1
|
Takeuchi LE, Kalia LV, Simmons CA. Vascular models of Alzheimer's disease: An overview of recent in vitro models of the blood-brain barrier. Neurobiol Dis 2025; 208:106864. [PMID: 40089165 DOI: 10.1016/j.nbd.2025.106864] [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: 11/03/2024] [Revised: 02/18/2025] [Accepted: 03/06/2025] [Indexed: 03/17/2025] Open
Abstract
Alzheimer's disease (AD) remains an overwhelming epidemiologic and economic burden on our healthcare systems, affecting an estimate of 11 % of individuals aged 65 years and older. Increasing evidence of the role of the blood-brain barrier (BBB) in AD pathology lends support to the vascular hypothesis of AD, which posits that damage to cerebral vasculature and impairments to cerebral blood flow are major contributors to neurodegeneration in AD. While the question remains whether the dysfunction of the BBB is the cause or consequence of the disease, understanding of the relationship between vascular pathology and AD is growing increasingly complex, warranting the need for better tools to study vasculature in AD. This review provides an overview of AD models in the context of studying vascular impairments and their relevance in pathology. Specifically, we summarize opportunities in in vitro models, cell sources, and phenotypic observations in sporadic and familial forms of AD. Further, we describe recent advances in generating models which recapitulate in vivo characteristics of the BBB in AD through the use of microfluidics, induced pluripotent stem cells (iPSC), and organoid technologies. Finally, we provide a searchable database of reported cell-based models of pathogenic AD gene variants.
Collapse
Affiliation(s)
- Lily E Takeuchi
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5G 3G9, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada.
| | - Lorraine V Kalia
- Division of Neurology, Department of Medicine, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, ON M5T 2S8, Canada; Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada; Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Craig A Simmons
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5G 3G9, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada.
| |
Collapse
|
2
|
Dai Z, Yu Y, Chen R, Zhu H, Fong H, Kuang J, Jiang Y, Chen Y, Niu Y, Chen T, Shi L. Selenium promotes neural development through the regulation of GPX4 and SEPP1 in an iPSC-derived neuronal model. Biomaterials 2025; 316:123011. [PMID: 39708777 DOI: 10.1016/j.biomaterials.2024.123011] [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: 02/27/2024] [Revised: 12/05/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
Selenium (Se) is incorporated into selenoproteins in the form of selenocysteine, which has biological functions associated with neural development. Unfortunately, the specific roles and mechanisms of selenoproteins at different stages of neuronal development are still unclear. Therefore, in this study, we successfully established a neuronal model derived from induced pluripotent stem cells (iPSC-iNeuron) and used Se nanoparticles (SeNPs@LNT) with high bioavailability to intervene at different stages of neural development in iPSC-iNeuron model. Interestingly, our results showed that SeNPs@LNT could not only accelerate the proliferation of neural progenitor cells (NPCs) by upregulating glutathione peroxidase 4 (GPX4) during the NPC stage, but also can promote neuronal differentiation by increasing selenoprotein P (SEPP1) during the neuronal stage, resulting in efficient and rapid neural development. In addition, further mechanistic studies showed that SeNPs@LNT can regulate selenoproteins by activating the PI3K/Akt/Nrf2 signaling pathway, thereby affecting neuronal development. Notably, Further analysis of ASD patients in National Center for Biotechnology Information single-cell RNA-seq datasets also revealed significantly lower GPX4 expression within NRGN-expressing neurons in ASD patients, and GO enrichment analysis of genes in NRGN-expressing neurons from ASD patients showed that the downregulation of selenoproteins due to aberrant selenoprotein synthesis may be closely associated with decreased ATP synthesis resulting from abnormal mitochondrial and respiratory chain signaling pathways. Taken together, this study provides evidence that SeNPs@LNT exerts a beneficial effect on early neural development through the regulation of selenoproteins.
Collapse
Affiliation(s)
- Zhenzhu Dai
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non-Human Primate Research, GHM Institute of CNS Regeneration, Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Yanzi Yu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non-Human Primate Research, GHM Institute of CNS Regeneration, Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Ruhai Chen
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non-Human Primate Research, GHM Institute of CNS Regeneration, Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Hongyao Zhu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non-Human Primate Research, GHM Institute of CNS Regeneration, Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Hin Fong
- Faculty of Medicine, International School, Jinan University, Guangzhou, 510632, China
| | - Junxin Kuang
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Yunbo Jiang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non-Human Primate Research, GHM Institute of CNS Regeneration, Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Yalan Chen
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non-Human Primate Research, GHM Institute of CNS Regeneration, Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Yimei Niu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non-Human Primate Research, GHM Institute of CNS Regeneration, Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Tianfeng Chen
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non-Human Primate Research, GHM Institute of CNS Regeneration, Department of Chemistry, Jinan University, Guangzhou, 510632, China; Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China.
| | - Lingling Shi
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non-Human Primate Research, GHM Institute of CNS Regeneration, Department of Chemistry, Jinan University, Guangzhou, 510632, China; Department of Psychiatry, The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China; Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226019, China; Department of Neurology, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, 570100, China.
| |
Collapse
|
3
|
Zhou P, Chao Q, Li C, Wang N, Guo S, Wang P, Ge P, Li C, Guo R, Yang N, Lu Z, Tang Z, Zhang Q, Fu T, Xiao Q, Zhu H. Microglia-targeting nanosystems that cooperatively deliver Chinese herbal ingredients alleviate behavioral and cognitive deficits in Alzheimer's disease model mice. J Nanobiotechnology 2025; 23:313. [PMID: 40275381 PMCID: PMC12020378 DOI: 10.1186/s12951-025-03385-z] [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: 01/27/2025] [Accepted: 04/11/2025] [Indexed: 04/26/2025] Open
Abstract
The effective treatment of Alzheimer's disease (AD) is challenging because of its complex and controversial pathological mechanisms. Moreover, multiple barriers, such as the blood-brain barrier (BBB), reduce drug delivery efficiency. Microglia-related neuroinflammation has recently attracted increasing attention as a possible cause of AD and has become a novel therapeutic target. Therefore, overcoming the BBB and targeted delivery of anti-inflammatory agents to microglia seem to be effective practical strategies for treating AD. A large proportion of natural active extracts possess exceptional immunomodulating capabilities. In this study, the cooperative delivery of berberine (Ber) and palmatine (Pal) by transferrin-decorated extracellular vesicles (Tf-hEVs-Ber/Pal), which can cross the BBB and precisely target microglia, was performed. This nanosystem effectively cleared amyloid β-protein (Aβ) aggregates, significantly regulated the neuroinflammatory environment both in vitro and in vivo and markedly altered the behavior and improved the cognitive and learning abilities of AD model mice. The efficacy of a microglia-targeting combined therapeutic approach for AD was demonstrated, which broadens the potential application of Chinese herbal ingredients.
Collapse
Affiliation(s)
- Peng Zhou
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Quan Chao
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chen Li
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ningjing Wang
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Siqi Guo
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Pingping Wang
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Pingyuan Ge
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Caihong Li
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Rui Guo
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Nianyun Yang
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhangdi Lu
- Polymer Science and Materials Chemistry, Exponent Ltd, Hong Kong, 999077, China
| | - Zhishu Tang
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources, Shannxi University of Chinese Medicine, Xianyang, 712038, China
- China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Qichun Zhang
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Tingming Fu
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Qingqing Xiao
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Huaxu Zhu
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
4
|
Yu P, Liu B, Dong C, Chang Y. Induced Pluripotent Stem Cells-Based Regenerative Therapies in Treating Human Aging-Related Functional Decline and Diseases. Cells 2025; 14:619. [PMID: 40277944 PMCID: PMC12025799 DOI: 10.3390/cells14080619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 04/26/2025] Open
Abstract
A significant increase in life expectancy worldwide has resulted in a growing aging population, accompanied by a rise in aging-related diseases that pose substantial societal, economic, and medical challenges. This trend has prompted extensive efforts within many scientific and medical communities to develop and enhance therapies aimed at delaying aging processes, mitigating aging-related functional decline, and addressing aging-associated diseases to extend health span. Research in aging biology has focused on unraveling various biochemical and genetic pathways contributing to aging-related changes, including genomic instability, telomere shortening, and cellular senescence. The advent of induced pluripotent stem cells (iPSCs), derived through reprogramming human somatic cells, has revolutionized disease modeling and understanding in humans by addressing the limitations of conventional animal models and primary human cells. iPSCs offer significant advantages over other pluripotent stem cells, such as embryonic stem cells, as they can be obtained without the need for embryo destruction and are not restricted by the availability of healthy donors or patients. These attributes position iPSC technology as a promising avenue for modeling and deciphering mechanisms that underlie aging and associated diseases, as well as for studying drug effects. Moreover, iPSCs exhibit remarkable versatility in differentiating into diverse cell types, making them a promising tool for personalized regenerative therapies aimed at replacing aged or damaged cells with healthy, functional equivalents. This review explores the breadth of research in iPSC-based regenerative therapies and their potential applications in addressing a spectrum of aging-related conditions.
Collapse
Affiliation(s)
- Peijie Yu
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hunghom, Hong Kong 999077, China; (P.Y.); (B.L.)
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
| | - Bin Liu
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hunghom, Hong Kong 999077, China; (P.Y.); (B.L.)
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
| | - Cheng Dong
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hunghom, Hong Kong 999077, China; (P.Y.); (B.L.)
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
| | - Yun Chang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hunghom, Hong Kong 999077, China; (P.Y.); (B.L.)
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
| |
Collapse
|
5
|
Ma QL, Ebright B, Li B, Li J, Galvan J, Sanchez A, Renteln M, Dikeman D, Wang S, Kerman BE, Seidler P, Gutierrez-Grebenkova B, Hjelm BE, Hawes D, Hiniker AE, Hurth KM, Bennett DA, Louie SG, Chui HC, Limon A, Arvanitakis Z, Yassine HN. Evidence for cPLA2 activation in Alzheimer's Disease Synaptic Pathology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.27.645605. [PMID: 40236242 PMCID: PMC11996316 DOI: 10.1101/2025.03.27.645605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Background Synapses are essential for learning and memory, and their loss predicts cognitive decline in Alzheimer's disease (AD). Synaptic loss is associated with excitotoxicity, neuroinflammation, amyloid-β, and tau pathology, but the molecular mechanisms remain unclear. There is an urgent need to identify new targets to modify the disease and slow synaptic loss and cognitive decline. This study examines if calcium-dependent phospholipase A2 (cPLA2) is implicated in AD synaptic loss. cPLA2 catalyzes membrane phospholipids to release arachidonic acid, which can be metabolized into inflammatory eicosanoids. Methods cPLA2 levels were examined in synaptosomes isolated from the postmortem frontal cortex of individuals with no cognitive impairment (NCI), mild cognitive impairment (MCI), and AD dementia from the Religious Orders Study (ROS). Eicosanoids in synaptosomes were analyzed using lipidomics. Immunofluorescent staining investigated cPLA2 interactions with synaptic markers. Human iPSCs-derived neurons were used to study cPLA2 overactivation after exposure to amyloid-β 42 oligomers (Aβ42O), its relationships with synaptic markers, and the effects of cPLA2 inhibitors. Results We observed elevated cPLA2 (cPLA2α and cPLA2β) in AD synaptosomes and positive correlations with postsynaptic density protein 95 (PSD-95) and cognitive dysfunction. Eicosanoids were increased in AD synaptosomes and correlated with cPLA2, indicating cPLA2 activity at synapses/synaptosomes. Phosphorylated cPLA2α (p-cPLA2α) colocalized with PSD-95 in synaptosomes, and with postsynaptic Ca 2+ /calmodulin-dependent protein kinase IIα (CaMKIIα) and dendritic microtubule-associated protein 2 (MAP2) in NCI and AD brains, where their levels were reduced in AD. P-cPLA2α colocalizes with MAP2 at the neuronal soma associated with neuritic plaques and neurodegeneration in AD. Aβ42O activates cPLA2α in human iPSCs-derived neurons, leading to p-cPLA2α relocation from the cytosol to synaptic and dendritic sites to colocalize with CaMKIIα and MAP2, resulting in their reduction. P-cPLA2α also colocalized with PSD-95 in Aβ42O-exposed neurons, accompanied with increased PSD-95 intensity at soma membrane. These processes were reversed by the cPLA2 inhibitor ASB14780. Conclusions cPLA2 overactivation at synapses, dendrites, and excitatory neuronal somas is associated with synaptic loss, neuritic plaques and neurodegeneration, potentially contributing to cognitive decline in AD. Future research needs to explore the role of cPLA2 as a disease-modifying target for AD.
Collapse
|
6
|
Rao C, Semrau S, Fossati V. Decoding microglial functions in Alzheimer's disease: insights from human models. Trends Immunol 2025; 46:310-323. [PMID: 40113535 PMCID: PMC11993344 DOI: 10.1016/j.it.2025.02.011] [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: 01/06/2025] [Revised: 02/14/2025] [Accepted: 02/19/2025] [Indexed: 03/22/2025]
Abstract
Microglia, key orchestrators of the brain's immune responses, play a pivotal role in the progression of Alzheimer's disease (AD). Emerging human models, including stem cell-derived microglia and cerebral organoids, are transforming our understanding of microglial contributions to AD pathology. In this review, we highlight how these models have uncovered human-specific microglial responses to amyloid plaques and their regulation of neuroinflammation, which are not recapitulated in animal models. We also illustrate how advanced human models that better mimic brain physiology and AD pathology are providing unprecedented insights into the multifaceted roles of microglia. These innovative approaches, combined with sophisticated technologies for cell editing and analysis, are shaping AD research and opening new avenues for therapeutic interventions targeting microglia.
Collapse
Affiliation(s)
- Chandrika Rao
- The New York Stem Cell Foundation Research Institute, New York, NY, USA
| | - Stefan Semrau
- The New York Stem Cell Foundation Research Institute, New York, NY, USA
| | - Valentina Fossati
- The New York Stem Cell Foundation Research Institute, New York, NY, USA.
| |
Collapse
|
7
|
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
|
8
|
Cai Y, Kanyo J, Wilson R, Bathla S, Cardozo PL, Tong L, Qin S, Fuentes LA, Pinheiro-de-Sousa I, Huynh T, Sun L, Mansuri MS, Tian Z, Gan HR, Braker A, Trinh HK, Huttner A, Lam TT, Petsalaki E, Brennand KJ, Nairn AC, Grutzendler J. Subcellular proteomics and iPSC modeling uncover reversible mechanisms of axonal pathology in Alzheimer's disease. NATURE AGING 2025; 5:504-527. [PMID: 40065072 PMCID: PMC11922768 DOI: 10.1038/s43587-025-00823-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 01/29/2025] [Indexed: 03/21/2025]
Abstract
Dystrophic neurites (also termed axonal spheroids) are found around amyloid deposits in Alzheimer's disease (AD), where they impair axonal electrical conduction, disrupt neural circuits and correlate with AD severity. Despite their importance, the mechanisms underlying spheroid formation remain incompletely understood. To address this, we developed a proximity labeling approach to uncover the proteome of spheroids in human postmortem and mouse brains. Additionally, we established a human induced pluripotent stem cell (iPSC)-derived AD model enabling mechanistic investigation and optical electrophysiology. These complementary approaches revealed the subcellular molecular architecture of spheroids and identified abnormalities in key biological processes, including protein turnover, cytoskeleton dynamics and lipid transport. Notably, the PI3K/AKT/mTOR pathway, which regulates these processes, was activated in spheroids. Furthermore, phosphorylated mTOR levels in spheroids correlated with AD severity in humans. Notably, mTOR inhibition in iPSC-derived neurons and mice ameliorated spheroid pathology. Altogether, our study provides a multidisciplinary toolkit for investigating mechanisms and therapeutic targets for axonal pathology in neurodegeneration.
Collapse
Affiliation(s)
- Yifei Cai
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA.
| | - Jean Kanyo
- Keck MS & Proteomics Resource, Yale School of Medicine, New Haven, CT, USA
| | - Rashaun Wilson
- Keck MS & Proteomics Resource, Yale School of Medicine, New Haven, CT, USA
| | - Shveta Bathla
- Yale/NIDA Neuroproteomics Center, Yale University, New Haven, CT, USA
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | | | - Lei Tong
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Shanshan Qin
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Lukas A Fuentes
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
| | | | - Tram Huynh
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Liyuan Sun
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Mohammad Shahid Mansuri
- Yale/NIDA Neuroproteomics Center, Yale University, New Haven, CT, USA
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Zichen Tian
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Hao-Ran Gan
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Amber Braker
- Yale College, Department of Neuroscience, Yale University, New Haven, CT, USA
| | - Hoang Kim Trinh
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Anita Huttner
- Department of Pathology, Yale University, New Haven, CT, USA
| | - TuKiet T Lam
- Keck MS & Proteomics Resource, Yale School of Medicine, New Haven, CT, USA
- Yale/NIDA Neuroproteomics Center, Yale University, New Haven, CT, USA
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
| | - Evangelia Petsalaki
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, UK
| | - Kristen J Brennand
- Department of Psychiatry, Yale University, New Haven, CT, USA
- Department of Genetics, Yale University, New Haven, CT, USA
| | - Angus C Nairn
- Yale/NIDA Neuroproteomics Center, Yale University, New Haven, CT, USA
- Department of Psychiatry, Yale University, New Haven, CT, USA
- Department of Pharmacology, Yale University, New Haven, CT, USA
| | - Jaime Grutzendler
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA.
- Department of Neuroscience, Yale University, New Haven, CT, USA.
- Wu Tsai Institute, Yale University, New Haven, CT, USA.
| |
Collapse
|
9
|
Calvo B, Schembri-Wismayer P, Durán-Alonso MB. Age-Related Neurodegenerative Diseases: A Stem Cell's Perspective. Cells 2025; 14:347. [PMID: 40072076 PMCID: PMC11898746 DOI: 10.3390/cells14050347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 02/22/2025] [Accepted: 02/24/2025] [Indexed: 03/15/2025] Open
Abstract
Neurodegenerative diseases encompass a number of very heterogeneous disorders, primarily characterized by neuronal loss and a concomitant decline in neurological function. Examples of this type of clinical condition are Alzheimer's Disease, Parkinson's Disease, Huntington's Disease and Amyotrophic Lateral Sclerosis. Age has been identified as a major risk in the etiology of these disorders, which explains their increased incidence in developed countries. Unfortunately, despite continued and intensive efforts, no cure has yet been found for any of these diseases; reliable markers that allow for an early diagnosis of the disease and the identification of key molecular events leading to disease onset and progression are lacking. Altered adult neurogenesis appears to precede the appearance of severe symptoms. Given the scarcity of human samples and the considerable differences with model species, increasingly complex human stem-cell-based models are being developed. These are shedding light on the molecular alterations that contribute to disease development, facilitating the identification of new clinical targets and providing a screening platform for the testing of candidate drugs. Moreover, the secretome and other promising features of these cell types are being explored, to use them as replacement cells of high plasticity or as co-adjuvant therapy in combinatorial treatments.
Collapse
Affiliation(s)
- Belén Calvo
- Faculty of Health Sciences, Catholic University of Ávila, 05005 Ávila, Spain;
| | - Pierre Schembri-Wismayer
- Department of Anatomy, Faculty of Medicine and Surgery, University of Malta, MSD 2080 Msida, Malta;
| | - María Beatriz Durán-Alonso
- Department of Biochemistry and Molecular Biology and Physiology, Faculty of Medicine, University of Valladolid, 47005 Valladolid, Spain
| |
Collapse
|
10
|
Rodriguez-Iglesias N, Paris I, Valero J, Cañas-Zabala L, Carretero A, Hatje K, Zhang JD, Patsch C, Britschgi M, Gutbier S, Sierra A. A bottom-up approach identifies the antipsychotic and antineoplastic trifluoperazine and the ribose derivative deoxytubercidin as novel microglial phagocytosis inhibitors. Glia 2025; 73:330-351. [PMID: 39495090 DOI: 10.1002/glia.24637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 10/14/2024] [Accepted: 10/15/2024] [Indexed: 11/05/2024]
Abstract
Phagocytosis is an indispensable function of microglia, the brain professional phagocytes. Microglia is particularly efficient phagocytosing cells that undergo programmed cell death (apoptosis) in physiological conditions. However, mounting evidence suggests microglial phagocytosis dysfunction in multiple brain disorders. These observations prompted us to search for phagocytosis modulators (enhancers or inhibitors) with therapeutic potential. We used a bottom-up strategy that consisted on the identification of phagocytosis modulators using phenotypic high throughput screenings (HTSs) in cell culture and validation in organotypic cultures and in vivo. We performed two complementary HTS campagnes: at Achucarro, we used primary cultures of mouse microglia and compounds of the Prestwick Chemical Library; at Roche, we used human iPSC derived macrophage-like cells and a proprietary chemo-genomic library with 2200 compounds with known mechanism-of-action. Next, we validated the more robust compounds using hippocampal organotypic cultures and identified two phagocytosis inhibitors: trifluoperazine, a dopaminergic and adrenergic antagonist used as an antipsychotic and antineoplastic; and deoxytubercidin, a ribose derivative. Finally, we tested whether these compounds were able to modulate phagocytosis of apoptotic newborn cells in the adult hippocampal neurogenic niche in vivo by administering them into the mouse hippocampus using osmotic minipumps. We confirmed that both trifluoperazine and deoxytubercidin have anti-phagocytic activity in vivo, and validated our bottom-up strategy to identify novel phagocytosis modulators. These results show that chemical libraries with annotated mechanism of action are an starting point for the pharmacological modulation of microglia in drug discovery projects aiming at the therapeutic manipulation of phagocytosis in brain diseases.
Collapse
Affiliation(s)
- Noelia Rodriguez-Iglesias
- Glial Cell Biology Lab, Achucarro Basque Center for Neuroscience, Leioa, Spain
- Department of Neuroscience, University of the Basque Country EHU/UPV, Leioa, Spain
| | - Iñaki Paris
- Glial Cell Biology Lab, Achucarro Basque Center for Neuroscience, Leioa, Spain
- Department of Neuroscience, University of the Basque Country EHU/UPV, Leioa, Spain
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Jorge Valero
- Glial Cell Biology Lab, Achucarro Basque Center for Neuroscience, Leioa, Spain
- Department of Neuroscience, University of the Basque Country EHU/UPV, Leioa, Spain
- Ikerbasque Foundation, Bilbao, Spain
| | - Lorena Cañas-Zabala
- Glial Cell Biology Lab, Achucarro Basque Center for Neuroscience, Leioa, Spain
- Department of Neuroscience, University of the Basque Country EHU/UPV, Leioa, Spain
| | - Alejandro Carretero
- Glial Cell Biology Lab, Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Klas Hatje
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Jitao David Zhang
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Christoph Patsch
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Markus Britschgi
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Simon Gutbier
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Amanda Sierra
- Glial Cell Biology Lab, Achucarro Basque Center for Neuroscience, Leioa, Spain
- Ikerbasque Foundation, Bilbao, Spain
- Department of Biochemistry and Molecular Biology, University of the Basque Country EHU/UPV, Leioa, Spain
| |
Collapse
|
11
|
Park JJ, Rim YA, Sohn Y, Nam Y, Ju JH. Prospects of induced pluripotent stem cells in treating advancing Alzheimer's disease: A review. Histol Histopathol 2025; 40:157-170. [PMID: 38847077 DOI: 10.14670/hh-18-766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
Abstract
The World Health Organization has identified Alzheimer's disease (AD), the leading cause of dementia globally, as a public health priority. However, the complex multifactorial pathology of AD means that its etiology remains incompletely understood. Despite being recognized a century ago, incomplete knowledge has hindered the development of effective treatments for AD. Recent scientific advancements, particularly in induced pluripotent stem cell (iPSC) technology, show great promise in elucidating the fundamental mechanisms of AD. iPSCs play a dual role in regenerating damaged cells for therapeutic purposes and creating disease models to understand AD pathology and aid in drug screening. Nevertheless, as an emerging field, iPSC technology requires further technological advancement to develop effective AD treatments in the future. Thus, this review summarizes recent advances in stem cell therapies, specifically iPSCs, aimed at understanding AD pathology and developing treatments.
Collapse
Affiliation(s)
- Juyoun Janis Park
- YiPSCELL Inc, Seocho-gu, Seoul, South Korea
- Johns Hopkins University, Baltimore, Maryland, USA
| | - Yeri Alice Rim
- YiPSCELL Inc, Seocho-gu, Seoul, South Korea
- CiSTEM Laboratory, Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Yeowon Sohn
- Department of Biohealth Regulatory Science, Sungkyunkwan University, Suwon, South Korea
| | - Yoojun Nam
- YiPSCELL Inc, Seocho-gu, Seoul, South Korea
- Department of Biohealth Regulatory Science, Sungkyunkwan University, Suwon, South Korea.
| | - Ji Hyeon Ju
- YiPSCELL Inc, Seocho-gu, Seoul, South Korea
- CiSTEM Laboratory, Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea.
- Department of Biomedicine and Health Sciences, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
12
|
Kala S, Strutz AG, Katt ME. The Rise of Pluripotent Stem Cell-Derived Glia Models of Neuroinflammation. Neurol Int 2025; 17:6. [PMID: 39852770 PMCID: PMC11767680 DOI: 10.3390/neurolint17010006] [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: 11/20/2024] [Revised: 01/02/2025] [Accepted: 01/09/2025] [Indexed: 01/26/2025] Open
Abstract
Neuroinflammation is a blanket term that describes the body's complex inflammatory response in the central nervous system (CNS). It encompasses a phenotype shift to a proinflammatory state, the release of cytokines, the recruitment of peripheral immune cells, and a wide variety of other processes. Neuroinflammation has been implicated in nearly every major CNS disease ranging from Alzheimer's disease to brain cancer. Understanding and modeling neuroinflammation is critical for the identification of novel therapeutic targets in the treatment of CNS diseases. Unfortunately, the translation of findings from non-human models has left much to be desired. This review systematically discusses the role of human pluripotent stem cell (hPSC)-derived glia and supporting cells within the CNS, including astrocytes, microglia, oligodendrocyte precursor cells, pericytes, and endothelial cells, to describe the state of the field and hope for future discoveries. hPSC-derived cells offer an expanded potential to study the pathobiology of neuroinflammation and immunomodulatory cascades that impact disease progression. While much progress has been made in the development of models, there is much left to explore in the application of these models to understand the complex inflammatory response in the CNS.
Collapse
Affiliation(s)
- Srishti Kala
- Cancer Cell Biology Graduate Education Program, School of Medicine, West Virginia University Health Science Center, Morgantown, WV 26506, USA;
| | - Andrew G. Strutz
- Department of Microbiology, Immunology, and Cell Biology, School of Medicine, West Virginia University Health Science Center, Morgantown, WV 26506, USA;
| | - Moriah E. Katt
- Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, WV 26506, USA
- Department of Neuroscience, School of Medicine, West Virginia University Health Science Center, Morgantown, WV 26506, USA
| |
Collapse
|
13
|
Cheng Y, Xu M, Wu J, Qian K, Yang P, Zhou L, Meng R, Li Y, Wang T, Sheng D, Wei Y, Zhang Q. Carcinoma-Astrocyte Gap Junction Interruption by a Dual-Targeted Biomimetic Liposomal System to Attenuate Chemoresistance and Treat Brain Metastasis. ACS NANO 2024; 18:34107-34125. [PMID: 39626120 DOI: 10.1021/acsnano.4c09996] [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: 12/18/2024]
Abstract
Brain metastasis contributes substantially to the morbidity and mortality of various malignancies and is characterized by high chemoresistance. Intracellular communication between carcinoma cells and astrocytes through gap junctions, which are assembled mainly by the connexin 43 protein, has been shown to play a vital role in this process. However, effectively blocking the gap junctions between the two cell types remains extremely challenging because of insufficient drug delivery to the target site. Herein, we designed a connexin blocker-carbenoxolone (CBX)-loaded biomimetic liposomal system with artificial liposomes fused with brain metastatic cell and reactive astrocyte membranes (LAsomes) to block gap junctions and attenuate chemoresistance. LAsomes effectively penetrated the blood-brain barrier via semaphorin 4D (SEMA 4D)─Plexin B1 interactions and actively migrated to their source cells via homotypic recognition. Consequently, LAsomes effectively inhibited material transfer and Ca2+ flow from metastatic cells to astrocytes via gap junctions, thereby markedly increasing the sensitivity of metastatic tumor cells to chemotherapy. These results reveal that closing the gap junctions may be a promising therapeutic strategy for intractable brain metastasis.
Collapse
Affiliation(s)
- Yunlong Cheng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
- Institute of Traditional Chinese Medicine, & Key Laboratory of TCM Drug Delivery, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, Shaanxi 710003, China
| | - Minjun Xu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jing Wu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Kang Qian
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Peng Yang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Lingling Zhou
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Ran Meng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yixian Li
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Tianying Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Dongyu Sheng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yan Wei
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
| | - Qizhi Zhang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| |
Collapse
|
14
|
Peethambaran Mallika A. Tri-culture modeling of neuroinflammation, neurodegeneration, and neuroprotection. J Alzheimers Dis 2024; 102:742-744. [PMID: 39506313 DOI: 10.1177/13872877241294181] [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: 11/08/2024]
Abstract
The study of neurodegenerative diseases, such as Alzheimer's disease (AD), has long been a complex and challenging task. One of the major hurdles in understanding these diseases is the difficulty in recapitulating the complex interactions between neurons, astrocytes, and microglia in a laboratory setting. In recent years, researchers have made significant progress in developing triculture models that combine these three cell types, allowing for a more accurate representation of the cellular context of the human brain. This commentary discusses the recent advancements and importance of using tri-culture model systems in clarifying the pathophysiology of AD and discusses the recent article by Kim et al. (2024) published in the Journal of Alzheimer's Disease.
Collapse
|
15
|
Fujiyama S, Asano H, Namatame I. Integration of a fully automated flow cytometry system with high robustness into a Screening Station. SLAS Technol 2024; 29:100215. [PMID: 39454873 DOI: 10.1016/j.slast.2024.100215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 10/01/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024]
Abstract
In recent years, there has been an increasing demand for the detection of rare cells in drug discovery research, such as cells that have differentiated off-purpose or are required for immunogenicity evaluation. Since detection and quantification limits depend on the robustness of the experiment, inter-human differences in technique have a significant impact on the performance of the assay system. Here, we integrated flow cytometry into a cell experiment platform, Screening Station, to construct a robust assay system, examined each step of the flow cytometric pretreatment using Jurkat cells, and finally evaluated the overall assay performance. Cell detection rate when the experiment was performed manually was 48.8 % ± 5.7 % (CV=11.6 %) versus 73.7 %±2.0 % (CV=2.8 %) with the automated method. To further clarify the analytical performance of the automated method, 1-100 PD-1 expressing Jurkat cells were spiked with 1 × 105 Jurkat cells, and the lower limit of detection, linearity, and CV% were evaluated. Average detection rate was 69 %, decision count was 0.985, and lower limit of detection was 4 cells (0.004 %). We evaluated the CV% value of the number of detected cells per spiked cell and found our system to be highly robust, approximating a binomial distribution with a 69 % recovery rate. In conclusion, we have integrated the Novocyte flow cytometry system into an automated experimental platform, Screening Station, to create a fully automated flow cytometric assay system with high robustness. Our platform can fulfill the technology needs of drug discovery for rare cell detection, which have intensified in recent years.
Collapse
Affiliation(s)
- Shingo Fujiyama
- Astellas Pharma Inc., 21, Miyukigaoka, Tsukuba-shi 305-8585, Ibaraki, Japan.
| | - Hidemitsu Asano
- Rorze Lifescience Inc., 430-1, Kamiyokoba, Tsukuba-shi 305-0854, Ibaraki, Japan
| | - Ichiji Namatame
- Astellas Pharma Inc., 21, Miyukigaoka, Tsukuba-shi 305-8585, Ibaraki, Japan
| |
Collapse
|
16
|
Summers RA, Fagiani F, Rowitch DH, Absinta M, Reich DS. Novel human iPSC models of neuroinflammation in neurodegenerative disease and regenerative medicine. Trends Immunol 2024; 45:799-813. [PMID: 39307583 PMCID: PMC11471369 DOI: 10.1016/j.it.2024.08.004] [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: 07/27/2024] [Revised: 08/23/2024] [Accepted: 08/25/2024] [Indexed: 10/13/2024]
Abstract
The importance of neuroinflammation in neurodegenerative diseases is becoming increasingly evident, and, in parallel, human induced pluripotent stem cell (hiPSC) models of physiology and pathology are emerging. Here, we review new advancements in the differentiation of hiPSCs into glial, neural, and blood-brain barrier (BBB) cell types, and the integration of these cells into complex organoids and chimeras. These advancements are relevant for modeling neuroinflammation in the context of prevalent neurodegenerative disorders, such as Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). With awareness of current limitations, recent progress in the development and application of various hiPSC-derived models shows potential for aiding the identification of candidate therapeutic targets and immunotherapy approaches.
Collapse
Affiliation(s)
- Rose Ana Summers
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK; Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Francesca Fagiani
- Translational Neuropathology Unit, Institute of Experimental Neurology, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - David H Rowitch
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Martina Absinta
- Translational Neuropathology Unit, Institute of Experimental Neurology, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Daniel S Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
17
|
Mohite P, Puri A, Dave R, Budar A, Munde S, Ghosh SB, Alqahtani T, Shmrany HA, Kumer A, Dhara B. Unlocking the therapeutic potential: odyssey of induced pluripotent stem cells in precision cell therapies. Int J Surg 2024; 110:6432-6455. [PMID: 38963728 PMCID: PMC11487032 DOI: 10.1097/js9.0000000000001892] [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: 02/08/2024] [Accepted: 06/17/2024] [Indexed: 07/06/2024]
Abstract
This review explores the application of induced pluripotent stem cells (iPSCs) in regenerative medicine. The therapeutic significance of iPSC-derived cell therapy within regenerative medicine, emphasizes their reprogramming process and crucial role in cellular differentiation while setting the purpose and scope for the comprehensive exploration of iPSC-derived cell therapy. The subsequent sections intricately examine iPSC-derived cell therapy, unraveling the diverse derivatives of iPSCs and striking a delicate balance between advantages and limitations in therapeutic applications. Mechanisms of action, revealing how iPSC-derived cells seamlessly integrate into tissues, induce regeneration, and contribute to disease modeling and drug screening advancements is discussed. The analysis extends to clinical trials, shedding light on outcomes, safety considerations, and ethical dimensions. Challenges and concerns, including the risk of tumorigenesis and scalability issues, are explored. The focus extends to disease-specific applications, showcasing iPSC-derived cell therapy as a promising avenue for various medical conditions, supported by illustrative case studies. Future directions and research needs are outlined, identifying areas for further exploration, safety considerations and potential enhancements that will shape the future landscape of iPSC-derived therapies. In conclusion, this review provides a significant understanding of iPSC-derived cell therapy's status that contemplates the implications for regenerative medicine and personalized treatment using iPSCs, offering a comprehensive perspective on the evolving field within the confines of a dynamic and promising scientific frontier.
Collapse
Affiliation(s)
- Popat Mohite
- AETs St. John Institute of Pharmacy and Research, Palghar, Maharashtra
| | - Abhijeet Puri
- AETs St. John Institute of Pharmacy and Research, Palghar, Maharashtra
| | - Roshan Dave
- AETs St. John Institute of Pharmacy and Research, Palghar, Maharashtra
| | - Aarati Budar
- AETs St. John Institute of Pharmacy and Research, Palghar, Maharashtra
| | - Shubham Munde
- AETs St. John Institute of Pharmacy and Research, Palghar, Maharashtra
| | - Shruti Bagchi Ghosh
- Department of Pharmaceutical Chemistry, Calcutta Institute of Pharmaceutical Technology and Allied Health Science, Uluberia, Howrah
| | - Taha Alqahtani
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha
| | - Humood Al Shmrany
- Department of Medical Laboratory Sciences, College of Applied medical sciences, Prince Sattam bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Ajoy Kumer
- Department of Chemistry, IUBAT-International University of Business Agriculture & Technology, Dhaka, Bangladesh
| | - Bikram Dhara
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, India
- Department of Health Sciences, Novel Global Community and Educational Foundation. Hebersham, NSW, Australia
| |
Collapse
|
18
|
Park JC, Han JW, Lee W, Kim J, Lee SE, Lee D, Choi H, Han J, Kang YJ, Diep YN, Cho H, Kang R, Yu WJ, Lee J, Choi M, Im SW, Kim JI, Mook-Jung I. Microglia Gravitate toward Amyloid Plaques Surrounded by Externalized Phosphatidylserine via TREM2. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400064. [PMID: 38981007 PMCID: PMC11425970 DOI: 10.1002/advs.202400064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 05/08/2024] [Indexed: 07/11/2024]
Abstract
Microglia play a crucial role in synaptic elimination by engulfing dystrophic neurons via triggering receptors expressed on myeloid cells 2 (TREM2). They are also involved in the clearance of beta-amyloid (Aβ) plaques in Alzheimer's disease (AD); nonetheless, the driving force behind TREM2-mediated phagocytosis of beta-amyloid (Aβ) plaques remains unknown. Here, using advanced 2D/3D/4D co-culture systems with loss-of-function mutations in TREM2 (a frameshift mutation engineered in exon 2) brain organoids/microglia/assembloids, it is identified that the clearance of Aβ via TREM2 is accelerated by externalized phosphatidylserine (ePtdSer) generated from dystrophic neurons surrounding the Aβ plaques. Moreover, it is investigated whether microglia from both sporadic (CRISPR-Cas9-based APOE4 lines) and familial (APPNL-G-F/MAPT double knock-in mice) AD models show reduced levels of TREM2 and lack of phagocytic activity toward ePtdSer-positive Aβ plaques. Herein new insight is provided into TREM2-dependent microglial phagocytosis of Aβ plaques in the context of the presence of ePtdSer during AD progression.
Collapse
Affiliation(s)
- Jong-Chan Park
- Department of Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Metabiohealth, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jong Won Han
- Department of Biochemistry and Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
| | - Woochan Lee
- Department of Biochemistry and Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
- Genome Medicine Institute, Medical Research Center, Seoul National University, Seoul, 03080, Republic of Korea
| | - Jieun Kim
- Department of Biochemistry and Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
| | - Sang-Eun Lee
- Department of Physiology and Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
- BK21 FOUR Biomedical Science Program, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, UK
- Neuroscience Research Institute, Seoul National University Medical Research Center, Seoul, 03080, Republic of Korea
| | - Dongjoon Lee
- Department of Biochemistry and Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
| | - Hayoung Choi
- Department of Biochemistry and Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
| | - Jihui Han
- Department of Biochemistry and Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
| | - You Jung Kang
- Department of Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Yen N Diep
- Department of Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hansang Cho
- Department of Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Rian Kang
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Metabiohealth, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Won Jong Yu
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Metabiohealth, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jean Lee
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
| | - Murim Choi
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
| | - Sun-Wha Im
- Department of Biochemistry and Molecular Biology, Kangwon National University School of Medicine, Gangwon, Seoul, 24341, Republic of Korea
| | - Jong-Il Kim
- Genome Medicine Institute, Medical Research Center, Seoul National University, Seoul, 03080, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
- Cancer Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Inhee Mook-Jung
- Department of Biochemistry and Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
- Convergence Dementia Research Center, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
| |
Collapse
|
19
|
Pascoal T, Rohden F, Ferreira P, Bellaver B, Ferrari-Souza JP, Aguzzoli C, Soares C, Abbas S, Zalzale H, Povala G, Lussier F, Leffa D, Bauer-Negrini G, Rahmouni N, Tissot C, Therriault JT, Servaes S, Stevenson J, Benedet A, Ashton N, Karikari T, Tudorascu D, Zetterberg H, Blennow K, Zimmer E, Souza D, Rosa-Neto P. Glial reactivity is linked to synaptic dysfunction across the aging and Alzheimer's disease spectrum. RESEARCH SQUARE 2024:rs.3.rs-4782732. [PMID: 39184102 PMCID: PMC11343173 DOI: 10.21203/rs.3.rs-4782732/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Previous studies have shown that glial and neuronal changes may trigger synaptic dysfunction in Alzheimer's disease(AD). However, the link between glial and neuronal markers and synaptic abnormalities in the living brain is poorly understood. Here, we investigated the association between biomarkers of astrocyte and microglial reactivity and synaptic dysfunction in 478 individuals across the aging and AD spectrum from two cohorts with available CSF measures of amyloid-β(Aβ), phosphorylated tau(pTau181), astrocyte reactivity(GFAP), microglial activation(sTREM2), and synaptic biomarkers(GAP43 and neurogranin). Elevated CSF GFAP levels were linked to presynaptic and postsynaptic dysfunction, regardless of cognitive status or Aβ presence. CSF sTREM2 levels were associated with presynaptic biomarkers in cognitively unimpaired and impaired Aβ + individuals and postsynaptic biomarkers in cognitively impaired Aβ + individuals. Notably, CSF pTau181 levels mediated all associations between GFAP or sTREM2 levels and synaptic dysfunction biomarkers. These results suggest that neuronal-related synaptic biomarkers could be used in clinical trials targeting glial reactivity in AD.
Collapse
|
20
|
Sierra A, Miron VE, Paolicelli RC, Ransohoff RM. Microglia in Health and Diseases: Integrative Hubs of the Central Nervous System (CNS). Cold Spring Harb Perspect Biol 2024; 16:a041366. [PMID: 38438189 PMCID: PMC11293550 DOI: 10.1101/cshperspect.a041366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
Microglia are usually referred to as "the innate immune cells of the brain," "the resident macrophages of the central nervous system" (CNS), or "CNS parenchymal macrophages." These labels allude to their inherent immune function, related to their macrophage lineage. However, beyond their classic innate immune responses, microglia also play physiological roles crucial for proper brain development and maintenance of adult brain homeostasis. Microglia sense both external and local stimuli through a variety of surface receptors. Thus, they might serve as integrative hubs at the interface between the external environment and the CNS, able to decode, filter, and buffer cues from outside, with the aim of preserving and maintaining brain homeostasis. In this perspective, we will cast a critical look at how these multiple microglial functions are acquired and coordinated, and we will speculate on their impact on human brain physiology and pathology.
Collapse
Affiliation(s)
- Amanda Sierra
- Achucarro Basque Center for Neuroscience, Glial Cell Biology Laboratory, Science Park of UPV/EHU, E-48940 Leioa, Bizkaia, Spain
- Department of Biochemistry and Molecular Biology, University of the Basque Country EHU/UPV, 48940 Leioa, Spain
- Ikerbasque Foundation, Bilbao 48009, Spain
| | - Veronique E Miron
- BARLO Multiple Sclerosis Centre, Keenan Research Centre for Biomedical Science at St. Michael's Hospital, Toronto M5B 1T8, Canada
- Department of Immunology, University of Toronto, Toronto M5S 1A8, Canada
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh BioQuarter, Edinburgh EH16 4TJ, United Kingdom
| | - Rosa C Paolicelli
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, CH-1005 Lausanne, Switzerland
| | | |
Collapse
|
21
|
Etxeberria A, Shen YAA, Vito S, Silverman SM, Imperio J, Lalehzadeh G, Soung AL, Du C, Xie L, Choy MK, Hsiao YC, Ngu H, Cho CH, Ghosh S, Novikova G, Rezzonico MG, Leahey R, Weber M, Gogineni A, Elstrott J, Xiong M, Greene JJ, Stark KL, Chan P, Roth GA, Adrian M, Li Q, Choi M, Wong WR, Sandoval W, Foreman O, Nugent AA, Friedman BA, Sadekar S, Hötzel I, Hansen DV, Chih B, Yuen TJ, Weimer RM, Easton A, Meilandt WJ, Bohlen CJ. Neutral or Detrimental Effects of TREM2 Agonist Antibodies in Preclinical Models of Alzheimer's Disease and Multiple Sclerosis. J Neurosci 2024; 44:e2347232024. [PMID: 38830764 PMCID: PMC11255434 DOI: 10.1523/jneurosci.2347-23.2024] [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: 12/18/2023] [Revised: 05/06/2024] [Accepted: 05/25/2024] [Indexed: 06/05/2024] Open
Abstract
Human genetics and preclinical studies have identified key contributions of TREM2 to several neurodegenerative conditions, inspiring efforts to modulate TREM2 therapeutically. Here, we characterize the activities of three TREM2 agonist antibodies in multiple mixed-sex mouse models of Alzheimer's disease (AD) pathology and remyelination. Receptor activation and downstream signaling are explored in vitro, and active dose ranges are determined in vivo based on pharmacodynamic responses from microglia. For mice bearing amyloid-β (Aβ) pathology (PS2APP) or combined Aβ and tau pathology (TauPS2APP), chronic TREM2 agonist antibody treatment had limited impact on microglia engagement with pathology, overall pathology burden, or downstream neuronal damage. For mice with demyelinating injuries triggered acutely with lysolecithin, TREM2 agonist antibodies unexpectedly disrupted injury resolution. Likewise, TREM2 agonist antibodies limited myelin recovery for mice experiencing chronic demyelination from cuprizone. We highlight the contributions of dose timing and frequency across models. These results introduce important considerations for future TREM2-targeting approaches.
Collapse
Affiliation(s)
- Ainhoa Etxeberria
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Yun-An A Shen
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Stephen Vito
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Sean M Silverman
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Jose Imperio
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Guita Lalehzadeh
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Allison L Soung
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Changchun Du
- Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, California 94080
| | - Luke Xie
- Translational Imaging, Genentech, Inc., South San Francisco, California 94080
| | - Man Kin Choy
- Translational Imaging, Genentech, Inc., South San Francisco, California 94080
| | - Yi-Chun Hsiao
- Antibody Engineering, Genentech, Inc., South San Francisco, California 94080
| | - Hai Ngu
- Pathology, Genentech, Inc., South San Francisco, California 94080
| | - Chang Hoon Cho
- Human Pathobiology and OMNI Reverse Translation, Genentech, Inc., South San Francisco, California 94080
| | - Soumitra Ghosh
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Gloriia Novikova
- Bioinformatics, Genentech, Inc., South San Francisco, California 94080
| | | | - Rebecca Leahey
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Martin Weber
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Alvin Gogineni
- Translational Imaging, Genentech, Inc., South San Francisco, California 94080
| | - Justin Elstrott
- Translational Imaging, Genentech, Inc., South San Francisco, California 94080
| | - Monica Xiong
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Jacob J Greene
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Kimberly L Stark
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Pamela Chan
- Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, California 94080
| | - Gillie A Roth
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, Inc., South San Francisco, California 94080
| | - Max Adrian
- Pathology, Genentech, Inc., South San Francisco, California 94080
| | - Qingling Li
- Microchemistry Lipidomics and Proteomics, Genentech, Inc., South San Francisco, California 94080
| | - Meena Choi
- Microchemistry Lipidomics and Proteomics, Genentech, Inc., South San Francisco, California 94080
| | - Weng Ruh Wong
- Microchemistry Lipidomics and Proteomics, Genentech, Inc., South San Francisco, California 94080
| | - Wendy Sandoval
- Microchemistry Lipidomics and Proteomics, Genentech, Inc., South San Francisco, California 94080
| | - Oded Foreman
- Pathology, Genentech, Inc., South San Francisco, California 94080
| | - Alicia A Nugent
- Human Pathobiology and OMNI Reverse Translation, Genentech, Inc., South San Francisco, California 94080
| | - Brad A Friedman
- Bioinformatics, Genentech, Inc., South San Francisco, California 94080
| | - Shraddha Sadekar
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, Inc., South San Francisco, California 94080
| | - Isidro Hötzel
- Antibody Engineering, Genentech, Inc., South San Francisco, California 94080
| | - David V Hansen
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Ben Chih
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
- Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, California 94080
| | - Tracy J Yuen
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Robby M Weimer
- Translational Imaging, Genentech, Inc., South San Francisco, California 94080
| | - Amy Easton
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - William J Meilandt
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Christopher J Bohlen
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| |
Collapse
|
22
|
Sun Z, Zhang X, So KF, Jiang W, Chiu K. Targeting Microglia in Alzheimer's Disease: Pathogenesis and Potential Therapeutic Strategies. Biomolecules 2024; 14:833. [PMID: 39062547 PMCID: PMC11274940 DOI: 10.3390/biom14070833] [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: 05/03/2024] [Revised: 07/01/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Microglia, as resident macrophages in the central nervous system, play a multifunctional role in the pathogenesis of Alzheimer's disease (AD). Their clustering around amyloid-β (Aβ) deposits is a core pathological feature of AD. Recent advances in single-cell RNA sequencing (scRNA-seq) and single-nucleus RNA sequencing (snRNA-seq) have revealed dynamic changes in microglial phenotypes over time and across different brain regions during aging and AD progression. As AD advances, microglia primarily exhibit impaired phagocytosis of Aβ and tau, along with the release of pro-inflammatory cytokines that damage synapses and neurons. Targeting microglia has emerged as a potential therapeutic approach for AD. Treatment strategies involving microglia can be broadly categorized into two aspects: (1) enhancing microglial function: This involves augmenting their phagocytic ability against Aβ and cellular debris and (2) mitigating neuroinflammation: Strategies include inhibiting TNF-α signaling to reduce the neuroinflammatory response triggered by microglia. Clinical trials exploring microglia-related approaches for AD treatment have garnered attention. Additionally, natural products show promise in enhancing beneficial effects and suppressing inflammatory responses. Clarifying microglial dynamics, understanding their roles, and exploring novel therapeutic approaches will advance our fight against AD.
Collapse
Affiliation(s)
- Zhongqing Sun
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
- Department of Ophthalmology, School of Clinical Medicine, Li Kai Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- State Key Lab of Brain and Cognitive Sciences, Li Kai Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Xin Zhang
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Kwok-Fai So
- State Key Lab of Brain and Cognitive Sciences, Li Kai Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Key Laboratory of CNS Regeneration (Ministry of Education), Jinan University, Guangzhou 510632, China
- Department of Psychology, The University of Hong Kong, Hong Kong SAR, China
| | - Wen Jiang
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Kin Chiu
- Department of Ophthalmology, School of Clinical Medicine, Li Kai Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- State Key Lab of Brain and Cognitive Sciences, Li Kai Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Psychology, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
23
|
Pereira MF, Shyti R, Testa G. In and out: Benchmarking in vitro, in vivo, ex vivo, and xenografting approaches for an integrative brain disease modeling pipeline. Stem Cell Reports 2024; 19:767-795. [PMID: 38865969 PMCID: PMC11390705 DOI: 10.1016/j.stemcr.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 05/09/2024] [Accepted: 05/11/2024] [Indexed: 06/14/2024] Open
Abstract
Human cellular models and their neuronal derivatives have afforded unprecedented advances in elucidating pathogenic mechanisms of neuropsychiatric diseases. Notwithstanding their indispensable contribution, animal models remain the benchmark in neurobiological research. In an attempt to harness the best of both worlds, researchers have increasingly relied on human/animal chimeras by xenografting human cells into the animal brain. Despite the unparalleled potential of xenografting approaches in the study of the human brain, literature resources that systematically examine their significance and advantages are surprisingly lacking. We fill this gap by providing a comprehensive account of brain diseases that were thus far subjected to all three modeling approaches (transgenic rodents, in vitro human lineages, human-animal xenografting) and provide a critical appraisal of the impact of xenografting approaches for advancing our understanding of those diseases and brain development. Next, we give our perspective on integrating xenografting modeling pipeline with recent cutting-edge technological advancements.
Collapse
Affiliation(s)
- Marlene F Pereira
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Via Adamello 16, 20139 Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Via Santa Sofia 9, 20122 Milan, Italy; Neurogenomics Centre, Human Technopole, Viale Rita Levi-Montalcini 1, 20157 Milan, Italy.
| | - Reinald Shyti
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Via Adamello 16, 20139 Milan, Italy; Neurogenomics Centre, Human Technopole, Viale Rita Levi-Montalcini 1, 20157 Milan, Italy.
| | - Giuseppe Testa
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Via Adamello 16, 20139 Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Via Santa Sofia 9, 20122 Milan, Italy; Neurogenomics Centre, Human Technopole, Viale Rita Levi-Montalcini 1, 20157 Milan, Italy.
| |
Collapse
|
24
|
Eskandari-Sedighi G, Crichton M, Zia S, Gomez-Cardona E, Cortez LM, Patel ZH, Takahashi-Yamashiro K, St Laurent CD, Sidhu G, Sarkar S, Aghanya V, Sim VL, Tan Q, Julien O, Plemel JR, Macauley MS. Alzheimer's disease associated isoforms of human CD33 distinctively modulate microglial cell responses in 5XFAD mice. Mol Neurodegener 2024; 19:42. [PMID: 38802940 PMCID: PMC11129479 DOI: 10.1186/s13024-024-00734-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 05/16/2024] [Indexed: 05/29/2024] Open
Abstract
Microglia play diverse pathophysiological roles in Alzheimer's disease (AD), with genetic susceptibility factors skewing microglial cell function to influence AD risk. CD33 is an immunomodulatory receptor associated with AD susceptibility through a single nucleotide polymorphism that modulates mRNA splicing, skewing protein expression from a long protein isoform (CD33M) to a short isoform (CD33m). Understanding how human CD33 isoforms differentially impact microglial cell function in vivo has been challenging due to functional divergence of CD33 between mice and humans. We address this challenge by studying transgenic mice expressing either of the human CD33 isoforms crossed with the 5XFAD mouse model of amyloidosis and find that human CD33 isoforms have opposing effects on the response of microglia to amyloid-β (Aβ) deposition. Mice expressing CD33M have increased Aβ levels, more diffuse plaques, fewer disease-associated microglia, and more dystrophic neurites compared to 5XFAD control mice. Conversely, CD33m promotes plaque compaction and microglia-plaque contacts, and minimizes neuritic plaque pathology, highlighting an AD protective role for this isoform. Protective phenotypes driven by CD33m are detected at an earlier timepoint compared to the more aggressive pathology in CD33M mice that appears at a later timepoint, suggesting that CD33m has a more prominent impact on microglia cell function at earlier stages of disease progression. In addition to divergent roles in modulating phagocytosis, scRNAseq and proteomics analyses demonstrate that CD33m+ microglia upregulate nestin, an intermediate filament involved in cell migration, at plaque contact sites. Overall, our work provides new functional insights into how CD33, as a top genetic susceptibility factor for AD, modulates microglial cell function.
Collapse
Affiliation(s)
| | | | - Sameera Zia
- Division of Neurology, Department of Medicine, University of Alberta, Edmonton, Canada
| | | | - Leonardo M Cortez
- Division of Neurology, Department of Medicine, University of Alberta, Edmonton, Canada
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Zain H Patel
- Department of Cell Biology, University of Alberta, Edmonton, Canada
| | | | | | - Gaurav Sidhu
- Department of Chemistry, University of Alberta, Edmonton, Canada
| | - Susmita Sarkar
- Department of Chemistry, University of Alberta, Edmonton, Canada
| | - Vivian Aghanya
- Department of Chemistry, University of Alberta, Edmonton, Canada
| | - Valerie L Sim
- Division of Neurology, Department of Medicine, University of Alberta, Edmonton, Canada
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Qiumin Tan
- Department of Cell Biology, University of Alberta, Edmonton, Canada
| | - Olivier Julien
- Department of Biochemistry, University of Alberta, Edmonton, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Jason R Plemel
- Division of Neurology, Department of Medicine, University of Alberta, Edmonton, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Canada
| | - Matthew S Macauley
- Department of Chemistry, University of Alberta, Edmonton, Canada.
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada.
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Canada.
| |
Collapse
|
25
|
Jovanovic VM, Mesch KT, Tristan CA. hPSC-Derived Astrocytes at the Forefront of Translational Applications in Neurological Disorders. Cells 2024; 13:903. [PMID: 38891034 PMCID: PMC11172187 DOI: 10.3390/cells13110903] [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: 04/02/2024] [Revised: 05/17/2024] [Accepted: 05/23/2024] [Indexed: 06/20/2024] Open
Abstract
Astrocytes, the most abundant glial cell type in the brain, play crucial roles in maintaining homeostasis within the central nervous system (CNS). Impairment or abnormalities of typical astrocyte functions in the CNS serve as a causative or contributing factor in numerous neurodevelopmental, neurodegenerative, and neuropsychiatric disorders. Currently, disease-modeling and drug-screening approaches, primarily focused on human astrocytes, rely on human pluripotent stem cell (hPSC)-derived astrocytes. However, it is important to acknowledge that these hPSC-derived astrocytes exhibit notable differences across studies and when compared to their in vivo counterparts. These differences may potentially compromise translational outcomes if not carefully accounted for. This review aims to explore state-of-the-art in vitro models of human astrocyte development, focusing on the developmental processes, functional maturity, and technical aspects of various hPSC-derived astrocyte differentiation protocols. Additionally, it summarizes their successful application in modeling neurological disorders. The discussion extends to recent advancements in the large-scale production of human astrocytes and their application in developing high-throughput assays conducive to therapeutic drug discovery.
Collapse
Affiliation(s)
- Vukasin M. Jovanovic
- Stem Cell Translation Laboratory (SCTL), Division of Preclinical Innovation (DPI), National Center for Advancing Translational Sciences (NCATS), NIH, Rockville, MD 20850, USA (C.A.T.)
| | | | | |
Collapse
|
26
|
Fruhwürth S, Zetterberg H, Paludan SR. Microglia and amyloid plaque formation in Alzheimer's disease - Evidence, possible mechanisms, and future challenges. J Neuroimmunol 2024; 390:578342. [PMID: 38640827 DOI: 10.1016/j.jneuroim.2024.578342] [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: 02/01/2024] [Revised: 03/21/2024] [Accepted: 04/03/2024] [Indexed: 04/21/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by cognitive decline that severely affects patients and their families. Genetic and environmental risk factors, such as viral infections, synergize to accelerate the aging-associated neurodegeneration. Genetic risk factors for late-onset AD (LOAD), which accounts for most AD cases, are predominantly implicated in microglial and immune cell functions. As such, microglia play a major role in formation of amyloid beta (Aβ) plaques, the major pathological hallmark of AD. This review aims to provide an overview of the current knowledge regarding the role of microglia in Aβ plaque formation, as well as their impact on morphological and functional diversity of Aβ plaques. Based on this discussion, we seek to identify challenges and opportunities in this field with potential therapeutic implications.
Collapse
Affiliation(s)
- Stefanie Fruhwürth
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, Institute of Neurology, University College London Queen Square, London, UK; UK Dementia Research Institute at UCL, London, UK; Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China; Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, USA
| | - Søren R Paludan
- Department of Rheumatology and Inflammatory Research, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden; Department of Biomedicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
27
|
Tsering W, Prokop S. Neuritic Plaques - Gateways to Understanding Alzheimer's Disease. Mol Neurobiol 2024; 61:2808-2821. [PMID: 37940777 PMCID: PMC11043180 DOI: 10.1007/s12035-023-03736-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 10/21/2023] [Indexed: 11/10/2023]
Abstract
Extracellular deposits of amyloid-β (Aβ) in the form of plaques are one of the main pathological hallmarks of Alzheimer's disease (AD). Over the years, many different Aβ plaque morphologies such as neuritic plaques, dense cored plaques, cotton wool plaques, coarse-grain plaques, and diffuse plaques have been described in AD postmortem brain tissues, but correlation of a given plaque type with AD progression or AD symptoms is not clear. Furthermore, the exact trigger causing the development of one Aβ plaque morphological subtype over the other is still unknown. Here, we review the current knowledge about neuritic plaques, a subset of Aβ plaques surrounded by swollen or dystrophic neurites, which represent the most detrimental and consequential Aβ plaque morphology. Neuritic plaques have been associated with local immune activation, neuronal network dysfunction, and cognitive decline. Given that neuritic plaques are at the interface of Aβ deposition, tau aggregation, and local immune activation, we argue that understanding the exact mechanism of neuritic plaque formation is crucial to develop targeted therapies for AD.
Collapse
Affiliation(s)
- Wangchen Tsering
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, USA
| | - Stefan Prokop
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA.
- McKnight Brain Institute, University of Florida, Gainesville, USA.
- Department of Pathology, University of Florida, Gainesville, USA.
- Fixel Institute for Neurological Diseases, University of Florida, Gainesville, USA.
| |
Collapse
|
28
|
Birkle TJ, Willems HM, Skidmore J, Brown GC. Disease phenotypic screening in neuron-glia cocultures identifies blockers of inflammatory neurodegeneration. iScience 2024; 27:109454. [PMID: 38550989 PMCID: PMC10973195 DOI: 10.1016/j.isci.2024.109454] [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: 08/02/2023] [Revised: 10/09/2023] [Accepted: 03/06/2024] [Indexed: 01/30/2025] Open
Abstract
Neuropathology is often mediated by interactions between neurons and glia that cannot be modeled by monocultures. However, cocultures are difficult to use and analyze for high-content screening. Here, we perform compound screening using primary neuron-glia cultures to model inflammatory neurodegeneration, live-cell stains, and automated classification of neurons, astrocytes or microglia using open-source software. Out of 227 compounds with known bioactivities, 29 protected against lipopolysaccharide-induced neuronal loss, including drugs affecting adrenergic, steroid, inflammatory and MAP kinase signaling. The screen also identified physiological compounds, such as noradrenaline and progesterone, that protected and identified neurotoxic compounds, such as a TLR7 agonist, that induced microglial proliferation. Most compounds used here have not been tested in a neuron-glia coculture neurodegeneration assay previously. Thus, combining a complex cellular disease model with high-content screening of known compounds and automated image analysis allows identification of important biology, as well as potential targets and drugs for treatment.
Collapse
Affiliation(s)
| | | | - John Skidmore
- ALBORADA Drug Discovery Institute, Cambridge CB2 0AH, UK
| | - Guy C. Brown
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| |
Collapse
|
29
|
Duan Y, He K, Lan W, Luo Y, Fan H, Lin P, Wang W, Tang Y. Noninvasive Assessment of hiPSC Differentiation toward Cardiomyocytes Using Pretrained Convolutional Neural Networks and the Channel Pruning Algorithm. ACS Biomater Sci Eng 2024; 10:2498-2509. [PMID: 38531866 DOI: 10.1021/acsbiomaterials.3c01938] [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: 03/28/2024]
Abstract
Human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (hiPSC-CMs) offer versatile applications in tissue engineering and drug screening. To facilitate the monitoring of hiPSC cardiac differentiation, a noninvasive approach using convolutional neural networks (CNNs) was explored. HiPSCs were differentiated into cardiomyocytes and analyzed using the quantitative real-time polymerase chain reaction (qRT-PCR). The bright-field images of the cells at different time points were captured to create the dataset. Six pretrained models (AlexNet, GoogleNet, ResNet 18, ResNet 50, DenseNet 121, VGG 19-BN) were employed to identify different stages in differentiation. VGG 19-BN outperformed the other five CNNs and exhibited remarkable performance with 99.2% accuracy, recall, precision, and F1 score and 99.8% specificity. The pruning process was then applied to the optimal model, resulting in a significant reduction of model parameters while maintaining high accuracy. Finally, an automation application using the pruned VGG 19-BN model was developed, facilitating users in assessing the cell status during the myocardial differentiation of hiPSCs.
Collapse
Affiliation(s)
- Yujie Duan
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Kaitong He
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Wei Lan
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Yuli Luo
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Hao Fan
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Peiran Lin
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Wenlong Wang
- School of Mechanical and Electrical Engineering, Guangzhou University, Guangzhou 510006, China
| | - Yadong Tang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| |
Collapse
|
30
|
Yu Y, Chen R, Mao K, Deng M, Li Z. The Role of Glial Cells in Synaptic Dysfunction: Insights into Alzheimer's Disease Mechanisms. Aging Dis 2024; 15:459-479. [PMID: 37548934 PMCID: PMC10917533 DOI: 10.14336/ad.2023.0718] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 07/18/2023] [Indexed: 08/08/2023] Open
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder that impacts a substantial number of individuals globally. Despite its widespread prevalence, there is currently no cure for AD. It is widely acknowledged that normal synaptic function holds a key role in memory, cognitive abilities, and the interneuronal transfer of information. As AD advances, symptoms including synaptic impairment, decreased synaptic density, and cognitive decline become increasingly noticeable. The importance of glial cells in the formation of synapses, the growth of neurons, brain maturation, and safeguarding the microenvironment of the central nervous system is well recognized. However, during AD progression, overactive glial cells can cause synaptic dysfunction, neuronal death, and abnormal neuroinflammation. Both neuroinflammation and synaptic dysfunction are present in the early stages of AD. Therefore, focusing on the changes in glia-synapse communication could provide insights into the mechanisms behind AD. In this review, we aim to provide a summary of the role of various glial cells, including microglia, astrocytes, oligodendrocytes, and oligodendrocyte precursor cells, in regulating synaptic dysfunction. This may offer a new perspective on investigating the underlying mechanisms of AD.
Collapse
Affiliation(s)
- Yang Yu
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.
| | - Ran Chen
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.
- School of Medicine, Sun Yat-sen University, Shenzhen, China.
| | - Kaiyue Mao
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.
- School of Medicine, Sun Yat-sen University, Shenzhen, China.
| | - Maoyan Deng
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.
- School of Medicine, Sun Yat-sen University, Shenzhen, China.
| | - Zhigang Li
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Shenzhen, China.
| |
Collapse
|
31
|
Zheng YW, Yu SY, Li Z, Xu YT, Zhao WW, Jiang D, Chen HY, Xu JJ. High-Precision Single-Cell microRNA Therapy by a Functional Nanopipette with Sensitive Photoelectrochemical Feedback. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307067. [PMID: 37972263 DOI: 10.1002/smll.202307067] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/15/2023] [Indexed: 11/19/2023]
Abstract
This work proposes the concept of single-cell microRNA (miR) therapy and proof-of-concept by engineering a nanopipette for high-precision miR-21-targeted therapy in a single HeLa cell with sensitive photoelectrochemical (PEC) feedback. Targeting the representative oncogenic miR-21, the as-functionalized nanopipette permits direct intracellular drug administration with precisely controllable dosages, and the corresponding therapeutic effects can be sensitively transduced by a PEC sensing interface that selectively responds to the indicator level of cytosolic caspase-3. The experimental results reveal that injection of ca. 4.4 × 10-20 mol miR-21 inhibitor, i.e., 26488 copies, can cause the obvious therapeutic action in the targeted cell. This work features a solution to obtain the accurate knowledge of how a certain miR-drug with specific dosages treats the cells and thus provides an insight into futuristic high-precision clinical miR therapy using personalized medicine, provided that the prerequisite single-cell experiments are courses of personalized customization.
Collapse
Affiliation(s)
- You-Wei Zheng
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Si-Yuan Yu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Zheng Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Yi-Tong Xu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Wei-Wei Zhao
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Dechen Jiang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Hong-Yuan Chen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Jing-Juan Xu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| |
Collapse
|
32
|
Supakul S, Murakami R, Oyama C, Shindo T, Hatakeyama Y, Itsuno M, Bannai H, Shibata S, Maeda S, Okano H. Mutual interaction of neurons and astrocytes derived from iPSCs with APP V717L mutation developed the astrocytic phenotypes of Alzheimer's disease. Inflamm Regen 2024; 44:8. [PMID: 38419091 PMCID: PMC10900748 DOI: 10.1186/s41232-023-00310-5] [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: 09/05/2023] [Accepted: 11/22/2023] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND The development of induced pluripotent stem cells (iPSCs) technology has enabled human cellular disease modeling for inaccessible cell types, such as neural cells in the brain. However, many of the iPSC-derived disease models established to date typically involve only a single cell type. These monoculture models are inadequate for accurately simulating the brain environment, where multiple cell types interact. The limited cell type diversity in monoculture models hinders the accurate recapitulation of disease phenotypes resulting from interactions between different cell types. Therefore, our goal was to create cell models that include multiple interacting cell types to better recapitulate disease phenotypes. METHODS To establish a co-culture model of neurons and astrocytes, we individually induced neurons and astrocytes from the same iPSCs using our novel differentiation methods, and then co-cultured them. We evaluated the effects of co-culture on neurons and astrocytes using immunocytochemistry, immuno-electron microscopy, and Ca2+ imaging. We also developed a co-culture model using iPSCs from a patient with familial Alzheimer's disease (AD) patient (APP V717L mutation) to investigate whether this model would manifest disease phenotypes not seen in the monoculture models. RESULTS The co-culture of the neurons and astrocytes increased the branching of astrocyte processes, the number of GFAP-positive cells, neuronal activities, the number of synapses, and the density of presynaptic vesicles. In addition, immuno-electron microscopy confirmed the formation of a tripartite synaptic structure in the co-culture model, and inhibition of glutamate transporters increased neuronal activity. Compared to the co-culture model of the control iPSCs, the co-culture model of familial AD developed astrogliosis-like phenotype, which was not observed in the monoculture model of astrocytes. CONCLUSIONS Co-culture of iPSC-derived neurons and astrocytes enhanced the morphological changes mimicking the in vivo condition of both cell types. The formation of the functional tripartite synaptic structures in the co-culture model suggested the mutual interaction between the cells. Furthermore, the co-culture model with the APP V717L mutation expressed in neurons exhibited an astrocytic phenotype reminiscent of AD brain pathology. These results suggest that our co-culture model is a valuable tool for disease modeling of neurodegenerative diseases.
Collapse
Affiliation(s)
- Sopak Supakul
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Rei Murakami
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Chisato Oyama
- Department of Electrical Engineering and Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, 169-8555, Japan
| | - Tomoko Shindo
- Electron Microscope Laboratory, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Yuki Hatakeyama
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Maika Itsuno
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Hiroko Bannai
- Department of Electrical Engineering and Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, 169-8555, Japan
| | - Shinsuke Shibata
- Electron Microscope Laboratory, Keio University School of Medicine, Tokyo, 160-8582, Japan
- Division of Microscopic Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 951-8510, Japan
| | - Sumihiro Maeda
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan.
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan.
| |
Collapse
|
33
|
Bobotis BC, Halvorson T, Carrier M, Tremblay MÈ. Established and emerging techniques for the study of microglia: visualization, depletion, and fate mapping. Front Cell Neurosci 2024; 18:1317125. [PMID: 38425429 PMCID: PMC10902073 DOI: 10.3389/fncel.2024.1317125] [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: 10/10/2023] [Accepted: 01/15/2024] [Indexed: 03/02/2024] Open
Abstract
The central nervous system (CNS) is an essential hub for neuronal communication. As a major component of the CNS, glial cells are vital in the maintenance and regulation of neuronal network dynamics. Research on microglia, the resident innate immune cells of the CNS, has advanced considerably in recent years, and our understanding of their diverse functions continues to grow. Microglia play critical roles in the formation and regulation of neuronal synapses, myelination, responses to injury, neurogenesis, inflammation, and many other physiological processes. In parallel with advances in microglial biology, cutting-edge techniques for the characterization of microglial properties have emerged with increasing depth and precision. Labeling tools and reporter models are important for the study of microglial morphology, ultrastructure, and dynamics, but also for microglial isolation, which is required to glean key phenotypic information through single-cell transcriptomics and other emerging approaches. Strategies for selective microglial depletion and modulation can provide novel insights into microglia-targeted treatment strategies in models of neuropsychiatric and neurodegenerative conditions, cancer, and autoimmunity. Finally, fate mapping has emerged as an important tool to answer fundamental questions about microglial biology, including their origin, migration, and proliferation throughout the lifetime of an organism. This review aims to provide a comprehensive discussion of these established and emerging techniques, with applications to the study of microglia in development, homeostasis, and CNS pathologies.
Collapse
Affiliation(s)
- Bianca Caroline Bobotis
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Centre for Advanced Materials and Related Technology, Victoria, BC, Canada
| | - Torin Halvorson
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
- British Columbia Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - Micaël Carrier
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Département de Psychiatrie et de Neurosciences, Faculté de Médecine, Université Laval, Québec City, QC, Canada
- Axe neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Centre for Advanced Materials and Related Technology, Victoria, BC, Canada
- Axe neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
- Department of Molecular Medicine, Université Laval, Québec City, QC, Canada
| |
Collapse
|
34
|
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
|
35
|
Yadav N, Purow BW. Understanding current experimental models of glioblastoma-brain microenvironment interactions. J Neurooncol 2024; 166:213-229. [PMID: 38180686 PMCID: PMC11056965 DOI: 10.1007/s11060-023-04536-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 12/07/2023] [Indexed: 01/06/2024]
Abstract
Glioblastoma (GBM) is a common and devastating primary brain tumor, with median survival of 16-18 months after diagnosis in the setting of substantial resistance to standard-of-care and inevitable tumor recurrence. Recent work has implicated the brain microenvironment as being critical for GBM proliferation, invasion, and resistance to treatment. GBM does not operate in isolation, with neurons, astrocytes, and multiple immune populations being implicated in GBM tumor progression and invasiveness. The goal of this review article is to provide an overview of the available in vitro, ex vivo, and in vivo experimental models for assessing GBM-brain interactions, as well as discuss each model's relative strengths and limitations. Current in vitro models discussed will include 2D and 3D co-culture platforms with various cells of the brain microenvironment, as well as spheroids, whole organoids, and models of fluid dynamics, such as interstitial flow. An overview of in vitro and ex vivo organotypic GBM brain slices is also provided. Finally, we conclude with a discussion of the various in vivo rodent models of GBM, including xenografts, syngeneic grafts, and genetically-engineered models of GBM.
Collapse
Affiliation(s)
- Niket Yadav
- Department of Neurology, University of Virginia Comprehensive Cancer Center, University of Virginia Health System, Charlottesville, VA, 22903, USA
- Medical Scientist Training Program, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Benjamin W Purow
- Department of Neurology, University of Virginia Comprehensive Cancer Center, University of Virginia Health System, Charlottesville, VA, 22903, USA.
| |
Collapse
|
36
|
Ohta A, Kawai S, Pretemer Y, Nishio M, Nagata S, Fuse H, Yamagishi Y, Toguchida J. Automated cell culture system for the production of cell aggregates with growth plate-like structure from induced pluripotent stem cells. SLAS Technol 2023; 28:433-441. [PMID: 37562511 DOI: 10.1016/j.slast.2023.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 07/02/2023] [Accepted: 08/08/2023] [Indexed: 08/12/2023]
Abstract
Programmable liquid handling devices for cell culture systems have dramatically enhanced scalability and reproducibility. We previously reported a protocol to produce cell aggregates demonstrating growth plate-like structures containing hypertrophic chondrocytes from human induced pluripotent stem cells (hiPSCs). To apply this protocol to large-scale drug screening for growth plate-related diseases, we adapted it to the automated cell culture system (ACCS) consisting of programmable liquid handling devices connected to CO2 incubators, a refrigerator, and labware feeders, designed for up to 4 batches with several cell culture plates culturing for several months. We developed a new program preparing culture media with growth factors at final concentration immediately before dispensing them to each well and precisely positioning the tip for the medium change without damaging cell aggregates. Using these programs on the ACCS, we successfully cultured cell aggregates for 56 days, only needing to replenish the labware, medium, and growth factors twice a week. The size of cell aggregates in each well increased over time, with low well-to-well variability. Cell aggregates on day 56 showed histochemical, immunohistochemical, and gene expression properties of growth plate-like structures containing hypertrophic chondrocytes, indicating proper quality as materials for basic research and drug discovery of growth plate related diseases. The established program will be a suitable reference for making programs of experiments requiring long term and complex culture procedures using ACCS.
Collapse
Affiliation(s)
- Akira Ohta
- Department of Fundamental Cell Technology, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan.
| | - Shunsuke Kawai
- Department of Fundamental Cell Technology, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Yann Pretemer
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Megumi Nishio
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Sanae Nagata
- Department of Fundamental Cell Technology, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Hiromitsu Fuse
- Department of Fundamental Cell Technology, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Yukiko Yamagishi
- Department of Fundamental Cell Technology, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan; Drug Discovery Research, Astellas Pharma Inc., Tsukuba-shi, Ibaraki, Japan
| | - Junya Toguchida
- Department of Fundamental Cell Technology, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan; Department of Regeneration Science and Engineering, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan.
| |
Collapse
|
37
|
Tsering W, Hery GP, Phillips JL, Lolo K, Bathe T, Villareal JA, Ruan IY, Prokop S. Transformation of non-neuritic into neuritic plaques during AD progression drives cortical spread of tau pathology via regenerative failure. Acta Neuropathol Commun 2023; 11:190. [PMID: 38037144 PMCID: PMC10691154 DOI: 10.1186/s40478-023-01688-6] [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: 11/08/2023] [Accepted: 11/10/2023] [Indexed: 12/02/2023] Open
Abstract
Extracellular amyloid-β (Aβ) plaques and intracellular aggregates of tau protein in form of neurofibrillary tangles (NFT) are pathological hallmarks of Alzheimer's disease (AD). The exact mechanism how these two protein aggregates interact in AD is still a matter of debate. Neuritic plaques (NP), a subset of Aβ plaques containing dystrophic neurites (DN), are suggested to be unique to AD and might play a role in the interaction of Aβ and tau. Quantifying NP and non-NP in postmortem brain specimens from patients with increasing severity of AD neuropathological changes (ADNC), we demonstrate that the total number of Aβ plaques and NP increase, while the number of non-NP stagnates. Furthermore, investigating the correlation between NP and NFT, we identified unexpected brain region-specific differences when comparing cases with increasingly more severe ADNC. In neocortical regions NFT counts increase in parallel with NP counts during the progression of ADNC, while this correlation is not observed in hippocampus. These data support the notion that non-NP are transformed into NP during the progression of ADNC and indicate that NP might drive cortical NFT formation. Next, using spatial transcriptomics, we analyzed the gene expression profile of the microenvironment around non-NP and NP. We identified an upregulation of neuronal systems and Ca-dependent event pathways around NP compared to non-NP. We speculate that the upregulation of these transcripts may hint at a compensatory mechanism underlying NP formation. Our studies suggest that the transformation of non-NP to NP is a key event in ADNC progression and points to regenerative failure as a potential driving force of this process.
Collapse
Affiliation(s)
- Wangchen Tsering
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- College of Medicine, Mcknight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Gabriela P Hery
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Pathology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Jennifer L Phillips
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Pathology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Kiara Lolo
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Tim Bathe
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Pathology, College of Medicine, University of Florida, Gainesville, FL, USA
- College of Medicine, Mcknight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Jonathan A Villareal
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Isabelle Y Ruan
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Stefan Prokop
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA.
- Department of Pathology, College of Medicine, University of Florida, Gainesville, FL, USA.
- College of Medicine, Mcknight Brain Institute, University of Florida, Gainesville, FL, USA.
- Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
38
|
Marei HE, Khan MUA, Hasan A. Potential use of iPSCs for disease modeling, drug screening, and cell-based therapy for Alzheimer's disease. Cell Mol Biol Lett 2023; 28:98. [PMID: 38031028 PMCID: PMC10687886 DOI: 10.1186/s11658-023-00504-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Alzheimer's disease (AD) is a chronic illness marked by increasing cognitive decline and nervous system deterioration. At this time, there is no known medication that will stop the course of Alzheimer's disease; instead, most symptoms are treated. Clinical trial failure rates for new drugs remain high, highlighting the urgent need for improved AD modeling for improving understanding of the underlying pathophysiology of disease and improving drug development. The development of induced pluripotent stem cells (iPSCs) has made it possible to model neurological diseases like AD, giving access to an infinite number of patient-derived cells capable of differentiating neuronal fates. This advance will accelerate Alzheimer's disease research and provide an opportunity to create more accurate patient-specific models of Alzheimer's disease to support pathophysiological research, drug development, and the potential application of stem cell-based therapeutics. This review article provides a complete summary of research done to date on the potential use of iPSCs from AD patients for disease modeling, drug discovery, and cell-based therapeutics. Current technological developments in AD research including 3D modeling, genome editing, gene therapy for AD, and research on familial (FAD) and sporadic (SAD) forms of the disease are discussed. Finally, we outline the issues that need to be elucidated and future directions for iPSC modeling in AD.
Collapse
Affiliation(s)
- Hany E Marei
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35116, Egypt.
| | - Muhammad Umar Aslam Khan
- Biomedical Research Center, Qatar University, 2713, Doha, Qatar
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha, Qatar
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha, Qatar
| |
Collapse
|
39
|
Lavekar SS, Patel MD, Montalvo-Parra MD, Krencik R. Asteroid impact: the potential of astrocytes to modulate human neural networks within organoids. Front Neurosci 2023; 17:1305921. [PMID: 38075269 PMCID: PMC10702564 DOI: 10.3389/fnins.2023.1305921] [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: 10/02/2023] [Accepted: 11/08/2023] [Indexed: 02/12/2024] Open
Abstract
Astrocytes are a vital cellular component of the central nervous system that impact neuronal function in both healthy and pathological states. This includes intercellular signals to neurons and non-neuronal cells during development, maturation, and aging that can modulate neural network formation, plasticity, and maintenance. Recently, human pluripotent stem cell-derived neural aggregate cultures, known as neurospheres or organoids, have emerged as improved experimental platforms for basic and pre-clinical neuroscience compared to traditional approaches. Here, we summarize the potential capability of using organoids to further understand the mechanistic role of astrocytes upon neural networks, including the production of extracellular matrix components and reactive signaling cues. Additionally, we discuss the application of organoid models to investigate the astrocyte-dependent aspects of neuropathological diseases and to test astrocyte-inspired technologies. We examine the shortcomings of organoid-based experimental platforms and plausible improvements made possible by cutting-edge neuroengineering technologies. These advancements are expected to enable the development of improved diagnostic strategies and high-throughput translational applications regarding neuroregeneration.
Collapse
Affiliation(s)
| | | | | | - R. Krencik
- Department of Neurosurgery, Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, United States
| |
Collapse
|
40
|
Stöberl N, Maguire E, Salis E, Shaw B, Hall-Roberts H. Human iPSC-derived glia models for the study of neuroinflammation. J Neuroinflammation 2023; 20:231. [PMID: 37817184 PMCID: PMC10566197 DOI: 10.1186/s12974-023-02919-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 10/02/2023] [Indexed: 10/12/2023] Open
Abstract
Neuroinflammation is a complex biological process that plays a significant role in various brain disorders. Microglia and astrocytes are the key cell types involved in inflammatory responses in the central nervous system. Neuroinflammation results in increased levels of secreted inflammatory factors, such as cytokines, chemokines, and reactive oxygen species. To model neuroinflammation in vitro, various human induced pluripotent stem cell (iPSC)-based models have been utilized, including monocultures, transfer of conditioned media between cell types, co-culturing multiple cell types, neural organoids, and xenotransplantation of cells into the mouse brain. To induce neuroinflammatory responses in vitro, several stimuli have been established that can induce responses in either microglia, astrocytes, or both. Here, we describe and critically evaluate the different types of iPSC models that can be used to study neuroinflammation and highlight how neuroinflammation has been induced and measured in these cultures.
Collapse
Affiliation(s)
- Nina Stöberl
- UK Dementia Research Institute (UK DRI), School of Medicine, Cardiff University, Cardiff, CF10 3AT UK
| | - Emily Maguire
- UK Dementia Research Institute (UK DRI), School of Medicine, Cardiff University, Cardiff, CF10 3AT UK
| | - Elisa Salis
- UK Dementia Research Institute (UK DRI), School of Medicine, Cardiff University, Cardiff, CF10 3AT UK
| | - Bethany Shaw
- UK Dementia Research Institute (UK DRI), School of Medicine, Cardiff University, Cardiff, CF10 3AT UK
| | - Hazel Hall-Roberts
- UK Dementia Research Institute (UK DRI), School of Medicine, Cardiff University, Cardiff, CF10 3AT UK
| |
Collapse
|
41
|
Menduti G, Boido M. Recent Advances in High-Content Imaging and Analysis in iPSC-Based Modelling of Neurodegenerative Diseases. Int J Mol Sci 2023; 24:14689. [PMID: 37834135 PMCID: PMC10572296 DOI: 10.3390/ijms241914689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 09/24/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
In the field of neurodegenerative pathologies, the platforms for disease modelling based on patient-derived induced pluripotent stem cells (iPSCs) represent a valuable molecular diagnostic/prognostic tool. Indeed, they paved the way for the in vitro recapitulation of the pathological mechanisms underlying neurodegeneration and for characterizing the molecular heterogeneity of disease manifestations, also enabling drug screening approaches for new therapeutic candidates. A major challenge is related to the choice and optimization of the morpho-functional study designs in human iPSC-derived neurons to deeply detail the cell phenotypes as markers of neurodegeneration. In recent years, the specific combination of high-throughput screening with subcellular resolution microscopy for cell-based high-content imaging (HCI) screening allowed in-depth analyses of cell morphology and neurite trafficking in iPSC-derived neuronal cells by using specific cutting-edge microscopes and automated computational assays. The present work aims to describe the main recent protocols and advances achieved with the HCI analysis in iPSC-based modelling of neurodegenerative diseases, highlighting technical and bioinformatics tips and tricks for further uses and research. To this end, microscopy requirements and the latest computational pipelines to analyze imaging data will be explored, while also providing an overview of the available open-source high-throughput automated platforms.
Collapse
Affiliation(s)
- Giovanna Menduti
- Department of Neuroscience “Rita Levi Montalcini”, Neuroscience Institute Cavalieri Ottolenghi, University of Turin, Regione Gonzole 10, Orbassano, 10043 Turin, TO, Italy;
| | | |
Collapse
|
42
|
Dolan MJ, Therrien M, Jereb S, Kamath T, Gazestani V, Atkeson T, Marsh SE, Goeva A, Lojek NM, Murphy S, White CM, Joung J, Liu B, Limone F, Eggan K, Hacohen N, Bernstein BE, Glass CK, Leinonen V, Blurton-Jones M, Zhang F, Epstein CB, Macosko EZ, Stevens B. Exposure of iPSC-derived human microglia to brain substrates enables the generation and manipulation of diverse transcriptional states in vitro. Nat Immunol 2023; 24:1382-1390. [PMID: 37500887 PMCID: PMC10382323 DOI: 10.1038/s41590-023-01558-2] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/09/2023] [Indexed: 07/29/2023]
Abstract
Microglia, the macrophages of the brain parenchyma, are key players in neurodegenerative diseases such as Alzheimer's disease. These cells adopt distinct transcriptional subtypes known as states. Understanding state function, especially in human microglia, has been elusive owing to a lack of tools to model and manipulate these cells. Here, we developed a platform for modeling human microglia transcriptional states in vitro. We found that exposure of human stem-cell-differentiated microglia to synaptosomes, myelin debris, apoptotic neurons or synthetic amyloid-beta fibrils generated transcriptional diversity that mapped to gene signatures identified in human brain microglia, including disease-associated microglia, a state enriched in neurodegenerative diseases. Using a new lentiviral approach, we demonstrated that the transcription factor MITF drives a disease-associated transcriptional signature and a highly phagocytic state. Together, these tools enable the manipulation and functional interrogation of human microglial states in both homeostatic and disease-relevant contexts.
Collapse
Affiliation(s)
- Michael-John Dolan
- Stanley Center for Psychiatric Research, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Boston Children's Hospital, F.M. Kirby Neurobiology Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Martine Therrien
- Stanley Center for Psychiatric Research, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Boston Children's Hospital, F.M. Kirby Neurobiology Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Saša Jereb
- Stanley Center for Psychiatric Research, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Boston Children's Hospital, F.M. Kirby Neurobiology Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Tushar Kamath
- Stanley Center for Psychiatric Research, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Vahid Gazestani
- Stanley Center for Psychiatric Research, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Trevor Atkeson
- Stanley Center for Psychiatric Research, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Samuel E Marsh
- Boston Children's Hospital, F.M. Kirby Neurobiology Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Aleksandrina Goeva
- Stanley Center for Psychiatric Research, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Neal M Lojek
- Stanley Center for Psychiatric Research, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sarah Murphy
- Boston Children's Hospital, F.M. Kirby Neurobiology Center, Boston, MA, USA
| | | | - Julia Joung
- Broad Institute of MIT and Harvard Cambridge, Cambridge, MA, USA
- Department of Biological Engineering, MIT, Cambridge, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
- Department of Brain and Cognitive Science, MIT, Cambridge, MA, USA
- McGovern Institute for Brain Research at MIT, Cambridge, MA, USA
| | - Bingxu Liu
- Broad Institute of MIT and Harvard Cambridge, Cambridge, MA, USA
| | - Francesco Limone
- Stanley Center for Psychiatric Research, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Leiden University Medical Center, LUMC, Leiden, the Netherlands
| | - Kevin Eggan
- Stanley Center for Psychiatric Research, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Nir Hacohen
- Broad Institute of MIT and Harvard Cambridge, Cambridge, MA, USA
- Department of Medicine, Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Bradley E Bernstein
- Broad Institute of MIT and Harvard Cambridge, Cambridge, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, USA
- Departments of Cell Biology and Pathology, Harvard Medical School, Boston, MA, USA
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Ville Leinonen
- Department of Neurosurgery, Kuopio University Hospital and Institute of Clinical Medicine - Neurosurgery, University of Eastern Finland, Kuopio, Finland
| | - Mathew Blurton-Jones
- Department of Neurobiology and Behavior, Sue and Bill Gross Stem Cell Research Center, UCI Institute for Memory Impairments and Neurological Disorders, Institute for Immunology, University of California, Irvine, CA, USA
| | - Feng Zhang
- Broad Institute of MIT and Harvard Cambridge, Cambridge, MA, USA
- Department of Biological Engineering, MIT, Cambridge, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
- Department of Brain and Cognitive Science, MIT, Cambridge, MA, USA
- McGovern Institute for Brain Research at MIT, Cambridge, MA, USA
| | | | - Evan Z Macosko
- Stanley Center for Psychiatric Research, The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Massachusetts General Hospital, Department of Psychiatry, Boston, MA, USA.
| | - Beth Stevens
- Stanley Center for Psychiatric Research, The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Boston Children's Hospital, F.M. Kirby Neurobiology Center, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard Cambridge, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Boston, MA, USA.
| |
Collapse
|
43
|
Muñoz Herrera OM, Hong BV, Ruiz Mendiola U, Maezawa I, Jin LW, Lebrilla CB, Harvey DJ, Zivkovic AM. Cholesterol, Amyloid Beta, Fructose, and LPS Influence ROS and ATP Concentrations and the Phagocytic Capacity of HMC3 Human Microglia Cell Line. Int J Mol Sci 2023; 24:10396. [PMID: 37373543 PMCID: PMC10299308 DOI: 10.3390/ijms241210396] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/16/2023] [Accepted: 06/17/2023] [Indexed: 06/29/2023] Open
Abstract
Research has found that genes specific to microglia are among the strongest risk factors for Alzheimer's disease (AD) and that microglia are critically involved in the etiology of AD. Thus, microglia are an important therapeutic target for novel approaches to the treatment of AD. High-throughput in vitro models to screen molecules for their effectiveness in reversing the pathogenic, pro-inflammatory microglia phenotype are needed. In this study, we used a multi-stimulant approach to test the usefulness of the human microglia cell 3 (HMC3) cell line, immortalized from a human fetal brain-derived primary microglia culture, in duplicating critical aspects of the dysfunctional microglia phenotype. HMC3 microglia were treated with cholesterol (Chol), amyloid beta oligomers (AβO), lipopolysaccharide (LPS), and fructose individually and in combination. HMC3 microglia demonstrated changes in morphology consistent with activation when treated with the combination of Chol + AβO + fructose + LPS. Multiple treatments increased the cellular content of Chol and cholesteryl esters (CE), but only the combination treatment of Chol + AβO + fructose + LPS increased mitochondrial Chol content. Microglia treated with combinations containing Chol + AβO had lower apolipoprotein E (ApoE) secretion, with the combination of Chol + AβO + fructose + LPS having the strongest effect. Combination treatment with Chol + AβO + fructose + LPS also induced APOE and TNF-α expression, reduced ATP production, increased reactive oxygen species (ROS) concentration, and reduced phagocytosis events. These findings suggest that HMC3 microglia treated with the combination of Chol + AβO + fructose + LPS may be a useful high-throughput screening model amenable to testing on 96-well plates to test potential therapeutics to improve microglial function in the context of AD.
Collapse
Affiliation(s)
- Oscar M. Muñoz Herrera
- Department of Nutrition, University of California, Davis, CA 95616, USA; (O.M.M.H.); (B.V.H.)
| | - Brian V. Hong
- Department of Nutrition, University of California, Davis, CA 95616, USA; (O.M.M.H.); (B.V.H.)
| | - Ulises Ruiz Mendiola
- Department of Pathology and Laboratory Medicine, University of California, Davis Medical Center, Sacramento, CA 95817, USA; (U.R.M.); (I.M.); (L.-W.J.)
| | - Izumi Maezawa
- Department of Pathology and Laboratory Medicine, University of California, Davis Medical Center, Sacramento, CA 95817, USA; (U.R.M.); (I.M.); (L.-W.J.)
| | - Lee-Way Jin
- Department of Pathology and Laboratory Medicine, University of California, Davis Medical Center, Sacramento, CA 95817, USA; (U.R.M.); (I.M.); (L.-W.J.)
| | | | - Danielle J. Harvey
- Department of Public Health Sciences, University of California, Davis, CA 95616, USA;
| | - Angela M. Zivkovic
- Department of Nutrition, University of California, Davis, CA 95616, USA; (O.M.M.H.); (B.V.H.)
| |
Collapse
|
44
|
Qu W, Canoll P, Hargus G. Molecular Insights into Cell Type-specific Roles in Alzheimer's Disease: Human Induced Pluripotent Stem Cell-based Disease Modelling. Neuroscience 2023; 518:10-26. [PMID: 35569647 PMCID: PMC9974106 DOI: 10.1016/j.neuroscience.2022.05.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 05/04/2022] [Accepted: 05/06/2022] [Indexed: 10/18/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia resulting in widespread degeneration of the central nervous system with severe cognitive impairment. Despite the devastating toll of AD, the incomplete understanding of the complex molecular mechanisms hinders the expeditious development of effective cures. Emerging evidence from animal studies has shown that different brain cell types play distinct roles in the pathogenesis of AD. Glutamatergic neurons are preferentially affected in AD and pronounced gliosis contributes to the progression of AD in both a cell-autonomous and a non-cell-autonomous manner. Much has been discovered through genetically modified animal models, yet frequently failed translational attempts to clinical applications call for better disease models. Emerging evidence supports the significance of human-induced pluripotent stem cell (iPSC) derived brain cells in modeling disease development and progression, opening new avenues for the discovery of molecular mechanisms. This review summarizes the function of different cell types in the pathogenesis of AD, such as neurons, microglia, and astrocytes, and recognizes the potential of utilizing the rapidly growing iPSC technology in modeling AD.
Collapse
Affiliation(s)
- Wenhui Qu
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States; Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, United States
| | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States
| | - Gunnar Hargus
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States; Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, United States.
| |
Collapse
|
45
|
Romei MG, Leonard B, Kim I, Kim HS, Lazar GA. Antibody-guided proteases enable selective and catalytic degradation of challenging therapeutic targets. J Biol Chem 2023; 299:104685. [PMID: 37031819 DOI: 10.1016/j.jbc.2023.104685] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 03/08/2023] [Accepted: 03/23/2023] [Indexed: 04/11/2023] Open
Abstract
The exquisite specificity, natural biological functions, and favorable development properties of antibodies make them highly effective agents as drugs. Monoclonal antibodies are particularly strong as inhibitors of systemically accessible targets where trough-level concentrations can sustain full target occupancy. Yet beyond this pharmacologic wheelhouse, antibodies perform suboptimally for targets of high abundance and those not easily accessible from circulation. Fundamentally, this restraint on broader application is due largely to the stoichiometric nature of their activity - one drug molecule is generally able to inhibit a maximum of two target molecules at a time. Enzymes in contrast are able to catalytically turnover multiple substrates, making them a natural sub-stoichiometric solution for targets of high abundance or in poorly accessible sites of action. However, enzymes have their own limitations as drugs, including, in particular the polypharmacology and broad specificity often seen with native enzymes. In this study, we introduce antibody-guided proteolytic enzymes to enable selective sub-stoichiometric turnover of therapeutic targets. We demonstrate that antibody-mediated substrate targeting can enhance enzyme activity and specificity, with proof of concept for two challenging target proteins, amyloid-β (Aβ) and immunoglobulin G (IgG). This work advances a new biotherapeutic platform that combines the favorable properties of antibodies and proteolytic enzymes to more effectively suppress high-bar therapeutic targets.
Collapse
Affiliation(s)
- Matthew G Romei
- Department of Antibody Engineering, Genentech Inc., South San Francisco, CA, USA.
| | - Brandon Leonard
- Department of Antibody Engineering, Genentech Inc., South San Francisco, CA, USA
| | - Ingrid Kim
- Department of Antibody Engineering, Genentech Inc., South San Francisco, CA, USA
| | - Hok Seon Kim
- Department of Antibody Engineering, Genentech Inc., South San Francisco, CA, USA
| | - Greg A Lazar
- Department of Antibody Engineering, Genentech Inc., South San Francisco, CA, USA
| |
Collapse
|
46
|
Whitehouse C, Corbett N, Brownlees J. 3D models of neurodegeneration: implementation in drug discovery. Trends Pharmacol Sci 2023; 44:208-221. [PMID: 36822950 DOI: 10.1016/j.tips.2023.01.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/25/2023] [Accepted: 01/27/2023] [Indexed: 02/24/2023]
Abstract
A lack of in vitro models that robustly represent the complex cellular pathologies underlying neurodegeneration has resulted in a translational gap between in vitro and in vivo results, creating a bottleneck in the development of new therapeutics. In the past decade, new and complex 3D models of the brain have been published at an exponential rate. However, many novel 3D models of neurodegeneration overlook the validation and throughput requirements for implementation in drug discovery. This therefore represents a knowledge gap that could hinder the translation of these models to drug discovery efforts. We review the recent progress in the development of 3D models of neurodegeneration, examining model design benefits and validation techniques, and discuss opportunities and standards for 3D models of neurodegeneration to be implemented in drug discovery and development.
Collapse
Affiliation(s)
| | - Nicola Corbett
- MSD R&D Innovation Centre, 120 Moorgate, London EC2M 6UR, UK
| | - Janet Brownlees
- MSD R&D Innovation Centre, 120 Moorgate, London EC2M 6UR, UK
| |
Collapse
|
47
|
Hebisch M, Klostermeier S, Wolf K, Boccaccini AR, Wolf SE, Tanzi RE, Kim DY. The Impact of the Cellular Environment and Aging on Modeling Alzheimer's Disease in 3D Cell Culture Models. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205037. [PMID: 36642841 PMCID: PMC10015857 DOI: 10.1002/advs.202205037] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/29/2022] [Indexed: 06/13/2023]
Abstract
Creating a cellular model of Alzheimer's disease (AD) that accurately recapitulates disease pathology has been a longstanding challenge. Recent studies showed that human AD neural cells, integrated into three-dimensional (3D) hydrogel matrix, display key features of AD neuropathology. Like in the human brain, the extracellular matrix (ECM) plays a critical role in determining the rate of neuropathogenesis in hydrogel-based 3D cellular models. Aging, the greatest risk factor for AD, significantly alters brain ECM properties. Therefore, it is important to understand how age-associated changes in ECM affect accumulation of pathogenic molecules, neuroinflammation, and neurodegeneration in AD patients and in vitro models. In this review, mechanistic hypotheses is presented to address the impact of the ECM properties and their changes with aging on AD and AD-related dementias. Altered ECM characteristics in aged brains, including matrix stiffness, pore size, and composition, will contribute to disease pathogenesis by modulating the accumulation, propagation, and spreading of pathogenic molecules of AD. Emerging hydrogel-based disease models with differing ECM properties provide an exciting opportunity to study the impact of brain ECM aging on AD pathogenesis, providing novel mechanistic insights. Understanding the role of ECM aging in AD pathogenesis should also improve modeling AD in 3D hydrogel systems.
Collapse
Affiliation(s)
- Matthias Hebisch
- Genetics and Aging Research UnitMcCance Center for Brain health, MassGeneral Institute for Neurodegenerative DiseaseMassachusetts General HospitalHarvard Medical SchoolCharlestownMA02129USA
| | - Stefanie Klostermeier
- Institute of Medical PhysicsFriedrich‐Alexander Universität Erlangen‐Nürnberg91052ErlangenGermany
- Max‐Planck‐Zentrum für Physik und Medizin91054ErlangenGermany
| | - Katharina Wolf
- Department of Medicine 1Friedrich‐Alexander‐Universität Erlangen‐Nürnberg91054ErlangenGermany
| | - Aldo R. Boccaccini
- Institute of BiomaterialsDepartment of Materials Science and EngineeringFriedrich‐Alexander‐Universität Erlangen‐Nürnberg91058ErlangenGermany
| | - Stephan E. Wolf
- Institute of Glass and CeramicsDepartment of Materials Science and EngineeringFriedrich‐Alexander‐Universität Erlangen‐Nürnberg91058ErlangenGermany
| | - Rudolph E. Tanzi
- Genetics and Aging Research UnitMcCance Center for Brain health, MassGeneral Institute for Neurodegenerative DiseaseMassachusetts General HospitalHarvard Medical SchoolCharlestownMA02129USA
| | - Doo Yeon Kim
- Genetics and Aging Research UnitMcCance Center for Brain health, MassGeneral Institute for Neurodegenerative DiseaseMassachusetts General HospitalHarvard Medical SchoolCharlestownMA02129USA
| |
Collapse
|
48
|
Cai W, Wu T, Chen N. The Amyloid-Beta Clearance: From Molecular Targets to Glial and Neural Cells. Biomolecules 2023; 13:313. [PMID: 36830682 PMCID: PMC9953441 DOI: 10.3390/biom13020313] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 02/03/2023] [Accepted: 02/04/2023] [Indexed: 02/11/2023] Open
Abstract
The deposition of amyloid-beta (Aβ) plaques in the brain is one of the primary pathological characteristics of Alzheimer's disease (AD). It can take place 20-30 years before the onset of clinical symptoms. The imbalance between the production and the clearance of Aβ is one of the major causes of AD. Enhancing Aβ clearance at an early stage is an attractive preventive and therapeutic strategy of AD. Direct inhibition of Aβ production and aggregation using small molecules, peptides, and monoclonal antibody drugs has not yielded satisfactory efficacy in clinical trials for decades. Novel approaches are required to understand and combat Aβ deposition. Neurological dysfunction is a complex process that integrates the functions of different types of cells in the brain. The role of non-neurons in AD has not been fully elucidated. An in-depth understanding of the interactions between neurons and non-neurons can contribute to the elucidation of Aβ formation and the identification of effective drug targets. AD patient-derived pluripotent stem cells (PSCs) contain complete disease background information and have the potential to differentiate into various types of neurons and non-neurons in vitro, which may bring new insight into the treatment of AD. Here, we systematically review the latest studies on Aβ clearance and clarify the roles of cell interactions among microglia, astroglia and neurons in response to Aβ plaques, which will be beneficial to explore methods for reconstructing AD disease models using inducible PSCs (iPSCs) through cell differentiation techniques and validating the applications of models in understanding the formation of Aβ plaques. This review may provide the most promising directions of finding the clues for preventing and delaying the development of AD.
Collapse
Affiliation(s)
| | | | - Ning Chen
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| |
Collapse
|
49
|
Sahlgren Bendtsen KM, Hall VJ. The Breakthroughs and Caveats of Using Human Pluripotent Stem Cells in Modeling Alzheimer's Disease. Cells 2023; 12:cells12030420. [PMID: 36766763 PMCID: PMC9913971 DOI: 10.3390/cells12030420] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
Modeling Alzheimer's disease (AD) using human-induced pluripotent stem cells (iPSCs) is a field now spanning 15 years. Developments in the field have shown a shift in using simple 2D cortical neuron models to more advanced tri-cultures and 3D cerebral organoids that recapitulate more features of the disease. This is largely due to development and optimization of new cell protocols. In this review, we highlight recent major breakthroughs in the AD field and the implications this has in modeling AD using iPSCs (AD-iPSCs). To date, AD-iPSCs have been largely used to recapitulate and study impaired amyloid precursor protein (APP) processing and tau phosphorylation in both familial and sporadic AD. AD-iPSCs have also been studied for varying neuronal and glial dysfunctions. Moreover, they have been useful for discovering new molecular mechanisms, such as identifying proteins that bridge APP processing with tau phosphorylation and for identifying molecular pathways that bridge APP processing dysfunction with impaired cholesterol biosynthesis. Perhaps the greatest use of AD-iPSCs has been in discovering compounds via drug screening, that reduce amyloid beta (Aβ) in neurons, such as the anti-inflammatory compound, cromolyn, and antiparasitic drugs, avermectins. In addition, high content screening using AD-iPSCs has led to the identification of statins that can reduce levels of phosphorylated tau (p-Tau) in neurons. Some of these compounds have made it through to testing in human clinical trials. Improvements in omic technologies including single cell RNA sequencing and proteomics as well as advances in production of iPSC-cerebral organoids and tri-cultures is likely to result in the further discovery of new drugs and treatments for AD. Some caveats remain in the field, including, long experimental conditions to create mature neurons, high costs of media that limit research capabilities, and a lack of reproducibility using current iPSC-cerebral organoid protocols. Despite these current limitations, AD-iPSCs remain an excellent cellular model for studying AD mechanisms and for drug discovery.
Collapse
|
50
|
Warden AS, Han C, Hansen E, Trescott S, Nguyen C, Kim R, Schafer D, Johnson A, Wright M, Ramirez G, Lopez-Sanchez M, Coufal NG. Tools for studying human microglia: In vitro and in vivo strategies. Brain Behav Immun 2023; 107:369-382. [PMID: 36336207 PMCID: PMC9810377 DOI: 10.1016/j.bbi.2022.10.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 09/11/2022] [Accepted: 10/13/2022] [Indexed: 11/06/2022] Open
Abstract
Microglia may only represent 10% of central nervous system (CNS) cells but they perform critical roles in development, homeostasis and neurological disease. Microglia are also environmentally regulated, quickly losing their transcriptomic and epigenetic signature after leaving the CNS. This facet of microglia biology is both fascinating and technically challenging influencing the study of the genetics and function of human microglia in a manner that recapitulates the CNS environment. In this review we provide a comprehensive overview of existing in vitro and in vivo methodology to study human microglia, such as immortalized cells lines, stem cell-derived microglia, cerebral organoids and xenotransplantation. Since there is currently no single method that completely recapitulates all hallmarks of human ex vivo adult homeostatic microglia, we also discuss the advantages and limitations of each existing model as a practical guide for researchers.
Collapse
Affiliation(s)
- Anna S Warden
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Claudia Han
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Emily Hansen
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Samantha Trescott
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Celina Nguyen
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Roy Kim
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Danielle Schafer
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Avalon Johnson
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Madison Wright
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Gabriela Ramirez
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Mark Lopez-Sanchez
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Nicole G Coufal
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|