impact in the drug development process, and a roadmap for realizing that potential.
Collapse
Affiliation(s)
- Serah Kang
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Eugene C Chen
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Helen Cifuentes
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Julia Y Co
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Gabrielle Cole
- Investigative Toxicology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Jessica Graham
- Product Quality & Occupational Toxicology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of Americaica
| | - Rebecca Hsia
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Tomomi Kiyota
- Investigative Toxicology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Jessica A Klein
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Katharina T Kroll
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Lenitza M Nieves Lopez
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Leah M Norona
- Investigative Toxicology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Heshan Peiris
- Human Genetics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Ratnakar Potla
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Monica Romero-Lopez
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Julien G Roth
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Min Tseng
- Investigative Toxicology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Aaron M Fullerton
- Investigative Toxicology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Kimberly A Homan
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| |
Collapse
|
12
|
Strasser AS, Gonzalez-Reiche AS, Zhou X, Valdebenito-Maturana B, Ye X, Zhang B, Wu M, van Bakel H, Jabs EW. Limb reduction in an Esco2 cohesinopathy mouse model is mediated by p53-dependent apoptosis and vascular disruption. Nat Commun 2024; 15:7154. [PMID: 39168984 PMCID: PMC11339411 DOI: 10.1038/s41467-024-51328-3] [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: 09/01/2023] [Accepted: 08/01/2024] [Indexed: 08/23/2024] Open
Abstract
Roberts syndrome (RBS) is an autosomal recessive disorder with profound growth deficiency and limb reduction caused by ESCO2 loss-of-function variants. Here, we elucidate the pathogenesis of limb reduction in an Esco2fl/fl;Prrx1-CreTg/0 mouse model using bulk- and single-cell-RNA-seq and gene co-expression network analyses during embryogenesis. Our results reveal morphological and vascular defects culminating in hemorrhage of mutant limbs at E12.5. Underlying this abnormal developmental progression is a pre-apoptotic, mesenchymal cell population specific to mutant limb buds enriched for p53-related signaling beginning at E9.5. We then characterize these p53-related processes of cell cycle arrest, DNA damage, cell death, and the inflammatory leukotriene signaling pathway in vivo. In utero treatment with pifithrin-α, a p53 inhibitor, rescued the hemorrhage in mutant limbs. Lastly, significant enrichments were identified among genes associated with RBS, thalidomide embryopathy, and other genetic limb reduction disorders, suggesting a common vascular etiology among these conditions.
Collapse
Affiliation(s)
- Arielle S Strasser
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
| | - Ana Silvia Gonzalez-Reiche
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
| | - Xianxiao Zhou
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
| | - Braulio Valdebenito-Maturana
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
| | - Xiaoqian Ye
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
| | - Meng Wu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA.
- Department of Clinical Genomics, Mayo Clinic, 200 First Street, Rochester, MN, USA.
- Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 First Street, Rochester, MN, USA.
| | - Harm van Bakel
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA.
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA.
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA.
- Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA.
| | - Ethylin Wang Jabs
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA.
- Department of Clinical Genomics, Mayo Clinic, 200 First Street, Rochester, MN, USA.
- Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 First Street, Rochester, MN, USA.
- Department of Cell, Development and Regenerative Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA.
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA.
| |
Collapse
|
13
|
Lopez I, Truskey GA. Multi-cellular engineered living systems to assess reproductive toxicology. Reprod Toxicol 2024; 127:108609. [PMID: 38759876 PMCID: PMC11179964 DOI: 10.1016/j.reprotox.2024.108609] [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: 12/21/2023] [Revised: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 05/19/2024]
Abstract
Toxicants and some drugs can negatively impact reproductive health. Many toxicants haven't been tested due to lack of available models. The impact of many drugs taken during pregnancy to address maternal health may adversely affect fetal development with life-long effects and clinical trials do not examine toxicity effects on the maternal-fetal interface, requiring indirect assessment of safety and efficacy. Due to current gaps in reproductive toxicological knowledge and limitations of animal models, multi-cellular engineered living systems may provide solutions for modeling reproductive physiology and pathology for chemical and xenobiotic toxicity studies. Multi-cellular engineered living systems, such as microphysiological systems (MPS) and organoids, model of functional units of tissues. In this review, we highlight the key functions and structures of human reproductive organs and well-known representative toxicants afflicting these systems. We then discuss current approaches and specific studies where scientists have used MPS or organoids to recreate in vivo markers and cellular responses of the female and male reproductive system, as well as pregnancy-associated placenta formation and embryo development. We provide specific examples of organoids and organ-on-chip that have been used for toxicological purposes with varied success. Finally, we address current issues related to usage of MPS, emerging techniques for improving upon these complications, and improvements needed to make MPS more capable in assessing reproductive toxicology. Overall, multi-cellular engineered living systems have considerable promise to serve as a suitable, alternative reproductive biological model compared to animal studies and 2D culture.
Collapse
Affiliation(s)
- Isabella Lopez
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, United States
| | - George A Truskey
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, United States.
| |
Collapse
|
14
|
Cadavid JL, Li NT, McGuigan AP. Bridging systems biology and tissue engineering: Unleashing the full potential of complex 3D in vitro tissue models of disease. BIOPHYSICS REVIEWS 2024; 5:021301. [PMID: 38617201 PMCID: PMC11008916 DOI: 10.1063/5.0179125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 03/12/2024] [Indexed: 04/16/2024]
Abstract
Rapid advances in tissue engineering have resulted in more complex and physiologically relevant 3D in vitro tissue models with applications in fundamental biology and therapeutic development. However, the complexity provided by these models is often not leveraged fully due to the reductionist methods used to analyze them. Computational and mathematical models developed in the field of systems biology can address this issue. Yet, traditional systems biology has been mostly applied to simpler in vitro models with little physiological relevance and limited cellular complexity. Therefore, integrating these two inherently interdisciplinary fields can result in new insights and move both disciplines forward. In this review, we provide a systematic overview of how systems biology has been integrated with 3D in vitro tissue models and discuss key application areas where the synergies between both fields have led to important advances with potential translational impact. We then outline key directions for future research and discuss a framework for further integration between fields.
Collapse
|
15
|
Rengel BD, Schuler-Faccini L, Fraga LR, Vianna FSL, Kowalski TW. Possible New Candidates Involved to Thalidomide-Related Limbs and Cardiac Defects: A Systems Biology Approach. Biochem Genet 2024:10.1007/s10528-024-10790-w. [PMID: 38689186 DOI: 10.1007/s10528-024-10790-w] [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: 06/22/2023] [Accepted: 03/19/2024] [Indexed: 05/02/2024]
Abstract
Thalidomide is a known teratogen that causes malformations especially in heart and limbs. Its mechanism of teratogenicity is still not fully elucidated. Recently, a new target of thalidomide was described, TBX5, and was observed a new interaction between HAND2 and TBX5 that is disrupted in the presence of thalidomide. Therefore, our study aimed to raise potential candidates for thalidomide teratogenesis, through systems biology, evaluating HAND2 and TBX5 interaction and heart and limbs malformations of thalidomide. Genes and proteins related to TBX5 and HAND2 were selected through TF2DNA, REACTOME, Human Phenotype Ontology, and InterPro databases. Networks were assembled using STRING © database. Network analysis were performed in Cytoscape © and R v3.6.2. Differential gene expression (DGE) analysis was performed through gene expression omnibus. We constructed a network for HAND2 and TBX5 interaction; a network for heart and limbs malformations of TE; and the two joined networks. We observed that EP300 protein seemed to be important in all networks. We also looked for proteins containing C2H2 domain in the assembled networks. ZIC3, GLI1, GLI3, ZNF148, and PRDM16 were the ones present in both heart and limbs malformations of TE networks. Furthermore, in the DGE analysis after treatment with thalidomide, we observed that FANCB, ESCO2, and XRCC2 were downregulated and present both in heart and limbs networks. Through systems biology, we were able to point to different new proteins and genes, and selected specially EP300, which was important in all the analyzed networks, to be further evaluated in the TE teratogenicity.
Collapse
Affiliation(s)
- Bruna Duarte Rengel
- Laboratory of Medical Genetics and Evolution, Genetics Department, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Brazilian Teratogen Information Service (SIAT), Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Graduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Lavínia Schuler-Faccini
- Laboratory of Medical Genetics and Evolution, Genetics Department, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- National Institute of Population Medical Genetics (INAGEMP), Porto Alegre, Brazil
- Brazilian Teratogen Information Service (SIAT), Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Lucas Rosa Fraga
- National Institute of Population Medical Genetics (INAGEMP), Porto Alegre, Brazil
- Brazilian Teratogen Information Service (SIAT), Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Department of Morphological Sciences, Institute of Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Graduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Fernanda Sales Luiz Vianna
- Laboratory of Medical Genetics and Evolution, Genetics Department, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
- Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
- National Institute of Population Medical Genetics (INAGEMP), Porto Alegre, Brazil.
- Brazilian Teratogen Information Service (SIAT), Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.
- Graduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
- Genomic Medicine Laboratory, Hospital de Clínicas de Porto Alegre (HCPA), Ramiro Barcelos Street, 2350, Porto Alegre, CEP 90035-903, Brazil.
| | - Thayne Woycinck Kowalski
- Laboratory of Medical Genetics and Evolution, Genetics Department, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
- Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
- National Institute of Population Medical Genetics (INAGEMP), Porto Alegre, Brazil.
- Brazilian Teratogen Information Service (SIAT), Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.
- Graduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
- Genomic Medicine Laboratory, Hospital de Clínicas de Porto Alegre (HCPA), Ramiro Barcelos Street, 2350, Porto Alegre, CEP 90035-903, Brazil.
- Bioinformatics Core, Hospital de Clínicas de Porto Alegre, HCPA, Porto Alegre, Brazil.
| |
Collapse
|
16
|
Giorgi C, Castelli V, d’Angelo M, Cimini A. Organoids Modeling Stroke in a Petri Dish. Biomedicines 2024; 12:877. [PMID: 38672231 PMCID: PMC11048104 DOI: 10.3390/biomedicines12040877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Stroke is a common neurological disorder, the second leading cause of death, and the third leading cause of disability. Unfortunately, the only approved drug for it is tissue plasminogen, but the therapeutic window is limited. In this context, preclinical studies are relevant to better dissect the underlying mechanisms of stroke and for the drug screening of potential therapies. Brain organoids could be relevant in this setting. They are derived from pluripotent stem cells or isolated organ progenitors that differentiate to form an organ-like tissue, exhibiting multiple cell types that self-organize to form a structure not unlike the organ in vivo. Brain organoids mimic many key features of early human brain development at molecular, cellular, structural, and functional levels and have emerged as novel model systems that can be used to investigate human brain diseases including stroke. Brain organoids are a promising and powerful tool for ischemic stroke studies; however, there are a few concerns that need to be addressed, including the lack of vascularization and the many cell types that are typically present in the human brain. The aim of this review is to discuss the potential of brain organoids as a novel model system for studying ischemic stroke, highlighting both the advantages and disadvantages in the use of this technology.
Collapse
Affiliation(s)
| | | | - Michele d’Angelo
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (C.G.); (V.C.)
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (C.G.); (V.C.)
| |
Collapse
|
17
|
Giorgi C, Lombardozzi G, Ammannito F, Scenna MS, Maceroni E, Quintiliani M, d’Angelo M, Cimini A, Castelli V. Brain Organoids: A Game-Changer for Drug Testing. Pharmaceutics 2024; 16:443. [PMID: 38675104 PMCID: PMC11054008 DOI: 10.3390/pharmaceutics16040443] [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: 02/07/2024] [Revised: 03/12/2024] [Accepted: 03/20/2024] [Indexed: 04/28/2024] Open
Abstract
Neurological disorders are the second cause of death and the leading cause of disability worldwide. Unfortunately, no cure exists for these disorders, but the actual therapies are only able to ameliorate people's quality of life. Thus, there is an urgent need to test potential therapeutic approaches. Brain organoids are a possible valuable tool in the study of the brain, due to their ability to reproduce different brain regions and maturation stages; they can be used also as a tool for disease modelling and target identification of neurological disorders. Recently, brain organoids have been used in drug-screening processes, even if there are several limitations to overcome. This review focuses on the description of brain organoid development and drug-screening processes, discussing the advantages, challenges, and limitations of the use of organoids in modeling neurological diseases. We also highlighted the potential of testing novel therapeutic approaches. Finally, we examine the challenges and future directions to improve the drug-screening process.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Annamaria Cimini
- Department of Life, Health and Environmental Science, University of L’Aquila, 67100 L’Aquila, Italy; (C.G.); (G.L.); (F.A.); (M.S.S.); (E.M.); (M.Q.); (M.d.)
| | - Vanessa Castelli
- Department of Life, Health and Environmental Science, University of L’Aquila, 67100 L’Aquila, Italy; (C.G.); (G.L.); (F.A.); (M.S.S.); (E.M.); (M.Q.); (M.d.)
| |
Collapse
|
18
|
Maramraju S, Kowalczewski A, Kaza A, Liu X, Singaraju JP, Albert MV, Ma Z, Yang H. AI-organoid integrated systems for biomedical studies and applications. Bioeng Transl Med 2024; 9:e10641. [PMID: 38435826 PMCID: PMC10905559 DOI: 10.1002/btm2.10641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 12/07/2023] [Accepted: 12/13/2023] [Indexed: 03/05/2024] Open
Abstract
In this review, we explore the growing role of artificial intelligence (AI) in advancing the biomedical applications of human pluripotent stem cell (hPSC)-derived organoids. Stem cell-derived organoids, these miniature organ replicas, have become essential tools for disease modeling, drug discovery, and regenerative medicine. However, analyzing the vast and intricate datasets generated from these organoids can be inefficient and error-prone. AI techniques offer a promising solution to efficiently extract insights and make predictions from diverse data types generated from microscopy images, transcriptomics, metabolomics, and proteomics. This review offers a brief overview of organoid characterization and fundamental concepts in AI while focusing on a comprehensive exploration of AI applications in organoid-based disease modeling and drug evaluation. It provides insights into the future possibilities of AI in enhancing the quality control of organoid fabrication, label-free organoid recognition, and three-dimensional image reconstruction of complex organoid structures. This review presents the challenges and potential solutions in AI-organoid integration, focusing on the establishment of reliable AI model decision-making processes and the standardization of organoid research.
Collapse
Affiliation(s)
- Sudhiksha Maramraju
- Department of Biomedical EngineeringUniversity of North TexasDentonTexasUSA
- Texas Academy of Mathematics and ScienceUniversity of North TexasDentonTexasUSA
| | - Andrew Kowalczewski
- Department of Biomedical & Chemical EngineeringSyracuse UniversitySyracuseNew YorkUSA
- BioInspired Institute for Material and Living SystemsSyracuse UniversitySyracuseNew YorkUSA
| | - Anirudh Kaza
- Department of Biomedical EngineeringUniversity of North TexasDentonTexasUSA
- Texas Academy of Mathematics and ScienceUniversity of North TexasDentonTexasUSA
| | - Xiyuan Liu
- Department of Mechanical & Aerospace EngineeringSyracuse UniversitySyracuseNew YorkUSA
| | - Jathin Pranav Singaraju
- Department of Biomedical EngineeringUniversity of North TexasDentonTexasUSA
- Texas Academy of Mathematics and ScienceUniversity of North TexasDentonTexasUSA
| | - Mark V. Albert
- Department of Biomedical EngineeringUniversity of North TexasDentonTexasUSA
- Department of Computer Science and EngineeringUniversity of North TexasDentonTexasUSA
| | - Zhen Ma
- Department of Biomedical & Chemical EngineeringSyracuse UniversitySyracuseNew YorkUSA
- BioInspired Institute for Material and Living SystemsSyracuse UniversitySyracuseNew YorkUSA
| | - Huaxiao Yang
- Department of Biomedical EngineeringUniversity of North TexasDentonTexasUSA
| |
Collapse
|
19
|
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
|
20
|
Jäntti H, Kistemaker L, Buonfiglioli A, De Witte LD, Malm T, Hol EM. Emerging Models to Study Human Microglia In vitro. ADVANCES IN NEUROBIOLOGY 2024; 37:545-568. [PMID: 39207712 DOI: 10.1007/978-3-031-55529-9_30] [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: 09/04/2024]
Abstract
New in vitro models provide an exciting opportunity to study live human microglia. Previously, a major limitation in understanding human microglia in health and disease has been their limited availability. Here, we provide an overview of methods to obtain human stem cell or blood monocyte-derived microglia-like cells that provide a nearly unlimited source of live human microglia for research. We address how understanding microglial ontogeny can help modeling microglial identity and function in a dish with increased accuracy. Moreover, we categorize stem cell-derived differentiation methods into embryoid body based, growth factor driven, and coculture-driven approaches, and review novel viral approaches to reprogram stem cells directly into microglia-like cells. Furthermore, we review typical readouts used in the field to verify microglial identity and characterize functional microglial phenotypes. We provide an overview of methods used to study microglia in environments more closely resembling the (developing) human CNS, such as cocultures and brain organoid systems with incorporated or innately developing microglia. We highlight how microglia-like cells can be utilized to reveal molecular and functional mechanisms in human disease context, focusing on Alzheimer's disease and other neurodegenerative diseases as well as neurodevelopmental diseases. Finally, we provide a critical overview of challenges and future opportunities to more accurately model human microglia in a dish and conclude that novel in vitro microglia-like cells provide an exciting potential to bring preclinical research of microglia to a new era.
Collapse
Affiliation(s)
- Henna Jäntti
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Lois Kistemaker
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Alice Buonfiglioli
- Department of Psychiatry, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lot D De Witte
- Department of Psychiatry, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tarja Malm
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Elly M Hol
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
21
|
Majumder J, Torr EE, Aisenbrey EA, Lebakken CS, Favreau PF, Richards WD, Yin Y, Chang Q, Murphy WL. Human induced pluripotent stem cell-derived planar neural organoids assembled on synthetic hydrogels. J Tissue Eng 2024; 15:20417314241230633. [PMID: 38361535 PMCID: PMC10868488 DOI: 10.1177/20417314241230633] [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: 10/30/2023] [Accepted: 01/20/2024] [Indexed: 02/17/2024] Open
Abstract
The tailorable properties of synthetic polyethylene glycol (PEG) hydrogels make them an attractive substrate for human organoid assembly. Here, we formed human neural organoids from iPSC-derived progenitor cells in two distinct formats: (i) cells seeded on a Matrigel surface; and (ii) cells seeded on a synthetic PEG hydrogel surface. Tissue assembly on synthetic PEG hydrogels resulted in three dimensional (3D) planar neural organoids with greater neuronal diversity, greater expression of neurovascular and neuroinflammatory genes, and reduced variability when compared with tissues assembled upon Matrigel. Further, our 3D human tissue assembly approach occurred in an open cell culture format and created a tissue that was sufficiently translucent to allow for continuous imaging. Planar neural organoids formed on PEG hydrogels also showed higher expression of neural, vascular, and neuroinflammatory genes when compared to traditional brain organoids grown in Matrigel suspensions. Further, planar neural organoids contained functional microglia that responded to pro-inflammatory stimuli, and were responsive to anti-inflammatory drugs. These results demonstrate that the PEG hydrogel neural organoids can be used as a physiologically relevant in vitro model of neuro-inflammation.
Collapse
Affiliation(s)
- Joydeb Majumder
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, USA
| | - Elizabeth E Torr
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, USA
| | - Elizabeth A Aisenbrey
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, USA
| | | | | | | | - Yanhong Yin
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Qiang Chang
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
- Departments of Medical Genetics and Neurology, University of Wisconsin-Madison, Madison, WI, USA
| | - William L Murphy
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
22
|
Banerjee M, Das A, Chatterjee P, Banerjee S. Human Pluripotent Stem Cell-Based Assays to Predict Developmental Toxicity. Methods Mol Biol 2024; 2753:181-199. [PMID: 38285339 DOI: 10.1007/978-1-0716-3625-1_8] [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: 01/30/2024]
Abstract
Human beings are continuously exposed to various toxic substances throughout their lives, which affect their reproductive health and eventually the offspring they give birth to. Mainly, these toxins damage the heart and neurological development of the newborn, but most recently, they have begun to affect the musculoskeletal system as well. These toxins are usually present in food, pharmaceuticals, cosmetics, or even the polluted air that people breathe; as a result, the prevalence of birth defects is steadily rising. For this reason, it becomes a necessity to deploy a new set of assays to test for such toxins in industries to decrease the occurrence of developmental toxicity. These assays are exceedingly expensive when carried out conventionally using animal models or cells from such sources and have a lower predictive value due to the vast variation between animals and humans. To overcome such major problems, human pluripotent stem cells are now frequently used for these assays. These cells are easily available, are quickly generated from somatic cells (induce pluripotent stem cells), can be of human origin without harming people, and eliminate animal harm, which makes them the top choice of scientists for carrying out any in vitro developmental toxicity assays.This chapter, therefore, provides an overview of several steps that can be used to predict a compound's developmental toxicity by utilizing human pluripotent stem cells. Here, the easiest and most effective procedure has been outlined that can screen many compounds simultaneously.
Collapse
Affiliation(s)
- Madhura Banerjee
- School of BioSciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Aritrika Das
- School of BioSciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Prarthana Chatterjee
- School of BioSciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Satarupa Banerjee
- School of BioSciences and Technology, Vellore Institute of Technology, Vellore, India.
| |
Collapse
|
23
|
Di Credico A, Weiss A, Corsini M, Gaggi G, Ghinassi B, Wilbertz JH, Di Baldassarre A. Machine learning identifies phenotypic profile alterations of human dopaminergic neurons exposed to bisphenols and perfluoroalkyls. Sci Rep 2023; 13:21907. [PMID: 38081991 PMCID: PMC10713827 DOI: 10.1038/s41598-023-49364-y] [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: 09/21/2023] [Accepted: 12/07/2023] [Indexed: 12/18/2023] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease and is characterized by the loss of midbrain dopaminergic neurons. Endocrine disrupting chemicals (EDCs) are active substances that interfere with hormonal signaling. Among EDCs, bisphenols (BPs) and perfluoroalkyls (PFs) are chemicals leached from plastics and other household products, and humans are unavoidably exposed to these xenobiotics. Data from animal studies suggest that EDCs exposure may play a role in PD, but data about the effect of BPs and PFs on human models of the nervous system are lacking. Previous studies demonstrated that machine learning (ML) applied to microscopy data can classify different cell phenotypes based on image features. In this study, the effect of BPs and PFs at different concentrations within the real-life exposure range (0.01, 0.1, 1, and 2 µM) on the phenotypic profile of human stem cell-derived midbrain dopaminergic neurons (mDANs) was analyzed. Cells exposed for 72 h to the xenobiotics were stained with neuronal markers and evaluated using high content microscopy yielding 126 different phenotypic features. Three different ML models (LDA, XGBoost and LightGBM) were trained to classify EDC-treated versus control mDANs. EDC treated mDANs were identified with high accuracies (0.88-0.96). Assessment of the phenotypic feature contribution to the classification showed that EDCs induced a significant increase of alpha-synuclein (αSyn) and tyrosine hydroxylase (TH) staining intensity within the neurons. Moreover, microtubule-associated protein 2 (MAP2) neurite length and branching were significantly diminished in treated neurons. Our study shows that human mDANs are adversely impacted by exposure to EDCs, causing their phenotype to shift and exhibit more characteristics of PD. Importantly, ML-supported high-content imaging can identify concrete but subtle subcellular phenotypic changes that can be easily overlooked by visual inspection alone and that define EDCs effects in mDANs, thus enabling further pathological characterization in the future.
Collapse
Affiliation(s)
- Andrea Di Credico
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies, and Technology (CAST), 66100, Chieti, Italy
- Department of Medicine and Aging Sciences, "G. D'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
- UdATech Lab Center (UdATech), 66100, Chieti, Italy
| | | | - Massimo Corsini
- Dipartimento Di Neuroscienze Umane, "Sapienza" University of Rome, Chieti, Italy
| | - Giulia Gaggi
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies, and Technology (CAST), 66100, Chieti, Italy
- Department of Medicine and Aging Sciences, "G. D'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
- UdATech Lab Center (UdATech), 66100, Chieti, Italy
| | - Barbara Ghinassi
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies, and Technology (CAST), 66100, Chieti, Italy
- Department of Medicine and Aging Sciences, "G. D'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
- UdATech Lab Center (UdATech), 66100, Chieti, Italy
| | | | - Angela Di Baldassarre
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies, and Technology (CAST), 66100, Chieti, Italy
- Department of Medicine and Aging Sciences, "G. D'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
- UdATech Lab Center (UdATech), 66100, Chieti, Italy
| |
Collapse
|
24
|
Borenstein JT, Cummins G, Dutta A, Hamad E, Hughes MP, Jiang X, Lee HH, Lei KF, Tang XS, Zheng Y, Chen J. Bionanotechnology and bioMEMS (BNM): state-of-the-art applications, opportunities, and challenges. LAB ON A CHIP 2023; 23:4928-4949. [PMID: 37916434 DOI: 10.1039/d3lc00296a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
The development of micro- and nanotechnology for biomedical applications has defined the cutting edge of medical technology for over three decades, as advancements in fabrication technology developed originally in the semiconductor industry have been applied to solving ever-more complex problems in medicine and biology. These technologies are ideally suited to interfacing with life sciences, since they are on the scale lengths as cells (microns) and biomacromolecules (nanometers). In this paper, we review the state of the art in bionanotechnology and bioMEMS (collectively BNM), including developments and challenges in the areas of BNM, such as microfluidic organ-on-chip devices, oral drug delivery, emerging technologies for managing infectious diseases, 3D printed microfluidic devices, AC electrokinetics, flexible MEMS devices, implantable microdevices, paper-based microfluidic platforms for cellular analysis, and wearable sensors for point-of-care testing.
Collapse
Affiliation(s)
| | - Gerard Cummins
- School of Engineering, University of Birmingham, Edgbaston, B15 2TT, UK.
| | - Abhishek Dutta
- Department of Electrical & Computer Engineering, University of Connecticut, USA.
| | - Eyad Hamad
- Biomedical Engineering Department, School of Applied Medical Sciences, German Jordanian University, Amman, Jordan.
| | - Michael Pycraft Hughes
- Department of Biomedical Engineering, Khalifa University, Abu Dhabi, United Arab Emirates.
| | - Xingyu Jiang
- Department of Biomedical Engineering, Southern University of Science and Technology, China.
| | - Hyowon Hugh Lee
- Weldon School of Biomedical Engineering, Center for Implantable Devices, Purdue University, West Lafayette, IN, USA.
| | | | | | | | - Jie Chen
- Department of Electrical and Computer Engineering, University of Alberta, Edmonton, AB T6G 2R3, Canada.
| |
Collapse
|
25
|
Benwood C, Walters-Shumka J, Scheck K, Willerth SM. 3D bioprinting patient-derived induced pluripotent stem cell models of Alzheimer's disease using a smart bioink. Bioelectron Med 2023; 9:10. [PMID: 37221543 DOI: 10.1186/s42234-023-00112-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/09/2023] [Indexed: 05/25/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD), a progressive neurodegenerative disorder, is becoming increasingly prevalent as our population ages. It is characterized by the buildup of amyloid beta plaques and neurofibrillary tangles containing hyperphosphorylated-tau. The current treatments for AD do not prevent the long-term progression of the disease and pre-clinical models often do not accurately represent its complexity. Bioprinting combines cells and biomaterials to create 3D structures that replicate the native tissue environment and can be used as a tool in disease modeling or drug screening. METHODS This work differentiated both healthy and diseased patient-derived human induced pluripotent stems cells (hiPSCs) into neural progenitor cells (NPCs) that were bioprinted using the Aspect RX1 microfluidic printer into dome-shaped constructs. The combination of cells, bioink, and puromorphamine (puro)-releasing microspheres were used to mimic the in vivo environment and direct the differentiation of the NPCs into basal forebrain-resembling cholinergic neurons (BFCN). These tissue models were then characterized for cell viability, immunocytochemistry, and electrophysiology to evaluate their functionality and physiology for use as disease-specific neural models. RESULTS Tissue models were successfully bioprinted and the cells were viable for analysis after 30- and 45-day cultures. The neuronal and cholinergic markers β-tubulin III (Tuj1), forkhead box G1 (FOXG1), and choline acetyltransferase (ChAT) were identified as well as the AD markers amyloid beta and tau. Further, immature electrical activity was observed when the cells were excited with potassium chloride and acetylcholine. CONCLUSIONS This work shows the successful development of bioprinted tissue models incorporating patient derived hiPSCs. Such models can potentially be used as a tool to screen promising drug candidates for treating AD. Further, this model could be used to increase the understanding of AD progression. The use of patient derived cells also shows the potential of this model for use in personalized medicine applications.
Collapse
Affiliation(s)
- Claire Benwood
- Department of Mechanical Engineering, University of Victoria, Victoria, BC, V8P 5C2, Canada
| | | | - Kali Scheck
- Division of Medical Sciences, University of Victoria, Victoria, BC, V8P 5C2, Canada
| | - Stephanie M Willerth
- Department of Mechanical Engineering, University of Victoria, Victoria, BC, V8P 5C2, Canada.
- Division of Medical Sciences, University of Victoria, Victoria, BC, V8P 5C2, Canada.
- School of Biomedical Engineering, The University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
26
|
Hinojosa MG, Johansson Y, Cediel-Ulloa A, Ivanova E, Gabring N, Gliga A, Forsby A. Evaluation of mRNA markers in differentiating human SH-SY5Y cells for estimation of developmental neurotoxicity. Neurotoxicology 2023; 97:65-77. [PMID: 37210002 DOI: 10.1016/j.neuro.2023.05.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 05/05/2023] [Accepted: 05/17/2023] [Indexed: 05/22/2023]
Abstract
Current guidelines for developmental neurotoxicity (DNT) evaluation are based on animal models. These have limitations so more relevant, efficient and robust approaches for DNT assessment are needed. We have used the human SH-SY5Y neuroblastoma cell model to evaluate a panel of 93 mRNA markers that are frequent in Neuronal diseases and functional annotations and also differentially expressed during retinoic acid-induced differentiation in the cell model. Rotenone, valproic acid (VPA), acrylamide (ACR) and methylmercury chloride (MeHg) were used as DNT positive compounds. Tolbutamide, D-mannitol and clofibrate were used as DNT negative compounds. To determine concentrations for exposure for gene expression analysis, we developed a pipeline for neurite outgrowth assessment by live-cell imaging. In addition, cell viability was measured by the resazurin assay. Gene expression was analyzed by RT-qPCR after 6 days of exposure during differentiation to concentrations of the DNT positive compounds that affected neurite outgrowth, but with no or minimal effect on cell viability. Methylmercury affected cell viability at lower concentrations than neurite outgrowth, hence the cells were exposed with the highest non-cytotoxic concentration. Rotenone (7.3nM) induced 32 differentially expressed genes (DEGs), ACR (70µM) 8 DEGs, and VPA (75µM) 16 DEGs. No individual genes were significantly dysregulated by all 3 DNT positive compounds (p<0.05), but 9 genes were differentially expressed by 2 of them. Methylmercury (0.8nM) was used to validate the 9 DEGs. The expression of SEMA5A (encoding semaphorin 5A) and CHRNA7 (encoding nicotinic acetylcholine receptor subunit α7) was downregulated by all 4 DNT positive compounds. None of the DNT negative compounds dysregulated any of the 9 DEGs in common for the DNT positive compounds. We suggest that SEMA5A or CHRNA7 should be further evaluated as biomarkers for DNT studies in vitro since they also are involved in neurodevelopmental adverse outcomes in humans.
Collapse
Affiliation(s)
- M G Hinojosa
- Department of Biochemistry and Biophysics, Stockholm University, 106 91, Stockholm, Sweden
| | - Y Johansson
- Department of Biochemistry and Biophysics, Stockholm University, 106 91, Stockholm, Sweden
| | - A Cediel-Ulloa
- Department of Organismal Biology, Environmental Toxicology, Uppsala University, 752 36, Uppsala, Sweden
| | - E Ivanova
- Department of Biochemistry and Biophysics, Stockholm University, 106 91, Stockholm, Sweden
| | - N Gabring
- Department of Biochemistry and Biophysics, Stockholm University, 106 91, Stockholm, Sweden
| | - A Gliga
- Institute of Environmental Medicine, Karolinska Institute, Stockholm, 171 77, Sweden
| | - A Forsby
- Department of Biochemistry and Biophysics, Stockholm University, 106 91, Stockholm, Sweden
| |
Collapse
|
27
|
Analysis of Aβ-induced neurotoxicity and microglial responses in simple two- and three-dimensional human iPSC-derived cortical culture systems. Tissue Cell 2023; 81:102023. [PMID: 36709697 DOI: 10.1016/j.tice.2023.102023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 12/26/2022] [Accepted: 01/11/2023] [Indexed: 01/15/2023]
Abstract
The extracellular accumulation of amyloid-β (Aβ) in plaques and associated neurodegeneration are the pathological hallmarks of Alzheimer's disease (AD). These plaques are surrounded by microglia-the resident tissue macrophages of the brain parenchyma that originate from primitive macrophages from the embryonic yolk sac. Microglia, including a unique subpopulation called "disease-associated microglia" (DAM), are strongly implicated in AD pathology; however, their exact function and physiology remain largely unknown. Notably, simple cell and tissue culture systems that adequately recreate the brain microenvironment and can simulate critical aspects of AD pathology could fundamentally contribute to elucidating microglial function in disease development and progression. Thus, we added human-induced pluripotent stem cell (hiPSC)-induced primitive macrophages (hiMacs) to hiPSC-induced cortical neurons (cell model) and cortical organoids (tissue model). The treatment of these culture systems with the O-acyl isopeptide of Aβ1-42, which reverts to natural extracellular Aβ1-42 at neutral pH and starts self-aggregation, caused the degeneration of hiPSC-induced cortical neurons in 2D culture and within cortical organoid cultures. Notably, the hiMacs phagocytosed extracellular Aβ and exhibited a DAM-like phenotype. In both cell and tissue organoid culture systems, neurodegeneration was attenuated by the addition of hiMacs. Moreover, in cortical organoids, Aβ plaques formed more circular and fewer hotspot-like morphological structures in the vicinity of hiMacs. These findings demonstrate the utility of simple hiPSC-induced cortical cell and tissue culture systems supplemented with hiMacs for elucidating critical aspects of AD pathology, such as microglial function and physiology. Adopting such systems in routine research practice may lead to the development of novel therapeutic strategies for AD.
Collapse
|
28
|
Zilio F, Lavazza A. Consciousness in a Rotor? Science and Ethics of Potentially Conscious Human Cerebral Organoids. AJOB Neurosci 2023; 14:178-196. [PMID: 36794285 DOI: 10.1080/21507740.2023.2173329] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Human cerebral organoids are three-dimensional biological cultures grown in the laboratory to mimic as closely as possible the cellular composition, structure, and function of the corresponding organ, the brain. For now, cerebral organoids lack blood vessels and other characteristics of the human brain, but are also capable of having coordinated electrical activity. They have been usefully employed for the study of several diseases and the development of the nervous system in unprecedented ways. Research on human cerebral organoids is proceeding at a very fast pace and their complexity is bound to improve. This raises the question of whether cerebral organoids will also be able to develop the unique feature of the human brain, consciousness. If this is the case, some ethical issues would arise. In this article, we discuss the necessary neural correlates and constraints for the emergence of consciousness according to some of the most debated neuroscientific theories. Based on this, we consider what the moral status of a potentially conscious brain organoid might be, in light of ethical and ontological arguments. We conclude by proposing a precautionary principle and some leads for further investigation. In particular, we consider the outcomes of some very recent experiments as entities of a potential new kind.
Collapse
|
29
|
Rajput A, Sevalkar G, Pardeshi K, Pingale P. COMPUTATIONAL NANOSCIENCE AND TECHNOLOGY. OPENNANO 2023. [DOI: 10.1016/j.onano.2023.100147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
|
30
|
Abdal Dayem A, Lee SB, Lim KM, Kim A, Shin HJ, Vellingiri B, Kim YB, Cho SG. Bioactive peptides for boosting stem cell culture platform: Methods and applications. Biomed Pharmacother 2023; 160:114376. [PMID: 36764131 DOI: 10.1016/j.biopha.2023.114376] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
Peptides, short protein fragments, can emulate the functions of their full-length native counterparts. Peptides are considered potent recombinant protein alternatives due to their specificity, high stability, low production cost, and ability to be easily tailored and immobilized. Stem cell proliferation and differentiation processes are orchestrated by an intricate interaction between numerous growth factors and proteins and their target receptors and ligands. Various growth factors, functional proteins, and cellular matrix-derived peptides efficiently enhance stem cell adhesion, proliferation, and directed differentiation. For that, peptides can be immobilized on a culture plate or conjugated to scaffolds, such as hydrogels or synthetic matrices. In this review, we assess the applications of a variety of peptides in stem cell adhesion, culture, organoid assembly, proliferation, and differentiation, describing the shortcomings of recombinant proteins and their full-length counterparts. Furthermore, we discuss the challenges of peptide applications in stem cell culture and materials design, as well as provide a brief outlook on future directions to advance peptide applications in boosting stem cell quality and scalability for clinical applications in tissue regeneration.
Collapse
Affiliation(s)
- Ahmed Abdal Dayem
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea
| | - Soo Bin Lee
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea
| | - Kyung Min Lim
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea; R&D Team, StemExOne co., ltd. 303, Life Science Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Aram Kim
- Department of Urology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05029, Republic of Korea; R&D Team, StemExOne co., ltd. 303, Life Science Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Hyun Jin Shin
- Department of Ophthalmology, Research Institute of Medical Science, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05029, Republic of Korea; R&D Team, StemExOne co., ltd. 303, Life Science Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Balachandar Vellingiri
- Stem cell and Regenerative Medicine/Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab (CUPB), Bathinda 151401, Punjab, India
| | - Young Bong Kim
- Department of Biomedical Science & Engineering, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea; R&D Team, StemExOne co., ltd. 303, Life Science Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| |
Collapse
|
31
|
Whalen S, Inoue F, Ryu H, Fair T, Markenscoff-Papadimitriou E, Keough K, Kircher M, Martin B, Alvarado B, Elor O, Laboy Cintron D, Williams A, Hassan Samee MA, Thomas S, Krencik R, Ullian EM, Kriegstein A, Rubenstein JL, Shendure J, Pollen AA, Ahituv N, Pollard KS. Machine learning dissection of human accelerated regions in primate neurodevelopment. Neuron 2023; 111:857-873.e8. [PMID: 36640767 PMCID: PMC10023452 DOI: 10.1016/j.neuron.2022.12.026] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 09/29/2022] [Accepted: 12/18/2022] [Indexed: 01/15/2023]
Abstract
Using machine learning (ML), we interrogated the function of all human-chimpanzee variants in 2,645 human accelerated regions (HARs), finding 43% of HARs have variants with large opposing effects on chromatin state and 14% on neurodevelopmental enhancer activity. This pattern, consistent with compensatory evolution, was confirmed using massively parallel reporter assays in chimpanzee and human neural progenitor cells. The species-specific enhancer activity of HARs was accurately predicted from the presence and absence of transcription factor footprints in each species. Despite these striking cis effects, activity of a given HAR sequence was nearly identical in human and chimpanzee cells. This suggests that HARs did not evolve to compensate for changes in the trans environment but instead altered their ability to bind factors present in both species. Thus, ML prioritized variants with functional effects on human neurodevelopment and revealed an unexpected reason why HARs may have evolved so rapidly.
Collapse
Affiliation(s)
- Sean Whalen
- Gladstone Institutes, San Francisco, CA 94158, USA
| | - Fumitaka Inoue
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA; Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - Hane Ryu
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA; Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA; Pharmaceutical Sciences and Pharmacogenomics Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Tyler Fair
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | | | - Kathleen Keough
- Gladstone Institutes, San Francisco, CA 94158, USA; Pharmaceutical Sciences and Pharmacogenomics Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Martin Kircher
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 10117 Berlin, Germany; Institute of Human Genetics, University Medical Center Schleswig-Holstein, University of Lübeck, 23562 Lübeck, Germany
| | - Beth Martin
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Beatriz Alvarado
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Orry Elor
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA; Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - Dianne Laboy Cintron
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA; Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | | | | | - Sean Thomas
- Gladstone Institutes, San Francisco, CA 94158, USA
| | - Robert Krencik
- Department of Neurosurgery, Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, USA
| | - Erik M Ullian
- Departments of Ophthalmology and Physiology, University of California, San Francisco, San Francisco, CA, USA; Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, USA
| | - Arnold Kriegstein
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - John L Rubenstein
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA, USA
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA; Howard Hughes Medical Institute, Seattle, WA 98195, USA; Brotman Baty Institute for Precision Medicine, Seattle, WA 98195, USA
| | - Alex A Pollen
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Nadav Ahituv
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA; Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA.
| | - Katherine S Pollard
- Gladstone Institutes, San Francisco, CA 94158, USA; Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA; Department of Epidemiology and Biostatistics and Institute for Computational Health Sciences, University of California, San Francisco, San Francisco, CA, USA; Chan-Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
32
|
Wang Z, Luo X, Luo Z, Tan Y, He G, Li P, Yang X. Transcriptome sequencing reveals neurotoxicity in embryonic neural stem/progenitor cells under heat stress. Toxicol In Vitro 2023; 86:105486. [DOI: 10.1016/j.tiv.2022.105486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 09/19/2022] [Accepted: 10/16/2022] [Indexed: 12/05/2022]
|
33
|
Wang X, Gao Z, Zhou W. Mass spectrometry-based quantitation combined with time-dependent metabolomics to discover metabolic features in human neurogenesis using neural constructs generated from neural progenitor cells. Analyst 2023; 148:609-617. [PMID: 36594636 DOI: 10.1039/d2an01162j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Direct studies focusing on the human brain are difficult to plan and conduct due to ethical and practical reasons. The advent of human pluripotent stem cell (hPSC)-derived neurons has revolutionized the research of the human brain and central nervous system, but relevant analytical techniques have been much less explored. Herein, we have designed a novel bioanalytical strategy to discover the characteristics of human neurogenesis using liquid chromatography-mass spectrometry-based quantitation and time-dependent metabolomics in combination with hPSC-derived neural constructs. To examine the growth of neurons in vitro, a quantitative method for the simultaneous measurement of N-acetylaspartic acid (NAA) and N-acetylglutamic acid (NAG) in a culture medium was established. The analysis of endogenous NAA and NAG concentrations over 28 days of neural cell culture not only illustrated the growth and maturation process of neural progenitors, but also confirmed the successful achievement of human neural constructs. Depending on the quantitative results, day 0, 10, 18, and 28 samples representing different growth phases were selected for further investigation of the global metabolic changes in developing human neurons. A versatile non-targeted, time-dependent metabolomics study identified 17 significantly changed metabolites and revealed the altered metabolic pathways including amino acid metabolism (tryptophan, phenylalanine, aspartate and beta-alanine metabolisms), pantothenate and coenzyme A biosynthesis, fatty acid metabolism, and purine and pyrimidine metabolism. The new metabolite profiles and overall metabolic pathways advance our understanding of human neurodevelopment. Additionally, the bioanalytical approach proposed in this study opens an interesting window for the capture and evaluation of the complex metabolic states of human neural cells, which would potentially be utilized in other in vitro models relevant to pathophysiology and treatment of neurological disorders, benefiting biomarker discovery and metabolic mechanism interpretation.
Collapse
Affiliation(s)
- Xin Wang
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China.
| | - Zhenye Gao
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China.
| | - Wenxiu Zhou
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China.
| |
Collapse
|
34
|
Sokolowski DJ, Ahn J, Erdman L, Hou H, Ellis K, Wang L, Goldenberg A, Wilson M. Differential Expression Enrichment Tool (DEET): an interactive atlas of human differential gene expression. NAR Genom Bioinform 2023; 5:lqad003. [PMID: 36694664 PMCID: PMC9869326 DOI: 10.1093/nargab/lqad003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 12/14/2022] [Accepted: 01/19/2023] [Indexed: 01/24/2023] Open
Abstract
Differential gene expression analysis using RNA sequencing (RNA-seq) data is a standard approach for making biological discoveries. Ongoing large-scale efforts to process and normalize publicly available gene expression data enable rapid and systematic reanalysis. While several powerful tools systematically process RNA-seq data, enabling their reanalysis, few resources systematically recompute differentially expressed genes (DEGs) generated from individual studies. We developed a robust differential expression analysis pipeline to recompute 3162 human DEG lists from The Cancer Genome Atlas, Genotype-Tissue Expression Consortium, and 142 studies within the Sequence Read Archive. After measuring the accuracy of the recomputed DEG lists, we built the Differential Expression Enrichment Tool (DEET), which enables users to interact with the recomputed DEG lists. DEET, available through CRAN and RShiny, systematically queries which of the recomputed DEG lists share similar genes, pathways, and TF targets to their own gene lists. DEET identifies relevant studies based on shared results with the user's gene lists, aiding in hypothesis generation and data-driven literature review.
Collapse
Affiliation(s)
| | - Jedid Ahn
- Genetics and Genome Biology, SickKids Research Institute, Toronto, ON, Canada,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada,Department of Computer Science, University of Toronto, Toronto, ON, Canada
| | - Lauren Erdman
- Genetics and Genome Biology, SickKids Research Institute, Toronto, ON, Canada,Department of Computer Science, University of Toronto, Toronto, ON, Canada
| | - Huayun Hou
- Genetics and Genome Biology, SickKids Research Institute, Toronto, ON, Canada
| | - Kai Ellis
- Genetics and Genome Biology, SickKids Research Institute, Toronto, ON, Canada,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Liangxi Wang
- Genetics and Genome Biology, SickKids Research Institute, Toronto, ON, Canada,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Anna Goldenberg
- Genetics and Genome Biology, SickKids Research Institute, Toronto, ON, Canada,Department of Computer Science, University of Toronto, Toronto, ON, Canada,Vector Institute, Toronto, ON, Canada,CIFAR, Toronto, ON, Canada
| | - Michael D Wilson
- To whom correspondence should be addressed. Tel: +1 416 813 7654 (Ext 328699); Fax: +1 416 813 4931;
| |
Collapse
|
35
|
Sullivan KM, Ko E, Kim EM, Ballance WC, Ito JD, Chalifoux M, Kim YJ, Bashir R, Kong H. Extracellular Microenvironmental Control for Organoid Assembly. TISSUE ENGINEERING. PART B, REVIEWS 2022; 28:1209-1222. [PMID: 35451330 PMCID: PMC9836674 DOI: 10.1089/ten.teb.2021.0186] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 03/04/2022] [Indexed: 01/22/2023]
Abstract
Organoids, which are multicellular clusters with similar physiological functions to living organs, have gained increasing attention in bioengineering. As organoids become more advanced, methods to form complex structures continue to develop. There is evidence that the extracellular microenvironment can regulate organoid quality. The extracellular microenvironment consists of soluble bioactive molecules, extracellular matrix, and biofluid flow. However, few efforts have been made to discuss the microenvironment optimal to engineer specific organoids. Therefore, this review article examines the extent to which engineered extracellular microenvironments regulate organoid quality. First, we summarize the natural tissue and organ's unique chemical and mechanical properties, guiding researchers to design an extracellular microenvironment used for organoid engineering. Then, we summarize how the microenvironments contribute to the formation and growth of the brain, lung, intestine, liver, retinal, and kidney organoids. The approaches to forming and evaluating the resulting organoids are also discussed in detail. Impact statement Organoids, which are multicellular clusters with similar physiological function to living organs, have been gaining increasing attention in bioengineering. As organoids become more advanced, methods to form complex structures continue to develop. This review article focuses on recent efforts to engineer the extracellular microenvironment in organoid research. We summarized the natural organ's microenvironment, which informs researchers of key factors that can influence organoid formation. Then, we summarize how these microenvironmental controls significantly contribute to the formation and growth of the corresponding brain, lung, intestine, liver, retinal, and kidney organoids. The approaches to forming and evaluating the resulting organoids are discussed in detail, including extracellular matrix choice and properties, culture methods, and the evaluation of the morphology and functionality through imaging and biochemical analysis.
Collapse
Affiliation(s)
- Kathryn M. Sullivan
- Department of Bioengineering, and University of Illinois at Urbana−Champaign, Urbana, Illinois, USA
| | - Eunkyung Ko
- Department of Bioengineering, and University of Illinois at Urbana−Champaign, Urbana, Illinois, USA
| | - Eun Mi Kim
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana−Champaign, Urbana, Illinois, USA
| | - William C. Ballance
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana−Champaign, Urbana, Illinois, USA
| | - John D. Ito
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana−Champaign, Urbana, Illinois, USA
| | - Madeleine Chalifoux
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana−Champaign, Urbana, Illinois, USA
| | - Young Jun Kim
- Environmental Safety Group, Korea Institute of Science and Technology (KIST–Europe), Saarbrucken, Germany
| | - Rashid Bashir
- Department of Bioengineering, and University of Illinois at Urbana−Champaign, Urbana, Illinois, USA
| | - Hyunjoon Kong
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana−Champaign, Urbana, Illinois, USA
| |
Collapse
|
36
|
Jiang L, Li Q, Liang W, Du X, Yang Y, Zhang Z, Xu L, Zhang J, Li J, Chen Z, Gu Z. Organ-On-A-Chip Database Revealed-Achieving the Human Avatar in Silicon. Bioengineering (Basel) 2022; 9:685. [PMID: 36421086 PMCID: PMC9687773 DOI: 10.3390/bioengineering9110685] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/08/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Organ-on-a-chip (OOC) provides microphysiological conditions on a microfluidic chip, which makes up for the shortcomings of traditional in vitro cellular culture models and animal models. It has broad application prospects in drug development and screening, toxicological mechanism research, and precision medicine. A large amount of data could be generated through its applications, including image data, measurement data from sensors, ~omics data, etc. A database with proper architecture is required to help scholars in this field design experiments, organize inputted data, perform analysis, and promote the future development of novel OOC systems. In this review, we overview existing OOC databases that have been developed, including the BioSystics Analytics Platform (BAP) developed by the University of Pittsburgh, which supports study design as well as data uploading, storage, visualization, analysis, etc., and the organ-on-a-chip database (Ocdb) developed by Southeast University, which has collected a large amount of literature and patents as well as relevant toxicological and pharmaceutical data and provides other major functions. We used examples to overview how the BAP database has contributed to the development and applications of OOC technology in the United States for the MPS consortium and how the Ocdb has supported researchers in the Chinese Organoid and Organs-On-A-Chip society. Lastly, the characteristics, advantages, and limitations of these two databases were discussed.
Collapse
Affiliation(s)
- Lincao Jiang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, SiPaiLou # 2, Nanjing 210096, China
| | - Qiwei Li
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, SiPaiLou # 2, Nanjing 210096, China
| | - Weicheng Liang
- School of Life Science and Technology, Southeast University, Nanjing 210096, China
| | - Xuan Du
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, SiPaiLou # 2, Nanjing 210096, China
| | - Yi Yang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, SiPaiLou # 2, Nanjing 210096, China
| | - Zilin Zhang
- Jiangsu Avartarget Biotechnology Corp., Suzhou 215163, China
| | - Lili Xu
- Jiangsu Avartarget Biotechnology Corp., Suzhou 215163, China
| | - Jing Zhang
- Jiangsu Avartarget Biotechnology Corp., Suzhou 215163, China
| | - Jian Li
- School of Life Science and Technology, Southeast University, Nanjing 210096, China
| | - Zaozao Chen
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, SiPaiLou # 2, Nanjing 210096, China
| | - Zhongze Gu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, SiPaiLou # 2, Nanjing 210096, China
| |
Collapse
|
37
|
Cakir B, Kiral FR, Park IH. Advanced in vitro models: Microglia in action. Neuron 2022; 110:3444-3457. [PMID: 36327894 DOI: 10.1016/j.neuron.2022.10.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/28/2022] [Accepted: 10/04/2022] [Indexed: 11/05/2022]
Abstract
In the central nervous system (CNS), microglia carry out multiple tasks related to brain development, maintenance of brain homeostasis, and function of the CNS. Recent advanced in vitro model systems allow us to perform more detailed and specific analyses of microglial functions in the CNS. The development of human pluripotent stem cells (hPSCs)-based 2D and 3D cell culture methods, particularly advancements in brain organoid models, offers a better platform to dissect microglial function in various contexts. Despite the improvement of these methods, there are still definite restrictions. Understanding their drawbacks and benefits ensures their proper use. In this primer, we review current developments regarding in vitro microglial production and characterization and their use to address fundamental questions about microglial function in healthy and diseased states, and we discuss potential future improvements with a particular emphasis on brain organoid models.
Collapse
Affiliation(s)
- Bilal Cakir
- Department of Genetics, Yale Stem Cell Center, Child Study Center, Yale School of Medicine, New Haven, CT 06520, USA.
| | - Ferdi Ridvan Kiral
- Department of Genetics, Yale Stem Cell Center, Child Study Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - In-Hyun Park
- Department of Genetics, Yale Stem Cell Center, Child Study Center, Yale School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
38
|
Damianidou E, Mouratidou L, Kyrousi C. Research models of neurodevelopmental disorders: The right model in the right place. Front Neurosci 2022; 16:1031075. [PMID: 36340790 PMCID: PMC9630472 DOI: 10.3389/fnins.2022.1031075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 10/07/2022] [Indexed: 11/25/2022] Open
Abstract
Neurodevelopmental disorders (NDDs) are a heterogeneous group of impairments that affect the development of the central nervous system leading to abnormal brain function. NDDs affect a great percentage of the population worldwide, imposing a high societal and economic burden and thus, interest in this field has widely grown in recent years. Nevertheless, the complexity of human brain development and function as well as the limitations regarding human tissue usage make their modeling challenging. Animal models play a central role in the investigation of the implicated molecular and cellular mechanisms, however many of them display key differences regarding human phenotype and in many cases, they partially or completely fail to recapitulate them. Although in vitro two-dimensional (2D) human-specific models have been highly used to address some of these limitations, they lack crucial features such as complexity and heterogeneity. In this review, we will discuss the advantages, limitations and future applications of in vivo and in vitro models that are used today to model NDDs. Additionally, we will describe the recent development of 3-dimensional brain (3D) organoids which offer a promising approach as human-specific in vitro models to decipher these complex disorders.
Collapse
Affiliation(s)
- Eleni Damianidou
- University Mental Health, Neurosciences and Precision Medicine Research Institute “Costas Stefanis”, Athens, Greece
| | - Lidia Mouratidou
- University Mental Health, Neurosciences and Precision Medicine Research Institute “Costas Stefanis”, Athens, Greece
- First Department of Psychiatry, Medical School, Eginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina Kyrousi
- University Mental Health, Neurosciences and Precision Medicine Research Institute “Costas Stefanis”, Athens, Greece
- First Department of Psychiatry, Medical School, Eginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
- *Correspondence: Christina Kyrousi,
| |
Collapse
|
39
|
Quality criteria for in vitro human pluripotent stem cell-derived models of tissue-based cells. Reprod Toxicol 2022; 112:36-50. [PMID: 35697279 DOI: 10.1016/j.reprotox.2022.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/27/2022] [Accepted: 06/07/2022] [Indexed: 12/21/2022]
Abstract
The advent of the technology to isolate or generate human pluripotent stem cells provided the potential to develop a wide range of human models that could enhance understanding of mechanisms underlying human development and disease. These systems are now beginning to mature and provide the basis for the development of in vitro assays suitable to understand the biological processes involved in the multi-organ systems of the human body, and will improve strategies for diagnosis, prevention, therapies and precision medicine. Induced pluripotent stem cell lines are prone to phenotypic and genotypic changes and donor/clone dependent variability, which means that it is important to identify the most appropriate characterization markers and quality control measures when sourcing new cell lines and assessing differentiated cell and tissue culture preparations for experimental work. This paper considers those core quality control measures for human pluripotent stem cell lines and evaluates the state of play in the development of key functional markers for their differentiated cell derivatives to promote assurance of reproducibility of scientific data derived from pluripotent stem cell-based systems.
Collapse
|
40
|
Cuní-López C, Stewart R, Quek H, White AR. Recent Advances in Microglia Modelling to Address Translational Outcomes in Neurodegenerative Diseases. Cells 2022; 11:cells11101662. [PMID: 35626698 PMCID: PMC9140031 DOI: 10.3390/cells11101662] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/13/2022] [Accepted: 05/16/2022] [Indexed: 12/12/2022] Open
Abstract
Neurodegenerative diseases are deteriorating conditions of the nervous system that are rapidly increasing in the aging population. Increasing evidence suggests that neuroinflammation, largely mediated by microglia, the resident immune cells of the brain, contributes to the onset and progression of neurodegenerative diseases. Hence, microglia are considered a major therapeutic target that could potentially yield effective disease-modifying treatments for neurodegenerative diseases. Despite the interest in studying microglia as drug targets, the availability of cost-effective, flexible, and patient-specific microglia cellular models is limited. Importantly, the current model systems do not accurately recapitulate important pathological features or disease processes, leading to the failure of many therapeutic drugs. Here, we review the key roles of microglia in neurodegenerative diseases and provide an update on the current microglia platforms utilised in neurodegenerative diseases, with a focus on human microglia-like cells derived from peripheral blood mononuclear cells as well as human-induced pluripotent stem cells. The described microglial platforms can serve as tools for investigating disease biomarkers and improving the clinical translatability of the drug development process in neurodegenerative diseases.
Collapse
Affiliation(s)
- Carla Cuní-López
- Cell & Molecular Biology Department, Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; (C.C.-L.); (R.S.)
- Faculty of Medicine, The University of Queensland, Brisbane, QLD 4006, Australia
| | - Romal Stewart
- Cell & Molecular Biology Department, Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; (C.C.-L.); (R.S.)
- UQ Centre for Clinical Research, The University of Queensland, Royal Brisbane & Women’s Hospital, Brisbane, QLD 4006, Australia
| | - Hazel Quek
- Cell & Molecular Biology Department, Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; (C.C.-L.); (R.S.)
- Correspondence: (H.Q.); (A.R.W.)
| | - Anthony R. White
- Cell & Molecular Biology Department, Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; (C.C.-L.); (R.S.)
- Correspondence: (H.Q.); (A.R.W.)
| |
Collapse
|
41
|
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: 37] [Impact Index Per Article: 12.3] [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
|
42
|
Rathore RS, R Ayyannan S, Mahto SK. Emerging three-dimensional neuronal culture assays for neurotherapeutics drug discovery. Expert Opin Drug Discov 2022; 17:619-628. [DOI: 10.1080/17460441.2022.2061458] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Rahul S Rathore
- Pharmaceutical Chemistry Research Laboratory II, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi-221005, UP, India
| | - Senthil R Ayyannan
- Pharmaceutical Chemistry Research Laboratory II, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi-221005, UP, India
| | - Sanjeev K Mahto
- Tissue Engineering and Biomicrofluidics Laboratory, School of Biomedical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi-221005, UP, India
| |
Collapse
|
43
|
Hu B, Cheng Z, Liang S. Advantages and prospects of stem cells in nanotoxicology. CHEMOSPHERE 2022; 291:132861. [PMID: 34774913 DOI: 10.1016/j.chemosphere.2021.132861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 11/06/2021] [Accepted: 11/08/2021] [Indexed: 06/13/2023]
Abstract
Nanomaterials have been widely used in many fields, especially in biomedical and stem cell therapy. However, the potential risks associated with nanomaterials applications are also gradually increasing. Therefore, effective and robust toxicology models are critical to evaluate the developmental toxicity of nanomaterials. The development of stem cell research provides a new idea of developmental toxicology. Recently, many researchers actively investigated the effects of nanomaterials with different sizes and surface modifications on various stem cells (such as embryonic stem cells (ESCs), adult stem cells, etc.) to study the toxic effects and toxic mechanisms. In this review, we summarized the effects of nanomaterials on the proliferation and differentiation of ESCs, mesenchymal stem cells and neural stem cells. Moreover, we discussed the advantages of stem cells in nanotoxicology compared with other cell lines. Finally, combined with the latest research methods and new molecular mechanisms, we analyzed the application of stem cells in nanotoxicology.
Collapse
Affiliation(s)
- Bowen Hu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, Xinjiang, 830017, China.
| | - Zhanwen Cheng
- School of Environmental Science and Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Shengxian Liang
- Institute of Life Sciences and Green Development, College of Life Sciences, Hebei University, Baoding, 071000, China
| |
Collapse
|
44
|
Youhanna S, Kemas AM, Preiss L, Zhou Y, Shen JX, Cakal SD, Paqualini FS, Goparaju SK, Shafagh RZ, Lind JU, Sellgren CM, Lauschke VM. Organotypic and Microphysiological Human Tissue Models for Drug Discovery and Development-Current State-of-the-Art and Future Perspectives. Pharmacol Rev 2022; 74:141-206. [PMID: 35017176 DOI: 10.1124/pharmrev.120.000238] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 10/12/2021] [Indexed: 12/11/2022] Open
Abstract
The number of successful drug development projects has been stagnant for decades despite major breakthroughs in chemistry, molecular biology, and genetics. Unreliable target identification and poor translatability of preclinical models have been identified as major causes of failure. To improve predictions of clinical efficacy and safety, interest has shifted to three-dimensional culture methods in which human cells can retain many physiologically and functionally relevant phenotypes for extended periods of time. Here, we review the state of the art of available organotypic culture techniques and critically review emerging models of human tissues with key importance for pharmacokinetics, pharmacodynamics, and toxicity. In addition, developments in bioprinting and microfluidic multiorgan cultures to emulate systemic drug disposition are summarized. We close by highlighting important trends regarding the fabrication of organotypic culture platforms and the choice of platform material to limit drug absorption and polymer leaching while supporting the phenotypic maintenance of cultured cells and allowing for scalable device fabrication. We conclude that organotypic and microphysiological human tissue models constitute promising systems to promote drug discovery and development by facilitating drug target identification and improving the preclinical evaluation of drug toxicity and pharmacokinetics. There is, however, a critical need for further validation, benchmarking, and consolidation efforts ideally conducted in intersectoral multicenter settings to accelerate acceptance of these novel models as reliable tools for translational pharmacology and toxicology. SIGNIFICANCE STATEMENT: Organotypic and microphysiological culture of human cells has emerged as a promising tool for preclinical drug discovery and development that might be able to narrow the translation gap. This review discusses recent technological and methodological advancements and the use of these systems for hit discovery and the evaluation of toxicity, clearance, and absorption of lead compounds.
Collapse
Affiliation(s)
- Sonia Youhanna
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Aurino M Kemas
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Lena Preiss
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Yitian Zhou
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Joanne X Shen
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Selgin D Cakal
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Francesco S Paqualini
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Sravan K Goparaju
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Reza Zandi Shafagh
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Johan Ulrik Lind
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Carl M Sellgren
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| |
Collapse
|
45
|
Kofman S, Mohan N, Sun X, Ibric L, Piermarini E, Qiang L. Human mini brains and spinal cords in a dish: Modeling strategies, current challenges, and prospective advances. J Tissue Eng 2022; 13:20417314221113391. [PMID: 35898331 PMCID: PMC9310295 DOI: 10.1177/20417314221113391] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/27/2022] [Indexed: 11/15/2022] Open
Abstract
Engineered three-dimensional (3D) in vitro and ex vivo neural tissues, also known as "mini brains and spinal cords in a dish," can be derived from different types of human stem cells via several differentiation protocols. In general, human mini brains are micro-scale physiological systems consisting of mixed populations of neural progenitor cells, glial cells, and neurons that may represent key features of human brain anatomy and function. To date, these specialized 3D tissue structures can be characterized into spheroids, organoids, assembloids, organ-on-a-chip and their various combinations based on generation procedures and cellular components. These 3D CNS models incorporate complex cell-cell interactions and play an essential role in bridging the gap between two-dimensional human neuroglial cultures and animal models. Indeed, they provide an innovative platform for disease modeling and therapeutic cell replacement, especially shedding light on the potential to realize personalized medicine for neurological disorders when combined with the revolutionary human induced pluripotent stem cell technology. In this review, we highlight human 3D CNS models developed from a variety of experimental strategies, emphasize their advances and remaining challenges, evaluate their state-of-the-art applications in recapitulating crucial phenotypic aspects of many CNS diseases, and discuss the role of contemporary technologies in the prospective improvement of their composition, consistency, complexity, and maturation.
Collapse
Affiliation(s)
- Simeon Kofman
- Department of Neurobiology and Anatomy, Drexel
University College of Medicine, Philadelphia, PA, USA
| | - Neha Mohan
- Department of Neurobiology and Anatomy, Drexel
University College of Medicine, Philadelphia, PA, USA
| | - Xiaohuan Sun
- Department of Neurobiology and Anatomy, Drexel
University College of Medicine, Philadelphia, PA, USA
| | - Larisa Ibric
- Department of Neurobiology and Anatomy, Drexel
University College of Medicine, Philadelphia, PA, USA
| | - Emanuela Piermarini
- Department of Neurobiology and Anatomy, Drexel
University College of Medicine, Philadelphia, PA, USA
| | - Liang Qiang
- Department of Neurobiology and Anatomy, Drexel
University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
46
|
Ryan AR, Cleaver O. Plumbing our organs: Lessons from vascular development to instruct lab generated tissues. Curr Top Dev Biol 2022; 148:165-194. [DOI: 10.1016/bs.ctdb.2022.02.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
47
|
Functional Characterization of Human Pluripotent Stem Cell-Derived Models of the Brain with Microelectrode Arrays. Cells 2021; 11:cells11010106. [PMID: 35011667 PMCID: PMC8750870 DOI: 10.3390/cells11010106] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/22/2021] [Accepted: 12/24/2021] [Indexed: 12/26/2022] Open
Abstract
Human pluripotent stem cell (hPSC)-derived neuron cultures have emerged as models of electrical activity in the human brain. Microelectrode arrays (MEAs) measure changes in the extracellular electric potential of cell cultures or tissues and enable the recording of neuronal network activity. MEAs have been applied to both human subjects and hPSC-derived brain models. Here, we review the literature on the functional characterization of hPSC-derived two- and three-dimensional brain models with MEAs and examine their network function in physiological and pathological contexts. We also summarize MEA results from the human brain and compare them to the literature on MEA recordings of hPSC-derived brain models. MEA recordings have shown network activity in two-dimensional hPSC-derived brain models that is comparable to the human brain and revealed pathology-associated changes in disease models. Three-dimensional hPSC-derived models such as brain organoids possess a more relevant microenvironment, tissue architecture and potential for modeling the network activity with more complexity than two-dimensional models. hPSC-derived brain models recapitulate many aspects of network function in the human brain and provide valid disease models, but certain advancements in differentiation methods, bioengineering and available MEA technology are needed for these approaches to reach their full potential.
Collapse
|
48
|
Abstract
Organoids-cellular aggregates derived from stem or progenitor cells that recapitulate organ function in miniature-are of growing interest in developmental biology and medicine. Organoids have been developed for organs and tissues such as the liver, gut, brain, and pancreas; they are used as organ surrogates to study a wide range of questions in basic and developmental biology, genetic disorders, and therapies. However, many organoids reported to date have been cultured in Matrigel, which is prepared from the secretion of Engelbreth-Holm-Swarm mouse sarcoma cells; Matrigel is complex and poorly defined. This complexity makes it difficult to elucidate Matrigel-specific factors governing organoid development. In this review, we discuss promising Matrigel-free methods for the generation and maintenance of organoids that use decellularized extracellular matrix (ECM), synthetic hydrogels, or gel-forming recombinant proteins.
Collapse
Affiliation(s)
- Mark T Kozlowski
- DEVCOM US Army Research Laboratory, Weapons and Materials Research Directorate, Science of Extreme Materials Division, Polymers Branch, 6300 Rodman Rd. Building 4600, Aberdeen Proving Ground, Aberdeen, MD, 21005, USA.
| | - Christiana J Crook
- Department of Translational Research and Cellular Therapeutics, Diabetes and Metabolism Research Institute, City of Hope National Medical Center, 1500 Duarte Rd., Duarte, CA, 91010, USA
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, 1500 Duarte Rd., Duarte, CA, 91010, USA
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, 1500 Duarte Rd., Duarte, CA, 91010, USA
| | - Hsun Teresa Ku
- Department of Translational Research and Cellular Therapeutics, Diabetes and Metabolism Research Institute, City of Hope National Medical Center, 1500 Duarte Rd., Duarte, CA, 91010, USA
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, 1500 Duarte Rd., Duarte, CA, 91010, USA
| |
Collapse
|
49
|
Capturing the third dimension in drug discovery: Spatially-resolved tools for interrogation of complex 3D cell models. Biotechnol Adv 2021; 55:107883. [PMID: 34875362 DOI: 10.1016/j.biotechadv.2021.107883] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 11/22/2021] [Accepted: 11/30/2021] [Indexed: 02/07/2023]
Abstract
Advanced three-dimensional (3D) cell models have proven to be capable of depicting architectural and microenvironmental features of several tissues. By providing data of higher physiological and pathophysiological relevance, 3D cell models have been contributing to a better understanding of human development, pathology onset and progression mechanisms, as well as for 3D cell-based assays for drug discovery. Nonetheless, the characterization and interrogation of these tissue-like structures pose major challenges on the conventional analytical methods, pushing the development of spatially-resolved technologies. Herein, we review recent advances and pioneering technologies suitable for the interrogation of multicellular 3D models, while capable of retaining biological spatial information. We focused on imaging technologies and omics tools, namely transcriptomics, proteomics and metabolomics. The advantages and shortcomings of these novel methodologies are discussed, alongside the opportunities to intertwine data from the different tools.
Collapse
|
50
|
Ma S, Cong Z, Chen H, Wen H, Cao L, Liu C, Yang F, Liao Y. Velvet antler polypeptide-loaded polyvinyl alcohol-sodium alginate hydrogels promote the differentiation of neural progenitor cells in 3D towards oligodendrocytes in vitro. Eur J Pharm Sci 2021; 167:106003. [PMID: 34517106 DOI: 10.1016/j.ejps.2021.106003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 08/03/2021] [Accepted: 09/08/2021] [Indexed: 01/10/2023]
Abstract
Three-dimensional (3D) culture of neural progenitor cells (NPCs) in hydrogels represents a powerful means for recapitulating neurodevelopment, disease modelling and drug discovery. However, the differentiation of NPCs to oligodendrocytes in 3D scaffolds remains a great challenge. In this study, polyvinyl alcohol (PVA) - sodium alginate (SA) composite hydrogels intended for NPC culture in 3D were fabricated by ionic crosslinking between SA and calcium ions. It was demonstrated that adding PVA to the composite hydrogels resulted in increases in pore size and swelling rate and decreases in elastic moduli as the PVA proportion was enhanced. In addition, the composite hydrogels were biocompatible with mouse NPCs and improved the proliferation of the encapsulated NPCs compared with SA hydrogels. Moreover, when velvet antler polypeptides (VAPs), which were capable of facilitating the differentiation of NPCs to oligodendrocyte fate in 2D, were loaded into PVA-SA hydrogels, NPCs differentiated into neurons, astrocytes and oligodendrocytes, with the presence of VAPs promoting oligodendrogenesis in a dose-dependant manner. The present composite hydrogels provide a suitable scaffold for the construction of neural tissue engineering and neurological disease modelling.
Collapse
Affiliation(s)
- Siqi Ma
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151 Malianwa North Road, Haidian District, Beijing 100193, PR China
| | - Zhaoqing Cong
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151 Malianwa North Road, Haidian District, Beijing 100193, PR China
| | - Huan Chen
- Life Sciences and Environmental Sciences Center, Harbin University of Commerce, Harbin, 150076, PR China
| | - Han Wen
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151 Malianwa North Road, Haidian District, Beijing 100193, PR China
| | - Li Cao
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151 Malianwa North Road, Haidian District, Beijing 100193, PR China
| | - Chunyu Liu
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151 Malianwa North Road, Haidian District, Beijing 100193, PR China
| | - Feifei Yang
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151 Malianwa North Road, Haidian District, Beijing 100193, PR China.
| | - Yonghong Liao
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151 Malianwa North Road, Haidian District, Beijing 100193, PR China.
| |
Collapse
|