1
|
Beltrami S, Rizzo S, Schiuma G, Cianci G, Narducci M, Baroni M, Di Luca D, Rizzo R, Bortolotti D. West Nile virus non-structural protein 1 promotes amyloid Beta deposition and neurodegeneration. Int J Biol Macromol 2025; 305:141032. [PMID: 39954900 DOI: 10.1016/j.ijbiomac.2025.141032] [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: 11/14/2024] [Revised: 02/04/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025]
Abstract
Recent observations highlight a notable surge in West Nile Virus (WNV) infections in Europe that can lead to neuroinvasive consequences associated with neurodegeneration, mainly triggered by WNV Non-Structural protein 1 (NS1). During viral replication, various protein-protein interactions take place, allowing viral proteins to interact with host factors. NS1 is actively secreted in the bloodstream by infected cells and is known to affect endothelial permeability and host immune response. Focusing on the recently discovered antimicrobial roles of Amyloid-Beta (Aβ) in the context Central Nervous System (CNS), we connected WNV late pathology to overlapping features encountered in neurodegenerative diseases. In fact, CNS viral infections, or presence of specific viral components, activate glial cells, which in turn increase Aβ expression as an antiviral mechanism, leading to Aβ accumulation and neuronal damage. Considering West Nile neuroinvasive disease (WNND) as a possible complication of WNV infection, we investigated the impact of soluble WNV (s)NS1 on glial and neuronal cells, in 2D and 3D in vitro models. We reported an increased Aβ deposition after WNV sNS1 treatment, particularly of Aβ-142 isoform, and increased glial activation with a subsequent neurotoxicity. These findings underscore the crucial role of sNS1 in CNS-related effects during WNV infection, suggesting a novel pathogenetic role.
Collapse
Affiliation(s)
- Silvia Beltrami
- Department of Environmental and Prevention Sciences, University of Ferrara, Ferrara 44121, Italy.
| | - Sabrina Rizzo
- Department of Environmental and Prevention Sciences, University of Ferrara, Ferrara 44121, Italy.
| | - Giovanna Schiuma
- Department of Environmental and Prevention Sciences, University of Ferrara, Ferrara 44121, Italy.
| | - Giorgia Cianci
- Department of Environmental and Prevention Sciences, University of Ferrara, Ferrara 44121, Italy.
| | - Marco Narducci
- Department of Environmental and Prevention Sciences, University of Ferrara, Ferrara 44121, Italy; Temple University, Japan Campus, 1 Chome-14-29 Taishido, Setagaya City, Tokyo 154-0004, Japan.
| | - Marcello Baroni
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara 44121, Italy.
| | - Dario Di Luca
- Department of Medical Sciences, University of Ferrara, Ferrara 44121, Italy.
| | - Roberta Rizzo
- Department of Environmental and Prevention Sciences, University of Ferrara, Ferrara 44121, Italy.
| | - Daria Bortolotti
- Department of Environmental and Prevention Sciences, University of Ferrara, Ferrara 44121, Italy.
| |
Collapse
|
2
|
Wendt S, Lin AJ, Ebert SN, Brennan DJ, Cai W, Bai Y, Kong DY, Sorrentino S, Groten CJ, Lee C, Frew J, Choi HB, Karamboulas K, Delhaye M, Mackenzie IR, Kaplan DR, Miller FD, MacVicar BA, Nygaard HB. A 3D human iPSC-derived multi-cell type neurosphere system to model cellular responses to chronic amyloidosis. J Neuroinflammation 2025; 22:119. [PMID: 40275379 PMCID: PMC12023538 DOI: 10.1186/s12974-025-03433-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 03/31/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is characterized by progressive amyloid beta (Aβ) deposition in the brain, with eventual widespread neurodegeneration. While the cell-specific molecular signature of end-stage AD is reasonably well characterized through autopsy material, less is known about the molecular pathways in the human brain involved in the earliest exposure to Aβ. Human model systems that not only replicate the pathological features of AD but also the transcriptional landscape in neurons, astrocytes and microglia are crucial for understanding disease mechanisms and for identifying novel therapeutic targets. METHODS In this study, we used a human 3D iPSC-derived neurosphere model to explore how resident neurons, microglia and astrocytes and their interplay are modified by chronic amyloidosis induced over 3-5 weeks by supplementing media with synthetic Aβ1 - 42 oligomers. Neurospheres under chronic Aβ exposure were grown with or without microglia to investigate the functional roles of microglia. Neuronal activity and oxidative stress were monitored using genetically encoded indicators, including GCaMP6f and roGFP1, respectively. Single nuclei RNA sequencing (snRNA-seq) was performed to profile Aβ and microglia driven transcriptional changes in neurons and astrocytes, providing a comprehensive analysis of cellular responses. RESULTS Microglia efficiently phagocytosed Aβ inside neurospheres and significantly reduced neurotoxicity, mitigating amyloidosis-induced oxidative stress and neurodegeneration following different exposure times to Aβ. The neuroprotective effects conferred by the presence of microglia was associated with unique gene expression profiles in astrocytes and neurons, including several known AD-associated genes such as APOE. These findings reveal how microglia can directly alter the molecular landscape of AD. CONCLUSIONS Our human 3D neurosphere culture system with chronic Aβ exposure reveals how microglia may be essential for the cellular and transcriptional responses in AD pathogenesis. Microglia are not only neuroprotective in neurospheres but also act as key drivers of Aβ-dependent APOE expression suggesting critical roles for microglia in regulating APOE in the AD brain. This novel, well characterized, functional in vitro platform offers unique opportunities to study the roles and responses of microglia to Aβ modelling key aspects of human AD. This tool will help identify new therapeutic targets, accelerating the transition from discovery to clinical applications.
Collapse
Affiliation(s)
- Stefan Wendt
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada.
| | - Ada J Lin
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Sarah N Ebert
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, V6 T 1Z4, Canada
| | - Declan J Brennan
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Wenji Cai
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Yanyang Bai
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Da Young Kong
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Stefano Sorrentino
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Christopher J Groten
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Christopher Lee
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Jonathan Frew
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
- Opalia Co, Montreal, QC, H2X 3Y7, Canada
| | - Hyun B Choi
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Konstantina Karamboulas
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON, M5G 0 A4, Canada
| | - Mathias Delhaye
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Ian R Mackenzie
- Department of Pathology, Vancouver General Hospital, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - David R Kaplan
- Department of Medical Genetics, University of British Columbia, Vancouver, V6 T 1Z3, Canada
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON, M5G 0 A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1 A8, Canada
| | - Freda D Miller
- Department of Medical Genetics, University of British Columbia, Vancouver, V6 T 1Z3, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, V6 T 1Z4, Canada
| | - Brian A MacVicar
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Haakon B Nygaard
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada.
| |
Collapse
|
3
|
Boschen SL, A Mukerjee A, H Faroqi A, E Rabichow B, Fryer J. Research models to study lewy body dementia. Mol Neurodegener 2025; 20:46. [PMID: 40269912 PMCID: PMC12020038 DOI: 10.1186/s13024-025-00837-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 04/09/2025] [Indexed: 04/25/2025] Open
Abstract
Lewy body dementia (LBD) encompasses neurodegenerative dementias characterized by cognitive fluctuations, visual hallucinations, and parkinsonism. Clinical differentiation of LBD from Alzheimer's disease (AD) remains complex due to symptom overlap, yet approximately 25% of dementia cases are diagnosed as LBD postmortem, primarily identified by the presence of α-synuclein aggregates, tau tangles, and amyloid plaques. These pathological features position LBD as a comorbid condition of both Parkinson's disease (PD) and AD, with over 50% of LBD cases exhibiting co-pathologies. LBD's mixed pathology complicates the development of comprehensive models that reflect the full spectrum of LBD's etiological, clinical, and pathological features. While existing animal and cellular models have facilitated significant discoveries in PD and AD research, they lack specificity in capturing LBD's unique pathogenic mechanisms, limiting the exploration of therapeutic avenues for LBD specifically. This review assesses widely used PD and AD models in terms of their relevance to LBD, particularly focusing on their ability to replicate human disease pathology and assess treatment efficacy. Furthermore, we discuss potential modifications to these models to advance the understanding of LBD mechanisms and propose innovative research directions aimed at developing models with enhanced etiological, face, predictive, and construct validity.
Collapse
Affiliation(s)
- Suelen Lucio Boschen
- Department of Neuroscience, Mayo Clinic Jacksonville, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA.
- Department of Neurosurgery, Mayo Clinic Jacksonville, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA.
| | - Aarushi A Mukerjee
- Department of Neuroscience, Mayo Clinic Jacksonville, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA
| | - Ayman H Faroqi
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Ben E Rabichow
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - John Fryer
- Translational Genomics Research Institute, 445 N 5th St, Phoenix, AZ, 850054, USA
| |
Collapse
|
4
|
Kim JJ, Hebisch M, Kwak SS, Zheng M, Nuli S, Bae JS, Brand E, Tanzi RE, Irimia D, Kim DY. Cryopreserving 3D cell culture models of Alzheimer's disease in hydrogel microbeads. Sci Rep 2025; 15:12543. [PMID: 40216831 PMCID: PMC11992178 DOI: 10.1038/s41598-025-94810-8] [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: 09/09/2024] [Accepted: 03/17/2025] [Indexed: 04/14/2025] Open
Abstract
Long-term preservation of fully differentiated human neurons poses a longstanding challenge in neuroscience research. Numerous cellular disease models have been established using cultured human neuronal cells, including our three-dimensional (3D) human neural cell culture model of Alzheimer's disease (AD). However, the absence of a reliable method for preserving fully differentiated human neural cell cultures for a long time has hindered the sharing and standardization of these models. To address this critical limitation, we focused on cryopreservation, which is the gold standard for long-term preservation, and combined this with three key technological advancements. First, we employed parallelized microfluidic devices for the efficient generation of 3D cell cultures within uniform hydrogel microbeads (~ 220 μm), which facilitate the rapid exchange of media ingredients and cryoprotectants. Second, we implemented a cytophobic microwell system to safeguard neuron-encapsulated microbeads from fusion and aggregation. Third, we developed a novel inducible AD cell model optimized for cryopreservation and AD drug testing. We have successfully maintained encapsulated control and AD neural progenitor cells in microwells during differentiation for 12 days. Notably, fully differentiated human neural cells can be cryopreserved within Matrigel microbeads while retaining intact and mature neuronal processes, exhibiting no signs of damage to neurites following freeze/thaw cycles. Furthermore, we have demonstrated the successful cryopreservation, thawing, and induction of pathogenic Amyloid-β 42 (Aβ42) generation in fully differentiated AD neural progenitor cells. Our study offers a solution for one of the major challenges in neuroscience research, utilizing porous hydrogel microbead structures to facilitate rapid delivery of cryoprotectants and protect complex neuronal structures without undergoing damaging cell dissociation steps. The inducible "3D human microbead model of AD" enhances the speed, efficacy, and reproducibility of AD drug screening.
Collapse
Affiliation(s)
- Jae Jung Kim
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard School of Medicine, Shriners Children's Boston, Boston, MA, 02129, USA
- Department of Chemical Engineering, Hongik University, Seoul, 04066, South Korea
| | - Matthias Hebisch
- Genetics and Aging Research Unit, Institute for Neurodegenerative Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Sang Su Kwak
- Genetics and Aging Research Unit, Institute for Neurodegenerative Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Monica Zheng
- Genetics and Aging Research Unit, Institute for Neurodegenerative Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Shreya Nuli
- Genetics and Aging Research Unit, Institute for Neurodegenerative Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Jun-Seok Bae
- Genetics and Aging Research Unit, Institute for Neurodegenerative Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Emma Brand
- Genetics and Aging Research Unit, Institute for Neurodegenerative Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, Institute for Neurodegenerative Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA.
| | - Daniel Irimia
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard School of Medicine, Shriners Children's Boston, Boston, MA, 02129, USA.
| | - Doo Yeon Kim
- Genetics and Aging Research Unit, Institute for Neurodegenerative Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA.
| |
Collapse
|
5
|
Wu P, Chen D, Wang F, Lu K, Sigurdsson EM, Jin C. Formaldehyde induces and promotes Alzheimer's disease pathologies in a 3D human neural cell culture system. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.27.640690. [PMID: 40093146 PMCID: PMC11908216 DOI: 10.1101/2025.02.27.640690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Alzheimer's disease (AD) arises from complex multilevel interactions between genetic, epigenetic, and environmental factors. Recent studies suggest that exposure to the environmental and occupational toxicant formaldehyde (FA) may play a significant role in AD development. However, the effects of FA exposure on Aβ and tau pathologies in human neural cell 3D culture systems remain unexplored. To investigate FA's role in AD initiation, we differentiated 3D-cultured immortalized human neural progenitor ReN cells (ReNcell VM) into neurons and glial cells, followed by FA treatment. FA exposure for 12 weeks resulted in a dose-dependent increase in Aβ40, Aβ42, and phosphorylated tau levels. To further examine FA's role in AD progression, we established a 3D human neural cell culture AD model by transfecting ReN cells with AD-related mutant genes, including mutant APP and PSEN1, which recapitulate key AD pathological events. Our findings demonstrate that FA exposure significantly elevated Aβ40, Aβ42, and phosphorylated tau levels in this 3D-cultured AD model. These results suggest that FA exposure contributes to the initiation and progression of AD pathology in 3D-cultured human neural cells.
Collapse
|
6
|
Pavlou G, Spitz S, Pramotton FM, Tsai A, Li BM, Wang X, Barr OM, Ko EC, Zhang S, Ashley SJ, Maaser-Hecker A, Choi SH, Jorfi M, Tanzi RE, Kamm RD. Engineered 3D human neurovascular model of Alzheimer's disease to study vascular dysfunction. Biomaterials 2025; 314:122864. [PMID: 39357152 PMCID: PMC12012383 DOI: 10.1016/j.biomaterials.2024.122864] [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: 03/22/2024] [Revised: 09/18/2024] [Accepted: 09/26/2024] [Indexed: 10/04/2024]
Abstract
The blood-brain barrier (BBB) serves as a selective filter that prevents harmful substances from entering the healthy brain. Dysfunction of this barrier is implicated in several neurological diseases. In the context of Alzheimer's disease (AD), BBB breakdown plays a significant role in both the initiation and progression of the disease. This study introduces a three-dimensional (3D) self-assembled in vitro model of the human neurovascular unit to recapitulate some of the complex interactions between the BBB and AD pathologies. It incorporates primary human brain endothelial cells, pericytes and astrocytes, and stem cell-derived neurons and astrocytes harboring Familial AD (FAD) mutations. Over an extended co-culture period, the model demonstrates increased BBB permeability, dysregulation of key endothelial and pericyte markers, and morphological alterations mirroring AD pathologies. The model enables visualization of amyloid-beta (Aβ) accumulation in both neuronal and vascular compartments. This model may serve as a versatile tool for neuroscience research and drug development to provide insights into the dynamic relationship between vascular dysfunction and AD pathogenesis.
Collapse
Affiliation(s)
- Georgios Pavlou
- Department of Mechanical Engineering and Biological Engineering, Massachusetts Institute of Technology, Cambridge MA, 02139, USA
| | - Sarah Spitz
- Department of Mechanical Engineering and Biological Engineering, Massachusetts Institute of Technology, Cambridge MA, 02139, USA
| | - Francesca Michela Pramotton
- Department of Mechanical Engineering and Biological Engineering, Massachusetts Institute of Technology, Cambridge MA, 02139, USA
| | - Alice Tsai
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Brent M Li
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Xun Wang
- Department of Mechanical Engineering and Biological Engineering, Massachusetts Institute of Technology, Cambridge MA, 02139, USA
| | - Olivia M Barr
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Eunkyung Clare Ko
- Department of Mechanical Engineering and Biological Engineering, Massachusetts Institute of Technology, Cambridge MA, 02139, USA
| | - Shun Zhang
- Department of Mechanical Engineering and Biological Engineering, Massachusetts Institute of Technology, Cambridge MA, 02139, USA
| | - Savannah J Ashley
- Department of Mechanical Engineering and Biological Engineering, Massachusetts Institute of Technology, Cambridge MA, 02139, USA
| | - Anna Maaser-Hecker
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Se Hoon Choi
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Mehdi Jorfi
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA.
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA.
| | - Roger D Kamm
- Department of Mechanical Engineering and Biological Engineering, Massachusetts Institute of Technology, Cambridge MA, 02139, USA.
| |
Collapse
|
7
|
Kok LML, Helwegen K, Coveña NF, Heine VM. Human pluripotent stem cell-derived microglia shape neuronal morphology and enhance network activity in vitro. J Neurosci Methods 2025; 415:110354. [PMID: 39724963 DOI: 10.1016/j.jneumeth.2024.110354] [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: 10/23/2024] [Revised: 12/04/2024] [Accepted: 12/22/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND Microglia, the resident immune cells of the central nervous system, play a critical role in maintaining neuronal health, but are often overlooked in traditional neuron-focused in vitro models. NEW METHOD In this study, we developed a novel co-culture system of human pluripotent stem cell (hPSC)-derived microglia and neurons to investigate how hPSC-derived microglia influence neuronal morphology and network activity. Using high-content morphological analysis and multi-electrode arrays (MEA), we demonstrate that these microglia successfully incorporate into neuronal networks and modulate key aspects of neuronal function. RESULTS hPSC-derived microglia significantly reduced cellular debris and altered neuronal morphology by decreasing axonal and dendritic segments and reducing synapse density. Interestingly, despite the decrease in synapse density, neuronal network activity increased. CONCLUSION Our findings underscore the importance of including hPSC-derived microglia in in vitro models to better simulate in vivo neuroglial interactions and provide a platform for investigating neuron-glia dynamics in health and disease.
Collapse
Affiliation(s)
- L M L Kok
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, De Boelelaan, Amsterdam 1081 HV, The Netherlands
| | - K Helwegen
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, De Boelelaan, Amsterdam 1081 HV, The Netherlands
| | - N F Coveña
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, De Boelelaan, Amsterdam 1081 HV, The Netherlands; Laboratory Genetic Metabolic Diseases, Department of Laboratory Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam Neuroscience, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - V M Heine
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, De Boelelaan, Amsterdam 1081 HV, The Netherlands; Department of Child and Adolescent Psychiatry, Emma Center for Personalized Medicine, Emma Children's Hospital, Amsterdam UMC location Vrije Universiteit, Amsterdam Neuroscience, Amsterdam 1081 HV, The Netherlands.
| |
Collapse
|
8
|
Orr A, Kalantarnia F, Nazir S, Bolandi B, Alderson D, O'Grady K, Hoorfar M, Julian LM, Willerth SM. Recent advances in 3D bioprinted neural models: A systematic review on the applications to drug discovery. Adv Drug Deliv Rev 2025; 218:115524. [PMID: 39900293 DOI: 10.1016/j.addr.2025.115524] [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: 09/02/2024] [Revised: 12/12/2024] [Accepted: 01/26/2025] [Indexed: 02/05/2025]
Abstract
The design of neural tissue models with architectural and biochemical relevance to native tissues opens the way for the fundamental study and development of therapies for many disorders with limited treatment options. Here, we systematically review the most recent literature on 3D bioprinted neural models, including their potential for use in drug screening. Neural tissues that model the central nervous system (CNS) from the relevant literature are reviewed with comprehensive summaries of each study, and discussion of the model types, bioinks and additives, cell types used, bioprinted construct shapes and culture time, and the characterization methods used. In this review, we accentuate the lack of standardization among characterization methods to analyze the functionality (including chemical, metabolic and other pathways) and mechanical relevance of the 3D bioprinted constructs, and discuss this as a critical area for future exploration. These gaps must be addressed for this technology to be applied for effective drug screening applications, despite its enormous potential for rapid and efficient drug screening. The future of biomimetic, 3D printed neural tissues is promising and evaluation of the in vivo relevance on multiple levels should be sought to adequately compare model performance and develop viable treatment options for neurodegenerative diseases, or other conditions that affect the CNS.
Collapse
Affiliation(s)
- Amanda Orr
- Department of Mechanical Engineering, University of Victoria, Victoria, BC V8W 2Y2, Canada
| | | | - Shama Nazir
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Behzad Bolandi
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, ON, Canada
| | - Dominic Alderson
- Newcastle University Biosciences Institute, Newcastle-Upon-Tyne, NE2 4HH, UK
| | - Kerrin O'Grady
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, NY 13244, USA
| | - Mina Hoorfar
- Mechanical Engineering, University of Victoria, Victoria, BC V8W 2Y2, Canada
| | - Lisa M Julian
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Stephanie M Willerth
- Department of Mechanical Engineering, University of Victoria, Victoria, BC V8W 2Y2, Canada; Division of Medical Sciences, University of Victoria, Victoria, BC V8W 2Y2, Canada; Centre for Advanced Materials and Technology, University of Victoria, Victoria, BC V8W 2Y2, Canada; School of Biomedical Engineering, University of British Columbia, Victoria, BC V6T 1Z4, Canada.
| |
Collapse
|
9
|
Zellmer JC, Tarantino MB, Kim M, Lomoio S, Maesako M, Hajnóczky G, Bhattacharyya R. Stabilization of mitochondria-associated endoplasmic reticulum membranes regulates Aβ generation in a three-dimensional neural model of Alzheimer's disease. Alzheimers Dement 2025; 21:e14417. [PMID: 39713841 PMCID: PMC11848173 DOI: 10.1002/alz.14417] [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: 07/22/2024] [Revised: 09/24/2024] [Accepted: 10/21/2024] [Indexed: 12/24/2024]
Abstract
INTRODUCTION We previously demonstrated that regulating mitochondria-associated endoplasmic reticulum (ER) membranes (MAMs) affects axonal Aβ generation in a well-characterized three-dimensional (3D) neural Alzheimer's disease (AD) model. MAMs vary in thickness and length, impacting their functions. Here, we examined the effect of MAM thickness on Aβ in our 3D neural model of AD. METHODS We employed fluorescence resonance energy transfer (FRET) or fluorescence-based MAM stabilizers, electron microscopy, Aβ enzyme-linked immunosorbent assay (ELISA), and live-cell imaging with kymography to assess how stabilizing MAMs of different gap widths influence Aβ production and MAM axonal mobility. RESULTS Stabilizing tight MAMs (∼6 nm gap width) significantly increased Aβ levels, whereas basal (∼25 nm) and loose MAMs (∼40 nm) maintained or reduced Aβ levels, respectively. Tight MAMs reduced mitochondrial axonal velocity compared to basal MAMs, while loose MAMs showed severely reduced axonal distribution. DISCUSSION Our findings suggest that stabilizing MAMs of specific gap widths, particularly in axons, without complete destabilization could be an effective therapeutic strategy for AD. HIGHLIGHTS The stabilization of MAMs exacerbates or ameliorates Aβ generation from AD neurons in a MAM gap width-dependent manner. A specific stabilization threshold within the MAM gap width spectrum shifts the amyloidogenic process to non-amyloidogenic. Tight MAMs slow down mitochondrial axonal transport compared to lose MAMs offering a quantitative method for measuring MAM stabilization.
Collapse
Affiliation(s)
- Jacob C. Zellmer
- Genetics and Aging Research UnitMassGeneral Institute for Neurodegenerative DiseaseHenry and Allison McCance Center for Brain HealthDepartment of NeurologyMassachusetts General Hospital, Harvard Medical SchoolCharlestownMassachusettsUSA
| | - Marina B. Tarantino
- Genetics and Aging Research UnitMassGeneral Institute for Neurodegenerative DiseaseHenry and Allison McCance Center for Brain HealthDepartment of NeurologyMassachusetts General Hospital, Harvard Medical SchoolCharlestownMassachusettsUSA
| | - Michelle Kim
- Genetics and Aging Research UnitMassGeneral Institute for Neurodegenerative DiseaseHenry and Allison McCance Center for Brain HealthDepartment of NeurologyMassachusetts General Hospital, Harvard Medical SchoolCharlestownMassachusettsUSA
| | - Selene Lomoio
- Department of NeuroscienceTufts University School of MedicineBostonMassachusettsUSA
| | - Masato Maesako
- Alzheimer's Disease Research UnitMassGeneral Institute for Neurodegenerative DiseaseMassachusetts General Hospital/Harvard Medical SchoolCharlestownMassachusettsUSA
| | - György Hajnóczky
- MitoCare CenterDepartment of PathologyAnatomy & Cell BiologyThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Raja Bhattacharyya
- Genetics and Aging Research UnitMassGeneral Institute for Neurodegenerative DiseaseHenry and Allison McCance Center for Brain HealthDepartment of NeurologyMassachusetts General Hospital, Harvard Medical SchoolCharlestownMassachusettsUSA
| |
Collapse
|
10
|
Alsiraey N, Dewald HD. Nitroxidative stress in human neural progenitor cells: In situ measurement of nitric oxide/peroxynitrite imbalance using metalloporphyrin nanosensors. J Inorg Biochem 2025; 263:112785. [PMID: 39603147 DOI: 10.1016/j.jinorgbio.2024.112785] [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: 10/10/2024] [Revised: 11/18/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024]
Abstract
Nitric oxide (NO) is an essential inorganic signaling molecule produced by constitutive NO synthase (cNOS) in the neurological system. Under pathological conditions, NO rapidly reacts with superoxide (O2•-) to generate peroxynitrite (ONOO¯). Elevated ONOO¯ concentrations induce nitroxidative stress, potentially contributing to numerous pathological processes as observed in neurodegenerative diseases including Alzheimer's disease (AD). Metalloporphyrin nanosensors, (200-300 nm diameter), were applied to quantify the NO/ONOO¯ balance produced by a single human neural progenitor cell (hNPC), in situ. These nanosensors, positioned in proximity of 4-5 ± 1 μm from the hNPCs membrane, enabled real-time measurement of NO and ONOO¯ concentrations following calcium ionophore (CaI) stimulation. The ratio of NO to ONOO¯ concentration ([NO]/[ONOO¯]) was established for the purpose of quantifying nitroxidative stress levels. Normal hNPCs produced a maximum of 107 ± 1 nmol/L of NO and 451 ± 7 nmol/L of ONOO¯, yielding a [NO]/[ONOO¯] ratio of 0.25 ± 0.005. In contrast, the model of the dysfunctional hNPCs, for long-term (48 h) amyloid-beta 42 (Aβ42) exposure significantly altered NO/ONOO¯ production. The NO level decreased to 14 ± 0.1 nmol/L, while ONOO¯ increased to 843 ± 0.8 nmol/L, resulting in a 94 % reduction of the [NO]/[ONOO¯] ratio to 0.016 ± 0.0001. The [NO]/[ONOO¯] ratio is determined by this work as a possible biomarker of nNOS efficiency and hNPC dysfunction, with implications for neurodegenerative disorders such as AD. Promising applications in the early medical diagnosis of neurological illnesses, electrochemical metalloporphyrin nanosensors demonstrate efficacy in real-time nitroxidative stress monitoring.
Collapse
Affiliation(s)
- Nouf Alsiraey
- Department of Chemistry and Biochemistry, Ohio University, Athens, OH 45701, USA; Department of Chemistry, College of Science, Northern Border University, Arar 91431, Saudi Arabia
| | - Howard D Dewald
- Department of Chemistry and Biochemistry, Ohio University, Athens, OH 45701, USA.
| |
Collapse
|
11
|
Naderi Yeganeh P, Kwak SS, Jorfi M, Koler K, Kalatturu T, von Maydell D, Liu Z, Guo K, Choi Y, Park J, Abarca N, Bakiasi G, Cetinbas M, Sadreyev R, Griciuc A, Quinti L, Choi SH, Xia W, Tanzi RE, Hide W, Kim DY. Integrative pathway analysis across humans and 3D cellular models identifies the p38 MAPK-MK2 axis as a therapeutic target for Alzheimer's disease. Neuron 2025; 113:205-224.e8. [PMID: 39610246 PMCID: PMC11757051 DOI: 10.1016/j.neuron.2024.10.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 08/29/2024] [Accepted: 10/31/2024] [Indexed: 11/30/2024]
Abstract
Alzheimer's disease (AD) presents a complex pathological landscape, posing challenges to current therapeutic strategies that primarily target amyloid-β (Aβ). Using a novel integrative pathway activity analysis (IPAA), we identified 83 dysregulated pathways common between both post-mortem AD brains and three-dimensional AD cellular models showing robust Aβ42 accumulation. p38 mitogen-activated protein kinase (MAPK) was the most upregulated common pathway. Active p38 MAPK levels increased in the cellular models, human brains, and 5XFAD mice and selectively localized to presynaptic dystrophic neurites. Unbiased phosphoproteomics confirmed increased phosphorylation of p38 MAPK substrates. Downstream activation of MAPK-activated protein kinase 2 (MK2) plays a crucial role in Aβ42-p38 MAPK-mediated tau pathology. Therapeutic targeting of the p38 MAPK-MK2 axis with selective inhibitors significantly reduced Aβ42-driven tau pathology and neuronal loss. IPAA prioritizes the best models to derisk target-drug discovery by integrating human tissue gene expression with functional readouts from cellular models, enabling the identification and validation of high-confidence AD therapeutic targets.
Collapse
Affiliation(s)
- Pourya Naderi Yeganeh
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Sang Su Kwak
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Mehdi Jorfi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Katjuša Koler
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, Sheffield, UK
| | - Thejesh Kalatturu
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Djuna von Maydell
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Zhiqing Liu
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | | | - Younjung Choi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Joseph Park
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Nelson Abarca
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Grisilda Bakiasi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Murat Cetinbas
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Ruslan Sadreyev
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Ana Griciuc
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Luisa Quinti
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Se Hoon Choi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Weiming Xia
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Geriatric Research Education and Clinical Center, Bedford VA Healthcare System, Bedford, MA, USA; Department of Biological Sciences, University of Massachusetts Kennedy College of Science, Lowell, MA, USA
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA.
| | - Winston Hide
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, Sheffield, UK.
| | - Doo Yeon Kim
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA.
| |
Collapse
|
12
|
Liu E, Zhang Y, Wang JZ. Updates in Alzheimer's disease: from basic research to diagnosis and therapies. Transl Neurodegener 2024; 13:45. [PMID: 39232848 PMCID: PMC11373277 DOI: 10.1186/s40035-024-00432-x] [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: 03/12/2024] [Accepted: 07/11/2024] [Indexed: 09/06/2024] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder, characterized pathologically by extracellular deposition of β-amyloid (Aβ) into senile plaques and intracellular accumulation of hyperphosphorylated tau (pTau) as neurofibrillary tangles. Clinically, AD patients show memory deterioration with varying cognitive dysfunctions. The exact molecular mechanisms underlying AD are still not fully understood, and there are no efficient drugs to stop or reverse the disease progression. In this review, we first provide an update on how the risk factors, including APOE variants, infections and inflammation, contribute to AD; how Aβ and tau become abnormally accumulated and how this accumulation plays a role in AD neurodegeneration. Then we summarize the commonly used experimental models, diagnostic and prediction strategies, and advances in periphery biomarkers from high-risk populations for AD. Finally, we introduce current status of development of disease-modifying drugs, including the newly officially approved Aβ vaccines, as well as novel and promising strategies to target the abnormal pTau. Together, this paper was aimed to update AD research progress from fundamental mechanisms to the clinical diagnosis and therapies.
Collapse
Affiliation(s)
- Enjie Liu
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yao Zhang
- Department of Endocrine, Liyuan Hospital, Key Laboratory of Ministry of Education for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430077, China
| | - Jian-Zhi Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226000, China.
| |
Collapse
|
13
|
Kim Y, Kim H, Cho B, An S, Kang S, Kim S, Kim J. Modeling APOE ε4 familial Alzheimer's disease in directly converted 3D brain organoids. Front Aging Neurosci 2024; 16:1435445. [PMID: 39185458 PMCID: PMC11341472 DOI: 10.3389/fnagi.2024.1435445] [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: 05/20/2024] [Accepted: 07/25/2024] [Indexed: 08/27/2024] Open
Abstract
Brain organoids have become a valuable tool for studying human brain development, disease modeling, and drug testing. However, generating brain organoids with mature neurons is time-intensive and often incomplete, limiting their utility in studying age-related neurodegenerative diseases such as Alzheimer's disease (AD). Here, we report the generation of 3D brain organoids from human fibroblasts through direct reprogramming, with simplicity, efficiency, and reduced variability. We also demonstrate that induced brain organoids from APOE ε4 AD patient fibroblasts capture some disease-specific features and pathologies associated with APOE ε4 AD. Moreover, APOE ε4-induced brain organoids with mutant APP overexpression faithfully recapitulate the acceleration of AD-related pathologies, providing a more physiologically relevant and patient-specific model of familial AD. Importantly, transcriptome analysis reveals that gene sets specific to APOE ε4 patient-induced brain organoids are highly similar to those of APOE ε4 post-mortem AD brains. Overall, induced brain organoids from direct reprogramming offer a promising approach for more efficient and controlled studies of neurodegenerative disease modeling.
Collapse
Affiliation(s)
- Yunkyung Kim
- Department of Chemistry, Dongguk University, Seoul, Republic of Korea
| | - Hongwon Kim
- Department of Chemistry, Dongguk University, Seoul, Republic of Korea
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
| | - Byounggook Cho
- Department of Chemistry, Dongguk University, Seoul, Republic of Korea
| | - Saemin An
- Department of Chemistry, Dongguk University, Seoul, Republic of Korea
| | - Soi Kang
- Department of Chemistry, Dongguk University, Seoul, Republic of Korea
| | - Sumin Kim
- Department of Chemistry, Dongguk University, Seoul, Republic of Korea
| | - Jongpil Kim
- Department of Chemistry, Dongguk University, Seoul, Republic of Korea
| |
Collapse
|
14
|
Mulay AR, Hwang J, Kim DH. Microphysiological Blood-Brain Barrier Systems for Disease Modeling and Drug Development. Adv Healthc Mater 2024; 13:e2303180. [PMID: 38430211 PMCID: PMC11338747 DOI: 10.1002/adhm.202303180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 02/22/2024] [Indexed: 03/03/2024]
Abstract
The blood-brain barrier (BBB) is a highly controlled microenvironment that regulates the interactions between cerebral blood and brain tissue. Due to its selectivity, many therapeutics targeting various neurological disorders are not able to penetrate into brain tissue. Pre-clinical studies using animals and other in vitro platforms have not shown the ability to fully replicate the human BBB leading to the failure of a majority of therapeutics in clinical trials. However, recent innovations in vitro and ex vivo modeling called organs-on-chips have shown the potential to create more accurate disease models for improved drug development. These microfluidic platforms induce physiological stressors on cultured cells and are able to generate more physiologically accurate BBBs compared to previous in vitro models. In this review, different approaches to create BBBs-on-chips are explored alongside their application in modeling various neurological disorders and potential therapeutic efficacy. Additionally, organs-on-chips use in BBB drug delivery studies is discussed, and advances in linking brain organs-on-chips onto multiorgan platforms to mimic organ crosstalk are reviewed.
Collapse
Affiliation(s)
- Atharva R. Mulay
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland 21218
| | - Jihyun Hwang
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland 21218
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205
- Center for Microphysiological Systems, Johns Hopkins University School of Medicine, Baltimore, MD, 21205
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, 21218
| |
Collapse
|
15
|
Smirnova L, Hartung T. The Promise and Potential of Brain Organoids. Adv Healthc Mater 2024; 13:e2302745. [PMID: 38252094 DOI: 10.1002/adhm.202302745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 12/22/2023] [Indexed: 01/23/2024]
Abstract
Brain organoids are 3D in vitro culture systems derived from human pluripotent stem cells that self-organize to model features of the (developing) human brain. This review examines the techniques behind organoid generation, their current and potential applications, and future directions for the field. Brain organoids possess complex architecture containing various neural cell types, synapses, and myelination. They have been utilized for toxicology testing, disease modeling, infection studies, personalized medicine, and gene-environment interaction studies. An emerging concept termed Organoid Intelligence (OI) combines organoids with artificial intelligence systems to generate learning and memory, with the goals of modeling cognition and enabling biological computing applications. Brain organoids allow neuroscience studies not previously achievable with traditional techniques, and have the potential to transform disease modeling, drug development, and the understanding of human brain development and disorders. The aspirational vision of OI parallels the origins of artificial intelligence, and efforts are underway to map a roadmap toward its realization. In summary, brain organoids constitute a disruptive technology that is rapidly advancing and gaining traction across multiple disciplines.
Collapse
Affiliation(s)
- Lena Smirnova
- Center for Alternatives to Animal Testing (CAAT), Bloomberg School of Public Health and Whiting School of Engineering, Johns Hopkins University, 615 N Wolfe St, Baltimore, MD, 21205, USA
| | - Thomas Hartung
- Center for Alternatives to Animal Testing (CAAT), Bloomberg School of Public Health and Whiting School of Engineering, Johns Hopkins University, 615 N Wolfe St, Baltimore, MD, 21205, USA
- CAAT-Europe, University of Konstanz, Universitätsstr. 10, 78464, Konstanz, BW, Germany
| |
Collapse
|
16
|
Hou BY, Wu MH, Hsu HY, Lin YC, Yang DI. Polysaccharides from Basella alba Protect Post-Mitotic Neurons against Cell Cycle Re-Entry and Apoptosis Induced by the Amyloid-Beta Peptide by Blocking Sonic Hedgehog Expression. Int J Mol Sci 2024; 25:7316. [PMID: 39000427 PMCID: PMC11242684 DOI: 10.3390/ijms25137316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/25/2024] [Accepted: 07/01/2024] [Indexed: 07/16/2024] Open
Abstract
The amyloid-beta peptide (Aβ) is the neurotoxic component in senile plaques of Alzheimer's disease (AD) brains. Previously we have reported that Aβ toxicity is mediated by the induction of sonic hedgehog (SHH) to trigger cell cycle re-entry (CCR) and apoptosis in post-mitotic neurons. Basella alba is a vegetable whose polysaccharides carry immunomodulatory and anti-cancer actions, but their protective effects against neurodegeneration have never been reported. Herein, we tested whether polysaccharides derived from Basella alba (PPV-6) may inhibit Aβ toxicity and explored its underlying mechanisms. In differentiated rat cortical neurons, Aβ25-35 reduced cell viability, damaged neuronal structure, and compromised mitochondrial bioenergetic functions, all of which were recovered by PPV-6. Immunocytochemistry and western blotting revealed that Aβ25-35-mediated induction of cell cycle markers including cyclin D1, proliferating cell nuclear antigen (PCNA), and histone H3 phosphorylated at Ser-10 (p-Histone H3) in differentiated neurons was all suppressed by PPV-6, along with mitigation of caspase-3 cleavage. Further studies revealed that PPV-6 inhibited Aβ25-35 induction of SHH; indeed, PPV-6 was capable of suppressing neuronal CCR and apoptosis triggered by the exogenous N-terminal fragment of sonic hedgehog (SHH-N). Our findings demonstrated that, in the fully differentiated neurons, PPV-6 exerts protective actions against Aβ neurotoxicity via the downregulation of SHH to suppress neuronal CCR and apoptosis.
Collapse
Affiliation(s)
- Bo-Yu Hou
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; (B.-Y.H.); (M.-H.W.)
| | - Ming-Hsuan Wu
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; (B.-Y.H.); (M.-H.W.)
| | - Hui-Yu Hsu
- Mynature Biotech Inc., Yilan 260021, Taiwan;
| | - Yi-Chun Lin
- Department of Neurology, Taipei City Hospital Renai Branch, Taipei 106243, Taiwan
| | - Ding-I Yang
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; (B.-Y.H.); (M.-H.W.)
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| |
Collapse
|
17
|
Kang YJ, Hyeon SJ, McQuade A, Lim J, Baek SH, Diep YN, Do KV, Jeon Y, Jo D, Lee CJ, Blurton‐Jones M, Ryu H, Cho H. Neurotoxic Microglial Activation via IFNγ-Induced Nrf2 Reduction Exacerbating Alzheimer's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304357. [PMID: 38482922 PMCID: PMC11132036 DOI: 10.1002/advs.202304357] [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: 06/29/2023] [Revised: 01/08/2024] [Indexed: 05/29/2024]
Abstract
Microglial neuroinflammation appears to be neuroprotective in the early pathological stage, yet neurotoxic, which often precedes neurodegeneration in Alzheimer's disease (AD). However, it remains unclear how the microglial activities transit to the neurotoxic state during AD progression, due to complex neuron-glia interactions. Here, the mechanism of detrimental microgliosis in AD by employing 3D human AD mini-brains, brain tissues of AD patients, and 5XFAD mice is explored. In the human and animal AD models, amyloid-beta (Aβ)-overexpressing neurons and reactive astrocytes produce interferon-gamma (IFNγ) and excessive oxidative stress. IFNγ results in the downregulation of mitogen-activated protein kinase (MAPK) and the upregulation of Kelch-like ECH-associated Protein 1 (Keap1) in microglia, which inactivate nuclear factor erythroid-2-related factor 2 (Nrf2) and sensitize microglia to the oxidative stress and induces a proinflammatory microglia via nuclear factor kappa B (NFκB)-axis. The proinflammatory microglia in turn produce neurotoxic nitric oxide and proinflammatory mediators exacerbating synaptic impairment, phosphorylated-tau accumulation, and discernable neuronal loss. Interestingly, recovering Nrf2 in the microglia prevents the activation of proinflammatory microglia and significantly blocks the tauopathy in AD minibrains. Taken together, it is envisioned that IFNγ-driven Nrf2 downregulation in microglia as a key target to ameliorate AD pathology.
Collapse
Affiliation(s)
- You Jung Kang
- Institute of Quantum BiophysicsSungkyunkwan UniversitySuwonGyeonggi16419Republic of Korea
- Department of BiophysicsSungkyunkwan UniversitySuwonGyeonggi16419Republic of Korea
| | - Seung Jae Hyeon
- Center for Brain DisordersBrain Science InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
| | - Amanda McQuade
- Institute for Neurodegenerative DiseasesUniversity of CaliforniaSan FranciscoCA94158USA
- Department of Neurobiology & BehaviorUniversity of California IrvineIrvineCA92697USA
- Sue and Bill Gross Stem Cell Research CenterUniversity of California IrvineIrvineCA92697USA
- Institute for Memory Impairments and Neurological DisordersUniversity of California IrvineIrvineCA92697USA
| | - Jiwoon Lim
- IBS SchoolUniversity of Science and Technology (UST)Daejeon34114Republic of Korea
- Center for Cognition and SocialityInstitute for Basic Science (IBS)Daejeon34126Republic of Korea
| | - Seung Hyun Baek
- School of PharmacySungkyunkwan UniversitySuwonGyeonggi16419Republic of Korea
| | - Yen N. Diep
- Institute of Quantum BiophysicsSungkyunkwan UniversitySuwonGyeonggi16419Republic of Korea
- Department of BiophysicsSungkyunkwan UniversitySuwonGyeonggi16419Republic of Korea
- Department of Intelligent Precision Healthcare ConvergenceSungkyunkwan UniversitySuwonGyeonggi16419Republic of Korea
| | - Khanh V. Do
- Institute of Quantum BiophysicsSungkyunkwan UniversitySuwonGyeonggi16419Republic of Korea
- Department of Intelligent Precision Healthcare ConvergenceSungkyunkwan UniversitySuwonGyeonggi16419Republic of Korea
| | - Yeji Jeon
- School of PharmacySungkyunkwan UniversitySuwonGyeonggi16419Republic of Korea
| | - Dong‐Gyu Jo
- School of PharmacySungkyunkwan UniversitySuwonGyeonggi16419Republic of Korea
- Biomedical Institute for ConvergenceSungkyunkwan UniversitySuwonGyeonggi16419Republic of Korea
- Samsung Advanced Institute for Health Sciences and TechnologySungkyunkwan UniversitySeoul16419Republic of Korea
| | - C. Justin Lee
- Center for Cognition and SocialityInstitute for Basic Science (IBS)Daejeon34126Republic of Korea
| | - Mathew Blurton‐Jones
- Department of Neurobiology & BehaviorUniversity of California IrvineIrvineCA92697USA
- Sue and Bill Gross Stem Cell Research CenterUniversity of California IrvineIrvineCA92697USA
- Institute for Memory Impairments and Neurological DisordersUniversity of California IrvineIrvineCA92697USA
| | - Hoon Ryu
- Center for Brain DisordersBrain Science InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
| | - Hansang Cho
- Institute of Quantum BiophysicsSungkyunkwan UniversitySuwonGyeonggi16419Republic of Korea
- Department of BiophysicsSungkyunkwan UniversitySuwonGyeonggi16419Republic of Korea
- Department of Intelligent Precision Healthcare ConvergenceSungkyunkwan UniversitySuwonGyeonggi16419Republic of Korea
| |
Collapse
|
18
|
Park S, Laskow TC, Chen J, Guha P, Dawn B, Kim D. Microphysiological systems for human aging research. Aging Cell 2024; 23:e14070. [PMID: 38180277 PMCID: PMC10928588 DOI: 10.1111/acel.14070] [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: 06/30/2023] [Revised: 11/28/2023] [Accepted: 11/30/2023] [Indexed: 01/06/2024] Open
Abstract
Recent advances in microphysiological systems (MPS), also known as organs-on-a-chip (OoC), enable the recapitulation of more complex organ and tissue functions on a smaller scale in vitro. MPS therefore provide the potential to better understand human diseases and physiology. To date, numerous MPS platforms have been developed for various tissues and organs, including the heart, liver, kidney, blood vessels, muscle, and adipose tissue. However, only a few studies have explored using MPS platforms to unravel the effects of aging on human physiology and the pathogenesis of age-related diseases. Age is one of the risk factors for many diseases, and enormous interest has been devoted to aging research. As such, a human MPS aging model could provide a more predictive tool to understand the molecular and cellular mechanisms underlying human aging and age-related diseases. These models can also be used to evaluate preclinical drugs for age-related diseases and translate them into clinical settings. Here, we provide a review on the application of MPS in aging research. First, we offer an overview of the molecular, cellular, and physiological changes with age in several tissues or organs. Next, we discuss previous aging models and the current state of MPS for studying human aging and age-related conditions. Lastly, we address the limitations of current MPS and present future directions on the potential of MPS platforms for human aging research.
Collapse
Affiliation(s)
- Seungman Park
- Department of Mechanical EngineeringUniversity of Nevada, Las VegasLas VegasNevadaUSA
| | - Thomas C. Laskow
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Jingchun Chen
- Nevada Institute of Personalized MedicineUniversity of Nevada, Las VegasLas VegasNevadaUSA
| | - Prasun Guha
- Nevada Institute of Personalized MedicineUniversity of Nevada, Las VegasLas VegasNevadaUSA
- School of Life SciencesUniversity of Nevada, Las VegasLas VegasNevadaUSA
| | - Buddhadeb Dawn
- Department of Internal Medicine, Kirk Kerkorian School of MedicineUniversity of Nevada, Las VegasLas VegasNevadaUSA
| | - Deok‐Ho Kim
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Biomedical EngineeringJohns Hopkins UniversityBaltimoreMarylandUSA
- Center for Microphysiological SystemsJohns Hopkins UniversityBaltimoreMarylandUSA
| |
Collapse
|
19
|
Cuní-López C, Stewart R, Oikari LE, Nguyen TH, Roberts TL, Sun Y, Guo CC, Lupton MK, White AR, Quek H. Advanced patient-specific microglia cell models for pre-clinical studies in Alzheimer's disease. J Neuroinflammation 2024; 21:50. [PMID: 38365833 PMCID: PMC10870454 DOI: 10.1186/s12974-024-03037-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 02/01/2024] [Indexed: 02/18/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is an incurable neurodegenerative disorder with a rapidly increasing prevalence worldwide. Current approaches targeting hallmark pathological features of AD have had no consistent clinical benefit. Neuroinflammation is a major contributor to neurodegeneration and hence, microglia, the brain's resident immune cells, are an attractive target for potentially more effective therapeutic strategies. However, there is no current in vitro model system that captures AD patient-specific microglial characteristics using physiologically relevant and experimentally flexible culture conditions. METHODS To address this shortcoming, we developed novel 3D Matrigel-based monocyte-derived microglia-like cell (MDMi) mono-cultures and co-cultures with neuro-glial cells (ReNcell VM). We used single-cell RNA sequencing (scRNAseq) analysis to compare the transcriptomic signatures of MDMi between model systems (2D, 3D and 3D co-culture) and against published human microglia datasets. To demonstrate the potential of MDMi for use in personalized pre-clinical strategies, we generated and characterized MDMi models from sixteen AD patients and matched healthy controls, and profiled cytokine responses upon treatment with anti-inflammatory drugs (dasatinib and spiperone). RESULTS MDMi in 3D exhibited a more branched morphology and longer survival in culture compared to 2D. scRNAseq uncovered distinct MDMi subpopulations that exhibit higher functional heterogeneity and best resemble human microglia in 3D co-culture. AD MDMi in 3D co-culture showed altered cell-to-cell interactions, growth factor and cytokine secretion profiles and responses to amyloid-β. Drug testing assays revealed patient- and model-specific cytokine responses. CONCLUSION Our study presents a novel, physiologically relevant and AD patient-specific 3D microglia cell model that opens avenues towards improving personalized drug development strategies in AD.
Collapse
Affiliation(s)
- Carla Cuní-López
- Mental Health and Neuroscience Department, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
- Faculty of Medicine, The University of Queensland, Herston, QLD, 4006, Australia
| | - Romal Stewart
- Mental Health and Neuroscience Department, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
- UQ Centre for Clinical Research, The University of Queensland, Brisbane City, QLD, 4029, Australia
| | - Lotta E Oikari
- Mental Health and Neuroscience Department, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
- School of Biomedical Sciences, Queensland University of Technology, Brisbane City, QLD, 4000, Australia
| | - Tam Hong Nguyen
- Scientific Services, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | - Tara L Roberts
- UQ Centre for Clinical Research, The University of Queensland, Brisbane City, QLD, 4029, Australia
- Ingham Institute for Applied Medical Research and School of Medicine, Western Sydney University, Liverpool, NSW, 2170, Australia
| | - Yifan Sun
- Mental Health and Neuroscience Department, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | - Christine C Guo
- Mental Health and Neuroscience Department, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
- ActiGraph LLC, Pensacola, FL, 32502, USA
| | - Michelle K Lupton
- Mental Health and Neuroscience Department, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
- Faculty of Medicine, The University of Queensland, Herston, QLD, 4006, Australia
- School of Biomedical Sciences, Queensland University of Technology, Brisbane City, QLD, 4000, Australia
| | - Anthony R White
- Mental Health and Neuroscience Department, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia.
- Faculty of Medicine, The University of Queensland, Herston, QLD, 4006, Australia.
| | - Hazel Quek
- Mental Health and Neuroscience Department, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia.
- School of Biomedical Sciences, The University of Queensland, Lucia, QLD, 4072, Australia.
- School of Biomedical Sciences, Queensland University of Technology, Brisbane City, QLD, 4000, Australia.
| |
Collapse
|
20
|
Han S, Kim J, Kim SH, Youn W, Kim J, Ji GY, Yang S, Park J, Lee GM, Kim Y, Choi IS. In vitro induction of in vivo-relevant stellate astrocytes in 3D brain-derived, decellularized extracellular matrices. Acta Biomater 2023; 172:218-233. [PMID: 37788738 DOI: 10.1016/j.actbio.2023.09.046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 10/05/2023]
Abstract
In vitro fabrication of 3D cell culture systems that could provide in vivo tissue-like, structural, and biochemical environments to neural cells is essential not only for fundamental studies on brain function and behavior, but also for tissue engineering and regenerative medicine applicable to neural injury and neurodegenerative diseases. In particular, for astrocytes-which actively respond to the surroundings and exhibit varied morphologies based on stimuli (e.g., stiffness and chemicals) in vitro, as well as physiological or pathological conditions in vivo-it is crucial to establish an appropriate milieu in in vitro culture platforms. Herein, we report the induction of in vivo-relevant, stellate-shaped astrocytes derived from cortices of Rattus norvegicus by constructing the 3D cell culture systems of brain-derived, decellularized extracellular matrices (bdECMs). The bdECM hydrogels were mechanically stable and soft, and the bdECM-based 3D scaffolds supplied biochemically active environments that astrocytes could interact with, leading to the development of in vivo-like stellate structures. In addition to the distinct morphology with actively elongated endfeet, the astrocytes, cultured in 3D bdECM scaffolds, would have neurosupportive characteristics, indicated by the accelerated neurite outgrowth in the astrocyte-conditioned media. Furthermore, next-generation sequencing showed that the gene expression profiles of astrocytes cultured in bdECMs were significantly different from those cultured on 2D surfaces. The stellate-shaped astrocytes in the bdECMs were analyzed to have reached a more mature state, for instance, with decreased expression of genes for scaffold ECMs, actin filaments, and cell division. The results suggest that the bdECM-based 3D culture system offers an advanced platform for culturing primary cortical astrocytes and their mixtures with other neural cells, providing a brain-like, structural and biochemical milieu that promotes the maturity and in vivo-like characteristics of astrocytes in both form and gene expression. STATEMENT OF SIGNIFICANCE: Decellularized extracellular matrices (dECMs) have emerged as strong candidates for the construction of three-dimensional (3D) cell cultures in vitro, owing to the potential to provide native biochemical and physical environments. In this study, we fabricated hydrogels of brain-derived dECMs (bdECMs) and cultured primary astrocytes within the bdECM hydrogels in a 3D context. The cultured astrocytes exhibited a stellate morphology distinct from conventional 2D cultures, featuring tridimensionally elongated endfeet. qRT-PCR and NGS-based transcriptomic analyses revealed gene expression patterns indicative of a more mature state, compared with the 2D culture. Moreover, astrocytes cultured in bdECMs showed neurosupportive characteristics, as demonstrated by the accelerated neurite outgrowth in astrocyte-conditioned media. We believe that the bdECM hydrogel-based culture system can serve as an in vitro model system for astrocytes and their coculture with other neural cells, holding significant potential for neural engineering and therapeutic applications.
Collapse
Affiliation(s)
- Sol Han
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon 34141, South Korea
| | - Jungnam Kim
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon 34141, South Korea
| | - Su Hyun Kim
- Department of Biological Sciences, KAIST, Daejeon 34141, South Korea
| | - Wongu Youn
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon 34141, South Korea
| | - Jihoo Kim
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon 34141, South Korea
| | - Gil Yong Ji
- Cannabis Medical, Inc., Asan 31418, South Korea
| | - Seoin Yang
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon 34141, South Korea
| | - Joohyouck Park
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon 34141, South Korea
| | - Gyun Min Lee
- Department of Biological Sciences, KAIST, Daejeon 34141, South Korea
| | | | - Insung S Choi
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon 34141, South Korea; Department of Bio and Brain Engineering, KAIST, Daejeon 34141, South Korea.
| |
Collapse
|
21
|
Kim E, Kim H, Jedrychowski MP, Bakiasi G, Park J, Kruskop J, Choi Y, Kwak SS, Quinti L, Kim DY, Wrann CD, Spiegelman BM, Tanzi RE, Choi SH. Irisin reduces amyloid-β by inducing the release of neprilysin from astrocytes following downregulation of ERK-STAT3 signaling. Neuron 2023; 111:3619-3633.e8. [PMID: 37689059 PMCID: PMC10840702 DOI: 10.1016/j.neuron.2023.08.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/09/2023] [Accepted: 08/11/2023] [Indexed: 09/11/2023]
Abstract
A pathological hallmark of Alzheimer's disease (AD) is the deposition of amyloid-β (Aβ) protein in the brain. Physical exercise has been shown to reduce Aβ burden in various AD mouse models, but the underlying mechanisms have not been elucidated. Irisin, an exercise-induced hormone, is the secreted form of fibronectin type-III-domain-containing 5 (FNDC5). Here, using a three-dimensional (3D) cell culture model of AD, we show that irisin significantly reduces Aβ pathology by increasing astrocytic release of the Aβ-degrading enzyme neprilysin (NEP). This is mediated by downregulation of ERK-STAT3 signaling. Finally, we show that integrin αV/β5 acts as the irisin receptor on astrocytes required for irisin-induced release of astrocytic NEP, leading to clearance of Aβ. Our findings reveal for the first time a cellular and molecular mechanism by which exercise-induced irisin attenuates Aβ pathology, suggesting a new target pathway for therapies aimed at the prevention and treatment of AD.
Collapse
Affiliation(s)
- Eunhee Kim
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Hyeonwoo Kim
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Cell Biology, Harvard University Medical School, Boston, MA 02115, USA; Department of Biological Sciences, Korea Advanced Institute of Science & Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Mark P Jedrychowski
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Cell Biology, Harvard University Medical School, Boston, MA 02115, USA
| | - Grisilda Bakiasi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Joseph Park
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jane Kruskop
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Younjung Choi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Sang Su Kwak
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Luisa Quinti
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Doo Yeon Kim
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Christiane D Wrann
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA; Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Bruce M Spiegelman
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Cell Biology, Harvard University Medical School, Boston, MA 02115, USA
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA.
| | - Se Hoon Choi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA.
| |
Collapse
|
22
|
Xue H, Gate S, Gentry E, Losert W, Cao K. Development of an accelerated cellular model for early changes in Alzheimer's disease. Sci Rep 2023; 13:18384. [PMID: 37884611 PMCID: PMC10603068 DOI: 10.1038/s41598-023-45826-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 10/24/2023] [Indexed: 10/28/2023] Open
Abstract
Alzheimer's Disease (AD) is a leading cause of dementia characterized by amyloid plaques and neurofibrillary tangles, and its pathogenesis remains unclear. Current cellular models for AD often require several months to exhibit phenotypic features due to the lack of an aging environment in vitro. Lamin A is a key component of the nuclear lamina. Progerin, a truncated protein resulting from specific lamin A mutations, causes Hutchinson-Gilford Progeria Syndrome (HGPS), a disease that prematurely ages individuals. Studies have reported that lamin A expression is induced in the brains of AD patients, and overlapping cellular phenotypes have been observed between HGPS and AD cells. In this study, we investigated the effects of exogenous progerin expression on neural progenitor cells carrying familial AD mutations (FAD). Within three to four weeks of differentiation, these cells exhibited robust AD phenotypes, including increased tau phosphorylation, amyloid plaque accumulation, and an elevated Aβ42 to Aβ40 ratio. Additionally, progerin expression significantly increased AD cellular phenotypes such as cell death and cell cycle re-entry. Our results suggest that progerin expression could be used to create an accelerated model for AD development and drug screening.
Collapse
Affiliation(s)
- Huijing Xue
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, 20742, USA
| | - Sylvester Gate
- Institute of Physical Sciences, University of Maryland, College Park, MD, 20742, USA
| | - Emma Gentry
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, 20742, USA
| | - Wolfgang Losert
- Institute of Physical Sciences, University of Maryland, College Park, MD, 20742, USA
| | - Kan Cao
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, 20742, USA.
| |
Collapse
|
23
|
Ko EC, Spitz S, Pramotton FM, Barr OM, Xu C, Pavlou G, Zhang S, Tsai A, Maaser-Hecker A, Jorfi M, Choi SH, Tanzi RE, Kamm RD. Accelerating the in vitro emulation of Alzheimer's disease-associated phenotypes using a novel 3D blood-brain barrier neurosphere co-culture model. Front Bioeng Biotechnol 2023; 11:1251195. [PMID: 37901842 PMCID: PMC10600382 DOI: 10.3389/fbioe.2023.1251195] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 09/18/2023] [Indexed: 10/31/2023] Open
Abstract
High failure rates in clinical trials for neurodegenerative disorders such as Alzheimer's disease have been linked to an insufficient predictive validity of current animal-based disease models. This has created an increasing demand for alternative, human-based models capable of emulating key pathological phenotypes in vitro. Here, a three-dimensional Alzheimer's disease model was developed using a compartmentalized microfluidic device that combines a self-assembled microvascular network of the human blood-brain barrier with neurospheres derived from Alzheimer's disease-specific neural progenitor cells. To shorten microfluidic co-culture times, neurospheres were pre-differentiated for 21 days to express Alzheimer's disease-specific pathological phenotypes prior to the introduction into the microfluidic device. In agreement with post-mortem studies and Alzheimer's disease in vivo models, after 7 days of co-culture with pre-differentiated Alzheimer's disease-specific neurospheres, the three-dimensional blood-brain barrier network exhibited significant changes in barrier permeability and morphology. Furthermore, vascular networks in co-culture with Alzheimer's disease-specific microtissues displayed localized β-amyloid deposition. Thus, by interconnecting a microvascular network of the blood-brain barrier with pre-differentiated neurospheres the presented model holds immense potential for replicating key neurovascular phenotypes of neurodegenerative disorders in vitro.
Collapse
Affiliation(s)
- Eunkyung Clare Ko
- Department of Mechanical Engineering and Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Sarah Spitz
- Department of Mechanical Engineering and Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Francesca Michela Pramotton
- Department of Mechanical Engineering and Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Olivia M. Barr
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Ciana Xu
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Georgios Pavlou
- Department of Mechanical Engineering and Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Shun Zhang
- Department of Mechanical Engineering and Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Alice Tsai
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Anna Maaser-Hecker
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Mehdi Jorfi
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Se Hoon Choi
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Rudolph E. Tanzi
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Roger D. Kamm
- Department of Mechanical Engineering and Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| |
Collapse
|
24
|
Maniv I, Sarji M, Bdarneh A, Feldman A, Ankawa R, Koren E, Magid-Gold I, Reis N, Soteriou D, Salomon-Zimri S, Lavy T, Kesselman E, Koifman N, Kurz T, Kleifeld O, Michaelson D, van Leeuwen FW, Verheijen BM, Fuchs Y, Glickman MH. Altered ubiquitin signaling induces Alzheimer's disease-like hallmarks in a three-dimensional human neural cell culture model. Nat Commun 2023; 14:5922. [PMID: 37739965 PMCID: PMC10516951 DOI: 10.1038/s41467-023-41545-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 09/08/2023] [Indexed: 09/24/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by toxic protein accumulation in the brain. Ubiquitination is essential for protein clearance in cells, making altered ubiquitin signaling crucial in AD development. A defective variant, ubiquitin B + 1 (UBB+1), created by a non-hereditary RNA frameshift mutation, is found in all AD patient brains post-mortem. We now detect UBB+1 in human brains during early AD stages. Our study employs a 3D neural culture platform derived from human neural progenitors, demonstrating that UBB+1 alone induces extracellular amyloid-β (Aβ) deposits and insoluble hyperphosphorylated tau aggregates. UBB+1 competes with ubiquitin for binding to the deubiquitinating enzyme UCHL1, leading to elevated levels of amyloid precursor protein (APP), secreted Aβ peptides, and Aβ build-up. Crucially, silencing UBB+1 expression impedes the emergence of AD hallmarks in this model system. Our findings highlight the significance of ubiquitin signalling as a variable contributing to AD pathology and present a nonclinical platform for testing potential therapeutics.
Collapse
Affiliation(s)
- Inbal Maniv
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Mahasen Sarji
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Anwar Bdarneh
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Alona Feldman
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Roi Ankawa
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Elle Koren
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Inbar Magid-Gold
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Noa Reis
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Despina Soteriou
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Shiran Salomon-Zimri
- Department of Neurobiology, The George S. Wise Faculty of Life Sciences, The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Tali Lavy
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Ellina Kesselman
- The Wolfson Department of Chemical Engineering, The Technion Center for Electron Microscopy of Soft Matter, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Naama Koifman
- The Wolfson Department of Chemical Engineering, The Technion Center for Electron Microscopy of Soft Matter, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Thimo Kurz
- School of Molecular Biosciences, University of Glasgow, Glasgow, G12 8QQ, Scotland, UK
| | - Oded Kleifeld
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Daniel Michaelson
- Department of Neurobiology, The George S. Wise Faculty of Life Sciences, The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Fred W van Leeuwen
- Department of Neuroscience, Maastricht University, 6229 ER, Maastricht, the Netherlands
| | - Bert M Verheijen
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
- Department of Neuroscience, Maastricht University, 6229 ER, Maastricht, the Netherlands
| | - Yaron Fuchs
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel.
- Augmanity, Rehovot, 7670308, Israel.
| | - Michael H Glickman
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel.
| |
Collapse
|
25
|
Jorfi M, Park J, Hall CK, Lin CCJ, Chen M, von Maydell D, Kruskop JM, Kang B, Choi Y, Prokopenko D, Irimia D, Kim DY, Tanzi RE. Infiltrating CD8 + T cells exacerbate Alzheimer's disease pathology in a 3D human neuroimmune axis model. Nat Neurosci 2023; 26:1489-1504. [PMID: 37620442 PMCID: PMC11184920 DOI: 10.1038/s41593-023-01415-3] [Citation(s) in RCA: 71] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 07/18/2023] [Indexed: 08/26/2023]
Abstract
Brain infiltration of peripheral immune cells and their interactions with brain-resident cells may contribute to Alzheimer's disease (AD) pathology. To examine these interactions, in the present study we developed a three-dimensional human neuroimmune axis model comprising stem cell-derived neurons, astrocytes and microglia, together with peripheral immune cells. We observed an increase in the number of T cells (but not B cells) and monocytes selectively infiltrating into AD relative to control cultures. Infiltration of CD8+ T cells into AD cultures led to increased microglial activation, neuroinflammation and neurodegeneration. Using single-cell RNA-sequencing, we identified that infiltration of T cells into AD cultures led to induction of interferon-γ and neuroinflammatory pathways in glial cells. We found key roles for the C-X-C motif chemokine ligand 10 (CXCL10) and its receptor, CXCR3, in regulating T cell infiltration and neuronal damage in AD cultures. This human neuroimmune axis model is a useful tool to study the effects of peripheral immune cells in brain disease.
Collapse
Affiliation(s)
- Mehdi Jorfi
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Charlestown, MA, USA.
| | - Joseph Park
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Clare K Hall
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Chih-Chung Jerry Lin
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Meng Chen
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Djuna von Maydell
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Jane M Kruskop
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Byunghoon Kang
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Younjung Choi
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Dmitry Prokopenko
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Daniel Irimia
- Harvard Medical School, Boston, MA, USA
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Charlestown, MA, USA
- Shriners Burns Hospital, Boston, MA, USA
| | - Doo Yeon Kim
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
26
|
Kang YJ, Diep YN, Tran M, Tran VTA, Ambrin G, Ngo H, Cho H. Three-dimensional human neural culture on a chip recapitulating neuroinflammation and neurodegeneration. Nat Protoc 2023; 18:2838-2867. [PMID: 37542184 DOI: 10.1038/s41596-023-00861-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 05/30/2023] [Indexed: 08/06/2023]
Abstract
Neuroinflammation has either beneficial or detrimental effects, depending on risk factors and neuron-glia interactions in neurological disorders. However, studying neuroinflammation has been challenging due to the complexity of cell-cell interactions and lack of physio-pathologically relevant neuroinflammatory models. Here, we describe our three-dimensional microfluidic multicellular human neural culture model, referred to as a 'brain-on-a-chip' (BoC). This elucidates neuron-glia interactions in a controlled manner and recapitulates pathological signatures of the major neurological disorders: dementia, brain tumor and brain edema. This platform includes a chemotaxis module offering a week-long, stable chemo-gradient compared with the few hours in other chemotaxis models. Additionally, compared with conventional brain models cultured with mixed phenotypes of microglia, our BoC can separate the disease-associated microglia out of heterogeneous population and allow selective neuro-glial engagement in three dimensions. This provides benefits of interpreting the neuro-glia interactions while revealing that the prominent activation of innate immune cells is the risk factor leading to synaptic impairment and neuronal loss, validated in our BoC models of disorders. This protocol describes how to fabricate and implement our human BoC, manipulate in real time and perform end-point analyses. It takes 2 d to set up the device and cell preparations, 1-9 weeks to develop brain models under disease conditions and 2-3 d to carry out analyses. This protocol requires at least 1 month training for researchers with basic molecular biology techniques. Taken together, our human BoCs serve as reliable and valuable platforms to investigate pathological mechanisms involving neuroinflammation and to assess therapeutic strategies modulating neuroinflammation in neurological disorders.
Collapse
Affiliation(s)
- You Jung Kang
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
| | - Yen N Diep
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Republic of Korea
| | - Minh Tran
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Republic of Korea
| | - Van Thi Ai Tran
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
| | - Ghuncha Ambrin
- Department of Psychiatry, School of Medicine, University of California San Diego, San Diego, CA, USA
| | - Huyen Ngo
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Republic of Korea
| | - Hansang Cho
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea.
- Department of Biophysics, Sungkyunkwan University, Suwon, Republic of Korea.
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Republic of Korea.
| |
Collapse
|
27
|
Mullis AS, Kaplan DL. Functional bioengineered tissue models of neurodegenerative diseases. Biomaterials 2023; 298:122143. [PMID: 37146365 PMCID: PMC10209845 DOI: 10.1016/j.biomaterials.2023.122143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 04/27/2023] [Accepted: 05/01/2023] [Indexed: 05/07/2023]
Abstract
Aging-associated neurodegenerative diseases, such as Alzheimer's and Parkinson's diseases remain poorly understood and no disease-modifying treatments exist despite decades of investigation. Predominant in vitro (e.g., 2D cell culture, organoids) and in vivo (e.g., mouse) models of these diseases are insufficient mimics of human brain tissue structure and function and of human neurodegenerative pathobiology, and have thus contributed to this collective translational failure. This has been a longstanding challenge in the field, and new strategies are required to address both fundamental and translational needs. Bioengineered tissue culture models constitute a class of promising alternatives, as they can overcome the low cell density, poor nutrient exchange, and long term culturability limitations of existing in vitro models. Further, they can reconstruct the structural, mechanical, and biochemical cues of native brain tissue, providing a better mimic of human brain tissues for in vitro pathobiological investigation and drug development. We discuss bioengineering techniques for the generation of these neurodegenerative tissue models, including biomaterials-, organoid-, and microfluidics-based approaches, and design considerations for their construction. To aid the development of the next generation of functional neurodegenerative disease models, we discuss approaches to incorporate greater cellular diversity and simulate aging processes within bioengineered brain tissues.
Collapse
Affiliation(s)
- Adam S Mullis
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA.
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA; Allen Discovery Center, Tufts University, Medford, MA, 02155, USA.
| |
Collapse
|
28
|
D'Antoni C, Mautone L, Sanchini C, Tondo L, Grassmann G, Cidonio G, Bezzi P, Cordella F, Di Angelantonio S. Unlocking Neural Function with 3D In Vitro Models: A Technical Review of Self-Assembled, Guided, and Bioprinted Brain Organoids and Their Applications in the Study of Neurodevelopmental and Neurodegenerative Disorders. Int J Mol Sci 2023; 24:10762. [PMID: 37445940 DOI: 10.3390/ijms241310762] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/18/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Understanding the complexities of the human brain and its associated disorders poses a significant challenge in neuroscience. Traditional research methods have limitations in replicating its intricacies, necessitating the development of in vitro models that can simulate its structure and function. Three-dimensional in vitro models, including organoids, cerebral organoids, bioprinted brain models, and functionalized brain organoids, offer promising platforms for studying human brain development, physiology, and disease. These models accurately replicate key aspects of human brain anatomy, gene expression, and cellular behavior, enabling drug discovery and toxicology studies while providing insights into human-specific phenomena not easily studied in animal models. The use of human-induced pluripotent stem cells has revolutionized the generation of 3D brain structures, with various techniques developed to generate specific brain regions. These advancements facilitate the study of brain structure development and function, overcoming previous limitations due to the scarcity of human brain samples. This technical review provides an overview of current 3D in vitro models of the human cortex, their development, characterization, and limitations, and explores the state of the art and future directions in the field, with a specific focus on their applications in studying neurodevelopmental and neurodegenerative disorders.
Collapse
Affiliation(s)
- Chiara D'Antoni
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
- Center for Life Nano- and Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| | - Lorenza Mautone
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
- Center for Life Nano- and Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| | - Caterina Sanchini
- Center for Life Nano- and Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| | - Lucrezia Tondo
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
- Center for Life Nano- and Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| | - Greta Grassmann
- Center for Life Nano- and Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, 00185 Rome, Italy
| | - Gianluca Cidonio
- Center for Life Nano- and Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| | - Paola Bezzi
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
- Department of Fundamental Neurosciences, University of Lausanne, 1011 Lausanne, Switzerland
| | - Federica Cordella
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
- Center for Life Nano- and Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| | - Silvia Di Angelantonio
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
- Center for Life Nano- and Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
- D-Tails s.r.l., 00165 Rome, Italy
| |
Collapse
|
29
|
Salgado B, Sastre I, Bullido MJ, Aldudo J. Herpes Simplex Virus Type 1 Induces AD-like Neurodegeneration Markers in Human Progenitor and Differentiated ReNcell VM Cells. Microorganisms 2023; 11:1205. [PMID: 37317179 DOI: 10.3390/microorganisms11051205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 06/16/2023] Open
Abstract
An increasing body of evidence strongly suggests that infections or reactivations of herpes simplex virus type 1 (HSV-1) may be closely linked to Alzheimer's disease (AD). Promising results have been obtained using cell and animal models of HSV-1 infection, contributing to the understanding of the molecular mechanisms linking HSV-1 infection and AD neurodegeneration. ReNcell VM is a human neural stem cell line that has been used as a model system to study the impact of various infectious agents on the central nervous system. In this study, we demonstrate the suitability of the ReNcell VM cell line for developing a new in vitro model of HSV-1 infection. By following standard differentiation protocols, we were able to derive various nervous cell types, including neurons, astrocytes, and oligodendrocytes, from neural precursors. Additionally, we demonstrated the susceptibility of ReNcell VM cells, including precursor and differentiated cells, to HSV-1 infection and subsequent viral-induced AD-like neurodegeneration. Our findings support the use of this cell line to generate a new research platform for investigating AD neuropathology and its most significant risk factors, which may lead to important discoveries in the context of this highly impactful disease.
Collapse
Affiliation(s)
- Blanca Salgado
- Centro de Biologia Molecular "Severo Ochoa" (C.S.I.C.-U.A.M.), Universidad Autonoma de Madrid, 28049 Madrid, Spain
- Centro de Investigacion Biomedica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain
| | - Isabel Sastre
- Centro de Biologia Molecular "Severo Ochoa" (C.S.I.C.-U.A.M.), Universidad Autonoma de Madrid, 28049 Madrid, Spain
- Centro de Investigacion Biomedica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain
| | - Maria J Bullido
- Centro de Biologia Molecular "Severo Ochoa" (C.S.I.C.-U.A.M.), Universidad Autonoma de Madrid, 28049 Madrid, Spain
- Centro de Investigacion Biomedica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Universitario La Paz-IdiPAZ, 28046 Madrid, Spain
| | - Jesus Aldudo
- Centro de Biologia Molecular "Severo Ochoa" (C.S.I.C.-U.A.M.), Universidad Autonoma de Madrid, 28049 Madrid, Spain
- Centro de Investigacion Biomedica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Universitario La Paz-IdiPAZ, 28046 Madrid, Spain
| |
Collapse
|
30
|
Wittrahm R, Takalo M, Kuulasmaa T, Mäkinen PM, Mäkinen P, Končarević S, Fartzdinov V, Selzer S, Kokkola T, Antikainen L, Martiskainen H, Kemppainen S, Marttinen M, Jeskanen H, Rostalski H, Rahunen E, Kivipelto M, Ngandu T, Natunen T, Lambert JC, Tanzi RE, Kim DY, Rauramaa T, Herukka SK, Soininen H, Laakso M, Pike I, Leinonen V, Haapasalo A, Hiltunen M. Protective Alzheimer's disease-associated APP A673T variant predominantly decreases sAPPβ levels in cerebrospinal fluid and 2D/3D cell culture models. Neurobiol Dis 2023; 182:106140. [PMID: 37120095 DOI: 10.1016/j.nbd.2023.106140] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/23/2023] [Accepted: 04/25/2023] [Indexed: 05/01/2023] Open
Abstract
The rare A673T variant was the first variant found within the amyloid precursor protein (APP) gene conferring protection against Alzheimer's disease (AD). Thereafter, different studies have discovered that the carriers of the APP A673T variant show reduced levels of amyloid beta (Aβ) in the plasma and better cognitive performance at high age. Here, we analyzed cerebrospinal fluid (CSF) and plasma of APP A673T carriers and control individuals using a mass spectrometry-based proteomics approach to identify differentially regulated targets in an unbiased manner. Furthermore, the APP A673T variant was introduced into 2D and 3D neuronal cell culture models together with the pathogenic APP Swedish and London mutations. Consequently, we now report for the first time the protective effects of the APP A673T variant against AD-related alterations in the CSF, plasma, and brain biopsy samples from the frontal cortex. The CSF levels of soluble APPβ (sAPPβ) and Aβ42 were significantly decreased on average 9-26% among three APP A673T carriers as compared to three well-matched controls not carrying the protective variant. Consistent with these CSF findings, immunohistochemical assessment of cortical biopsy samples from the same APP A673T carriers did not reveal Aβ, phospho-tau, or p62 pathologies. We identified differentially regulated targets involved in protein phosphorylation, inflammation, and mitochondrial function in the CSF and plasma samples of APP A673T carriers. Some of the identified targets showed inverse levels in AD brain tissue with respect to increased AD-associated neurofibrillary pathology. In 2D and 3D neuronal cell culture models expressing APP with the Swedish and London mutations, the introduction of the APP A673T variant resulted in lower sAPPβ levels. Concomitantly, the levels of sAPPα were increased, while decreased levels of CTFβ and Aβ42 were detected in some of these models. Our findings emphasize the important role of APP-derived peptides in the pathogenesis of AD and demonstrate the effectiveness of the protective APP A673T variant to shift APP processing towards the non-amyloidogenic pathway in vitro even in the presence of two pathogenic mutations.
Collapse
Affiliation(s)
- Rebekka Wittrahm
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland.
| | - Mari Takalo
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland.
| | - Teemu Kuulasmaa
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland.
| | - Petra M Mäkinen
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland.
| | - Petri Mäkinen
- A.I. Virtanen Institute for Molecular Sciences, 70211 Kuopio, Finland.
| | | | | | - Stefan Selzer
- Proteome Sciences GmbH & Co. KG, 60438 Frankfurt, Germany.
| | - Tarja Kokkola
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, 70210 Kuopio, Finland.
| | - Leila Antikainen
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, 70210 Kuopio, Finland.
| | - Henna Martiskainen
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland.
| | - Susanna Kemppainen
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland.
| | - Mikael Marttinen
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; Structural and Computational Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany.
| | - Heli Jeskanen
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland.
| | - Hannah Rostalski
- A.I. Virtanen Institute for Molecular Sciences, 70211 Kuopio, Finland.
| | - Eija Rahunen
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland.
| | - Miia Kivipelto
- Population Health Unit, Finnish Institute for Health and Welfare, Helsinki, Finland; Division of Clinical Geriatrics, Department of Neurobiology, Center for Alzheimer Research, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; The Ageing Epidemiology Research Unit, School of Public Health, Imperial College London, London, United Kingdom; Theme Aging, Karolinska University Hospital, Stockholm, Sweden; Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland.
| | - Tiia Ngandu
- Population Health Unit, Finnish Institute for Health and Welfare, Helsinki, Finland; Division of Clinical Geriatrics, Department of Neurobiology, Center for Alzheimer Research, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Teemu Natunen
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland.
| | - Jean-Charles Lambert
- U1167, University of Lille, Inserm, Institut Pasteur de Lille, F-59000 Lille, France.
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States.
| | - Doo Yeon Kim
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States.
| | - Tuomas Rauramaa
- Department of Pathology, Kuopio University Hospital, 70211 Kuopio, Finland; Unit of Pathology, Institute of Clinical Medicine, University of Eastern Finland, 70210 Kuopio, Finland.
| | - Sanna-Kaisa Herukka
- Department of Neurology, University of Eastern Finland, 70210 Kuopio, Finland; NeuroCenter, Neurology, Kuopio University Hospital, Kuopio, Finland.
| | - Hilkka Soininen
- Department of Neurology, University of Eastern Finland, 70210 Kuopio, Finland.
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, 70210 Kuopio, Finland; Department of Medicine, Kuopio University Hospital, 70210 Kuopio, Finland.
| | - Ian Pike
- Proteome Sciences plc, Hamilton House, London, WC1H 9BB, UK.
| | - Ville Leinonen
- Department of Neurosurgery, Kuopio University Hospital, and Institute of Clinical Medicine, Unit of Neurosurgery, University of Eastern Finland, Kuopio, Finland.
| | | | - Mikko Hiltunen
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland.
| |
Collapse
|
31
|
Silva-Pedrosa R, Salgado AJ, Ferreira PE. Revolutionizing Disease Modeling: The Emergence of Organoids in Cellular Systems. Cells 2023; 12:930. [PMID: 36980271 PMCID: PMC10047824 DOI: 10.3390/cells12060930] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/03/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Cellular models have created opportunities to explore the characteristics of human diseases through well-established protocols, while avoiding the ethical restrictions associated with post-mortem studies and the costs associated with researching animal models. The capability of cell reprogramming, such as induced pluripotent stem cells (iPSCs) technology, solved the complications associated with human embryonic stem cells (hESC) usage. Moreover, iPSCs made significant contributions for human medicine, such as in diagnosis, therapeutic and regenerative medicine. The two-dimensional (2D) models allowed for monolayer cellular culture in vitro; however, they were surpassed by the three-dimensional (3D) cell culture system. The 3D cell culture provides higher cell-cell contact and a multi-layered cell culture, which more closely respects cellular morphology and polarity. It is more tightly able to resemble conditions in vivo and a closer approach to the architecture of human tissues, such as human organoids. Organoids are 3D cellular structures that mimic the architecture and function of native tissues. They are generated in vitro from stem cells or differentiated cells, such as epithelial or neural cells, and are used to study organ development, disease modeling, and drug discovery. Organoids have become a powerful tool for understanding the cellular and molecular mechanisms underlying human physiology, providing new insights into the pathogenesis of cancer, metabolic diseases, and brain disorders. Although organoid technology is up-and-coming, it also has some limitations that require improvements.
Collapse
Affiliation(s)
- Rita Silva-Pedrosa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; (A.J.S.); (P.E.F.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga, Portugal
- Centre of Biological Engineering (CEB), Department of Biological Engineering, University of Minho, 4710-057 Braga, Portugal
| | - António José Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; (A.J.S.); (P.E.F.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Pedro Eduardo Ferreira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; (A.J.S.); (P.E.F.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga, Portugal
| |
Collapse
|
32
|
Susceptibility of Ovine Bone Marrow-Derived Mesenchymal Stem Cell Spheroids to Scrapie Prion Infection. Animals (Basel) 2023; 13:ani13061043. [PMID: 36978584 PMCID: PMC10044354 DOI: 10.3390/ani13061043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/16/2023] [Accepted: 03/09/2023] [Indexed: 03/15/2023] Open
Abstract
In neurodegenerative diseases, including prion diseases, cellular in vitro models appear as fundamental tools for the study of pathogenic mechanisms and potential therapeutic compounds. Two-dimensional (2D) monolayer cell culture systems are the most used cell-based assays, but these platforms are not able to reproduce the microenvironment of in vivo cells. This limitation can be surpassed using three-dimensional (3D) culture systems such as spheroids that more effectively mimic in vivo cell interactions. Herein, we evaluated the effect of scrapie prion infection in monolayer-cultured ovine bone marrow-derived mesenchymal stem cells (oBM-MSCs) and oBM-MSC-derived spheroids in growth and neurogenic conditions, analyzing their cell viability and their ability to maintain prion infection. An MTT assay was performed in oBM-MSCs and spheroids subjected to three conditions: inoculated with brain homogenate from scrapie-infected sheep, inoculated with brain homogenate from healthy sheep, and non-inoculated controls. The 3D conditions improved the cell viability in most cases, although in scrapie-infected spheroids in growth conditions, a decrease in cell viability was observed. The levels of pathological prion protein (PrPSc) in scrapie-infected oBM-MSCs and spheroids were measured by ELISA. In neurogenic conditions, monolayer cells and spheroids maintained the levels of PrPSc over time. In growth conditions, however, oBM-MSCs showed decreasing levels of PrPSc throughout time, whereas spheroids were able to maintain stable PrPSc levels. The presence of PrPSc in spheroids was also confirmed by immunocytochemistry. Altogether, these results show that a 3D culture microenvironment improves the permissiveness of oBM-MSCs to scrapie infection in growth conditions and maintains the infection ability in neurogenic conditions, making this model of potential use for prion studies.
Collapse
|
33
|
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
|
34
|
Kang YJ, Xue Y, Shin JH, Cho H. Human mini-brains for reconstituting central nervous system disorders. LAB ON A CHIP 2023; 23:964-981. [PMID: 36644973 DOI: 10.1039/d2lc00897a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Neurological disorders in the central nervous system (CNS) are progressive and irreversible diseases leading to devastating impacts on patients' life as they cause cognitive impairment, dementia, and even loss of essential body functions. The development of effective medicines curing CNS disorders is, however, one of the most ambitious challenges due to the extremely complex functions and structures of the human brain. In this regard, there are unmet needs to develop simplified but physiopathologically-relevant brain models. Recent advances in the microfluidic techniques allow multicellular culture forming miniaturized 3D human brains by aligning parts of brain regions with specific cells serving suitable functions. In this review, we overview designs and strategies of microfluidics-based human mini-brains for reconstituting CNS disorders, particularly Alzheimer's disease (AD), Parkinson's disease (PD), traumatic brain injury (TBI), vascular dementia (VD), and environmental risk factor-driven dementia (ERFD). Afterward, the applications of the mini-brains in the area of medical science are introduced in terms of the clarification of pathogenic mechanisms and identification of promising biomarkers. We also present expanded model systems ranging from the CNS to CNS-connecting organ axes to study the entry pathways of pathological risk factors into the brain. Lastly, the advantages and potential challenges of current model systems are addressed with future perspectives.
Collapse
Affiliation(s)
- You Jung Kang
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea.
- Department of Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
| | - Yingqi Xue
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea.
- Department of Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jae Hee Shin
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea.
- Department of Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
| | - Hansang Cho
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea.
- Department of Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Republic of Korea
| |
Collapse
|
35
|
Park H, Kim J, Ryou C. A three-dimensional spheroid co-culture system of neurons and astrocytes derived from Alzheimer's disease patients for drug efficacy testing. Cell Prolif 2023:e13399. [PMID: 36628615 DOI: 10.1111/cpr.13399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/30/2022] [Accepted: 12/30/2022] [Indexed: 01/12/2023] Open
Abstract
Cell culture systems derived from the progenitor cells of human patients have many advantages over animal models for therapeutic drug testing and studies of disease pathogenesis. Here we describe a three-dimensional (3D) spheroid co-culture system of neurons and astrocytes derived from induced pluripotent stem cells-neural precursor cells (iPSCs-NPCs) of Alzheimer's disease (AD) patients or healthy individuals that can provide information on drug efficacy unobtainable by 2D co-culture or monoculture approaches. iPSCs-NPCs of healthy controls or AD patients were seeded onto 96-well U-bottom plates and incubated with neuronal differentiation medium for one week and with astrocytic medium for two weeks to replicate the temporal order of cell maturation during brain development. These 3D spheroid models expressed marker proteins for mature neurons and astrocytes. In particular, patient-derived spheroids showed beta-amyloid (Aβ) accumulation as revealed by thioflavin T (ThT) staining and ELISA. Aggregation of Aβ induced caspase activation and cell death, while the neuroprotectants nordihydroguaiaretic acid (NDGA) and curcumin (CU) reduced the levels of both ThT and caspase staining. Taken together, these results demonstrate the feasibility of our 3D spheroids combined with ThT and caspase staining as a patient-based anti-AD drug screening platform.
Collapse
Affiliation(s)
- HyunJung Park
- Department of Pharmacy, College of Pharmacy, and Institute of Pharmaceutical Science & Technology, Hanyang University, Ansan, Gyeonggi-do, Republic of Korea
| | - Jaehyeon Kim
- Department of Pharmacy, College of Pharmacy, and Institute of Pharmaceutical Science & Technology, Hanyang University, Ansan, Gyeonggi-do, Republic of Korea
| | - Chongsuk Ryou
- Department of Pharmacy, College of Pharmacy, and Institute of Pharmaceutical Science & Technology, Hanyang University, Ansan, Gyeonggi-do, Republic of Korea
| |
Collapse
|
36
|
In Vitro 3D Modeling of Neurodegenerative Diseases. BIOENGINEERING (BASEL, SWITZERLAND) 2023; 10:bioengineering10010093. [PMID: 36671665 PMCID: PMC9855033 DOI: 10.3390/bioengineering10010093] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/29/2022] [Accepted: 01/05/2023] [Indexed: 01/13/2023]
Abstract
The study of neurodegenerative diseases (such as Alzheimer's disease, Parkinson's disease, Huntington's disease, or amyotrophic lateral sclerosis) is very complex due to the difficulty in investigating the cellular dynamics within nervous tissue. Despite numerous advances in the in vivo study of these diseases, the use of in vitro analyses is proving to be a valuable tool to better understand the mechanisms implicated in these diseases. Although neural cells remain difficult to obtain from patient tissues, access to induced multipotent stem cell production now makes it possible to generate virtually all neural cells involved in these diseases (from neurons to glial cells). Many original 3D culture model approaches are currently being developed (using these different cell types together) to closely mimic degenerative nervous tissue environments. The aim of these approaches is to allow an interaction between glial cells and neurons, which reproduces pathophysiological reality by co-culturing them in structures that recapitulate embryonic development or facilitate axonal migration, local molecule exchange, and myelination (to name a few). This review details the advantages and disadvantages of techniques using scaffolds, spheroids, organoids, 3D bioprinting, microfluidic systems, and organ-on-a-chip strategies to model neurodegenerative diseases.
Collapse
|
37
|
Galiakberova AA, Brovkina OI, Kondratyev NV, Artyuhov AS, Momotyuk ED, Kulmukhametova ON, Lagunin AA, Shilov BV, Zadorozhny AD, Zakharov IS, Okorokova LS, Golimbet VE, Dashinimaev EB. Different iPSC-derived neural stem cells shows various spectrums of spontaneous differentiation during long term cultivation. Front Mol Neurosci 2023; 16:1037902. [PMID: 37201156 PMCID: PMC10186475 DOI: 10.3389/fnmol.2023.1037902] [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: 09/06/2022] [Accepted: 03/23/2023] [Indexed: 05/20/2023] Open
Abstract
Introduction Culturing of human neural stem cells (NSCs) derived from induced pluripotent stem cells (iPSC) is a promising area of research, as these cells have the potential to treat a wide range of neurological, neurodegenerative and psychiatric diseases. However, the development of optimal protocols for the production and long-term culturing of NSCs remains a challenge. One of the most important aspects of this problem is to determine the stability of NSCs during long-term in vitro passaging. To address this problem, our study was aimed at investigating the spontaneous differentiation profile in different iPSC-derived human NSCs cultures during long-term cultivation using. Methods Four different IPSC lines were used to generate NSC and spontaneously differentiated neural cultures using DUAL SMAD inhibition. These cells were analyzed at different passages using immunocytochemistry, qPCR, bulk transcriptomes and scRNA-seq. Results We found that various NSC lines generate significantly different spectrums of differentiated neural cells, which can also change significantly during long-term cultivation in vitro. Discussion Our results indicate that both internal (genetic and epigenetic) and external (conditions and duration of cultivation) factors influence the stability of NSCs. These results have important implications for the development of optimal NSCs culturing protocols and highlight the need to further investigate the factors influencing the stability of these cells in vitro.
Collapse
Affiliation(s)
- Adelya Albertovna Galiakberova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Olga Igorevna Brovkina
- Federal Research and Clinical Center, Federal Medical-Biological Agency of Russia, Moscow, Russia
| | | | - Alexander Sergeevich Artyuhov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Ekaterina Dmitrievna Momotyuk
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | | | - Alexey Aleksandrovich Lagunin
- Pirogov Russian National Research Medical University, Moscow, Russia
- Department of Bioinformatics, Institute of Biomedical Chemistry, Moscow, Russia
| | | | | | - Igor Sergeevitch Zakharov
- Department of Bioinformatics, Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia
| | | | | | - Erdem Bairovich Dashinimaev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Department of Bioinformatics, Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia
- Moscow Institute of Physics and Technology (State University), Dolgoprudny, Russia
- *Correspondence: Erdem Bairovich Dashinimaev,
| |
Collapse
|
38
|
Chang FC, Zhou Y, James MM, Zareie HM, Ando Y, Yang J, Zhang M. Effect of Degree of Deacetylation of Chitosan/Chitin on Human Neural Stem Cell Culture. Macromol Biosci 2023; 23:e2200389. [PMID: 36281904 DOI: 10.1002/mabi.202200389] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Indexed: 01/19/2023]
Abstract
Stem cell therapy and research for neural diseases depends on reliable reproduction of neural stem cells. Chitosan-based materials have been proposed as a substrate for culturing human neural stem cells (hNSCs) in the pursuit of clinically compatible culture conditions that are chemically defined and compliant with good manufacturing practices. The physical and biochemical properties of chitosan and chitin are strongly regulated by the degree of deacetylation (DD). However, the effect of DD on hNSC behavior has not been systematically investigated. In this study, films with DD ranging from 93% to 14% are fabricated with chitosan and chitin. Under xeno-free conditions, hNSCs proliferate preferentially on films with a higher DD, exhibiting adherent morphology and retaining multipotency. Lowering the DD leads to formation of neural stem cell spheroids due to unsteady adhesion. The neural spheroids present NSC multipotency protein expression reduction and cytoplasmic translocation. This study provides an insight into the influence of the DD on hNSCs behavior and may serve as a guideline for hNSC research using chitosan-based biomaterials. It demonstrates the capability of controlling hNSC fate by simply tailoring the DD of chitosan.
Collapse
Affiliation(s)
- Fei-Chien Chang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA, 98195, USA
| | - Yang Zhou
- Department of Materials Science and Engineering, University of Washington, Seattle, WA, 98195, USA
| | - Matthew Michael James
- Department of Materials Science and Engineering, University of Washington, Seattle, WA, 98195, USA
| | - Hadi M Zareie
- Department of Materials Science and Engineering, University of Washington, Seattle, WA, 98195, USA.,School of Mathematical and Physical Science, University of Technology, Ultimo, Sydney, NSW, 2007, Australia
| | - Yoshiki Ando
- Materials Department, Medical R&D Center, Corporate R&D Group, KYOCERA Corporation, Yasu, Shiga, 520-2362, Japan
| | - Jihui Yang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA, 98195, USA
| | - Miqin Zhang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA, 98195, USA
| |
Collapse
|
39
|
A Comprehensive Update of Cerebral Organoids between Applications and Challenges. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7264649. [DOI: 10.1155/2022/7264649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 10/30/2022] [Accepted: 11/17/2022] [Indexed: 12/12/2022]
Abstract
The basic technology of stem cells has been developed and created organoids, which have established a strong interest in regenerative medicine. Different cell types have been used to generate cerebral organoids, which include interneurons and oligodendrocytes (OLs). OLs are fundamental for brain development. Abundant studies have displayed that brain organoids can recapitulate fundamental and vital features of the human brain, such as cellular regulation and distribution, neuronal networks, electrical activities, and physiological structure. The organoids contain essential ventral brain domains and functional cortical interneurons, which are similar to the developing cortex and medial ganglionic eminence (MGE). So, brain organoids have provided a singular model to study and investigate neurological disorder mechanisms and therapeutics. Furthermore, the blood brain barrier (BBB) organoids modeling contributes to accelerate therapeutic discovery for the treatment of several neuropathologies. In this review, we summarized the advances of the brain organoids applications to investigate neurological disorder mechanisms such as neurodevelopmental and neurodegenerative disorders, mental disorders, brain cancer, and cerebral viral infections. We discussed brain organoids’ therapeutic application as a potential therapeutic unique method and highlighted in detail the challenges and hurdles of organoid models.
Collapse
|
40
|
Xue LL, Huangfu LR, Du RL, Chen L, Yu CY, Xiong LL, Wang TH. The age-specific pathological changes of β-amyloid plaques in the cortex and hippocampus of APP/PS1 transgenic AD mice. Neurol Res 2022; 44:1053-1065. [PMID: 35981107 DOI: 10.1080/01616412.2022.2112368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Numerous pathological variations and complex interactions are involved in the long period prior to cognitive decline in brains with Alzheimer's disease (AD). Thus, elucidation of the pathological disorders can facilitate early AD diagnosis. The aim of this study was to investigate the age-specific pathological changes of β-amyloid plaques in brain tissues of AD mice at different ages. METHODS We arranged the most widely available APP/PS1 transgenic AD models into six age groups: 3, 4 and 6 months (these three groups mimicked early-clinical stage AD), 9, 12 and 15 months (these three groups mimicked late-clinical stage AD). Cell morphology and arrangement in the cortex and hippocampus were observed by hematoxylin and eosin (HE) staining. Congo red staining and immunohistochemical staining were performed to exhibit the distribution of β-amyloid plaques in the cortex and hippocampus of AD brains. RESULTS Our results found that as age increased, the nuclei of cortical and hippocampal cells in AD mice were severely damaged. The number and area of β-amyloid plaques increased in AD mice in correspondence with age revealed by histological experiments. Importantly, β-amyloid plaques were detected in the cortex and hippocampus of 6-month-old AD mice shown by Congo red staining while detected in the cortex and hippocampus of 4-month-old AD mice shown by immunohistochemical staining. CONCLUSIONS The current study revealed the age-related pathological changes of β-amyloid plaques in the cortex and hippocampus of AD mice and displayed a higher specificity of immunohistochemical staining than Congo red staining when detecting pathological changes of brain tissues.
Collapse
Affiliation(s)
- Lu-Lu Xue
- State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Li-Ren Huangfu
- Animal Zoology Department, Institute of Neuroscience, Kunming medical University, Kunming, Yunnan, China
| | - Ruo-Lan Du
- Institute of Neurological Disease, Department of Anesthesiology, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Li Chen
- Institute of Neurological Disease, Department of Anesthesiology, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chang-Yin Yu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Liu-Lin Xiong
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Ting-Hua Wang
- State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China.,Animal Zoology Department, Institute of Neuroscience, Kunming medical University, Kunming, Yunnan, China.,Institute of Neurological Disease, Department of Anesthesiology, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
41
|
Alzheimer's Disease: Treatment Strategies and Their Limitations. Int J Mol Sci 2022; 23:ijms232213954. [PMID: 36430432 PMCID: PMC9697769 DOI: 10.3390/ijms232213954] [Citation(s) in RCA: 200] [Impact Index Per Article: 66.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/08/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer's disease (AD) is the most frequent case of neurodegenerative disease and is becoming a major public health problem all over the world. Many therapeutic strategies have been explored for several decades; however, there is still no curative treatment, and the priority remains prevention. In this review, we present an update on the clinical and physiological phase of the AD spectrum, modifiable and non-modifiable risk factors for AD treatment with a focus on prevention strategies, then research models used in AD, followed by a discussion of treatment limitations. The prevention methods can significantly slow AD evolution and are currently the best strategy possible before the advanced stages of the disease. Indeed, current drug treatments have only symptomatic effects, and disease-modifying treatments are not yet available. Drug delivery to the central nervous system remains a complex process and represents a challenge for developing therapeutic and preventive strategies. Studies are underway to test new techniques to facilitate the bioavailability of molecules to the brain. After a deep study of the literature, we find the use of soft nanoparticles, in particular nanoliposomes and exosomes, as an innovative approach for preventive and therapeutic strategies in reducing the risk of AD and solving problems of brain bioavailability. Studies show the promising role of nanoliposomes and exosomes as smart drug delivery systems able to penetrate the blood-brain barrier and target brain tissues. Finally, the different drug administration techniques for neurological disorders are discussed. One of the promising therapeutic methods is the intranasal administration strategy which should be used for preclinical and clinical studies of neurodegenerative diseases.
Collapse
|
42
|
Rickner HD, Jiang L, Hong R, O'Neill NK, Mojica CA, Snyder BJ, Zhang L, Shaw D, Medalla M, Wolozin B, Cheng CS. Single cell transcriptomic profiling of a neuron-astrocyte assembloid tauopathy model. Nat Commun 2022; 13:6275. [PMID: 36271092 PMCID: PMC9587045 DOI: 10.1038/s41467-022-34005-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 10/10/2022] [Indexed: 12/25/2022] Open
Abstract
The use of iPSC derived brain organoid models to study neurodegenerative disease has been hampered by a lack of systems that accurately and expeditiously recapitulate pathogenesis in the context of neuron-glial interactions. Here we report development of a system, termed AstTau, which propagates toxic human tau oligomers in iPSC derived neuron-astrocyte assembloids. The AstTau system develops much of the neuronal and astrocytic pathology observed in tauopathies including misfolded, phosphorylated, oligomeric, and fibrillar tau, strong neurodegeneration, and reactive astrogliosis. Single cell transcriptomic profiling combined with immunochemistry characterizes a model system that can more closely recapitulate late-stage changes in adult neurodegeneration. The transcriptomic studies demonstrate striking changes in neuroinflammatory and heat shock protein (HSP) chaperone systems in the disease process. Treatment with the HSP90 inhibitor PU-H71 is used to address the putative dysfunctional HSP chaperone system and produces a strong reduction of pathology and neurodegeneration, highlighting the potential of AstTau as a rapid and reproducible tool for drug discovery.
Collapse
Affiliation(s)
| | - Lulu Jiang
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Rui Hong
- Program in Bioinformatics, Boston University, Boston, MA, 02215, USA
| | | | - Chromewell A Mojica
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Benjamin J Snyder
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Lushuang Zhang
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Dipan Shaw
- Informatics Group, J. Craig Venter Institute, La Jolla, CA, 92037, USA
| | - Maria Medalla
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, 02118, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Benjamin Wolozin
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, 02118, USA.
- Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA.
- Center for Systems Neuroscience, Boston University, Boston, MA, 02118, USA.
| | - Christine S Cheng
- Department of Biology, Boston University, Boston, MA, 02215, USA.
- Program in Bioinformatics, Boston University, Boston, MA, 02215, USA.
- Informatics Group, J. Craig Venter Institute, La Jolla, CA, 92037, USA.
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
43
|
Shao X, Liu Z, Mao S, Han L. Unraveling the Mechanobiology Underlying Traumatic Brain Injury with Advanced Technologies and Biomaterials. Adv Healthc Mater 2022; 11:e2200760. [PMID: 35841392 DOI: 10.1002/adhm.202200760] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/27/2022] [Indexed: 01/27/2023]
Abstract
Traumatic brain injury (TBI) is a worldwide health and socioeconomic problem, associated with prolonged and complex neurological aftermaths, including a variety of functional deficits and neurodegenerative disorders. Research on the long-term effects has highlighted that TBI shall be regarded as a chronic health condition. The initiation and exacerbation of TBI involve a series of mechanical stimulations and perturbations, accompanied by mechanotransduction events within the brain tissues. Mechanobiology thus offers a unique perspective and likely promising approach to unravel the underlying molecular and biochemical mechanisms leading to neural cells dysfunction after TBI, which may contribute to the discovery of novel targets for future clinical treatment. This article investigates TBI and the subsequent brain dysfunction from a lens of neuromechanobiology. Following an introduction, the mechanobiological insights are examined into the molecular pathology of TBI, and then an overview is given of the latest research technologies to explore neuromechanobiology, with particular focus on microfluidics and biomaterials. Challenges and prospects in the current field are also discussed. Through this article, it is hoped that extensive technical innovation in biomedical devices and materials can be encouraged to advance the field of neuromechanobiology, paving potential ways for the research and rehabilitation of neurotrauma and neurological diseases.
Collapse
Affiliation(s)
- Xiaowei Shao
- Institute of Marine Science and Technology, Shandong University, Qingdao, Shandong, 266237, China.,Suzhou Research Institute, Shandong University, Suzhou, Jiangsu, 215123, China
| | - Zhongqian Liu
- Institute of Marine Science and Technology, Shandong University, Qingdao, Shandong, 266237, China
| | - Shijie Mao
- Institute of Marine Science and Technology, Shandong University, Qingdao, Shandong, 266237, China
| | - Lin Han
- Institute of Marine Science and Technology, Shandong University, Qingdao, Shandong, 266237, China
| |
Collapse
|
44
|
Mungenast L, Züger F, Selvi J, Faia-Torres AB, Rühe J, Suter-Dick L, Gullo MR. Directional Submicrofiber Hydrogel Composite Scaffolds Supporting Neuron Differentiation and Enabling Neurite Alignment. Int J Mol Sci 2022; 23:ijms231911525. [PMID: 36232822 PMCID: PMC9569964 DOI: 10.3390/ijms231911525] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/12/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
Cell cultures aiming at tissue regeneration benefit from scaffolds with physiologically relevant elastic moduli to optimally trigger cell attachment, proliferation and promote differentiation, guidance and tissue maturation. Complex scaffolds designed with guiding cues can mimic the anisotropic nature of neural tissues, such as spinal cord or brain, and recall the ability of human neural progenitor cells to differentiate and align. This work introduces a cost-efficient gelatin-based submicron patterned hydrogel–fiber composite with tuned stiffness, able to support cell attachment, differentiation and alignment of neurons derived from human progenitor cells. The enzymatically crosslinked gelatin-based hydrogels were generated with stiffnesses from 8 to 80 kPa, onto which poly(ε-caprolactone) (PCL) alignment cues were electrospun such that the fibers had a preferential alignment. The fiber–hydrogel composites with a modulus of about 20 kPa showed the strongest cell attachment and highest cell proliferation, rendering them an ideal differentiation support. Differentiated neurons aligned and bundled their neurites along the aligned PCL filaments, which is unique to this cell type on a fiber–hydrogel composite. This novel scaffold relies on robust and inexpensive technology and is suitable for neural tissue engineering where directional neuron alignment is required, such as in the spinal cord.
Collapse
Affiliation(s)
- Lena Mungenast
- Institute for Chemistry and Bioanalytics, University of Applied Sciences FHNW, Hofackerstrasse 30, 4132 Muttenz, Switzerland
- Correspondence: (L.M.); (M.R.G.)
| | - Fabian Züger
- Institute for Medical Engineering and Medical Informatics, University of Applied Sciences FHNW, Hofackerstrasse 30, 4132 Muttenz, Switzerland
| | - Jasmin Selvi
- Institute for Medical Engineering and Medical Informatics, University of Applied Sciences FHNW, Hofackerstrasse 30, 4132 Muttenz, Switzerland
| | - Ana Bela Faia-Torres
- Institute for Chemistry and Bioanalytics, University of Applied Sciences FHNW, Hofackerstrasse 30, 4132 Muttenz, Switzerland
| | - Jürgen Rühe
- Department of Microsystems Engineering, University of Freiburg–IMTEK, Georges-Koehler-Allee 103, 79110 Freiburg, Germany
| | - Laura Suter-Dick
- Institute for Chemistry and Bioanalytics, University of Applied Sciences FHNW, Hofackerstrasse 30, 4132 Muttenz, Switzerland
| | - Maurizio R. Gullo
- Institute for Medical Engineering and Medical Informatics, University of Applied Sciences FHNW, Hofackerstrasse 30, 4132 Muttenz, Switzerland
- Correspondence: (L.M.); (M.R.G.)
| |
Collapse
|
45
|
Kawakita S, Mandal K, Mou L, Mecwan MM, Zhu Y, Li S, Sharma S, Hernandez AL, Nguyen HT, Maity S, de Barros NR, Nakayama A, Bandaru P, Ahadian S, Kim HJ, Herculano RD, Holler E, Jucaud V, Dokmeci MR, Khademhosseini A. Organ-On-A-Chip Models of the Blood-Brain Barrier: Recent Advances and Future Prospects. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2201401. [PMID: 35978444 PMCID: PMC9529899 DOI: 10.1002/smll.202201401] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 06/22/2022] [Indexed: 05/09/2023]
Abstract
The human brain and central nervous system (CNS) present unique challenges in drug development for neurological diseases. One major obstacle is the blood-brain barrier (BBB), which hampers the effective delivery of therapeutic molecules into the brain while protecting it from blood-born neurotoxic substances and maintaining CNS homeostasis. For BBB research, traditional in vitro models rely upon Petri dishes or Transwell systems. However, these static models lack essential microenvironmental factors such as shear stress and proper cell-cell interactions. To this end, organ-on-a-chip (OoC) technology has emerged as a new in vitro modeling approach to better recapitulate the highly dynamic in vivo human brain microenvironment so-called the neural vascular unit (NVU). Such BBB-on-a-chip models have made substantial progress over the last decade, and concurrently there has been increasing interest in modeling various neurological diseases such as Alzheimer's disease and Parkinson's disease using OoC technology. In addition, with recent advances in other scientific technologies, several new opportunities to improve the BBB-on-a-chip platform via multidisciplinary approaches are available. In this review, an overview of the NVU and OoC technology is provided, recent progress and applications of BBB-on-a-chip for personalized medicine and drug discovery are discussed, and current challenges and future directions are delineated.
Collapse
Affiliation(s)
- Satoru Kawakita
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| | - Kalpana Mandal
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| | - Lei Mou
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
- Department of Clinical Laboratory, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, No. 63 Duobao Road, Liwan District, Guangzhou, Guangdong, 510150, P. R. China
| | | | - Yangzhi Zhu
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| | - Shaopei Li
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| | - Saurabh Sharma
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| | | | - Huu Tuan Nguyen
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| | - Surjendu Maity
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| | | | - Aya Nakayama
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| | - Praveen Bandaru
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| | - Samad Ahadian
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| | - Han-Jun Kim
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| | - Rondinelli Donizetti Herculano
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
- Department of Bioprocess and Biotechnology Engineering, School of Pharmaceutical Sciences, São Paulo State University (Unesp), Araraquara, SP, 14801-902, Brazil
| | - Eggehard Holler
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| | | | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| |
Collapse
|
46
|
Todd NK, Huang Y, Lee JY, Doruker P, Krieger JM, Salisbury R, MacDonald M, Bahar I, Thathiah A. GPCR kinases generate an APH1A phosphorylation barcode to regulate amyloid-β generation. Cell Rep 2022; 40:111110. [PMID: 35858570 PMCID: PMC9373432 DOI: 10.1016/j.celrep.2022.111110] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 05/05/2022] [Accepted: 06/28/2022] [Indexed: 11/03/2022] Open
Abstract
Emerging evidence suggests that G protein-coupled receptor (GPCR) kinases (GRKs) are associated with the pathophysiology of Alzheimer's disease (AD). However, GRKs have not been directly implicated in regulation of the amyloid-β (Aβ) pathogenic cascade in AD. Here, we determine that GRKs phosphorylate a non-canonical substrate, anterior pharynx-defective 1A (APH1A), an integral component of the γ-secretase complex. Significantly, we show that GRKs generate distinct phosphorylation barcodes in intracellular loop 2 (ICL2) and the C terminus of APH1A, which differentially regulate recruitment of the scaffolding protein β-arrestin 2 (βarr2) to APH1A and γ-secretase-mediated Aβ generation. Further molecular dynamics simulation studies reveal an interaction between the βarr2 finger loop domain and ICL2 and ICL3 of APH1A, similar to a GPCR-β-arrestin complex, which regulates γ-secretase activity. Collectively, these studies provide insight into the molecular and structural determinants of the APH1A-βarr2 interaction that critically regulate Aβ generation.
Collapse
Affiliation(s)
- Nicholas K Todd
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Graduate Program in Molecular Pharmacology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Yunhong Huang
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Ji Young Lee
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Pemra Doruker
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - James M Krieger
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Ryan Salisbury
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Matthew MacDonald
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Ivet Bahar
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Amantha Thathiah
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; University of Pittsburgh Brain Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| |
Collapse
|
47
|
Mehrabian M, Wang X, Eid S, Yan BQ, Grinberg M, Siegner M, Sackmann C, Sulman M, Zhao W, Williams D, Schmitt-Ulms G. Cardiac glycoside-mediated turnover of Na, K-ATPases as a rational approach to reducing cell surface levels of the cellular prion protein. PLoS One 2022; 17:e0270915. [PMID: 35776750 PMCID: PMC9249225 DOI: 10.1371/journal.pone.0270915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 06/17/2022] [Indexed: 01/16/2023] Open
Abstract
It is widely anticipated that a reduction of brain levels of the cellular prion protein (PrPC) can prolong survival in a group of neurodegenerative diseases known as prion diseases. To date, efforts to decrease steady-state PrPC levels by targeting this protein directly with small molecule drug-like compounds have largely been unsuccessful. Recently, we reported Na,K-ATPases to reside in immediate proximity to PrPC in the brain, unlocking an opportunity for an indirect PrPC targeting approach that capitalizes on the availability of potent cardiac glycosides (CGs). Here, we report that exposure of human co-cultures of neurons and astrocytes to non-toxic nanomolar levels of CGs causes profound reductions in PrPC levels. The mechanism of action underpinning this outcome relies primarily on a subset of CGs engaging the ATP1A1 isoform, one of three α subunits of Na,K-ATPases expressed in brain cells. Upon CG docking to ATP1A1, the ligand receptor complex, and PrPC along with it, is internalized by the cell. Subsequently, PrPC is channeled to the lysosomal compartment where it is digested in a manner that can be rescued by silencing the cysteine protease cathepsin B. These data signify that the repurposing of CGs may be beneficial for the treatment of prion disorders.
Collapse
Affiliation(s)
- Mohadeseh Mehrabian
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, Toronto, Ontario, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Xinzhu Wang
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, Toronto, Ontario, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Shehab Eid
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, Toronto, Ontario, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Bei Qi Yan
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, Toronto, Ontario, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Mark Grinberg
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, Toronto, Ontario, Canada
| | - Murdock Siegner
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, Toronto, Ontario, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Christopher Sackmann
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, Toronto, Ontario, Canada
| | - Muhammad Sulman
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, Toronto, Ontario, Canada
| | - Wenda Zhao
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, Toronto, Ontario, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Declan Williams
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, Toronto, Ontario, Canada
| | - Gerold Schmitt-Ulms
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, Toronto, Ontario, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
48
|
Roth A, Sander A, Oswald MS, Gärtner F, Knippschild U, Bischof J. Comprehensive Characterization of CK1δ-Mediated Tau Phosphorylation in Alzheimer’s Disease. Front Mol Biosci 2022; 9:872171. [PMID: 36203870 PMCID: PMC9531328 DOI: 10.3389/fmolb.2022.872171] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
A main pathological event in Alzheimer’s disease is the generation of neurofibrillary tangles originating from hyperphosphorylated and subsequently aggregated tau proteins. Previous reports demonstrated the critical involvement of members of the protein kinase family CK1 in the pathogenesis of Alzheimer’s disease by hyperphosphorylation of tau. However, precise mechanisms and effects of CK1-mediated tau phosphorylation are still not fully understood. In this study, we analyzed recombinant tau441 phosphorylated by CK1δ in vitro via mass spectrometry and identified ten potential phosphorylation sites, five of them are associated to Alzheimer’s disease. To confirm these results, in vitro kinase assays and two-dimensional phosphopeptide analyses were performed with tau441 phosphomutants confirming Alzheimer’s disease-associated residues Ser68/Thr71 and Ser289 as CK1δ-specific phosphorylation sites. Treatment of differentiated human neural progenitor cells with PF-670462 and Western blot analysis identified Ser214 as CK1δ-targeted phosphorylation site. The use of an in vitro tau aggregation assay demonstrated a possible role of CK1δ in tau aggregation. Results obtained in this study highlight the potential of CK1δ to be a promising target in the treatment of Alzheimer’s disease.
Collapse
|
49
|
Zhang SY, Zhao J, Ni JJ, Li H, Quan ZZ, Qing H. Application and prospects of high-throughput screening for in vitro neurogenesis. World J Stem Cells 2022; 14:393-419. [PMID: 35949394 PMCID: PMC9244953 DOI: 10.4252/wjsc.v14.i6.393] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 04/07/2022] [Accepted: 05/28/2022] [Indexed: 02/06/2023] Open
Abstract
Over the past few decades, high-throughput screening (HTS) has made great contributions to new drug discovery. HTS technology is equipped with higher throughput, minimized platforms, more automated and computerized operating systems, more efficient and sensitive detection devices, and rapid data processing systems. At the same time, in vitro neurogenesis is gradually becoming important in establishing models to investigate the mechanisms of neural disease or developmental processes. However, challenges remain in generating more mature and functional neurons with specific subtypes and in establishing robust and standardized three-dimensional (3D) in vitro models with neural cells cultured in 3D matrices or organoids representing specific brain regions. Here, we review the applications of HTS technologies on in vitro neurogenesis, especially aiming at identifying the essential genes, chemical small molecules and adaptive microenvironments that hold great prospects for generating functional neurons or more reproductive and homogeneous 3D organoids. We also discuss the developmental tendency of HTS technology, e.g., so-called next-generation screening, which utilizes 3D organoid-based screening combined with microfluidic devices to narrow the gap between in vitro models and in vivo situations both physiologically and pathologically.
Collapse
Affiliation(s)
- Shu-Yuan Zhang
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Juan Zhao
- Aerospace Medical Center, Aerospace Center Hospital, Beijing 100049, China
| | - Jun-Jun Ni
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Hui Li
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Zhen-Zhen Quan
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
50
|
Bose A, Petsko GA, Studer L. Induced pluripotent stem cells: a tool for modeling Parkinson's disease. Trends Neurosci 2022; 45:608-620. [PMID: 35667922 PMCID: PMC9576003 DOI: 10.1016/j.tins.2022.05.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 04/04/2022] [Accepted: 05/09/2022] [Indexed: 12/26/2022]
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disorder. Among its pathologies, progressive loss of dopaminergic (DA) neurons in the substantia nigra is characteristic and contributes to many of the most severe symptoms of PD. Recent advances in induced pluripotent stem cell (iPSC) technology have made it possible to generate patient-derived DA neuronal cell culture and organoid models of PD. These models have contributed to understanding disease mechanisms and the identification of novel targets and therapeutic candidates. Still needed are better ways to model the age-related aspects of PD, as well as a deeper understanding of the interactions among disease-modifying genes and between genetic and environmental contributions to the etiology and progression of PD.
Collapse
Affiliation(s)
- Anindita Bose
- Ann Romney Institute of Neurological Diseases, Harvard Medical School/Brigham and Women's Hospital, Boston, MA, USA; The Center for Stem Cell Biology, Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY, USA.
| | - Gregory A Petsko
- Ann Romney Institute of Neurological Diseases, Harvard Medical School/Brigham and Women's Hospital, Boston, MA, USA; The Center for Stem Cell Biology, Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY, USA
| | - Lorenz Studer
- The Center for Stem Cell Biology, Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY, USA
| |
Collapse
|