1
|
Lu SL, Pei Y, Liu WW, Han K, Cheng JCH, Li PC. Evaluating ECM stiffness and liver cancer radiation response via shear-wave elasticity in 3D culture models. Radiat Oncol 2024; 19:128. [PMID: 39334323 PMCID: PMC11430210 DOI: 10.1186/s13014-024-02513-7] [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: 12/16/2023] [Accepted: 08/26/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND The stiffness of the tumor microenvironment (TME) directly influences cellular behaviors. Radiotherapy (RT) is a common treatment for solid tumors, but the TME can impact its efficacy. In the case of liver cancer, clinical observations have shown that tumors within a cirrhotic, stiffer background respond less to RT, suggesting that the extracellular matrix (ECM) stiffness plays a critical role in the development of radioresistance. METHODS This study explored the effects of ECM stiffness and the inhibition of lysyl oxidase (LOX) isoenzymes on the radiation response of liver cancer in a millimeter-sized three-dimensional (3D) culture. We constructed a cube-shaped ECM-based millimeter-sized hydrogel containing Huh7 human liver cancer cells. By modulating the collagen concentration, we produced two groups of samples with different ECM stiffnesses to mimic the clinical scenarios of normal and cirrhotic livers. We used a single-transducer system for shear-wave-based elasticity measurement, to derive Young's modulus of the 3D cell culture to investigate how the ECM stiffness affects radiosensitivity. This is the first demonstration of a workflow for assessing radiation-induced response in a millimeter-sized 3D culture. RESULTS Increased ECM stiffness was associated with a decreased radiation response. Moreover, sonoporation-assisted LOX inhibition with BAPN (β-aminopropionitrile monofumarate) significantly decreased the initial ECM stiffness and increased RT-induced cell death. Inhibition of LOX was particularly effective in reducing ECM stiffness in stiffer matrices. Combining LOX inhibition with RT markedly increased radiation-induced DNA damage in cirrhotic liver cancer cells, enhancing their response to radiation. Furthermore, LOX inhibition can be combined with sonoporation to overcome stiffness-related radioresistance, potentially leading to better treatment outcomes for patients with liver cancer. CONCLUSIONS The findings underscore the significant influence of ECM stiffness on liver cancer's response to radiation. Sonoporation-aided LOX inhibition emerges as a promising strategy to mitigate stiffness-related resistance, offering potential improvements in liver cancer treatment outcomes.
Collapse
Affiliation(s)
- Shao-Lun Lu
- Department of Radiation Oncology, National Taiwan University Cancer Center, Taipei, Taiwan
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yu Pei
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | - Wei-Wen Liu
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
- Graduate of Institute of Oral Biology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Kun Han
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | - Jason Chia-Hsien Cheng
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
- Division of Radiation Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Pai-Chi Li
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan.
- Department of Electrical Engineering, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
2
|
Bergs J, Morr AS, Silva RV, Infante‐Duarte C, Sack I. The Networking Brain: How Extracellular Matrix, Cellular Networks, and Vasculature Shape the In Vivo Mechanical Properties of the Brain. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402338. [PMID: 38874205 PMCID: PMC11336943 DOI: 10.1002/advs.202402338] [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: 03/04/2024] [Revised: 05/22/2024] [Indexed: 06/15/2024]
Abstract
Mechanically, the brain is characterized by both solid and fluid properties. The resulting unique material behavior fosters proliferation, differentiation, and repair of cellular and vascular networks, and optimally protects them from damaging shear forces. Magnetic resonance elastography (MRE) is a noninvasive imaging technique that maps the mechanical properties of the brain in vivo. MRE studies have shown that abnormal processes such as neuronal degeneration, demyelination, inflammation, and vascular leakage lead to tissue softening. In contrast, neuronal proliferation, cellular network formation, and higher vascular pressure result in brain stiffening. In addition, brain viscosity has been reported to change with normal blood perfusion variability and brain maturation as well as disease conditions such as tumor invasion. In this article, the contributions of the neuronal, glial, extracellular, and vascular networks are discussed to the coarse-grained parameters determined by MRE. This reductionist multi-network model of brain mechanics helps to explain many MRE observations in terms of microanatomical changes and suggests that cerebral viscoelasticity is a suitable imaging marker for brain disease.
Collapse
Affiliation(s)
- Judith Bergs
- Department of RadiologyCharité – Universitätsmedizin BerlinCharitéplatz 110117BerlinGermany
| | - Anna S. Morr
- Department of RadiologyCharité – Universitätsmedizin BerlinCharitéplatz 110117BerlinGermany
| | - Rafaela V. Silva
- Experimental and Clinical Research Centera cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité – Universitätsmedizin BerlinLindenberger Weg 8013125BerlinGermany
- Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinECRC Experimental and Clinical Research CenterCharité – Universitätsmedizin BerlinCharitéplatz 110117BerlinGermany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC)Robert‐Rössle‐Straße 1013125BerlinGermany
| | - Carmen Infante‐Duarte
- Experimental and Clinical Research Centera cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité – Universitätsmedizin BerlinLindenberger Weg 8013125BerlinGermany
- Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinECRC Experimental and Clinical Research CenterCharité – Universitätsmedizin BerlinCharitéplatz 110117BerlinGermany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC)Robert‐Rössle‐Straße 1013125BerlinGermany
| | - Ingolf Sack
- Department of RadiologyCharité – Universitätsmedizin BerlinCharitéplatz 110117BerlinGermany
| |
Collapse
|
3
|
Francis KL, Zheng HB, Suskind DL, Murphree TA, Phan BA, Quah E, Hendrickson AS, Zhou X, Nuding M, Hudson AS, Guttman M, Morton GJ, Schwartz MW, Alonge KM, Scarlett JM. Characterizing the human intestinal chondroitin sulfate glycosaminoglycan sulfation signature in inflammatory bowel disease. Sci Rep 2024; 14:11839. [PMID: 38782973 PMCID: PMC11116513 DOI: 10.1038/s41598-024-60959-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: 12/21/2023] [Accepted: 04/29/2024] [Indexed: 05/25/2024] Open
Abstract
The intestinal extracellular matrix (ECM) helps maintain appropriate tissue barrier function and regulate host-microbial interactions. Chondroitin sulfate- and dermatan sulfate-glycosaminoglycans (CS/DS-GAGs) are integral components of the intestinal ECM, and alterations in CS/DS-GAGs have been shown to significantly influence biological functions. Although pathologic ECM remodeling is implicated in inflammatory bowel disease (IBD), it is unknown whether changes in the intestinal CS/DS-GAG composition are also linked to IBD in humans. Our aim was to characterize changes in the intestinal ECM CS/DS-GAG composition in intestinal biopsy samples from patients with IBD using mass spectrometry. We characterized intestinal CS/DS-GAGs in 69 pediatric and young adult patients (n = 13 control, n = 32 active IBD, n = 24 IBD in remission) and 6 adult patients. Here, we report that patients with active IBD exhibit a significant decrease in the relative abundance of CS/DS isomers associated with matrix stability (CS-A and DS) compared to controls, while isomers implicated in matrix instability and inflammation (CS-C and CS-E) were significantly increased. This imbalance of intestinal CS/DS isomers was restored among patients in clinical remission. Moreover, the abundance of pro-stabilizing CS/DS isomers negatively correlated with clinical disease activity scores, whereas both pro-inflammatory CS-C and CS-E content positively correlated with disease activity scores. Thus, pediatric patients with active IBD exhibited increased pro-inflammatory and decreased pro-stabilizing CS/DS isomer composition, and future studies are needed to determine whether changes in the CS/DS-GAG composition play a pathogenic role in IBD.
Collapse
Affiliation(s)
- Kendra L Francis
- Department of Pediatric Gastroenterology and Hepatology, Seattle Children's Hospital, Seattle, WA, USA
- Department of Medicine, University of Washington Medicine Diabetes Institute, 750 Republican St, Box 358062, Seattle, WA, 98195, USA
| | - Hengqi B Zheng
- Department of Pediatric Gastroenterology and Hepatology, Seattle Children's Hospital, Seattle, WA, USA
| | - David L Suskind
- Department of Pediatric Gastroenterology and Hepatology, Seattle Children's Hospital, Seattle, WA, USA
| | - Taylor A Murphree
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| | - Bao Anh Phan
- Department of Medicine, University of Washington Medicine Diabetes Institute, 750 Republican St, Box 358062, Seattle, WA, 98195, USA
| | - Emily Quah
- Department of Medicine, University of Washington Medicine Diabetes Institute, 750 Republican St, Box 358062, Seattle, WA, 98195, USA
| | - Aarun S Hendrickson
- Department of Medicine, University of Washington Medicine Diabetes Institute, 750 Republican St, Box 358062, Seattle, WA, 98195, USA
| | - Xisheng Zhou
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| | - Mason Nuding
- Department of Pediatric Gastroenterology and Hepatology, Seattle Children's Hospital, Seattle, WA, USA
| | - Alexandra S Hudson
- Department of Pediatric Gastroenterology and Hepatology, Seattle Children's Hospital, Seattle, WA, USA
| | - Miklos Guttman
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| | - Gregory J Morton
- Department of Medicine, University of Washington Medicine Diabetes Institute, 750 Republican St, Box 358062, Seattle, WA, 98195, USA
| | - Michael W Schwartz
- Department of Medicine, University of Washington Medicine Diabetes Institute, 750 Republican St, Box 358062, Seattle, WA, 98195, USA
| | - Kimberly M Alonge
- Department of Medicine, University of Washington Medicine Diabetes Institute, 750 Republican St, Box 358062, Seattle, WA, 98195, USA
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| | - Jarrad M Scarlett
- Department of Pediatric Gastroenterology and Hepatology, Seattle Children's Hospital, Seattle, WA, USA.
- Department of Medicine, University of Washington Medicine Diabetes Institute, 750 Republican St, Box 358062, Seattle, WA, 98195, USA.
| |
Collapse
|
4
|
Hendrickson AS, Francis KL, Kumar A, Le JP, Scarlett JM, Keene CD, Tovar DA, Alonge KM. Assessing translational applicability of perineuronal net dysfunction in Alzheimer's disease across species. Front Neurosci 2024; 18:1396101. [PMID: 38745932 PMCID: PMC11091253 DOI: 10.3389/fnins.2024.1396101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 04/12/2024] [Indexed: 05/16/2024] Open
Abstract
In the context of aging and age-associated neurodegenerative disorders, the brain's extracellular matrix (ECM) serves as a critical regulator for neuronal health and cognitive function. Within the extracellular space, proteoglycans and their glycosaminoglycan attachments play essential roles in forming, stabilizing, and protecting neural circuits throughout neurodevelopment and adulthood. Recent studies in rodents reveal that chondroitin sulfate-glycosaminoglycan (CS-GAG) containing perineuronal nets (PNNs) exhibit both structural and compositional differences throughout the brain. While animal studies are illuminating, additional research is required to translate these interregional PNN/CS-GAG variations to human brain tissue. In this perspective article, we first investigate the translational potential for interregional CS-GAG variances across species as novel targets for region-specific therapeutic development. We specifically focus on the observation that alterations in brain PNN-associated CS-GAGs have been linked with the progression of Alzheimer's disease (AD) neuropathology in humans, but these changes have not been fully recapitulated in rodent models of this disease. A second highlight of this perspective article investigates whether AD-associated shifts in CS-GAGs in humans may be dependent on region-specific baseline differences in CS-GAG sulfation patterning. The current findings begin to disentangle the intricate relationships between the interregional differences in brain PNN/CS-GAG matrices across species, while emphasizing the need to better understand the close relationship between dementia and changes in brain CS-GAG sulfation patterns in patients with AD and related dementias.
Collapse
Affiliation(s)
- Aarun S. Hendrickson
- Medicinal Chemistry, University of Washington, Seattle, WA, United States
- Department of Medicine, University of Washington Medicine Diabetes Institute, Seattle, WA, United States
| | - Kendra L. Francis
- Department of Medicine, University of Washington Medicine Diabetes Institute, Seattle, WA, United States
- Department of Pediatric Gastroenterology and Hepatology, Seattle Children’s Hospital, Seattle, WA, United States
| | - Asmit Kumar
- Medicinal Chemistry, University of Washington, Seattle, WA, United States
- Department of Medicine, University of Washington Medicine Diabetes Institute, Seattle, WA, United States
| | - Jaden P. Le
- Medicinal Chemistry, University of Washington, Seattle, WA, United States
- Department of Medicine, University of Washington Medicine Diabetes Institute, Seattle, WA, United States
| | - Jarrad M. Scarlett
- Department of Medicine, University of Washington Medicine Diabetes Institute, Seattle, WA, United States
- Department of Pediatric Gastroenterology and Hepatology, Seattle Children’s Hospital, Seattle, WA, United States
| | - C. Dirk Keene
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - David A. Tovar
- Department of Psychology, Vanderbilt University, Nashville, TN, United States
| | - Kimberly M. Alonge
- Medicinal Chemistry, University of Washington, Seattle, WA, United States
- Department of Medicine, University of Washington Medicine Diabetes Institute, Seattle, WA, United States
| |
Collapse
|
5
|
Kobayashi T. Protein homeostasis and degradation in quiescent neural stem cells. J Biochem 2024; 175:481-486. [PMID: 38299708 DOI: 10.1093/jb/mvae006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/27/2023] [Accepted: 01/15/2024] [Indexed: 02/02/2024] Open
Abstract
Tissue stem cells are maintained in the adult body throughout life and are crucial for tissue homeostasis as they supply newly functional cells. Quiescence is a reversible arrest in the G0/G1 phase of the cell cycle and a strategy to maintain the quality of tissue stem cells. Quiescence maintains stem cells in a self-renewable and differentiable state for a prolonged period by suppressing energy consumption and cell damage and depletion. Most adult neural stem cells in the brain maintain the quiescent state and produce neurons and glial cells through differentiation after activating from the quiescent state to the proliferating state. In this process, proteostasis, including proteolysis, is essential to transition between the quiescent and proliferating states associated with proteome remodeling. Recent reports have demonstrated that quiescent and proliferating neural stem cells have different expression patterns and roles as proteostatic molecules and are affected by age, indicating differing processes for protein homeostasis in these two states in the brain. This review discusses the multiple regulatory stages from protein synthesis (protein birth) to proteolysis (protein death) in quiescent neural stem cells.
Collapse
Affiliation(s)
- Taeko Kobayashi
- Department of Basic Medical Sciences, The Institute of Medical Science, The University of Tokyo, 108-8639, Japan
| |
Collapse
|
6
|
Pillai EK, Franze K. Mechanics in the nervous system: From development to disease. Neuron 2024; 112:342-361. [PMID: 37967561 DOI: 10.1016/j.neuron.2023.10.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/29/2023] [Accepted: 10/04/2023] [Indexed: 11/17/2023]
Abstract
Physical forces are ubiquitous in biological processes across scales and diverse contexts. This review highlights the significance of mechanical forces in nervous system development, homeostasis, and disease. We provide an overview of mechanical signals present in the nervous system and delve into mechanotransduction mechanisms translating these mechanical cues into biochemical signals. During development, mechanical cues regulate a plethora of processes, including cell proliferation, differentiation, migration, network formation, and cortex folding. Forces then continue exerting their influence on physiological processes, such as neuronal activity, glial cell function, and the interplay between these different cell types. Notably, changes in tissue mechanics manifest in neurodegenerative diseases and brain tumors, potentially offering new diagnostic and therapeutic target opportunities. Understanding the role of cellular forces and tissue mechanics in nervous system physiology and pathology adds a new facet to neurobiology, shedding new light on many processes that remain incompletely understood.
Collapse
Affiliation(s)
- Eva K Pillai
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK; Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117 Heidelberg, Germany; Developmental Biology Unit, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117 Heidelberg, Germany.
| | - Kristian Franze
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK; Institute of Medical Physics and Microtissue Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Henkestraße 91, 91052 Erlangen, Germany; Max-Planck-Zentrum für Physik und Medizin, Kussmaulallee 1, 91054 Erlangen, Germany.
| |
Collapse
|
7
|
Salmina AB, Alexandrova OP, Averchuk AS, Korsakova SA, Saridis MR, Illarioshkin SN, Yurchenko SO. Current progress and challenges in the development of brain tissue models: How to grow up the changeable brain in vitro? J Tissue Eng 2024; 15:20417314241235527. [PMID: 38516227 PMCID: PMC10956167 DOI: 10.1177/20417314241235527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 02/12/2024] [Indexed: 03/23/2024] Open
Abstract
In vitro modeling of brain tissue is a promising but not yet resolved problem in modern neurobiology and neuropharmacology. Complexity of the brain structure and diversity of cell-to-cell communication in (patho)physiological conditions make this task almost unachievable. However, establishment of novel in vitro brain models would ultimately lead to better understanding of development-associated or experience-driven brain plasticity, designing efficient approaches to restore aberrant brain functioning. The main goal of this review is to summarize the available data on methodological approaches that are currently in use, and to identify the most prospective trends in development of neurovascular unit, blood-brain barrier, blood-cerebrospinal fluid barrier, and neurogenic niche in vitro models. The manuscript focuses on the regulation of adult neurogenesis, cerebral microcirculation and fluids dynamics that should be reproduced in the in vitro 4D models to mimic brain development and its alterations in brain pathology. We discuss approaches that are critical for studying brain plasticity, deciphering the individual person-specific trajectory of brain development and aging, and testing new drug candidates in the in vitro models.
Collapse
Affiliation(s)
- Alla B Salmina
- Brain Science Institute, Research Center of Neurology, Moscow, Russia
- Bauman Moscow State Technical University, Moscow, Russia
| | - Olga P Alexandrova
- Brain Science Institute, Research Center of Neurology, Moscow, Russia
- Bauman Moscow State Technical University, Moscow, Russia
| | - Anton S Averchuk
- Brain Science Institute, Research Center of Neurology, Moscow, Russia
- Bauman Moscow State Technical University, Moscow, Russia
| | | | | | | | | |
Collapse
|
8
|
Bonucci M, Shu T, Holt LJ. How it feels in a cell. Trends Cell Biol 2023; 33:924-938. [PMID: 37286396 PMCID: PMC10592589 DOI: 10.1016/j.tcb.2023.05.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 05/01/2023] [Accepted: 05/02/2023] [Indexed: 06/09/2023]
Abstract
Life emerges from thousands of biochemical processes occurring within a shared intracellular environment. We have gained deep insights from in vitro reconstitution of isolated biochemical reactions. However, the reaction medium in test tubes is typically simple and diluted. The cell interior is far more complex: macromolecules occupy more than a third of the space, and energy-consuming processes agitate the cell interior. Here, we review how this crowded, active environment impacts the motion and assembly of macromolecules, with an emphasis on mesoscale particles (10-1000 nm diameter). We describe methods to probe and analyze the biophysical properties of cells and highlight how changes in these properties can impact physiology and signaling, and potentially contribute to aging, and diseases, including cancer and neurodegeneration.
Collapse
Affiliation(s)
- Martina Bonucci
- Institute for Systems Genetics, New York University Langone Medical Center, 435 E 30th Street, New York, NY 10016, USA
| | - Tong Shu
- Institute for Systems Genetics, New York University Langone Medical Center, 435 E 30th Street, New York, NY 10016, USA
| | - Liam J Holt
- Institute for Systems Genetics, New York University Langone Medical Center, 435 E 30th Street, New York, NY 10016, USA.
| |
Collapse
|
9
|
Li H, Ghorbani S, Ling CC, Yong VW, Xue M. The extracellular matrix as modifier of neuroinflammation and recovery in ischemic stroke and intracerebral hemorrhage. Neurobiol Dis 2023; 186:106282. [PMID: 37683956 DOI: 10.1016/j.nbd.2023.106282] [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: 05/28/2023] [Revised: 09/01/2023] [Accepted: 09/05/2023] [Indexed: 09/10/2023] Open
Abstract
Stroke is the second leading cause of death worldwide and has two major subtypes: ischemic stroke and hemorrhagic stroke. Neuroinflammation is a pathological hallmark of ischemic stroke and intracerebral hemorrhage (ICH), contributing to the extent of brain injury but also in its repair. Neuroinflammation is intricately linked to the extracellular matrix (ECM), which is profoundly altered after brain injury and in aging. In the early stages after ischemic stroke and ICH, immune cells are involved in the deposition and remodeling of the ECM thereby affecting processes such as blood-brain barrier and cellular integrity. ECM components regulate leukocyte infiltration into the central nervous system, activate a variety of immune cells, and induce the elevation of matrix metalloproteinases (MMPs) after stroke. In turn, excessive MMPs may degrade ECM into components that are pro-inflammatory and injurious. Conversely, in the later stages after stroke, several ECM molecules may contribute to tissue recovery. For example, thrombospondin-1 and biglycan may promote activity of regulatory T cells, inhibit the synthesis of proinflammatory cytokines, and aid regenerative processes. We highlight these roles of the ECM in ischemic stroke and ICH and discuss their potential cellular and molecular mechanisms. Finally, we discuss therapeutics that could be considered to normalize the ECM in stroke. Our goal is to spur research on the ECM in order to improve the prognosis of ischemic stroke and ICH.
Collapse
Affiliation(s)
- Hongmin Li
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, China; Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Alberta, Canada
| | - Samira Ghorbani
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Alberta, Canada
| | - Chang-Chun Ling
- Department of Chemistry, University of Calgary, Alberta, Canada
| | - V Wee Yong
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Alberta, Canada.
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
10
|
Zheng Q, Liu H, Yu W, Dong Y, Zhou L, Deng W, Hua F. Mechanical properties of the brain: Focus on the essential role of Piezo1-mediated mechanotransduction in the CNS. Brain Behav 2023; 13:e3136. [PMID: 37366640 PMCID: PMC10498085 DOI: 10.1002/brb3.3136] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 04/24/2023] [Accepted: 06/13/2023] [Indexed: 06/28/2023] Open
Abstract
BACKGROUND The brain is a highly mechanosensitive organ, and changes in the mechanical properties of brain tissue influence many physiological and pathological processes. Piezo type mechanosensitive ion channel component 1 (Piezo1), a protein found in metazoans, is highly expressed in the brain and involved in sensing changes of the mechanical microenvironment. Numerous studies have shown that Piezo1-mediated mechanotransduction is closely related to glial cell activation and neuronal function. However, the precise role of Piezo1 in the brain requires further elucidation. OBJECTIVE This review first discusses the roles of Piezo1-mediated mechanotransduction in regulating the functions of a variety of brain cells, and then briefly assesses the impact of Piezo1-mediated mechanotransduction on the progression of brain dysfunctional disorders. CONCLUSIONS Mechanical signaling contributes significantly to brain function. Piezo1-mediated mechanotransduction regulates processes such as neuronal differentiation, cell migration, axon guidance, neural regeneration, and oligodendrocyte axon myelination. Additionally, Piezo1-mediated mechanotransduction plays significant roles in normal aging and brain injury, as well as the development of various brain diseases, including demyelinating diseases, Alzheimer's disease, and brain tumors. Investigating the pathophysiological mechanisms through which Piezo1-mediated mechanotransduction affects brain function will give us a novel entry point for the diagnosis and treatment of numerous brain diseases.
Collapse
Affiliation(s)
- Qingcui Zheng
- Department of Anesthesiologythe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
- Key Laboratory of Anesthesiology of Jiangxi ProvinceThe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
- Jiangxi Province Key Laboratory of Molecular MedicineThe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
| | - Hailin Liu
- Department of Anesthesiologythe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
- Key Laboratory of Anesthesiology of Jiangxi ProvinceThe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
- Jiangxi Province Key Laboratory of Molecular MedicineThe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
| | - Wen Yu
- Department of Anesthesiologythe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
- Key Laboratory of Anesthesiology of Jiangxi ProvinceThe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
| | - Yao Dong
- Department of Anesthesiologythe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
- Key Laboratory of Anesthesiology of Jiangxi ProvinceThe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
- Jiangxi Province Key Laboratory of Molecular MedicineThe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
| | - Lanqian Zhou
- Department of Anesthesiologythe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
- Key Laboratory of Anesthesiology of Jiangxi ProvinceThe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
- Jiangxi Province Key Laboratory of Molecular MedicineThe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
| | - Wenze Deng
- Department of Anesthesiologythe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
- Key Laboratory of Anesthesiology of Jiangxi ProvinceThe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
| | - Fuzhou Hua
- Department of Anesthesiologythe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
- Key Laboratory of Anesthesiology of Jiangxi ProvinceThe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
| |
Collapse
|
11
|
Hall CM, Lasli S, Serwinski B, Djordjevic B, Sheridan GK, Moeendarbary E. Hippocampus of the APP NL-G-F mouse model of Alzheimer's disease exhibits region-specific tissue softening concomitant with elevated astrogliosis. Front Aging Neurosci 2023; 15:1212212. [PMID: 37547743 PMCID: PMC10398960 DOI: 10.3389/fnagi.2023.1212212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 07/03/2023] [Indexed: 08/08/2023] Open
Abstract
Widespread neurodegeneration, enlargement of cerebral ventricles, and atrophy of cortical and hippocampal brain structures are classic hallmarks of Alzheimer's disease (AD). Prominent macroscopic disturbances to the cytoarchitecture of the AD brain occur alongside changes in the mechanical properties of brain tissue, as reported in recent magnetic resonance elastography (MRE) measurements of human brain mechanics. Whilst MRE has many advantages, a significant shortcoming is its spatial resolution. Higher resolution "cellular scale" assessment of the mechanical alterations to brain regions involved in memory formation, such as the hippocampus, could provide fresh new insight into the etiology of AD. Characterization of brain tissue mechanics at the cellular length scale is the first stepping-stone to understanding how mechanosensitive neurons and glia are impacted by neurodegenerative disease-associated changes in their microenvironment. To provide insight into the microscale mechanics of aging brain tissue, we measured spatiotemporal changes in the mechanical properties of the hippocampus using high resolution atomic force microscopy (AFM) indentation tests on acute brain slices from young and aged wild-type mice and the APPNL-G-F mouse model. Several hippocampal regions in APPNL-G-F mice are significantly softer than age-matched wild-types, notably the dentate granule cell layer and the CA1 pyramidal cell layer. Interestingly, regional softening coincides with an increase in astrocyte reactivity, suggesting that amyloid pathology-mediated alterations to the mechanical properties of brain tissue may impact the function of mechanosensitive astrocytes. Our data also raise questions as to whether aberrant mechanotransduction signaling could impact the susceptibility of neurons to cellular stressors in their microenvironment.
Collapse
Affiliation(s)
- Chloe M. Hall
- Department of Mechanical Engineering, University College London, London, United Kingdom
- School of Applied Sciences, University of Brighton, Brighton, United Kingdom
| | - Soufian Lasli
- Department of Mechanical Engineering, University College London, London, United Kingdom
| | - Bianca Serwinski
- Department of Mechanical Engineering, University College London, London, United Kingdom
- 199 Biotechnologies Ltd., London, United Kingdom
- Faculty of Social Sciences, Northeastern University London, London, United Kingdom
| | - Boris Djordjevic
- Department of Mechanical Engineering, University College London, London, United Kingdom
- 199 Biotechnologies Ltd., London, United Kingdom
| | - Graham K. Sheridan
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Emad Moeendarbary
- Department of Mechanical Engineering, University College London, London, United Kingdom
| |
Collapse
|
12
|
Yan L, Dwiggins CW, Moriarty RA, Jung JW, Gupta U, Brandon KD, Stroka KM. Matrix stiffness regulates the tight junction phenotypes and local barrier properties in tricellular regions in an iPSC-derived BBB model. Acta Biomater 2023:S1742-7061(23)00327-6. [PMID: 37302732 DOI: 10.1016/j.actbio.2023.06.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 06/02/2023] [Accepted: 06/06/2023] [Indexed: 06/13/2023]
Abstract
The blood-brain barrier (BBB) can respond to various mechanical cues such as shear stress and substrate stiffness. In the human brain, the compromised barrier function of the BBB is closely associated with a series of neurological disorders that are often also accompanied by the alteration of brain stiffness. In many types of peripheral vasculature, higher matrix stiffness decreases barrier function of endothelial cells through mechanotransduction pathways that alter cell-cell junction integrity. However, human brain endothelial cells are specialized endothelial cells that largely resist changes in cell morphology and key BBB markers. Therefore, it has remained an open question how matrix stiffness affects barrier integrity in the human BBB. To gain insight into the effects of matrix stiffness on BBB permeability, we differentiated brain microvascular endothelial-like cells from human induced pluripotent stem cells (iBMEC-like cells) and cultured the cells on extracellular matrix-coated hydrogels of varying stiffness. We first detected and quantified the junction presentation of key tight junction (TJ) proteins. Our results show matrix-dependent junction phenotypes in iBMEC-like cells, where cells on softer gels (1 kPa) have significantly lower continuous and total TJ coverages. We also determined that these softer gels also lead to decreased barrier function in a local permeability assay. Furthermore, we found that matrix stiffness regulates the local permeability of iBMEC-like cells through the balance of continuous ZO-1 TJs and no junction regions ZO-1 in tricellular regions. Together, these findings provide valuable insights into the effects of matrix stiffness on TJ phenotypes and local permeability of iBMEC-like cells. STATEMENT OF SIGNIFICANCE: Brain mechanical properties, including stiffness, are particularly sensitive indicators for pathophysiological changes in neural tissue. The compromised function of the blood-brain barrier is closely associated with a series of neurological disorders often accompanied by altered brain stiffness. In this study, we use polymeric biomaterials and provide new evidence that biomaterial stiffness regulates the local permeability in iPSC-derived brain endothelial cells in tricellular regions through the tight junction protein ZO-1. Our findings provide valuable insights into the changes in junction architecture and barrier permeability in response to different substrate stiffnesses. Since BBB dysfunction has been linked to many diseases, understanding the influence of substrate stiffness on junction presentations and barrier permeability could lead to the development of new treatments for diseases associated with BBB dysfunction or drug delivery across BBB systems.
Collapse
Affiliation(s)
- Li Yan
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA.
| | - Cole W Dwiggins
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Rebecca A Moriarty
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Jae W Jung
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Udit Gupta
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Ken D Brandon
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Kimberly M Stroka
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; Biophysics Program, University of Maryland, College Park, MD 20742, USA; Center for Stem Cell Biology and Regenerative Medicine, University of Maryland, Baltimore, MD 21201, USA; Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD 21201, USA.
| |
Collapse
|
13
|
Bryniarska-Kubiak N, Kubiak A, Trojan E, Wesołowska J, Lekka M, Basta-Kaim A. Oxygen-Glucose Deprivation in Organotypic Hippocampal Cultures Leads to Cytoskeleton Rearrangement and Immune Activation: Link to the Potential Pathomechanism of Ischaemic Stroke. Cells 2023; 12:1465. [PMID: 37296586 PMCID: PMC10252361 DOI: 10.3390/cells12111465] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/18/2023] [Accepted: 05/19/2023] [Indexed: 06/12/2023] Open
Abstract
Ischaemic stroke is characterized by a sudden loss of blood circulation to an area of the brain, resulting in a corresponding loss of neurologic function. As a result of this process, neurons in the ischaemic core are deprived of oxygen and trophic substances and are consequently destroyed. Tissue damage in brain ischaemia results from a complex pathophysiological cascade comprising various distinct pathological events. Ischaemia leads to brain damage by stimulating many processes, such as excitotoxicity, oxidative stress, inflammation, acidotoxicity, and apoptosis. Nevertheless, less attention has been given to biophysical factors, including the organization of the cytoskeleton and the mechanical properties of cells. Therefore, in the present study, we sought to evaluate whether the oxygen-glucose deprivation (OGD) procedure, which is a commonly accepted experimental model of ischaemia, could affect cytoskeleton organization and the paracrine immune response. The abovementioned aspects were examined ex vivo in organotypic hippocampal cultures (OHCs) subjected to the OGD procedure. We measured cell death/viability, nitric oxide (NO) release, and hypoxia-inducible factor 1α (HIF-1α) levels. Next, the impact of the OGD procedure on cytoskeletal organization was evaluated using combined confocal fluorescence microscopy (CFM) and atomic force microscopy (AFM). Concurrently, to find whether there is a correlation between biophysical properties and the immune response, we examined the impact of OGD on the levels of crucial ischaemia cytokines (IL-1β, IL-6, IL-18, TNF-α, IL-10, IL-4) and chemokines (CCL3, CCL5, CXCL10) in OHCs and calculated Pearsons' and Spearman's rank correlation coefficients. The results of the current study demonstrated that the OGD procedure intensified cell death and nitric oxide release and led to the potentiation of HIF-1α release in OHCs. Moreover, we presented significant disturbances in the organization of the cytoskeleton (actin fibers, microtubular network) and cytoskeleton-associated protein 2 (MAP-2), which is a neuronal marker. Simultaneously, our study provided new evidence that the OGD procedure leads to the stiffening of OHCs and a malfunction in immune homeostasis. A negative linear correlation between tissue stiffness and branched IBA1 positive cells after the OGD procedure suggests the pro-inflammatory polarization of microglia. Moreover, the negative correlation of pro- and positive anti-inflammatory factors with actin fibers density indicates an opposing effect of the immune mediators on the rearrangement of cytoskeleton induced by OGD procedure in OHCs. Our study constitutes a basis for further research and provides a rationale for integrating biomechanical and biochemical methods in studying the pathomechanism of stroke-related brain damage. Furthermore, presented data pointed out the interesting direction of proof-of-concept studies, in which follow-up may establish new targets for brain ischemia therapy.
Collapse
Affiliation(s)
- Natalia Bryniarska-Kubiak
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland
| | - Andrzej Kubiak
- Department of Biophysical Microstructures, Institute of Nuclear Physics, Polish Academy of Sciences, 152 Radzikowskiego St., 31-342 Kraków, Poland
- Laboratory of Stem Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 7 Gronostajowa St., 30-387 Kraków, Poland
| | - Ewa Trojan
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland
| | - Julita Wesołowska
- Laboratory for In Vivo and In Vitro Imaging, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland
| | - Małgorzata Lekka
- Department of Biophysical Microstructures, Institute of Nuclear Physics, Polish Academy of Sciences, 152 Radzikowskiego St., 31-342 Kraków, Poland
| | - Agnieszka Basta-Kaim
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland
| |
Collapse
|
14
|
Akcay G, Luttge R. Microenvironments Matter: Advances in Brain-on-Chip. BIOSENSORS 2023; 13:551. [PMID: 37232912 PMCID: PMC10216565 DOI: 10.3390/bios13050551] [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: 03/23/2023] [Revised: 05/04/2023] [Accepted: 05/12/2023] [Indexed: 05/27/2023]
Abstract
To highlight the particular needs with respect to modeling the unique and complex organization of the human brain structure, we reviewed the state-of-the-art in devising brain models with engineered instructive microenvironments. To acquire a better perspective on the brain's working mechanisms, we first summarize the importance of regional stiffness gradients in brain tissue, varying per layer and the cellular diversities of the layers. Through this, one can acquire an understanding of the essential parameters in emulating the brain in vitro. In addition to the brain's organizational architecture, we addressed also how the mechanical properties have an impact on neuronal cell responses. In this respect, advanced in vitro platforms emerged and profoundly changed the methods of brain modeling efforts from the past, mainly focusing on animal or cell line research. The main challenges in imitating features of the brain in a dish are with regard to composition and functionality. In neurobiological research, there are now methods that aim to cope with such challenges by the self-assembly of human-derived pluripotent stem cells (hPSCs), i.e., brainoids. Alternatively, these brainoids can be used stand-alone or in conjunction with Brain-on-Chip (BoC) platform technology, 3D-printed gels, and other types of engineered guidance features. Currently, advanced in vitro methods have made a giant leap forward regarding cost-effectiveness, ease-of-use, and availability. We bring these recent developments together into one review. We believe our conclusions will give a novel perspective towards advancing instructive microenvironments for BoCs and the understanding of the brain's cellular functions either in modeling healthy or diseased states of the brain.
Collapse
Affiliation(s)
- Gulden Akcay
- Neuro-Nanoscale Engineering, Department of Mechanical Engineering/Microsystems, Institute of Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands;
| | - Regina Luttge
- Neuro-Nanoscale Engineering, Department of Mechanical Engineering/Microsystems, Institute of Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands;
- Eindhoven Artificial Intelligence Systems Institute, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
- Eindhoven Hendrik Casimir Institute, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| |
Collapse
|
15
|
Schaefer SD, Davies BM, Newcombe VF, Sutcliffe MP. Could spinal cord oscillation contribute to spinal cord injury in degenerative cervical myelopathy? BRAIN & SPINE 2023; 3:101743. [PMID: 37383476 PMCID: PMC10293319 DOI: 10.1016/j.bas.2023.101743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/20/2023] [Accepted: 04/12/2023] [Indexed: 06/30/2023]
Abstract
Introduction Degenerative Cervical Myelopathy [DCM] is a slow-motion spinal cord injury. Compression and dynamic compression have been considered disease hallmarks. However, this is likely an oversimplification, as compression is more commonly incidental and has only modest correlation to disease severity. MRI studies have recently suggested spinal cord oscillation could play a role. Research question To determine if spinal cord oscillation could contribute to spinal cord injury in degenerative cervical myelopathy. Material and methods A computational model of an oscillating spinal cord was developed from imaging of a healthy volunteer. Using finite element analysis, the observed implications of stress and strain, were measured in the context of a simulated disc herniation. The significance was bench marked by comparison to a more recognised dynamic injury mechanism; a flexion extension model of dynamic compression. Results Spinal cord oscillation altered both compressive and shear strain on the spinal cord. Following initial compression, compressive strain moves from within the spinal cord to the spinal cord surface, whilst shear strain is magnified by 0.1-0.2, depending on the amplitude of oscillation. These orders of magnitude are equivalent to a dynamic compression model. Discussion and conclusion Spinal cord oscillation could significantly contribute to spinal cord damage across DCM. Its repeated occurrence with every heartbeat, draws parallels to the concept of fatigue damage, which could reconcile differing theories on the origins of DCM. This remains hypothetical at this stage, and further investigations are required.
Collapse
|
16
|
Donnaloja F, Limonta E, Mancosu C, Morandi F, Boeri L, Albani D, Raimondi MT. Unravelling the mechanotransduction pathways in Alzheimer's disease. J Biol Eng 2023; 17:22. [PMID: 36978103 PMCID: PMC10045049 DOI: 10.1186/s13036-023-00336-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 03/02/2023] [Indexed: 03/30/2023] Open
Abstract
Alzheimer's disease (AD) represents one of the most common and debilitating neurodegenerative disorders. By the end of 2040, AD patients might reach 11.2 million in the USA, around 70% higher than 2022, with severe consequences on the society. As now, we still need research to find effective methods to treat AD. Most studies focused on the tau and amyloid hypothesis, but many other factors are likely involved in the pathophysiology of AD. In this review, we summarize scientific evidence dealing with the mechanotransduction players in AD to highlight the most relevant mechano-responsive elements that play a role in AD pathophysiology. We focused on the AD-related role of extracellular matrix (ECM), nuclear lamina, nuclear transport and synaptic activity. The literature supports that ECM alteration causes the lamin A increment in the AD patients, leading to the formation of nuclear blebs and invaginations. Nuclear blebs have consequences on the nuclear pore complexes, impairing nucleo-cytoplasmic transport. This may result in tau hyperphosphorylation and its consequent self-aggregation in tangles, which impairs the neurotransmitters transport. It all exacerbates in synaptic transmission impairment, leading to the characteristic AD patient's memory loss. Here we related for the first time all the evidence associating the mechanotransduction pathway with neurons. In addition, we highlighted the entire pathway influencing neurodegenerative diseases, paving the way for new research perspectives in the context of AD and related pathologies.
Collapse
Affiliation(s)
- Francesca Donnaloja
- Politecnico Di Milano, Department of Chemistry, Materials and Chemical Engineering "G. Natta", Campus Leonardo, Piazza Leonardo da Vinci 32, 20133, Milan, Italy.
| | - Emma Limonta
- Politecnico Di Milano, Department of Chemistry, Materials and Chemical Engineering "G. Natta", Campus Leonardo, Piazza Leonardo da Vinci 32, 20133, Milan, Italy
| | - Christian Mancosu
- Politecnico Di Milano, Department of Chemistry, Materials and Chemical Engineering "G. Natta", Campus Leonardo, Piazza Leonardo da Vinci 32, 20133, Milan, Italy
| | - Francesco Morandi
- Politecnico Di Milano, Department of Chemistry, Materials and Chemical Engineering "G. Natta", Campus Leonardo, Piazza Leonardo da Vinci 32, 20133, Milan, Italy
| | - Lucia Boeri
- Politecnico Di Milano, Department of Chemistry, Materials and Chemical Engineering "G. Natta", Campus Leonardo, Piazza Leonardo da Vinci 32, 20133, Milan, Italy
| | - Diego Albani
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Manuela Teresa Raimondi
- Politecnico Di Milano, Department of Chemistry, Materials and Chemical Engineering "G. Natta", Campus Leonardo, Piazza Leonardo da Vinci 32, 20133, Milan, Italy.
| |
Collapse
|
17
|
Bertalan G, Becker J, Tzschätzsch H, Morr A, Herthum H, Shahryari M, Greenhalgh RD, Guo J, Schröder L, Alzheimer C, Budday S, Franze K, Braun J, Sack I. Mechanical behavior of the hippocampus and corpus callosum: An attempt to reconcile ex vivo with in vivo and micro with macro properties. J Mech Behav Biomed Mater 2023; 138:105613. [PMID: 36549250 DOI: 10.1016/j.jmbbm.2022.105613] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 11/24/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022]
Abstract
Mechanical properties of brain tissue are very complex and vary with the species, region, method, and dynamic range, and between in vivo and ex vivo measurements. To reconcile this variability, we investigated in vivo and ex vivo stiffness properties of two distinct regions in the human and mouse brain - the hippocampus (HP) and the corpus callosum (CC) - using different methods. Under quasi-static conditions, we examined ex vivo murine HP and CC by atomic force microscopy (AFM). Between 16 and 40Hz, we investigated the in vivo brains of healthy volunteers by magnetic resonance elastography (MRE) in a 3-T clinical scanner. At high-frequency stimulation between 1000 and 1400Hz, we investigated the murine HP and CC ex vivo and in vivo with MRE in a 7-T preclinical system. HP and CC showed pronounced stiffness dispersion, as reflected by a factor of 32-36 increase in shear modulus from AFM to low-frequency human MRE and a 25-fold higher shear wave velocity in murine MRE than in human MRE. At low frequencies, HP was softer than CC, in both ex vivo mouse specimens (p < 0.05) and in vivo human brains (p < 0.01) while, at high frequencies, CC was softer than HP under in vivo (p < 0.01) and ex vivo (p < 0.05) conditions. The standard linear solid model comprising three elements reproduced the observed HP and CC stiffness dispersions, while other two- and three-element models failed. Our results indicate a remarkable consistency of brain stiffness across species, ex vivo and in vivo states, and different measurement techniques when marked viscoelastic dispersion properties combining equilibrium and non-equilibrium mechanical elements are considered.
Collapse
Affiliation(s)
- Gergerly Bertalan
- Department of Radiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Julia Becker
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Heiko Tzschätzsch
- Department of Radiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Anna Morr
- Department of Radiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Helge Herthum
- Department of Radiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Mehrgan Shahryari
- Department of Radiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ryan D Greenhalgh
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Jing Guo
- Department of Radiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Leif Schröder
- Translational Molecular Imaging, Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | - Christian Alzheimer
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Silvia Budday
- Institute of Applied Mechanics, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Kristian Franze
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom; Institute of Medical Physics, Friedrich-Alexander-Universität, Erlangen-Nürnberg, Erlangen, Germany; Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Jürgen Braun
- Institute of Medical Informatics, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ingolf Sack
- Department of Radiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
| |
Collapse
|
18
|
Yazdani N, Willits RK. Mimicking the neural stem cell niche: An engineer’s view of cell: material interactions. FRONTIERS IN CHEMICAL ENGINEERING 2023. [DOI: 10.3389/fceng.2022.1086099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Neural stem cells have attracted attention in recent years to treat neurodegeneration. There are two neurogenic regions in the brain where neural stem cells reside, one of which is called the subventricular zone (SVZ). The SVZ niche is a complicated microenvironment providing cues to regulate self-renewal and differentiation while maintaining the neural stem cell’s pool. Many scientists have spent years understanding the cellular and structural characteristics of the SVZ niche, both in homeostasis and pathological conditions. On the other hand, engineers focus primarily on designing platforms using the knowledge they acquire to understand the effect of individual factors on neural stem cell fate decisions. This review provides a general overview of what we know about the components of the SVZ niche, including the residing cells, extracellular matrix (ECM), growth factors, their interactions, and SVZ niche changes during aging and neurodegenerative diseases. Furthermore, an overview will be given on the biomaterials used to mimic neurogenic niche microenvironments and the design considerations applied to add bioactivity while meeting the structural requirements. Finally, it will discuss the potential gaps in mimicking the microenvironment.
Collapse
|
19
|
Baek J, Kumar S, Schaffer DV, Im SG. N-Cadherin adhesive ligation regulates mechanosensitive neural stem cell lineage commitment in 3D matrices. Biomater Sci 2022; 10:6768-6777. [PMID: 36314115 PMCID: PMC10195187 DOI: 10.1039/d2bm01349e] [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] [Indexed: 11/21/2022]
Abstract
During differentiation, neural stem cells (NSCs) encounter diverse cues from their niche, including not only biophysical cues from the extracellular matrix (ECM) but also cell-cell communication. However, it is still poorly understood how these cues cumulatively regulate mechanosensitive NSC fate commitment, especially in 3D matrices that better mimic in vivo systems. Here, we develop a click chemistry-based 3D hydrogel material system to fully decouple cell-cell and cell-ECM interactions by functionalizing small peptides: the HAVDI motif from N-cadherin and RGD motif from fibronectin. The hydrogel is engineered to range in stiffness from 75 Pa to 600 Pa. Interestingly, HAVDI-mediated interaction shows increased neurogenesis, except for the softest gel (75 Pa). Moreover, the HAVDI ligation attenuates the mechanosensing state of NSCs, exhibiting restricted cytoskeletal formation and RhoA signaling. Given that mechanosensitive neurogenesis has been reported to be regulated by cytoskeletal formation, our finding suggests that the enhanced neurogenesis in the HAVDI-modified gel may be highly associated with the HAVDI interaction-mediated attenuation of mechanosensing. Furthermore, NSCs in the HAVDI gel shows higher β-catenin activity, which has been known to promote neurogenesis. Our findings provide critical insights into how mechanosensitive NSC fate commitment is regulated as a consequence of diverse interactions in 3D microenvironments.
Collapse
Affiliation(s)
- Jieung Baek
- Dept. of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Dept. of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Sanjay Kumar
- Dept. of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Dept. of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Dept. of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - David V Schaffer
- Dept. of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Dept. of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Dept. of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Sung Gap Im
- Dept. of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea.
| |
Collapse
|
20
|
Morr AS, Nowicki M, Bertalan G, Vieira Silva R, Infante Duarte C, Koch SP, Boehm-Sturm P, Krügel U, Braun J, Steiner B, Käs JA, Fuhs T, Sack I. Mechanical properties of murine hippocampal subregions investigated by atomic force microscopy and in vivo magnetic resonance elastography. Sci Rep 2022; 12:16723. [PMID: 36202964 PMCID: PMC9537158 DOI: 10.1038/s41598-022-21105-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 09/22/2022] [Indexed: 12/02/2022] Open
Abstract
The hippocampus is a very heterogeneous brain structure with different mechanical properties reflecting its functional variety. In particular, adult neurogenesis in rodent hippocampus has been associated with specific viscoelastic properties in vivo and ex vivo. Here, we study the microscopic mechanical properties of hippocampal subregions using ex vivo atomic force microscopy (AFM) in correlation with the expression of GFP in presence of the nestin promoter, providing a marker of neurogenic activity. We further use magnetic resonance elastography (MRE) to investigate whether in vivo mechanical properties reveal similar spatial patterns, however, on a much coarser scale. AFM showed that tissue stiffness increases with increasing distance from the subgranular zone (p = 0.0069), and that stiffness is 39% lower in GFP than non-GFP regions (p = 0.0004). Consistently, MRE showed that dentate gyrus is, on average, softer than Ammon´s horn (shear wave speed = 3.2 ± 0.2 m/s versus 4.4 ± 0.3 m/s, p = 0.01) with another 3.4% decrease towards the subgranular zone (p = 0.0001). The marked reduction in stiffness measured by AFM in areas of high neurogenic activity is consistent with softer MRE values, indicating the sensitivity of macroscopic mechanical properties in vivo to micromechanical structures as formed by the neurogenic niche of the hippocampus.
Collapse
Affiliation(s)
- Anna S Morr
- Department of Radiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Marcin Nowicki
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Gergely Bertalan
- Department of Radiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Rafaela Vieira Silva
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC) and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Einstein Center for Neurosciences Berlin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Carmen Infante Duarte
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC) and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Stefan Paul Koch
- Department of Experimental Neurology and Center for Stroke Research, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Philipp Boehm-Sturm
- Department of Experimental Neurology and Center for Stroke Research, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ute Krügel
- Rudolf Boehm Institute of Pharmacology and Toxicology, University of Leipzig, Leipzig, Germany
| | - Jürgen Braun
- Institute of Medical Informatics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Barbara Steiner
- Clinic for Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Josef A Käs
- Section of Soft Matter Physics, Peter Debye Institute for Soft Matter Physics, Faculty of Physics and Geosciences, University of Leipzig, Leipzig, Germany
| | - Thomas Fuhs
- Section of Soft Matter Physics, Peter Debye Institute for Soft Matter Physics, Faculty of Physics and Geosciences, University of Leipzig, Leipzig, Germany
| | - Ingolf Sack
- Department of Radiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
21
|
Kwak D, Olsen PA, Danielsen A, Jensenius AR. A trio of biological rhythms and their relevance in rhythmic mechanical stimulation of cell cultures. Front Psychol 2022; 13:867191. [PMID: 35967633 PMCID: PMC9374063 DOI: 10.3389/fpsyg.2022.867191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 06/29/2022] [Indexed: 11/13/2022] Open
Abstract
The primary aim of this article is to provide a biological rhythm model based on previous theoretical and experimental findings to promote more comprehensive studies of rhythmic mechanical stimulation of cell cultures, which relates to tissue engineering and regenerative medicine fields. Through an interdisciplinary approach where different standpoints from biology and musicology are combined, we explore some of the core rhythmic features of biological and cellular rhythmic processes and present them as a trio model that aims to afford a basic but fundamental understanding of the connections between various biological rhythms. It is vital to highlight such links since rhythmic mechanical stimulation and its effect on cell cultures are vastly underexplored even though the cellular response to mechanical stimuli (mechanotransduction) has been studied widely and relevant experimental evidence suggests mechanotransduction processes are rhythmic.
Collapse
Affiliation(s)
- Dongho Kwak
- Department of Musicology, RITMO Centre for Interdisciplinary Studies in Rhythm, Time and Motion, University of Oslo, Oslo, Norway
| | - Petter Angell Olsen
- Hybrid Technology Hub-Centre for Organ on a Chip-Technology, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Unit for Cell Signaling, Department of Immunology and Transfusion Medicine, Oslo University Hospital, Oslo, Norway
| | - Anne Danielsen
- Department of Musicology, RITMO Centre for Interdisciplinary Studies in Rhythm, Time and Motion, University of Oslo, Oslo, Norway
| | - Alexander Refsum Jensenius
- Department of Musicology, RITMO Centre for Interdisciplinary Studies in Rhythm, Time and Motion, University of Oslo, Oslo, Norway
| |
Collapse
|
22
|
Maeda S, Yamada J, Iinuma KM, Nadanaka S, Kitagawa H, Jinno S. Chondroitin sulfate proteoglycan is a potential target of memantine to improve cognitive function via the promotion of adult neurogenesis. Br J Pharmacol 2022; 179:4857-4877. [PMID: 35797426 DOI: 10.1111/bph.15920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 06/02/2022] [Accepted: 06/28/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Chondroitin sulfate proteoglycan (CSPG) constitutes the neurogenic niche in the hippocampus. The reduction of hippocampal neurogenesis is involved in aging-related cognitive decline and dementia. The purpose of this study is to find candidates that improve cognitive function by analyzing the effects of memantine (MEM), a therapeutic agent for Alzheimer's disease, on CSPG and adult hippocampal neurogenesis. EXPERIMENTAL APPROACH The effects of MEM on neurogenesis-related cells and CSPG content were assessed in the hippocampus of middle-aged mice. The MEM-induced alterations in gene expressions of neurotrophins and enzymes associated with biosynthesis and degradation of CSPG in the hippocampus were also measured. The effects of MEM on cognitive function were estimated using a behavioral test battery. The same set of behavioral tests was applied to evaluate the effects of pharmacological depletion of CSPG in the hippocampus. KEY RESULTS The densities of newborn granule cells and content of CSPG in the hippocampus were increased by MEM. The expression levels of the enzyme responsible for the biosynthesis CSPG were increased by MEM. The neurotrophin-related molecules were activated by MEM. Short- and long-term memory performance was improved by MEM. Pharmacological depletion of CSPG impairs the effects of MEM on cognitive improvement in middle-aged mice. CONCLUSIONS AND IMPLICATIONS MEM regulates the biosynthesis and degradation of CSPG, which may underlie the improvement of cognitive function via the promotion of adult hippocampal neurogenesis. These results imply that CSPG-related enzymes may be potentially attractive candidates for the treatment of aging-related cognitive decline.
Collapse
Affiliation(s)
- Shoichiro Maeda
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Jun Yamada
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kyoko M Iinuma
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Satomi Nadanaka
- Department of Biochemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Hiroshi Kitagawa
- Department of Biochemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Shozo Jinno
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
23
|
Liu H, Hu J, Zheng Q, Feng X, Zhan F, Wang X, Xu G, Hua F. Piezo1 Channels as Force Sensors in Mechanical Force-Related Chronic Inflammation. Front Immunol 2022; 13:816149. [PMID: 35154133 PMCID: PMC8826255 DOI: 10.3389/fimmu.2022.816149] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/03/2022] [Indexed: 12/14/2022] Open
Abstract
Mechanical damage is one of the predisposing factors of inflammation, and it runs through the entire inflammatory pathological process. Repeated or persistent damaging mechanical irritation leads to chronic inflammatory diseases. The mechanism of how mechanical forces induce inflammation is not fully understood. Piezo1 is a newly discovered mechanically sensitive ion channel. The Piezo1 channel opens in response to mechanical stimuli, transducing mechanical signals into an inflammatory cascade in the cell leading to tissue inflammation. A large amount of evidence shows that Piezo1 plays a vital role in the occurrence and progression of chronic inflammatory diseases. This mini-review briefly presents new evidence that Piezo1 responds to different mechanical stresses to trigger inflammation in various tissues. The discovery of Piezo1 provides new insights for the treatment of chronic inflammatory diseases related to mechanical stress. Inhibiting the transduction of damaging mechanical signals into inflammatory signals can inhibit inflammation and improve the outcome of inflammation at an early stage. The pharmacology of Piezo1 has shown bright prospects. The development of tissue-specific Piezo1 drugs for clinical use may be a new target for treating chronic inflammation.
Collapse
Affiliation(s)
- Hailin Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jialing Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Qingcui Zheng
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaojin Feng
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Fenfang Zhan
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xifeng Wang
- Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Guohai Xu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Fuzhou Hua
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
24
|
Kofman S, Mohan N, Sun X, Ibric L, Piermarini E, Qiang L. Human mini brains and spinal cords in a dish: Modeling strategies, current challenges, and prospective advances. J Tissue Eng 2022; 13:20417314221113391. [PMID: 35898331 PMCID: PMC9310295 DOI: 10.1177/20417314221113391] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/27/2022] [Indexed: 11/15/2022] Open
Abstract
Engineered three-dimensional (3D) in vitro and ex vivo neural tissues, also known as "mini brains and spinal cords in a dish," can be derived from different types of human stem cells via several differentiation protocols. In general, human mini brains are micro-scale physiological systems consisting of mixed populations of neural progenitor cells, glial cells, and neurons that may represent key features of human brain anatomy and function. To date, these specialized 3D tissue structures can be characterized into spheroids, organoids, assembloids, organ-on-a-chip and their various combinations based on generation procedures and cellular components. These 3D CNS models incorporate complex cell-cell interactions and play an essential role in bridging the gap between two-dimensional human neuroglial cultures and animal models. Indeed, they provide an innovative platform for disease modeling and therapeutic cell replacement, especially shedding light on the potential to realize personalized medicine for neurological disorders when combined with the revolutionary human induced pluripotent stem cell technology. In this review, we highlight human 3D CNS models developed from a variety of experimental strategies, emphasize their advances and remaining challenges, evaluate their state-of-the-art applications in recapitulating crucial phenotypic aspects of many CNS diseases, and discuss the role of contemporary technologies in the prospective improvement of their composition, consistency, complexity, and maturation.
Collapse
Affiliation(s)
- Simeon Kofman
- Department of Neurobiology and Anatomy, Drexel
University College of Medicine, Philadelphia, PA, USA
| | - Neha Mohan
- Department of Neurobiology and Anatomy, Drexel
University College of Medicine, Philadelphia, PA, USA
| | - Xiaohuan Sun
- Department of Neurobiology and Anatomy, Drexel
University College of Medicine, Philadelphia, PA, USA
| | - Larisa Ibric
- Department of Neurobiology and Anatomy, Drexel
University College of Medicine, Philadelphia, PA, USA
| | - Emanuela Piermarini
- Department of Neurobiology and Anatomy, Drexel
University College of Medicine, Philadelphia, PA, USA
| | - Liang Qiang
- Department of Neurobiology and Anatomy, Drexel
University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|