1
|
Zhou Z, Olsson C, Gasser TC, Li X, Kleiven S. The White Matter Fiber Tract Deforms Most in the Perpendicular Direction During In Vivo Volunteer Impacts. J Neurotrauma 2024. [PMID: 39212616 DOI: 10.1089/neu.2024.0183] [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: 09/04/2024] Open
Abstract
White matter (WM) tract-related strains are increasingly used to quantify brain mechanical responses, but their dynamics in live human brains during in vivo impact conditions remain largely unknown. Existing research primarily looked into the normal strain along the WM fiber tracts (i.e., tract-oriented normal strain), but it is rarely the case that the fiber tract only endures tract-oriented normal strain during impacts. In this study, we aim to extend the in vivo measurement of WM fiber deformation by quantifying the normal strain perpendicular to the fiber tract (i.e., tract-perpendicular normal strain) and the shear strain along and perpendicular to the fiber tract (i.e., tract-oriented shear strain and tract-perpendicular shear strain, respectively). To achieve this, we combine the three-dimensional strain tensor from the tagged magnetic resonance imaging with the diffusion tensor imaging (DTI) from an open-access dataset, including 44 volunteer impacts under two head loading modes, i.e., neck rotations (N = 30) and neck extensions (N = 14). The strain tensor is rotated to the coordinate system with one axis aligned with DTI-revealed fiber orientation, and then four tract-related strain measures are calculated. The results show that tract-perpendicular normal strain peaks are the largest among the four strain types (p < 0.05, Friedman's test). The distribution of tract-related strains is affected by the head loading mode, of which laterally symmetric patterns with respect to the midsagittal plane are noted under neck extensions, but not under neck rotations. Our study presents a comprehensive in vivo strain quantification toward a multifaceted understanding of WM dynamics. We find that the WM fiber tract deforms most in the perpendicular direction, illuminating new fundamentals of brain mechanics. The reported strain images can be used to evaluate the fidelity of computational head models, especially those intended to predict fiber deformation under noninjurious conditions.
Collapse
Affiliation(s)
- Zhou Zhou
- Division of Neuronic Engineering, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Christoffer Olsson
- Division of Biomedical Imaging, KTH Royal Institute of Technology, Stockholm, Sweden
| | - T Christian Gasser
- Material and Structural Mechanics, Department of Engineering Mechanics, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Xiaogai Li
- Division of Neuronic Engineering, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Svein Kleiven
- Division of Neuronic Engineering, KTH Royal Institute of Technology, Stockholm, Sweden
| |
Collapse
|
2
|
Rubby MF, Fonder C, Uchayash S, Liang X, Sakaguchi DS, Que L. Assessment of the Behaviors of an In Vitro Brain Model On-Chip under Shockwave Impacts. ACS APPLIED MATERIALS & INTERFACES 2024; 16:33246-33258. [PMID: 38905518 DOI: 10.1021/acsami.4c08026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/23/2024]
Abstract
Herein we report the assessment of the effects of shockwave (SW) impacts on adult rat hippocampal progenitor cell (AHPC) neurospheres (NSs), which are used as in vitro brain models, for enhancing our understanding of the mechanisms of traumatic brain injury (TBI). The assessment has been achieved by using culture dishes and a new microchip. The microchip allows the chemicals released from the brain models cultured inside the cell culture chamber under SW impacts to diffuse to the nanosensors in adjacent sensor chambers through built-in diffusion barriers, which are used to prevent the cells from entering the sensor chambers, thereby mitigating the biofouling issues of the sensor surface. Experiments showed the negative impact of the SW on the viability, proliferation, and differentiation of the cells within the NSs. A qPCR gene expression analysis was performed and appeared to confirm some of the immunocytochemistry (ICC) results. Finally, we demonstrated that the microchip can be used to monitor lactate dehydrogenase (LDH) released from the AHPC-NSs subjected to SW impacts. As expected, LDH levels changed when AHPC-NSs were injured by SW impacts, verifying this chip can be used for assessing the degrees of injuries to AHPC-NSs by monitoring LDH levels. Taken together, these results suggest the feasibility of using the chip to better understand the interactions between SW impacts and in vitro brain models, paving the way for potentially establishing in vitro TBI models on a chip.
Collapse
Affiliation(s)
- Md Fazlay Rubby
- Department of Electrical and Computer Engineering, Iowa State University, Ames, Iowa 50011, United States
| | - Catherine Fonder
- Molecular, Cellular, and Developmental Biology Program, Iowa State University, Ames, Iowa 50011, United States
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa 50011, United States
- Nanovaccine Institute, Iowa State University, Ames, Iowa 50011, United States
| | - Sajid Uchayash
- Department of Electrical and Computer Engineering, Iowa State University, Ames, Iowa 50011, United States
| | - Xiaogan Liang
- Department of Mechanical Engineering, University of Michigan at Ann Arbor, Ann Arbor, Michigan 48109, United States
| | - Donald S Sakaguchi
- Molecular, Cellular, and Developmental Biology Program, Iowa State University, Ames, Iowa 50011, United States
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa 50011, United States
- Neuroscience Program, Iowa State University, Ames, Iowa 50011, United States
- Nanovaccine Institute, Iowa State University, Ames, Iowa 50011, United States
| | - Long Que
- Department of Electrical and Computer Engineering, Iowa State University, Ames, Iowa 50011, United States
| |
Collapse
|
3
|
Sullivan D, Vaglio BJ, Cararo-Lopes MM, Wong RDP, Graudejus O, Firestein BL. Stretch-Induced Injury Affects Cortical Neuronal Networks in a Time- and Severity-Dependent Manner. Ann Biomed Eng 2024; 52:1021-1038. [PMID: 38294641 DOI: 10.1007/s10439-023-03438-0] [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: 09/04/2023] [Accepted: 12/22/2023] [Indexed: 02/01/2024]
Abstract
Traumatic brain injury (TBI) is the leading cause of accident-related death and disability in the world and can lead to long-term neuropsychiatric symptoms, such as a decline in cognitive function and neurodegeneration. TBI includes primary and secondary injury, with head trauma and deformation of the brain caused by the physical force of the impact as primary injury, and cellular and molecular cascades that lead to cell death as secondary injury. Currently, there is no treatment for TBI-induced cell damage and neural circuit dysfunction in the brain, and thus, it is important to understand the underlying cellular mechanisms that lead to cell damage. In the current study, we use stretchable microelectrode arrays (sMEAs) to model the primary injury of TBI to study the electrophysiological effects of physically injuring cortical cells. We recorded electrophysiological activity before injury and then stretched the flexible membrane of the sMEAs to injure the cells to varying degrees. At 1, 24, and 72 h post-stretch, we recorded activity to analyze differences in spike rate, Fano factor, burstlet rate, burstlet width, synchrony of firing, local network efficiency, and Q statistic. Our results demonstrate that mechanical injury changes the firing properties of cortical neuron networks in culture in a time- and severity-dependent manner. Our results suggest that changes to electrophysiological properties after stretch are dependent on the strength of synchronization between neurons prior to injury.
Collapse
Affiliation(s)
- Dylan Sullivan
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
- Cell and Developmental Biology Graduate Program, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Brandon J Vaglio
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
- Biomedical Engineering Graduate Program, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Marina M Cararo-Lopes
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
- Cell and Developmental Biology Graduate Program, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Ruben D Ponce Wong
- BioMedical Sustainable Elastic Electronic Devices (BMSEED), Mesa, AZ, USA
| | - Oliver Graudejus
- BioMedical Sustainable Elastic Electronic Devices (BMSEED), Mesa, AZ, USA
- School of Molecular Science, Arizona State University, Tempe, AZ, USA
| | - Bonnie L Firestein
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ, 08854-8082, USA.
| |
Collapse
|
4
|
Ali HT, Sula I, AbuHamdia A, Elejla SA, Elrefaey A, Hamdar H, Elfil M. Nervous System Response to Neurotrauma: A Narrative Review of Cerebrovascular and Cellular Changes After Neurotrauma. J Mol Neurosci 2024; 74:22. [PMID: 38367075 PMCID: PMC10874332 DOI: 10.1007/s12031-024-02193-8] [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: 11/15/2023] [Accepted: 01/22/2024] [Indexed: 02/19/2024]
Abstract
Neurotrauma is a significant cause of morbidity and mortality worldwide. For instance, traumatic brain injury (TBI) causes more than 30% of all injury-related deaths in the USA annually. The underlying cause and clinical sequela vary among cases. Patients are liable to both acute and chronic changes in the nervous system after such a type of injury. Cerebrovascular disruption has the most common and serious effect in such cases because cerebrovascular autoregulation, which is one of the main determinants of cerebral perfusion pressure, can be effaced in brain injuries even in the absence of evident vascular injury. Disruption of the blood-brain barrier regulatory function may also ensue whether due to direct injury to its structure or metabolic changes. Furthermore, the autonomic nervous system (ANS) can be affected leading to sympathetic hyperactivity in many patients. On a cellular scale, the neuroinflammatory cascade medicated by the glial cells gets triggered in response to TBI. Nevertheless, cellular and molecular reactions involved in cerebrovascular repair are not fully understood yet. Most studies were done on animals with many drawbacks in interpreting results. Therefore, future studies including human subjects are necessarily needed. This review will be of relevance to clinicians and researchers interested in understanding the underlying mechanisms in neurotrauma cases and the development of proper therapies as well as those with a general interest in the neurotrauma field.
Collapse
Affiliation(s)
| | - Idris Sula
- College of Medicine, Sulaiman Al Rajhi University, Al Bukayriyah, Al Qassim, Saudi Arabia
| | - Abrar AbuHamdia
- Department of Medical Laboratory Science, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | | | | | - Hiba Hamdar
- Medical Learning Skills Academy, Beirut, Lebanon
- Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Mohamed Elfil
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
5
|
Xie Z, Li T, Su W, Lou Y, Zhang Y, Zhou X, Li Z, Bai X, Liu X. Extension domain of amyloid processor protein inhibits amyloidogenic cleavage and balances neural activity in a traumatic brain injury mouse model. CNS Neurosci Ther 2024; 30:e14402. [PMID: 37592823 PMCID: PMC10848085 DOI: 10.1111/cns.14402] [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: 01/31/2023] [Revised: 06/01/2023] [Accepted: 07/07/2023] [Indexed: 08/19/2023] Open
Abstract
BACKGROUND Mechanisms underlying cognitive dysfunction following traumatic brain injury (TBI) partially due to abnormal amyloid processor protein (APP) cleavage and neural hyperactivity. Binding of the extension domain of APP (ExD17) to the GABAbR1 receptor results in reduced neural activity, which might play a role in the mechanisms of cognitive dysfunction caused by TBI. METHODS Stretch-induced injury was utilized to establish a cell injury model in HT22 cells. The TBI model was created by striking the exposed brain tissue with a free-falling weight. Topical or intraperitoneal administration of ExD17 was performed. Cell viability was assessed through a cell counting kit-8 assay, while intracellular Ca2+ was measured using Fluo-4. Western blotting was used to investigate the expression of APP amyloidogenic cleavage proteins, GABAbR1, phospholipase C (PLC), PLCB3, and synaptic proteins. ELISA was performed to analyze the levels of Aβ42. Seizures were assessed using electroencephalography (EEG). Behaviors were evaluated through the novel object recognition test, open field test, elevated plus maze test, and nest-building test. RESULTS ExD17 improved cell viability and reduced intracellular calcium in the cell injury model. The treatment also suppressed the increased expression of APP amyloidogenic cleavage proteins and Aβ42 in both cell injury and TBI models. ExD17 treatment reversed the abnormal expression of GABAbR1, GRIA2, p-PLCG1/PLCG1 ratio, and p-PLCB3/PLCB3 ratio. In addition, ExD17 treatment reduced neural activity, seizure events, and their duration in TBI. Intraperitoneal injection of ExD17 improved behavioral outcomes in the TBI mouse model. CONCLUSIONS ExD17 treatment results in a reduction of amyloidogenic APP cleavage and neuroexcitotoxicity, ultimately leading to an improvement in the behavioral deficits observed in TBI mice.
Collapse
Affiliation(s)
- Zhenxing Xie
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Tianyu Li
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Wei Su
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yanyun Lou
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yongsheng Zhang
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiyuan Zhou
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Zhanfei Li
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiangjun Bai
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xinghua Liu
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
6
|
Mitevska A, Santacruz C, Martin EJ, Jones IE, Ghiacy A, Dixon S, Mostafazadeh N, Peng Z, Kiskinis E, Finan JD. Polyurethane Culture Substrates Enable Long-Term Neuron Monoculture in a Human in vitro Model of Neurotrauma. Neurotrauma Rep 2023; 4:682-692. [PMID: 37908320 PMCID: PMC10615064 DOI: 10.1089/neur.2023.0060] [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] [Indexed: 11/02/2023] Open
Abstract
Human induced pluripotent stem cell (hiPSC)-derived cells can reproduce human-specific pathophysiology, patient-specific vulnerability, and gene-environment interactions in neurological disease. Human in vitro models of neurotrauma therefore have great potential to advance the field. However, this potential cannot be realized until important biomaterials challenges are addressed. Status quo stretch injury models of neurotrauma culture cells on sheets of polydimethylsiloxane (PDMS) that are incompatible with long-term monoculture of hiPSC-derived neurons. Here, we overcame this challenge in an established human in vitro neurotrauma model by replacing PDMS with a highly biocompatible form of polyurethane (PU). This substitution allowed long-term monoculture of hiPSC-derived neurons. It also changed the biomechanics of stretch injury. We quantified these changes experimentally using high-speed videography and digital image correlation. We used finite element modeling to quantify the influence of the culture substrate's thickness, stiffness, and coefficient of friction on membrane stretch and concluded that the coefficient of friction explained most of the observed biomechanical changes. Despite these changes, we demonstrated that the modified model produced a robust, dose-dependent trauma phenotype in hiPSC-derived neuron monocultures. In summary, the introduction of this PU film makes it possible to maintain hiPSC-derived neurons in monoculture for long periods in a human in vitro neurotrauma model. In doing so, it opens new horizons in the field of neurotrauma by enabling the unique experimental paradigms (e.g., isogenic models) associated with hiPSC-derived neurons.
Collapse
Affiliation(s)
- Angela Mitevska
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Citlally Santacruz
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Eric J. Martin
- The Ken & Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Ian E. Jones
- Department of Mechanical and Industrial Engineering, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Arian Ghiacy
- Department of Mechanical and Industrial Engineering, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Simon Dixon
- Biomer Technology Ltd., Warrington, United Kingdom
| | - Nima Mostafazadeh
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Zhangli Peng
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Evangelos Kiskinis
- The Ken & Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - John D. Finan
- Department of Mechanical and Industrial Engineering, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
7
|
Bolden CT, Skibber MA, Olson SD, Zamorano Rojas M, Milewicz S, Gill BS, Cox CS. Validation and characterization of a novel blood-brain barrier platform for investigating traumatic brain injury. Sci Rep 2023; 13:16150. [PMID: 37752338 PMCID: PMC10522590 DOI: 10.1038/s41598-023-43214-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 09/21/2023] [Indexed: 09/28/2023] Open
Abstract
The Blood-Brain Barrier (BBB) is a highly-selective physiologic barrier responsible for maintaining cerebral homeostasis. Innovative in vitro models of the BBB are needed to provide useful insights into BBB function with CNS disorders like traumatic brain injury (TBI). TBI is a multidimensional and highly complex pathophysiological condition that requires intrinsic models to elucidate its mechanisms. Current models either lack fluidic shear stress, or neglect hemodynamic parameters important in recapitulating the human in vivo BBB phenotype. To address these limitations in the field, we developed a fluid dynamic novel platform which closely mimics these parameters. To validate our platform, Matrigel-coated Transwells were seeded with brain microvascular endothelial cells, both with and without co-cultured primary human astrocytes and bone-marrow mesenchymal stem cells. In this article we characterized BBB functional properties such as TEER and paracellular permeability. Our platform demonstrated physiologic relevant decreases in TEER in response to an ischemic environment, while directly measuring barrier fluid fluctuation. These recordings were followed with recovery, implying stability of the model. We also demonstrate that our dynamic platform is responsive to inflammatory and metabolic cues with resultant permeability coefficients. These results indicate that this novel dynamic platform will be a valuable tool for evaluating the recapitulating BBB function in vitro, screening potential novel therapeutics, and establishing a relevant paradigm to evaluate the pathophysiology of TBI.
Collapse
Affiliation(s)
- Christopher T Bolden
- Department of Pediatric Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.
- Center for Translational Injury Research, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.
| | - Max A Skibber
- Department of Pediatric Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Scott D Olson
- Department of Pediatric Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Miriam Zamorano Rojas
- Department of Pediatric Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Samantha Milewicz
- Department of Pediatric Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Brijesh S Gill
- Department of Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Charles S Cox
- Department of Pediatric Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.
- Center for Translational Injury Research, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.
| |
Collapse
|
8
|
Schlotterose L, Beldjilali-Labro M, Hagel M, Yadid M, Flaxer C, Flaxer E, Barnea AR, Hattermann K, Shohami E, Leichtmann-Bardoogo Y, Maoz BM. Inducing Mechanical Stimuli to Tissues Grown on a Magnetic Gel Allows Deconvoluting the Forces Leading to Traumatic Brain Injury. Neurotrauma Rep 2023; 4:560-572. [PMID: 37636339 PMCID: PMC10457614 DOI: 10.1089/neur.2023.0026] [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] [Indexed: 08/29/2023] Open
Abstract
Traumatic brain injury (TBI), which is characterized by damage to the brain resulting from a sudden traumatic event, is a major cause of death and disability worldwide. It has short- and long-term effects, including neuroinflammation, cognitive deficits, and depression. TBI consists of multiple steps that may sometimes have opposing effects or mechanisms, making it challenging to investigate and translate new knowledge into effective therapies. In order to better understand and address the underlying mechanisms of TBI, we have developed an in vitro platform that allows dynamic simulation of TBI conditions by applying external magnetic forces to induce acceleration and deceleration injury, which is often observed in human TBI. Endothelial and neuron-like cells were successfully grown on magnetic gels and applied to the platform. Both cell types showed an instant response to the TBI model, but the endothelial cells were able to recover quickly-in contrast to the neuron-like cells. In conclusion, the presented in vitro model mimics the mechanical processes of acceleration/deceleration injury involved in TBI and will be a valuable resource for further research on brain injury.
Collapse
Affiliation(s)
- Luise Schlotterose
- Institute of Anatomy, Kiel University, Kiel, Germany
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
| | | | - Mario Hagel
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Moran Yadid
- The Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
| | - Carina Flaxer
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Eli Flaxer
- AFEKA–Tel-Aviv Academic College of Engineering, Tel-Aviv, Israel
| | - A. Ronny Barnea
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
| | | | - Esther Shohami
- Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | | | - Ben M. Maoz
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
- Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel
- Sagol Center for Regenerative Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
9
|
Zhao Q, Zhang J, Li H, Li H, Xie F. Models of traumatic brain injury-highlights and drawbacks. Front Neurol 2023; 14:1151660. [PMID: 37396767 PMCID: PMC10309005 DOI: 10.3389/fneur.2023.1151660] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 05/26/2023] [Indexed: 07/04/2023] Open
Abstract
Traumatic brain injury (TBI) is the leading cause for high morbidity and mortality rates in young adults, survivors may suffer from long-term physical, cognitive, and/or psychological disorders. Establishing better models of TBI would further our understanding of the pathophysiology of TBI and develop new potential treatments. A multitude of animal TBI models have been used to replicate the various aspects of human TBI. Although numerous experimental neuroprotective strategies were identified to be effective in animal models, a majority of strategies have failed in phase II or phase III clinical trials. This failure in clinical translation highlights the necessity of revisiting the current status of animal models of TBI and therapeutic strategies. In this review, we elucidate approaches for the generation of animal models and cell models of TBI and summarize their strengths and limitations with the aim of exploring clinically meaningful neuroprotective strategies.
Collapse
Affiliation(s)
- Qinghui Zhao
- Institute of Physical Culture, Huanghuai University, Zhumadian, China
| | - Jianhua Zhang
- Institute of Physical Culture, Huanghuai University, Zhumadian, China
| | - Huige Li
- Institute of Physical Culture, Huanghuai University, Zhumadian, China
| | - Hongru Li
- Zhumadian Central Hospital, Zhumadian, China
| | - Fei Xie
- Faculty of Environment and Life, Beijing University of Technology, Beijing, China
| |
Collapse
|
10
|
Loussert-Fonta C, Stoppini L, Neuenschwander Y, Righini O, Prim D, Schmidt C, Heuschkel MO, Gomez Baisac L, Jovic´ M, Pfeifer ME, Extermann J, Roux A. Opening the black box of traumatic brain injury: a holistic approach combining human 3D neural tissue and an in vitro traumatic brain injury induction device. Front Neurosci 2023; 17:1189615. [PMID: 37397462 PMCID: PMC10308006 DOI: 10.3389/fnins.2023.1189615] [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: 03/19/2023] [Accepted: 05/09/2023] [Indexed: 07/04/2023] Open
Abstract
Traumatic brain injury (TBI) is caused by a wide range of physical events and can induce an even larger spectrum of short- to long-term pathophysiologies. Neuroscientists have relied on animal models to understand the relationship between mechanical damages and functional alterations of neural cells. These in vivo and animal-based in vitro models represent important approaches to mimic traumas on whole brains or organized brain structures but are not fully representative of pathologies occurring after traumas on human brain parenchyma. To overcome these limitations and to establish a more accurate and comprehensive model of human TBI, we engineered an in vitro platform to induce injuries via the controlled projection of a small drop of liquid onto a 3D neural tissue engineered from human iPS cells. With this platform, biological mechanisms involved in neural cellular injury are recorded through electrophysiology measurements, quantification of biomarkers released, and two imaging methods [confocal laser scanning microscope (CLSM) and optical projection tomography (OPT)]. The results showed drastic changes in tissue electrophysiological activities and significant releases of glial and neuronal biomarkers. Tissue imaging allowed us to reconstruct the injured area spatially in 3D after staining it with specific nuclear dyes and to determine TBI resulting in cell death. In future experiments, we seek to monitor the effects of TBI-induced injuries over a prolonged time and at a higher temporal resolution to better understand the subtleties of the biomarker release kinetics and the cell recovery phases.
Collapse
Affiliation(s)
- Céline Loussert-Fonta
- Tissue Engineering Laboratory, HEPIA HES-SO University of Applied Sciences and Arts Western Switzerland, Geneva, Switzerland
| | - Luc Stoppini
- Tissue Engineering Laboratory, HEPIA HES-SO University of Applied Sciences and Arts Western Switzerland, Geneva, Switzerland
| | - Yoan Neuenschwander
- Micro-Nanotechnology Group, HEPIA HES-SO University of Applied Sciences and Arts Western Switzerland, Geneva, Switzerland
| | - Ophélie Righini
- Diagnostic Systems Research Group, Institute of Life Technologies, School of Engineering, University of Applied Sciences and Arts Western Switzerland (HES-SO Valais-Wallis), Sion, Switzerland
| | - Denis Prim
- Diagnostic Systems Research Group, Institute of Life Technologies, School of Engineering, University of Applied Sciences and Arts Western Switzerland (HES-SO Valais-Wallis), Sion, Switzerland
| | - Cédric Schmidt
- Micro-Nanotechnology Group, HEPIA HES-SO University of Applied Sciences and Arts Western Switzerland, Geneva, Switzerland
| | - Marc O. Heuschkel
- Tissue Engineering Laboratory, HEPIA HES-SO University of Applied Sciences and Arts Western Switzerland, Geneva, Switzerland
| | - Loris Gomez Baisac
- Tissue Engineering Laboratory, HEPIA HES-SO University of Applied Sciences and Arts Western Switzerland, Geneva, Switzerland
| | - Milica Jovic´
- Diagnostic Systems Research Group, Institute of Life Technologies, School of Engineering, University of Applied Sciences and Arts Western Switzerland (HES-SO Valais-Wallis), Sion, Switzerland
| | - Marc E. Pfeifer
- Diagnostic Systems Research Group, Institute of Life Technologies, School of Engineering, University of Applied Sciences and Arts Western Switzerland (HES-SO Valais-Wallis), Sion, Switzerland
| | - Jérôme Extermann
- Micro-Nanotechnology Group, HEPIA HES-SO University of Applied Sciences and Arts Western Switzerland, Geneva, Switzerland
| | - Adrien Roux
- Tissue Engineering Laboratory, HEPIA HES-SO University of Applied Sciences and Arts Western Switzerland, Geneva, Switzerland
| |
Collapse
|
11
|
Hanna M, Ali A, Klienberger M, Pfister BJ. A Method for Evaluating Brain Deformation Under Sagittal Blunt Impacts Using a Half-Skull Human-Scale Surrogate. J Biomech Eng 2023; 145:1155772. [PMID: 36562120 DOI: 10.1115/1.4056547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 11/16/2022] [Indexed: 12/24/2022]
Abstract
Trauma to the brain is a biomechanical problem where the initiating event is a dynamic loading (blunt, inertial, blast) to the head. To understand the relationship between the mechanical parameters of the injury and the spatial and temporal deformation patterns in the brain, there is a need to develop a reusable and adaptable experimental traumatic brain injury (TBI) model that can measure brain motion under varying parameters. In this effort, we aim to directly measure brain deformation (strain and strain rates) in different brain regions in a human head model using a drop tower. METHODS Physical head models consisting of a half, sagittal plane skull, brain, and neck were constructed and subjected to crown and frontal impacts at two impact speeds. All tests were recorded with a high-speed camera at 1000 frames per second. Motion of visual markers within brain surrogates were used to track deformations and calculate spatial strain histories in 6 brain regions of interest. Principal strains, strain rates and strain impulses were calculated and reported. RESULTS Higher impact velocities corresponded to higher strain values across all impact scenarios. Crown impacts were characterized by high, long duration strains distributed across the parietal, frontal and hippocampal regions whereas frontal impacts were characterized by sharply rising and falling strains primarily found in the parietal, frontal, hippocampal and occipital regions. High strain rates were associated with short durations and impulses indicating fast but short-lived strains. 2.23 m/s (5 mph) crown impacts resulted in 53% of the brain with shear strains higher than 0.15 verses 32% for frontal impacts. CONCLUSIONS The results reveal large differences in the spatial and temporal strain responses between crown and forehead impacts. Overall, the results suggest that for the same speed, crown impact leads to higher magnitude strain patterns than a frontal impact. The data provided by this model provides unique insight into the spatial and temporal deformation patterns that have not been provided by alternate surrogate models. The model can be used to investigate how anatomical, material and loading features and parameters can affect deformation patterns in specific regions of interest in the brain.
Collapse
Affiliation(s)
- Michael Hanna
- Department of Biomedical Engineering, Center for Injury Biomechanics, Materials and Medicine, New Jersey Institute of Technology, Newark, NJ 07102
| | - Abdus Ali
- Department of Biomedical Engineering, Center for Injury Biomechanics, Materials and Medicine, New Jersey Institute of Technology, Newark, NJ 07102
| | | | - Bryan J Pfister
- Department of Biomedical Engineering, Center for Injury Biomechanics, Materials and Medicine, New Jersey Institute of Technology, Newark, NJ 07102
| |
Collapse
|
12
|
Adams AA, Li Y, Kim HA, Pfister BJ. Dorsal root ganglion neurons recapitulate the traumatic axonal injury of CNS neurons in response to a rapid stretch in vitro. Front Cell Neurosci 2023; 17:1111403. [PMID: 37066078 PMCID: PMC10090399 DOI: 10.3389/fncel.2023.1111403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/28/2023] [Indexed: 03/31/2023] Open
Abstract
Introduction: In vitro models of traumatic brain injury (TBI) commonly use neurons isolated from the central nervous system. Limitations with primary cortical cultures, however, can pose challenges to replicating some aspects of neuronal injury associated with closed head TBI. The known mechanisms of axonal degeneration from mechanical injury in TBI are in many ways similar to degenerative disease, ischemia, and spinal cord injury. It is therefore possible that the mechanisms that result in axonal degeneration in isolated cortical axons after in vitro stretch injury are shared with injured axons from different neuronal types. Dorsal root ganglia neurons (DRGN) are another neuronal source that may overcome some current limitations including remaining healthy in culture for long periods of time, ability to be isolated from adult sources, and myelinated in vitro. Methods: The current study sought to characterize the differential responses between cortical and DRGN axons to mechanical stretch injury associated with TBI. Using an in vitro model of traumatic axonal stretch injury, cortical and DRGN neurons were injured at a moderate (40% strain) and severe stretch (60% strain) and acute alterations in axonal morphology and calcium homeostasis were measured. Results: DRGN and cortical axons immediately form undulations in response to severe injury, experience similar elongation and recovery within 20 min after the initial injury, and had a similar pattern of degeneration over the first 24 h after injury. Additionally, both types of axons experienced comparable degrees of calcium influx after both moderate and severe injury that was prevented through pre-treatment with tetrodotoxin in cortical neurons and lidocaine in DRGNs. Similar to cortical axons, stretch injury also causes calcium activated proteolysis of sodium channel in DRGN axons that is prevented by treatment with lidocaine or protease inhibitors. Discussion: These findings suggest that DRGN axons share the early response of cortical neurons to a rapid stretch injury and the associated secondary injury mechanisms. The utility of a DRGN in vitro TBI model may allow future studies to explore TBI injury progression in myelinated and adult neurons.
Collapse
Affiliation(s)
- Alexandra A. Adams
- Center for Injury Biomechanics, Materials and Medicine, Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, United States
- Department of Biological Sciences, Rutgers University Newark, Newark, NJ, United States
| | - Ying Li
- Center for Injury Biomechanics, Materials and Medicine, Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, United States
| | - Haesun A. Kim
- Department of Biological Sciences, Rutgers University Newark, Newark, NJ, United States
| | - Bryan J. Pfister
- Center for Injury Biomechanics, Materials and Medicine, Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, United States
- Department of Biological Sciences, Rutgers University Newark, Newark, NJ, United States
| |
Collapse
|
13
|
Zargari M, Meyer LJ, Riess ML, Li Z, Barajas MB. P188 Therapy in In Vitro Models of Traumatic Brain Injury. Int J Mol Sci 2023; 24:3334. [PMID: 36834743 PMCID: PMC9961452 DOI: 10.3390/ijms24043334] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/26/2023] [Accepted: 01/30/2023] [Indexed: 02/11/2023] Open
Abstract
Traumatic brain injury (TBI) is a significant cause of morbidity and mortality worldwide. Varied mechanisms of injury contribute to the heterogeneity of this patient population as demonstrated by the multiple published grading scales and diverse required criteria leading to diagnoses from mild to severe. TBI pathophysiology is classically separated into a primary injury that is characterized by local tissue destruction as a result of the initial blow, followed by a secondary phase of injury constituted by a score of incompletely understood cellular processes including reperfusion injury, disruption to the blood-brain barrier, excitotoxicity, and metabolic dysregulation. There are currently no effective pharmacological treatments in the wide-spread use for TBI, in large part due to challenges associated with the development of clinically representative in vitro and in vivo models. Poloxamer 188 (P188), a Food and Drug Administration-approved amphiphilic triblock copolymer embeds itself into the plasma membrane of damaged cells. P188 has been shown to have neuroprotective properties on various cell types. The objective of this review is to provide a summary of the current literature on in vitro models of TBI treated with P188.
Collapse
Affiliation(s)
- Michael Zargari
- Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | | | - Matthias L. Riess
- TVHS VA Medical Center, Anesthesiology, Nashville, TN 37212, USA
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Zhu Li
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Matthew B. Barajas
- TVHS VA Medical Center, Anesthesiology, Nashville, TN 37212, USA
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
14
|
Establishment and Application of a Novel In Vitro Model of Microglial Activation in Traumatic Brain Injury. J Neurosci 2023; 43:319-332. [PMID: 36446585 PMCID: PMC9838700 DOI: 10.1523/jneurosci.1539-22.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 11/08/2022] [Accepted: 11/17/2022] [Indexed: 12/03/2022] Open
Abstract
Mechanical impact-induced primary injury after traumatic brain injury (TBI) leads to acute microglial pro-inflammatory activation and consequently mediates neurodegeneration, which is a major secondary brain injury mechanism. However, the detailed pathologic cascades have not been fully elucidated, partially because of the pathologic complexity in animal TBI models. Although there are several in vitro TBI models, none of them closely mimic post-TBI microglial activation. In the present study, we aimed to establish an in vitro TBI model, specifically reconstituting the pro-inflammatory activation and associated neurodegeneration following TBI. We proposed three sets of experiments. First, we established a needle scratch injured neuron-induced microglial activation and neurodegeneration in vitro model of TBI. Second, we compared microglial pro-inflammatory cytokines profiles between the in vitro TBI model and TBI in male mice. Additionally, we validated the role of injured neurons-derived damage-associated molecular patterns in amplifying microglial pro-inflammatory pathways using the in vitro TBI model. Third, we applied the in vitro model for the first time to characterize the cellular metabolic profile of needle scratch injured-neuron-activated microglia, and define the role of metabolic reprogramming in mediating pro-inflammatory microglial activation and mediated neurodegeneration. Our results showed that we successfully established a novel in vitro TBI model, which closely mimics primary neuronal injury-triggered microglial pro-inflammatory activation and mediated neurodegeneration after TBI. This in vitro model provides an advanced and highly translational platform for dissecting interactions in the pathologic processes of neuronal injury-microglial activation-neuronal degeneration cascade, and elucidating the detailed underlying cellular and molecular insights after TBI.SIGNIFICANCE STATEMENT Microglial activation is a key component of acute neuroinflammation that leads to neurodegeneration and long-term neurologic outcome deficits after TBI. However, it is not feasible to truly dissect primary neuronal injury-induced microglia activation, and consequently mediated neurodegeneration in vivo Furthermore, there is currently lacking of in vitro TBI models closely mimicking the TBI primary injury-mediated microglial activation. In this study, we successfully established and validated a novel in vitro TBI model of microglial activation, and for the first time, characterized the cellular metabolic profile of microglia in this model. This novel microglial activation in vitro TBI model will help in elucidating microglial inflammatory activation and consequently associated neurodegeneration after TBI.
Collapse
|
15
|
Kundu S, Singh S. What Happens in TBI? A Wide Talk on Animal Models and Future Perspective. Curr Neuropharmacol 2023; 21:1139-1164. [PMID: 35794772 PMCID: PMC10286592 DOI: 10.2174/1570159x20666220706094248] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 05/05/2022] [Accepted: 05/11/2022] [Indexed: 11/22/2022] Open
Abstract
Traumatic brain injury (TBI) is a global healthcare concern and a leading cause of death. The most common causes of TBI include road accidents, sports injuries, violence in warzones, and falls. TBI induces neuronal cell death independent of age, gender, and genetic background. TBI survivor patients often experience long-term behavioral changes like cognitive and emotional changes. TBI affects social activity, reducing the quality and duration of life. Over the last 40 years, several rodent models have been developed to mimic different clinical outcomes of human TBI for a better understanding of pathophysiology and to check the efficacy of drugs used for TBI. However, promising neuroprotective approaches that have been used preclinically have been found to be less beneficial in clinical trials. So, there is an urgent need to find a suitable animal model for establishing a new therapeutic intervention useful for TBI. In this review, we have demonstrated the etiology of TBI and post- TBI social life alteration, and also discussed various preclinical TBI models of rodents, zebrafish, and drosophila.
Collapse
Affiliation(s)
- Satyabrata Kundu
- Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Shamsher Singh
- Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| |
Collapse
|
16
|
Chari D, Basit R, Wiseman J, Chowdhury F. Simulating traumatic brain injury in vitro: developing high throughput models to test biomaterial based therapies. Neural Regen Res 2023; 18:289-292. [PMID: 35900405 PMCID: PMC9396524 DOI: 10.4103/1673-5374.346465] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Traumatic brain injuries are serious clinical incidents associated with some of the poorest outcomes in neurological practice. Coupled with the limited regenerative capacity of the brain, this has significant implications for patients, carers, and healthcare systems, and the requirement for life-long care in some cases. Clinical treatment currently focuses on limiting the initial neural damage with long-term care/support from multidisciplinary teams. Therapies targeting neuroprotection and neural regeneration are not currently available but are the focus of intensive research. Biomaterial-based interventions are gaining popularity for a range of applications including biomolecule and drug delivery, and to function as cellular scaffolds. Experimental investigations into the development of such novel therapeutics for traumatic brain injury will be critically underpinned by the availability of appropriate high throughput, facile, ethically viable, and pathomimetic biological model systems. This represents a significant challenge for researchers given the pathological complexity of traumatic brain injury. Specifically, there is a concerted post-injury response mounted by multiple neural cell types which includes microglial activation and astroglial scarring with the expression of a range of growth inhibitory molecules and cytokines in the lesion environment. Here, we review common models used for the study of traumatic brain injury (ranging from live animal models to in vitro systems), focusing on penetrating traumatic brain injury models. We discuss their relative advantages and drawbacks for the developmental testing of biomaterial-based therapies.
Collapse
|
17
|
Wang Y, Guo Q, Wang W, Wang Y, Fang K, Wan Q, Li H, Wu T. Potential use of bioactive nanofibrous dural substitutes with controlled release of IGF-1 for neuroprotection after traumatic brain injury. NANOSCALE 2022; 14:18217-18230. [PMID: 36468670 DOI: 10.1039/d2nr06081g] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
For patients suffering from traumatic brain injury (TBI), the closure of dural defects after decompressive craniectomy is the prerequisite to restoring normal physiological functions. It is also an urgent challenge to provide a neuroprotection effect against the primary and secondary nerve damage during long-term recovery. To solve these issues, we herein develop a class of bioactive, nanofibrous dural substitutes that can long-term release insulin-like growth factor 1 (IGF-1) for improving the survival and neurite outgrowth of neural cells after TBI. Such dural substitutes were polycaprolactone (PCL) nanofibers encapsulated with hyaluronic acid methacryloyl (HAMA)/IGF-1 by blend or coaxial electrospinning techniques, achieving bioactive PCL/HAMA/IGF nanofibrous dural substitutes with different release profiles of IGF-1. The nanofibrous dural substitutes exhibited good mechanical properties and hydrophobicity, which prevent cerebrospinal fluid leakage, maintain normal intracranial pressure, and avoid external impact on the brain. We also found that the viability and neurite outgrowth of SH-SY5Y cells and primary neurons were significantly enhanced after neurite transection or oxygen and glucose deprivation treatment. Taken together, such PCL/HAMA/IGF nanofibrous dural substitutes hold promising potential to provide neuroprotection effects after primary and secondary nerve damage in TBI, which would bring significant benefits to the field of neurosurgery involving the use of artificial dura mater.
Collapse
Affiliation(s)
- Yue Wang
- Department of Neurosurgery, Affiliated Hospital of Qingdao University, Qingdao 266071, China.
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao Medical College, Qingdao University, Qingdao 266071, China
| | - Qingxia Guo
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao Medical College, Qingdao University, Qingdao 266071, China
| | - Wei Wang
- Shandong Key Laboratory of Medical and Health Textile Materials, Collaborative Innovation Center for Eco-textiles of Shandong Province and the Ministry of Education, Qingdao 266071, China
| | - Yuanfei Wang
- Department of Central Laboratory, Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao 266001, China
| | - Kuanjun Fang
- Shandong Key Laboratory of Medical and Health Textile Materials, Collaborative Innovation Center for Eco-textiles of Shandong Province and the Ministry of Education, Qingdao 266071, China
| | - Qi Wan
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao Medical College, Qingdao University, Qingdao 266071, China
| | - Huanting Li
- Department of Neurosurgery, Affiliated Hospital of Qingdao University, Qingdao 266071, China.
| | - Tong Wu
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao Medical College, Qingdao University, Qingdao 266071, China
- Shandong Key Laboratory of Medical and Health Textile Materials, Collaborative Innovation Center for Eco-textiles of Shandong Province and the Ministry of Education, Qingdao 266071, China
| |
Collapse
|
18
|
Pan X, Li J, Li W, Wang H, Durisic N, Li Z, Feng Y, Liu Y, Zhao CX, Wang T. Axons-on-a-chip for mimicking non-disruptive diffuse axonal injury underlying traumatic brain injury. LAB ON A CHIP 2022; 22:4541-4555. [PMID: 36318066 DOI: 10.1039/d2lc00730d] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Diffuse axonal injury (DAI) is the most severe pathological feature of traumatic brain injury (TBI). However, how primary axonal injury is induced by transient mechanical impacts remains unknown, mainly due to the low temporal and spatial resolution of medical imaging approaches. Here we established an axon-on-a-chip (AoC) model for mimicking DAI and monitoring instant cellular responses. Integrating computational fluid dynamics and microfluidic techniques, DAI was induced by injecting a precisely controlled micro-flux in the transverse direction. The clear correlation between the flow speed of injecting flux and the severity of DAI was elucidated. We next used the AoC to investigate the instant intracellular responses underlying DAI and found that the dynamic formation of focal axonal swellings (FAS) accompanied by Ca2+ surge occurs during the flux. Surprisingly, periodic axonal cytoskeleton disruption also occurs rapidly after the flux. These instant injury responses are spatially restricted to the fluxed axon, not affecting the overall viability of the neuron in the acute stage. Compatible with high-resolution live microscopy, the AoC provides a versatile system to identify early mechanisms underlying DAI, offering a platform for screening effective treatments to alleviate TBI.
Collapse
Affiliation(s)
- Xiaorong Pan
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| | - Jie Li
- Division of Chemistry and Physical Biology, School of Physical Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Wei Li
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Haofei Wang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Nela Durisic
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Zhenyu Li
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| | - Yu Feng
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| | - Yifan Liu
- Division of Chemistry and Physical Biology, School of Physical Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Chun-Xia Zhao
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
- School of Chemical Engineering and Advanced Materials, The University of Adelaide, Adelaide, SA 5005, Australia.
| | - Tong Wang
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| |
Collapse
|
19
|
Hanna ME, Pfister BJ. Advancements in in vitro models of traumatic brain injury. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2022. [DOI: 10.1016/j.cobme.2022.100430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
20
|
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: 1.0] [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
|
21
|
Rue MC, Alonso LM, Marder E. Repeated applications of high potassium elicit long-term changes in a motor circuit from the crab, Cancer borealis. iScience 2022; 25:104919. [PMID: 36060056 PMCID: PMC9436765 DOI: 10.1016/j.isci.2022.104919] [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: 05/13/2022] [Revised: 07/12/2022] [Accepted: 08/08/2022] [Indexed: 12/04/2022] Open
Abstract
We examined the effects of altered extracellular potassium concentration on the output of the well-studied pyloric circuit in the crab, Cancer borealis. Pyloric neurons initially become quiescent, then recover spiking and bursting activity in high potassium saline (2.5x[K+]). These changes in circuit robustness are maintained after the perturbation is removed; pyloric neurons are more robust to subsequent potassium perturbations even after several hours of wash in control saline. Despite this long-term "memory" of the stimulus history, we found no differences in neuronal activity in control saline. The circuit's adaptation is erased by both low potassium saline (0.4x[K+]) and direct hyperpolarizing current. Initial sensitivity of PD neurons to high potassium saline also varies seasonally, indicating that changes in robustness may reflect natural changes in circuit states. Thus, perturbation, followed by recovery of normal activity, can hide cryptic changes in neuronal properties that are only revealed by subsequent challenges.
Collapse
Affiliation(s)
- Mara C.P. Rue
- Biology Department and Volen Center, Brandeis University, Waltham, MA 02454, USA
| | - Leandro M. Alonso
- Biology Department and Volen Center, Brandeis University, Waltham, MA 02454, USA
| | - Eve Marder
- Biology Department and Volen Center, Brandeis University, Waltham, MA 02454, USA,Corresponding author
| |
Collapse
|
22
|
Fiber orientation downsampling compromises the computation of white matter tract-related deformation. J Mech Behav Biomed Mater 2022; 132:105294. [DOI: 10.1016/j.jmbbm.2022.105294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 04/13/2022] [Accepted: 05/21/2022] [Indexed: 11/18/2022]
|
23
|
Xu D, Zhang N, Wang S, Yu Y, Zhang P, Li Y, Yang H. A Novel In Vitro Platform Development in the Lab for Modeling Blast Injury to Microglia. Front Bioeng Biotechnol 2022; 10:883545. [PMID: 35903797 PMCID: PMC9315251 DOI: 10.3389/fbioe.2022.883545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 06/10/2022] [Indexed: 11/23/2022] Open
Abstract
Traumatic brain injury (TBI), which is mainly caused by impact, often results in chronic neurological abnormalities. Since the pathological changes in vivo during primary biomechanical injury are quite complicated, the in-depth understanding of the pathophysiology and mechanism of TBI depends on the establishment of an effective experimental in vitro model. Usually, a bomb explosive blast was employed to establish the in vitro model, while the process is complex and unsuitable in the lab. Based on water-hammer, we have developed a device system to provide a single dynamic compression stress on living cells. A series of amplitude (∼5.3, ∼9.8, ∼13.5 MPa) were generated to explore the effects of dynamic compression loading on primary microglia within 48 h. Apoptosis experiments indicated that primary microglia had strong tolerance to blast waves. In addition, the generation of intercellular reactive oxygen species and secretory nitric oxide was getting strongly enhanced and recovered within 48 h. In addition, there is a notable release of pro-inflammatory cytokine by microglia. Our work provides a reproducible and peaceable method of loading single dynamic compression forces to cells in vitro. Microglia showed an acute inflammatory response to dynamic loadings, while no significant cell death was observed. This insight delivers a new technological approach that could open new areas to a better understanding of the mechanism of cell blast injuries.
Collapse
Affiliation(s)
- Dasen Xu
- School of Aeronautics, Northwestern Polytechnical University, Xi’an, China
- Center of Special Environmental Biomechanics and Biomedical Engineering, Northwestern Polytechnical University, Xi’an, China
| | - Nu Zhang
- Center of Special Environmental Biomechanics and Biomedical Engineering, Northwestern Polytechnical University, Xi’an, China
- School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Sijie Wang
- Center of Special Environmental Biomechanics and Biomedical Engineering, Northwestern Polytechnical University, Xi’an, China
- School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Yawei Yu
- School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Pan Zhang
- Center of Special Environmental Biomechanics and Biomedical Engineering, Northwestern Polytechnical University, Xi’an, China
- School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Yulong Li
- School of Aeronautics, Northwestern Polytechnical University, Xi’an, China
- Center of Special Environmental Biomechanics and Biomedical Engineering, Northwestern Polytechnical University, Xi’an, China
- *Correspondence: Yulong Li, ; Hui Yang,
| | - Hui Yang
- Center of Special Environmental Biomechanics and Biomedical Engineering, Northwestern Polytechnical University, Xi’an, China
- School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
- *Correspondence: Yulong Li, ; Hui Yang,
| |
Collapse
|
24
|
Weisz HA, Boone DR, Coggins WS, Edwards GA, Willey HE, Widen SG, Siegel D, Nelson AT, Prough DS, Hellmich HL. Mechanistic insights gained from cell and molecular analysis of the neuroprotective potential of bioactive natural compounds in an immortalized hippocampal cell line. PLoS One 2022; 17:e0267682. [PMID: 35657963 PMCID: PMC9165808 DOI: 10.1371/journal.pone.0267682] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 04/14/2022] [Indexed: 11/19/2022] Open
Abstract
Evaluating novel compounds for neuroprotective effects in animal models of traumatic brain injury (TBI) is a protracted, labor-intensive and costly effort. However, the present lack of effective treatment options for TBI, despite decades of research, shows the critical need for alternative methods for screening new drug candidates with neuroprotective properties. Because natural products have been a leading source of new therapeutic agents for human diseases, we used an in vitro model of stretch injury to rapidly assess pro-survival effects of three bioactive compounds, two isolated from natural products (clovanemagnolol [CM], vinaxanthone [VX]) and the third, a dietary compound (pterostilbene [PT]) found in blueberries. The stretch injury experiments were not used to validate drug efficacy in a comprehensive manner but used primarily, as proof-of-principle, to demonstrate that the neuroprotective potential of each bioactive agent can be quickly assessed in an immortalized hippocampal cell line in lieu of comprehensive testing in animal models of TBI. To gain mechanistic insights into potential molecular mechanisms of neuroprotective effects, we performed a pathway-specific PCR array analysis of the effects of CM on the rat hippocampus and microRNA sequencing analysis of the effects of VX and PT on cultured hippocampal progenitor neurons. We show that the neuroprotective properties of these natural compounds are associated with altered expression of several genes or microRNAs that have functional roles in neurodegeneration or cell survival. Our approach could help in quickly assessing multiple natural products for neuroprotective properties and expedite the process of new drug discovery for TBI therapeutics.
Collapse
Affiliation(s)
- Harris A. Weisz
- Department of Anesthesiology, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Deborah R. Boone
- Department of Anesthesiology, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - William S. Coggins
- Department of Neurosurgery, The University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Gabrielle A. Edwards
- Department of Anesthesiology, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Hannah E. Willey
- Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Steven G. Widen
- Department of Biochemistry & Molecular Biology, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Dionicio Siegel
- Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of California San Diego, San Diego, California, United States of America
| | - Andrew T. Nelson
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Donald S. Prough
- Department of Anesthesiology, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Helen L. Hellmich
- Department of Anesthesiology, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
- * E-mail:
| |
Collapse
|
25
|
Li Y, Chen W, Deng H, Li T, Liu Z, Liu X, Zhang Z, Chen X, Sheng J, Li K. TGF-β1 Protects Trauma-injured Murine Cortical Neurons by Upregulating L-type Calcium Channel Ca v1.2 via the p38 Pathway. Neuroscience 2022; 492:47-57. [PMID: 35460836 DOI: 10.1016/j.neuroscience.2022.04.010] [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: 02/10/2022] [Revised: 04/09/2022] [Accepted: 04/12/2022] [Indexed: 02/05/2023]
Abstract
Traumatic brain injury (TBI) is a leading cause of disability and death in adolescents, and there is a lack of effective methods of treatment. The neuroprotective effects exerted by TGF-β1 can ameliorate a range of neuronal lesions in multiple central nervous system diseases. In this study, we used an in-vitro TBI model of mechanical injury on murine primary cortical neurons and the neuro-2a cell line to investigate the neuroprotective role played by TGF-β1 in cortical neurons in TBI. Our results showed that TGF-β1 significantly increased neuronal viability and inhibited apoptosis for 24 h after trauma. The expression of Cav1.2, an L-type calcium channel (LTCC) isoform, decreased significantly after trauma injury, and this change was reversed by TGF-β1. Nimodipine, a classic LTCC blocker, abolished the protective effect of TGF-β1 on trauma-induced neuronal apoptosis. The knockdown of Cav1.2 in differentiated neuro-2a cells significantly inhibited the anti-apoptosis effect of TGF-β1 exerted on injured neuro-2a cells. Moreover, TGF-β1 rescued and enhanced the trauma-suppressed neuro-2a intracellular Ca2+ concentration, while the effect of TGF-β1 was partially inhibited by nimodipine. TGF-β1 significantly upregulated the expression of Cav1.2 by activating the p38 MAPK pathway and by inhibiting trauma-induced neuronal apoptosis. In conclusion, TGF-β1 increased trauma-injured murine cortical neuronal activity and inhibited apoptosis by upregulating Cav1.2 channels via activating the p38 MAPK pathway. Therefore, the TGF-β1/p38 MAPK/Cav 1.2 pathway has the potential to be used as a novel therapeutic target for TBI.
Collapse
Affiliation(s)
- Yanlei Li
- Department of Microbiology and Immunology, Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| | - Weiqiang Chen
- Department of Neurosurgery, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Huixiong Deng
- Department of Microbiology and Immunology, Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| | - Tian Li
- Department of Microbiology and Immunology, Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| | - Zhenning Liu
- Department of Laboratory, Guangzhou Chest Hospital, China
| | - Xueer Liu
- Department of Microbiology and Immunology, Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| | - Zelin Zhang
- Department of Microbiology and Immunology, Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| | - Xiaoxuan Chen
- Department of Microbiology and Immunology, Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| | - Jiangtao Sheng
- Department of Microbiology and Immunology, Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| | - Kangsheng Li
- Department of Microbiology and Immunology, Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| |
Collapse
|
26
|
Wu S, Zhao W, Ji S. Real-time dynamic simulation for highly accurate spatiotemporal brain deformation from impact. COMPUTER METHODS IN APPLIED MECHANICS AND ENGINEERING 2022; 394:114913. [PMID: 35572209 PMCID: PMC9097909 DOI: 10.1016/j.cma.2022.114913] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Real-time dynamic simulation remains a significant challenge for spatiotemporal data of high dimension and resolution. In this study, we establish a transformer neural network (TNN) originally developed for natural language processing and a separate convolutional neural network (CNN) to estimate five-dimensional (5D) spatiotemporal brain-skull relative displacement resulting from impact (isotropic spatial resolution of 4 mm with temporal resolution of 1 ms). Sequential training is applied to train (N = 5184 samples) the two neural networks for estimating the complete 5D displacement across a temporal duration of 60 ms. We find that TNN slightly but consistently outperforms CNN in accuracy for both displacement and the resulting voxel-wise four-dimensional (4D) maximum principal strain (e.g., root mean squared error (RMSE) of ~1.0% vs. ~1.6%, with coefficient of determination, R 2 >0.99 vs. >0.98, respectively, and normalized RMSE (NRMSE) at peak displacement of 2%-3%, based on an independent testing dataset; N = 314). Their accuracies are similar for a range of real-world impacts drawn from various published sources (dummy, helmet, football, soccer, and car crash; average RMSE/NRMSE of ~0.3 mm/~4%-5% and average R 2 of ~0.98 at peak displacement). Sequential training is effective for allowing instantaneous estimation of 5D displacement with high accuracy, although TNN poses a heavier computational burden in training. This work enables efficient characterization of the intrinsically dynamic brain strain in impact critical for downstream multiscale axonal injury model simulation. This is also the first application of TNN in biomechanics, which offers important insight into how real-time dynamic simulations can be achieved across diverse engineering fields.
Collapse
Affiliation(s)
- Shaoju Wu
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, United States of America
| | - Wei Zhao
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, United States of America
| | - Songbai Ji
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, United States of America
- Department of Mechanical Engineering, Worcester Polytechnic Institute, Worcester, MA, United States of America
| |
Collapse
|
27
|
Kim C, Choi WJ, Kang W. Cavitation nucleation and its ductile-to-brittle shape transition in soft gels under translational mechanical impact. Acta Biomater 2022; 142:160-173. [PMID: 35189381 DOI: 10.1016/j.actbio.2022.02.017] [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: 11/08/2021] [Revised: 02/08/2022] [Accepted: 02/14/2022] [Indexed: 02/05/2023]
Abstract
Cavitation bubbles in the human body, when subjected to impact, are being increasingly considered as a possible brain injury mechanism. However, the onset of cavitation and its complex dynamics in biological materials remain unclear. Our experimental results using soft gels as a tissue simulant show that the critical acceleration (acr) at cavitation nucleation monotonically increases with increasing stiffness of gelatin A/B, while acr for agarose and agar initially increases but is followed by a plateau or even decrease after stiffness reach to ∼100 kPa. Our image analyses of cavitation bubbles and theoretical work reveal that the observed trends in acr are directly linked to how bubbles grow in each gel. Gelatin A/B, regardless of their stiffness, form a localized damaged zone (tens of nanometers) at the gel-bubble interface during bubble growth. In contrary, the damaged zone in agar/agarose becomes significantly larger (> 100 times) with increasing shear modulus, which triggers the transition from formation of a small, damaged zone to activation of crack propagation. STATEMENT OF SIGNIFICANCE: We have studied cavitation nucleation and bubble growth in four different types of soft gels (i.e., tissue simulants) under translational impact. The critical linear acceleration for cavitation nucleation has been measured in the simulants by utilizing a recently developed method that mimics acceleration profiles of typical head blunt events. Each gel type exhibits significantly different trends in the critical acceleration and bubble shape (e.g., A gel-specific sphere-to-saucer transition) with increasing gel stiffness. Our theoretical framework, based on the concepts of a damaged zone and crack propagation in each gel, explains underlying mechanisms of the experimental observations. Our in-depth studies shed light on potential links between traumatic brain injuries and cavitation bubbles induced by translational acceleration, the overlooked mechanism in the literature.
Collapse
Affiliation(s)
- Chunghwan Kim
- Mechanical Engineering, School for Engineering of Matter, Transport and Energy, Arizona State University, Tempe, AZ 85281, United States
| | - Won June Choi
- Mechanical Engineering, School for Engineering of Matter, Transport and Energy, Arizona State University, Tempe, AZ 85281, United States
| | - Wonmo Kang
- Mechanical Engineering, School for Engineering of Matter, Transport and Energy, Arizona State University, Tempe, AZ 85281, United States.
| |
Collapse
|
28
|
Li WY, Fu XM, Wang ZD, Li ZG, Ma D, Sun P, Liu GB, Zhu XF, Wang Y. Krüppel-like factor 7 attenuates hippocampal neuronal injury after traumatic brain injury. Neural Regen Res 2022; 17:661-672. [PMID: 34380908 PMCID: PMC8504401 DOI: 10.4103/1673-5374.320991] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/20/2021] [Accepted: 05/06/2021] [Indexed: 11/25/2022] Open
Abstract
Our previous study has shown that the transcription factor Krüppel-like factor 7 (KLF7) promotes peripheral nerve regeneration and motor function recovery after spinal cord injury. KLF7 also participates in traumatic brain injury, but its regulatory mechanisms remain poorly understood. In the present study, an HT22 cell model of traumatic brain injury was established by stretch injury and oxygen-glucose deprivation. These cells were then transfected with an adeno-associated virus carrying KLF7 (AAV-KLF7). The results revealed that, after stretch injury and oxygen-glucose deprivation, KLF7 greatly reduced apoptosis, activated caspase-3 and lactate dehydrogenase, downregulated the expression of the apoptotic markers B-cell lymphoma 2 (Bcl-2)-associated X protein (Bax) and cleaved caspase-3, and increased the expression of βIII-tubulin and the antiapoptotic marker Bcl-2. Furthermore, KLF7 overexpression upregulated Janus kinase 2 (JAK2) and signal transducer and activator of transcription 3 (STAT3) phosphorylation in HT22 cells treated by stretch injury and oxygen-glucose deprivation. Immunoprecipitation assays revealed that KLF7 directly participated in the phosphorylation of STAT3. In addition, treatment with AG490, a selective inhibitor of JAK2/STAT3, weakened the protective effects of KLF7. A mouse controlled cortical impact model of traumatic brain injury was then established. At 30 minutes before modeling, AAV-KLF7 was injected into the ipsilateral lateral ventricle. The protein and mRNA levels of KLF7 in the hippocampus were increased at 1 day after injury and recovered to normal levels at 3 days after injury. KLF7 reduced ipsilateral hippocampal atrophy, decreased the injured cortex volume, downregulated Bax and cleaved caspase-3 expression, and increased the number of 5-bromo-2'-deoxyuridine-positive neurons and Bcl-2 protein expression. Moreover, KLF7 transfection greatly enhanced the phosphorylation of JAK2 and STAT3 in the ipsilateral hippocampus. These results suggest that KLF7 may protect hippocampal neurons after traumatic brain injury through activation of the JAK2/STAT3 signaling pathway. The study was approved by the Institutional Review Board of Mudanjiang Medical University, China (approval No. mdjyxy-2018-0012) on March 6, 2018.
Collapse
Affiliation(s)
- Wen-Yuan Li
- Institute of Neural Tissue Engineering, Mudanjiang Medical University, Mudanjiang, Heilongjiang Province, China
| | - Xiu-Mei Fu
- Department of Anatomy, College of Basic Medical Sciences, Chengde Medical University, Chengde, Hebei Province, China
- Hebei Key Laboratory of Nerve Injury and Repair, Chengde Medical University, Chengde, Hebei Province, China
| | - Zhen-Dong Wang
- Department of Otorhinolaryngology, Mudanjiang City Second People’s Hospital, Mudanjiang, Heilongjiang Province, China
| | - Zhi-Gang Li
- The First Department of General Surgery, Hongqi Hospital, Mudanjiang Medical University, Mudanjiang, Heilongjiang Province, China
| | - Duo Ma
- Institute of Neural Tissue Engineering, Mudanjiang Medical University, Mudanjiang, Heilongjiang Province, China
| | - Ping Sun
- Institute of Neural Tissue Engineering, Mudanjiang Medical University, Mudanjiang, Heilongjiang Province, China
| | - Gui-Bo Liu
- Institute of Neural Tissue Engineering, Mudanjiang Medical University, Mudanjiang, Heilongjiang Province, China
| | - Xiao-Feng Zhu
- Institute of Neural Tissue Engineering, Mudanjiang Medical University, Mudanjiang, Heilongjiang Province, China
| | - Ying Wang
- Institute of Neural Tissue Engineering, Mudanjiang Medical University, Mudanjiang, Heilongjiang Province, China
| |
Collapse
|
29
|
Meyer LJ, Lotze FP, Riess ML. Simulated traumatic brain injury in in-vitro mouse neuronal and brain endothelial cell culture models. J Pharmacol Toxicol Methods 2022; 114:107159. [PMID: 35149185 PMCID: PMC11151826 DOI: 10.1016/j.vascn.2022.107159] [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: 09/13/2021] [Revised: 01/02/2022] [Accepted: 02/04/2022] [Indexed: 11/18/2022]
Abstract
Traumatic brain injury can lead to fatal outcomes such as disability and death. Every year, it affects many patients all over the world. Not only the primary ischemic event, but also the subsequent reperfusion can cause severe brain injury. This so-called ischemia/reperfusion injury combined with mechanical forces lead to cellular disruption. Hence, this paper describes a special in-vitro model, mimicking traumatic brain injury by combining both hypoxia/reoxygenation and compression to simulate ischemia/reperfusion injury as well as the mechanical effects that occur concurrently when suffering traumatic brain injury. Through this approach, stroke, concussion, and traumatic brain injury can be studied on different cell lines in a simplified way. We used two primary mouse brain cell cultures, namely neurons and endothelial cells. Our results show that for the different cell types, different timelines of hypoxia and compression need to be explored to achieve the optimal amount of cellular damage in order to effectively mimic traumatic brain injury. Thus, this model will be useful to test potential treatments of brain injury in future in-vitro studies.
Collapse
Affiliation(s)
- Luise J Meyer
- Department of Anesthesiology, Vanderbilt University Medical Center, 1161 21(st) Avenue South, Nashville, TN 37232, USA; Department of Anesthesiology, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany
| | - Felicia P Lotze
- Department of Anesthesiology, Vanderbilt University Medical Center, 1161 21(st) Avenue South, Nashville, TN 37232, USA; Department of Anesthesiology, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany
| | - Matthias L Riess
- Department of Anesthesiology, Vanderbilt University Medical Center, 1161 21(st) Avenue South, Nashville, TN 37232, USA; Anesthesiology, TVHS VA Medical Center, 1310 24(th) Ave South, Nashville, TN 37212, USA; Department of Pharmacology, Vanderbilt University, 465 21(st) Avenue South, Nashville, TN 37232, USA.
| |
Collapse
|
30
|
Srinivasan G, Brafman DA. The Emergence of Model Systems to Investigate the Link Between Traumatic Brain Injury and Alzheimer's Disease. Front Aging Neurosci 2022; 13:813544. [PMID: 35211003 PMCID: PMC8862182 DOI: 10.3389/fnagi.2021.813544] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 12/20/2021] [Indexed: 12/12/2022] Open
Abstract
Numerous epidemiological studies have demonstrated that individuals who have sustained a traumatic brain injury (TBI) have an elevated risk for developing Alzheimer's disease and Alzheimer's-related dementias (AD/ADRD). Despite these connections, the underlying mechanisms by which TBI induces AD-related pathology, neuronal dysfunction, and cognitive decline have yet to be elucidated. In this review, we will discuss the various in vivo and in vitro models that are being employed to provide more definite mechanistic relationships between TBI-induced mechanical injury and AD-related phenotypes. In particular, we will highlight the strengths and weaknesses of each of these model systems as it relates to advancing the understanding of the mechanisms that lead to TBI-induced AD onset and progression as well as providing platforms to evaluate potential therapies. Finally, we will discuss how emerging methods including the use of human induced pluripotent stem cell (hiPSC)-derived cultures and genome engineering technologies can be employed to generate better models of TBI-induced AD.
Collapse
Affiliation(s)
| | - David A. Brafman
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, United States
| |
Collapse
|
31
|
Zhao W, Ji S. Cerebral vascular strains in dynamic head impact using an upgraded model with brain material property heterogeneity. J Mech Behav Biomed Mater 2022; 126:104967. [PMID: 34863650 PMCID: PMC8792345 DOI: 10.1016/j.jmbbm.2021.104967] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/27/2021] [Accepted: 11/06/2021] [Indexed: 02/03/2023]
Abstract
Cerebral vascular injury (CVI) is a frequent consequence of traumatic brain injury but has often been neglected. Substantial experimental work exists on vascular material properties and failure/subfailure thresholds. However, little is known about vascular in vivo loading conditions in dynamic head impact, which is necessary to investigate the risk, severity, and extent of CVI. In this study, we resort to the Worcester Head Injury Model (WHIM) V2.1 for investigation. The model embeds the cerebral vasculature network and is further upgraded to incorporate brain material property heterogeneity based on magnetic resonance elastography. The brain material property is calibrated to match with the previously validated anisotropic V1.0 version in terms of whole-brain strains against six experimental datasets of a wide range of blunt impact conditions. The upgraded WHIM is finally used to simulate five representative real-world head impacts drawn from contact sports and automotive crashes. We find that peak strains in veins are considerably higher than those in arteries and that peak circumferential strains are also higher than peak axial strains. For a typical concussive head impact, cerebral vascular axial strains reach the lowest reported yield strain of ∼7-8%. For severe automotive impacts, axial strains could reach ∼20%, which is on the order of the lowest reported ultimate failure strain of ∼24%. These results suggest in vivo mechanical loading conditions of the cerebral vasculature (excluding bridging veins not assessed here) due to rapid head rotation are at the lower end of failure/subfailure thresholds established from ex vivo experiments. This study provides some first insight into the risk, severity, and extent of CVI in real-world head impacts.
Collapse
Affiliation(s)
- Wei Zhao
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA
| | - Songbai Ji
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, Department of Mechanical Engineering, Worcester Polytechnic Institute, Worcester, MA,Corresponding author: Dr. Songbai Ji, 60 Prescott Street, Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA 01506, USA, ; (508) 831-4956
| |
Collapse
|
32
|
|
33
|
Streubel-Gallasch L, Zyśk M, Beretta C, Erlandsson A. Traumatic brain injury in the presence of Aβ pathology affects neuronal survival, glial activation and autophagy. Sci Rep 2021; 11:22982. [PMID: 34837024 PMCID: PMC8626479 DOI: 10.1038/s41598-021-02371-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 11/15/2021] [Indexed: 11/09/2022] Open
Abstract
Traumatic brain injury (TBI) presents a widespread health problem in the elderly population. In addition to the acute injury, epidemiological studies have observed an increased probability and earlier onset of dementias in the elderly following TBI. However, the underlying mechanisms of the connection between TBI and Alzheimer's disease in the aged brain and potential exacerbating factors is still evolving. The aim of this study was to investigate cellular injury-induced processes in the presence of amyloid β (Aβ) pathology. For this purpose, a co-culture system of cortical stem-cell derived astrocytes, neurons and oligodendrocytes were exposed to Aβ42 protofibrils prior to a mechanically induced scratch injury. Cellular responses, including neurodegeneration, glial activation and autophagy was assessed by immunoblotting, immunocytochemistry, ELISA and transmission electron microscopy. Our results demonstrate that the combined burden of Aβ exposure and experimental TBI causes a decline in the number of neurons, the differential expression of the key astrocytic markers glial fibrillary acidic protein and S100 calcium-binding protein beta, mitochondrial alterations and prevents the upregulation of autophagy. Our study provides valuable information about the impact of TBI sustained in the presence of Aβ deposits and helps to advance the understanding of geriatric TBI on the cellular level.
Collapse
Affiliation(s)
- Linn Streubel-Gallasch
- Department of Public Health and Caring Sciences/Molecular Geriatrics, Rudbeck Laboratory, Uppsala University, 751 85, Uppsala, Sweden
| | - Marlena Zyśk
- Department of Public Health and Caring Sciences/Molecular Geriatrics, Rudbeck Laboratory, Uppsala University, 751 85, Uppsala, Sweden
| | - Chiara Beretta
- Department of Public Health and Caring Sciences/Molecular Geriatrics, Rudbeck Laboratory, Uppsala University, 751 85, Uppsala, Sweden
| | - Anna Erlandsson
- Department of Public Health and Caring Sciences/Molecular Geriatrics, Rudbeck Laboratory, Uppsala University, 751 85, Uppsala, Sweden.
| |
Collapse
|
34
|
Mechanical Stretching-Induced Traumatic Brain Injury Is Mediated by the Formation of GSK-3β-Tau Complex to Impair Insulin Signaling Transduction. Biomedicines 2021; 9:biomedicines9111650. [PMID: 34829879 PMCID: PMC8615493 DOI: 10.3390/biomedicines9111650] [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: 09/16/2021] [Revised: 11/05/2021] [Accepted: 11/06/2021] [Indexed: 11/26/2022] Open
Abstract
Traumatic brain injury confers a significant and growing public health burden. It is a major environmental risk factor for dementia. Nonetheless, the mechanism by which primary mechanical injury leads to neurodegeneration and an increased risk of dementia-related diseases is unclear. Thus, we aimed to investigate the effect of stretching on SH-SY5Y neuroblastoma cells that proliferate in vitro. These cells retain the dopamine-β-hydroxylase activity, thus being suitable for neuromechanistic studies. SH-SY5Y cells were cultured on stretchable membranes. The culture conditions contained two groups, namely non-stretched (control) and stretched. They were subjected to cyclic stretching (6 and 24 h) and 25% elongation at 1 Hz. Following stretching at 25% and 1 Hz for 6 h, the mechanical injury changed the mitochondrial membrane potential and triggered oxidative DNA damage at 24 h. Stretching decreased the level of brain-derived neurotrophic factors and increased amyloid-β, thus indicating neuronal stress. Moreover, the mechanical injury downregulated the insulin pathway and upregulated glycogen synthase kinase 3β (GSK-3β)S9/p-Tau protein levels, which caused a neuronal injury. Following 6 and 24 h of stretching, GSK-3βS9 was directly bound to p-TauS396. In contrast, the neuronal injury was improved using GSK-3β inhibitor TWS119, which downregulated amyloid-β/p-Taus396 phosphorylation by enhancing ERK1/2T202/Y204 and AktS473 phosphorylation. Our findings imply that the neurons were under stress and that the inactivation of the GSK3β could alleviate this defect.
Collapse
|
35
|
Shoemaker AR, Jones IE, Jeffris KD, Gabrielli G, Togliatti AG, Pichika R, Martin E, Kiskinis E, Franz CK, Finan J. Biofidelic dynamic compression of human cortical spheroids reproduces neurotrauma phenotypes. Dis Model Mech 2021; 14:273823. [PMID: 34746950 PMCID: PMC8713991 DOI: 10.1242/dmm.048916] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 11/02/2021] [Indexed: 11/20/2022] Open
Abstract
Fundamental questions about patient heterogeneity and human-specific pathophysiology currently obstruct progress towards a therapy for traumatic brain injury (TBI). Human in vitro models have the potential to address these questions. 3D spheroidal cell culture protocols for human-origin neural cells have several important advantages over their 2D monolayer counterparts. Three dimensional spheroidal cultures may mature more quickly, develop more biofidelic electrophysiological activity and/or reproduce some aspects of brain architecture. Here, we present the first human in vitro model of non-penetrating TBI employing 3D spheroidal cultures. We used a custom-built device to traumatize these spheroids in a quantifiable, repeatable and biofidelic manner and correlated the heterogeneous, mechanical strain field with the injury phenotype. Trauma reduced cell viability, mitochondrial membrane potential and spontaneous, synchronous, electrophysiological activity in the spheroids. Electrophysiological deficits emerged at lower injury severities than changes in cell viability. Also, traumatized spheroids secreted lactate dehydrogenase, a marker of cell damage, and neurofilament light chain, a promising clinical biomarker of neurotrauma. These results demonstrate that 3D human in vitro models can reproduce important phenotypes of neurotrauma in vitro.
Collapse
Affiliation(s)
- Aaron R Shoemaker
- Department of Neurosurgery, NorthShore University Health System, Evanston, IL, USA
| | - Ian E Jones
- Department of Mechanical and Industrial Engineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Kira D Jeffris
- Department of Mechanical and Industrial Engineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Gina Gabrielli
- Department of Neurosurgery, NorthShore University Health System, Evanston, IL, USA
| | | | - Rajeswari Pichika
- Department of Physical Medicine and Rehabilitation, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Eric Martin
- Department of Physical Medicine and Rehabilitation, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Evangelos Kiskinis
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Colin K Franz
- Shirley Ryan AbilityLab, Chicago, IL, USA.,Department of Physical Medicine and Rehabilitation, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - John Finan
- Department of Mechanical and Industrial Engineering, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
36
|
Subramaniam DR, Unnikrishnan G, Sundaramurthy A, Rubio JE, Kote VB, Reifman J. Cerebral Vasculature Influences Blast-Induced Biomechanical Responses of Human Brain Tissue. Front Bioeng Biotechnol 2021; 9:744808. [PMID: 34805106 PMCID: PMC8599150 DOI: 10.3389/fbioe.2021.744808] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 10/18/2021] [Indexed: 11/13/2022] Open
Abstract
Multiple finite-element (FE) models to predict the biomechanical responses in the human brain resulting from the interaction with blast waves have established the importance of including the brain-surface convolutions, the major cerebral veins, and using non-linear brain-tissue properties to improve model accuracy. We hypothesize that inclusion of a more detailed network of cerebral veins and arteries can further enhance the model-predicted biomechanical responses and help identify correlates of blast-induced brain injury. To more comprehensively capture the biomechanical responses of human brain tissues to blast-wave exposure, we coupled a three-dimensional (3-D) detailed-vasculature human-head FE model, previously validated for blunt impact, with a 3-D shock-tube FE model. Using the coupled model, we computed the biomechanical responses of a human head facing an incoming blast wave for blast overpressures (BOPs) equivalent to 68, 83, and 104 kPa. We validated our FE model, which includes the detailed network of cerebral veins and arteries, the gyri and the sulci, and hyper-viscoelastic brain-tissue properties, by comparing the model-predicted intracranial pressure (ICP) values with previously collected data from shock-tube experiments performed on cadaver heads. In addition, to quantify the influence of including a more comprehensive network of brain vessels, we compared the biomechanical responses of our detailed-vasculature model with those of a reduced-vasculature model and a no-vasculature model for the same blast-loading conditions. For the three BOPs, the predicted ICP values matched well with the experimental results in the frontal lobe, with peak-pressure differences of 4-11% and phase-shift differences of 9-13%. As expected, incorporating the detailed cerebral vasculature did not influence the ICP, however, it redistributed the peak brain-tissue strains by as much as 30% and yielded peak strain differences of up to 7%. When compared to existing reduced-vasculature FE models that only include the major cerebral veins, our high-fidelity model redistributed the brain-tissue strains in most of the brain, highlighting the importance of including a detailed cerebral vessel network in human-head FE models to more comprehensively account for the biomechanical responses induced by blast exposure.
Collapse
Affiliation(s)
- Dhananjay Radhakrishnan Subramaniam
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, United States Army Medical Research and Development Command, Fort Detrick, MD, United States
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Ginu Unnikrishnan
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, United States Army Medical Research and Development Command, Fort Detrick, MD, United States
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Aravind Sundaramurthy
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, United States Army Medical Research and Development Command, Fort Detrick, MD, United States
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Jose E. Rubio
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, United States Army Medical Research and Development Command, Fort Detrick, MD, United States
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Vivek Bhaskar Kote
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, United States Army Medical Research and Development Command, Fort Detrick, MD, United States
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Jaques Reifman
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, United States Army Medical Research and Development Command, Fort Detrick, MD, United States
| |
Collapse
|
37
|
Palmieri M, Frati A, Santoro A, Frati P, Fineschi V, Pesce A. Diffuse Axonal Injury: Clinical Prognostic Factors, Molecular Experimental Models and the Impact of the Trauma Related Oxidative Stress. An Extensive Review Concerning Milestones and Advances. Int J Mol Sci 2021; 22:ijms221910865. [PMID: 34639206 PMCID: PMC8509530 DOI: 10.3390/ijms221910865] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 10/05/2021] [Accepted: 10/06/2021] [Indexed: 12/21/2022] Open
Abstract
Traumatic brain injury (TBI) is a condition burdened by an extremely high rate of morbidity and mortality and can result in an overall disability rate as high as 50% in affected individuals. Therefore, the importance of identifying clinical prognostic factors for diffuse axonal injury (DAI) in (TBI) is commonly recognized as critical. The aim of the present review paper is to evaluate the most recent contributions from the relevant literature in order to understand how each single prognostic factor determinates the severity of the clinical syndrome associated with DAI. The main clinical factors with an important impact on prognosis in case of DAI are glycemia, early GCS, the peripheral oxygen saturation, blood pressure, and time to recover consciousness. In addition, the severity of the lesion, classified on the ground of the cerebral anatomical structures involved after the trauma, has a strong correlation with survival after DAI. In conclusion, modern findings concerning the role of reactive oxygen species (ROS) and oxidative stress in DAI suggest that biomarkers such as GFAP, pNF-H, NF-L, microtubule associated protein tau, Aβ42, S-100β, NSE, AQP4, Drp-1, and NCX represent a possible critical target for future pharmaceutical treatments to prevent the damages caused by DAI.
Collapse
Affiliation(s)
- Mauro Palmieri
- Neurosurgery Division, A.O.U. “Policlinico Umberto I”, Human Neuroscience Department, “Sapienza” University, Viale Del Policlinico 155, 00161 Rome, Italy; (A.F.); (A.S.)
- Correspondence: ; Tel.: +39-063-377-5298
| | - Alessandro Frati
- Neurosurgery Division, A.O.U. “Policlinico Umberto I”, Human Neuroscience Department, “Sapienza” University, Viale Del Policlinico 155, 00161 Rome, Italy; (A.F.); (A.S.)
- IRCCS “Neuromed”, Via Atinense 18, 86077 Pozzilli, Italy
| | - Antonio Santoro
- Neurosurgery Division, A.O.U. “Policlinico Umberto I”, Human Neuroscience Department, “Sapienza” University, Viale Del Policlinico 155, 00161 Rome, Italy; (A.F.); (A.S.)
| | - Paola Frati
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences SAIMLAL, “Sapienza” University, Viale Regina Elena 336, 00185 Rome, Italy; (P.F.); (V.F.)
| | - Vittorio Fineschi
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences SAIMLAL, “Sapienza” University, Viale Regina Elena 336, 00185 Rome, Italy; (P.F.); (V.F.)
| | - Alessandro Pesce
- Neurosurgery Division, Santa Maria Goretti Hospital, Via Lucia Scaravelli, 04100 Latina, Italy;
| |
Collapse
|
38
|
Ramirez S, Mukherjee A, Sepulveda S, Becerra-Calixto A, Bravo-Vasquez N, Gherardelli C, Chavez M, Soto C. Modeling Traumatic Brain Injury in Human Cerebral Organoids. Cells 2021; 10:2683. [PMID: 34685663 PMCID: PMC8534257 DOI: 10.3390/cells10102683] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/30/2021] [Accepted: 10/01/2021] [Indexed: 01/22/2023] Open
Abstract
Traumatic brain injury (TBI) is a head injury that disrupts the normal brain structure and function. TBI has been extensively studied using various in vitro and in vivo models. Most of the studies have been done with rodent models, which may respond differently to TBI than human nerve cells. Taking advantage of the recent development of cerebral organoids (COs) derived from human induced pluripotent stem cells (iPSCs), which resemble the architecture of specific human brain regions, here, we adapted the controlled cortical impact (CCI) model to induce TBI in human COs as a novel in vitro platform. To adapt the CCI procedure into COs, we have developed a phantom brain matrix, matching the mechanical characteristics of the brain, altogether with an empty mouse skull as a platform to allow the use of the stereotactic CCI equipment on COs. After the CCI procedure, COs were histologically prepared to evaluate neurons and astrocyte populations using the microtubule-associated protein 2 (MAP2) and the glial fibrillary acidic protein (GFAP). Moreover, a marker of metabolic response, the neuron-specific enolase (NSE), and cellular death using cleaved caspase 3 were also analyzed. Our results show that human COs recapitulate the primary pathological changes of TBI, including metabolic alterations related to neuronal damage, neuronal loss, and astrogliosis. This novel approach using human COs to model TBI in vitro holds great potential and opens new alternatives for understanding brain abnormalities produced by TBI, and for the development and testing of new therapeutic approaches.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Claudio Soto
- Mitchell Center for Alzheimer’s Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School, University of Texas Health Science at Houston, Houston, TX 77030, USA; (S.R.); (A.M.); (S.S.); (A.B.-C.); (N.B.-V.); (C.G.); (M.C.)
| |
Collapse
|
39
|
Rationally designed drug delivery systems for the local treatment of resected glioblastoma. Adv Drug Deliv Rev 2021; 177:113951. [PMID: 34461201 DOI: 10.1016/j.addr.2021.113951] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/26/2021] [Accepted: 08/24/2021] [Indexed: 02/08/2023]
Abstract
Glioblastoma (GBM) is a particularly aggressive brain cancer associated with high recurrence and poor prognosis. The standard of care, surgical resection followed by concomitant radio- and chemotherapy, leads to low survival rates. The local delivery of active agents within the tumor resection cavity has emerged as an attractive means to initiate oncological treatment immediately post-surgery. This complementary approach bypasses the blood-brain barrier, increases the local concentration at the tumor site while reducing or avoiding systemic side effects. This review will provide a global overview on the local treatment for GBM with an emphasis on the lessons learned from past clinical trials. The main parameters to be considered to rationally design fit-of-purpose biomaterials and develop drug delivery systems for local administration in the GBM resection cavity to prevent the tumor recurrence will be described. The intracavitary local treatment of GBM should i) use materials that facilitate translation to the clinic; ii) be characterized by easy GMP effective scaling up and easy-handling application by the neurosurgeons; iii) be adaptable to fill the tumor-resected niche, mold to the resection cavity or adhere to the exposed brain parenchyma; iv) be biocompatible and possess mechanical properties compatible with the brain; v) deliver a therapeutic dose of rationally-designed or repurposed drug compound(s) into the GBM infiltrative margin. Proof of concept with high translational potential will be provided. Finally, future perspectives to facilitate the clinical translation of the local perisurgical treatment of GBM will be discussed.
Collapse
|
40
|
In vitro model of traumatic brain injury to screen neuro-regenerative biomaterials. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 128:112253. [PMID: 34474815 DOI: 10.1016/j.msec.2021.112253] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 05/21/2021] [Accepted: 06/11/2021] [Indexed: 11/24/2022]
Abstract
Penetrating traumatic brain injury (pTBI) causes serious neurological deficits with no clinical regenerative therapies currently available. Tissue engineering strategies using biomaterial-based 'structural bridges' offer high potential to promote neural regeneration post-injury. This includes surgical grade materials which can be repurposed as biological scaffolds to overcome challenges associated with long approval processes and scaleup for human application. However, high throughput, pathomimetic models of pTBI are lacking for the developmental testing of such neuro-materials, representing a bottleneck in this rapidly emergent field. We have established a high throughput and facile culture model containing the major neural cell types which govern biomaterial handling in the central nervous system. We show that induction of traumatic injuries was feasible in the model, with post-injury implantation of a surgical grade biomaterial. Cellular imaging in lesions was achievable using standard epifluorescence microscopy methods. Key pathological features of pTBI were evident in vitro namely immune cell infiltration of lesions/biomaterial, with responses characteristic of cell scarring, namely hypertrophic astrocytes with GFAP upregulation. Based on our observations, we consider the high-throughput, inexpensive and facile pTBI model can be used to study biomaterial 'implantation' and evaluate neural cell-biomaterial responses. The model is highly versatile to test a range of laboratory and clinical grade materials for neural regeneration.
Collapse
|
41
|
Wu YH, Rosset S, Lee TR, Dragunow M, Park T, Shim V. In Vitro Models of Traumatic Brain Injury: A Systematic Review. J Neurotrauma 2021; 38:2336-2372. [PMID: 33563092 DOI: 10.1089/neu.2020.7402] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Traumatic brain injury (TBI) is a major public health challenge that is also the third leading cause of death worldwide. It is also the leading cause of long-term disability in children and young adults worldwide. Despite a large body of research using predominantly in vivo and in vitro rodent models of brain injury, there is no medication that can reduce brain damage or promote brain repair mainly due to our lack of understanding in the mechanisms and pathophysiology of the TBI. The aim of this review is to examine in vitro TBI studies conducted from 2008-2018 to better understand the TBI in vitro model available in the literature. Specifically, our focus was to perform a detailed analysis of the in vitro experimental protocols used and their subsequent biological findings. Our review showed that the uniaxial stretch is the most frequently used way of load application, accounting for more than two-thirds of the studies reviewed. The rate and magnitude of the loading were varied significantly from study to study but can generally be categorized into mild, moderate, and severe injuries. The in vitro studies reviewed here examined key processes in TBI pathophysiology such as membrane disruptions leading to ionic dysregulation, inflammation, and the subsequent damages to the microtubules and axons, as well as cell death. Overall, the studies examined in this review contributed to the betterment of our understanding of TBI as a disease process. Yet, our review also revealed the areas where more work needs to be done such as: 1) diversification of load application methods that will include complex loading that mimics in vivo head impacts; 2) more widespread use of human brain cells, especially patient-matched human cells in the experimental set-up; and 3) need for building a more high-throughput system to be able to discover effective therapeutic targets for TBI.
Collapse
Affiliation(s)
- Yi-Han Wu
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
- Center for Brain Research, The University of Auckland, Auckland, New Zealand
| | - Samuel Rosset
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Tae-Rin Lee
- Advanced Institute of Convergence Technology, Seoul National University, Seoul, Korea
| | - Mike Dragunow
- Center for Brain Research, The University of Auckland, Auckland, New Zealand
- Department of Pharmacology, The University of Auckland, Auckland, New Zealand
| | - Thomas Park
- Center for Brain Research, The University of Auckland, Auckland, New Zealand
- Department of Pharmacology, The University of Auckland, Auckland, New Zealand
| | - Vickie Shim
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
42
|
Lotze FP, Riess ML. Poloxamer 188 Exerts Direct Protective Effects on Mouse Brain Microvascular Endothelial Cells in an In Vitro Traumatic Brain Injury Model. Biomedicines 2021; 9:1043. [PMID: 34440247 PMCID: PMC8393826 DOI: 10.3390/biomedicines9081043] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/13/2021] [Accepted: 08/16/2021] [Indexed: 12/04/2022] Open
Abstract
Traumatic Brain Injury (TBI), the main contributor to morbidity and mortality worldwide, can disrupt the cell membrane integrity of the vascular endothelial system, endangering blood-brain barrier function and threatening cellular subsistence. Protection of the vascular endothelial system might enhance clinical outcomes after TBI. Poloxamer 188 (P188) has been shown to improve neuronal function after ischemia/reperfusion (I/R) injury as well as after TBI. We aimed to establish an in vitro compression-type TBI model, comparing mild-to-moderate and severe injury, to observe the direct effects of P188 on Mouse Brain Microvascular Endothelial Cells (MBEC). Confluent MBEC were exposed to normoxic or hypoxic conditions for either 5 or 15 h (hours). 1 h compression was added, and P188 was administered during 2 h reoxygenation. A direct effect of P188 on MBEC was tested by assessing cell number/viability, cytotoxicity/membrane damage, metabolic activity, and total nitric oxide production (tNOp). While P188 enhanced cell number/viability, metabolic activity, and tNOp, an increase in cytotoxicity/membrane damage after mild-to-moderate injury was prevented. In severely injured MBEC, P188 improved metabolic activity only. P188, present during reoxygenation, influenced MBEC function directly in simulated I/R and compression-type TBI.
Collapse
Affiliation(s)
- Felicia P. Lotze
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
- Department of Anesthesiology, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Matthias L. Riess
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
- Anesthesiology, TVHS VA Medical Center, Nashville, TN 37212, USA
| |
Collapse
|
43
|
Braun NJ, Liao D, Alford PW. Orientation of neurites influences severity of mechanically induced tau pathology. Biophys J 2021; 120:3272-3282. [PMID: 34293301 PMCID: PMC8392125 DOI: 10.1016/j.bpj.2021.07.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 06/11/2021] [Accepted: 07/13/2021] [Indexed: 01/03/2023] Open
Abstract
Chronic traumatic encephalopathy is a neurodegenerative disease associated with repeated traumatic brain injury (TBI). Chronic traumatic encephalopathy is a tauopathy, in which cognitive decline is accompanied by the accumulation of neurofibrillary tangles of the protein tau in patients' brains. We recently found that mechanical force alone can induce tau mislocalization to dendritic spines and loss of synaptic function in in vitro neuronal cultures with random cell organization. However, in the brain, neurons are highly aligned, so here we aimed to determine how neuronal organization influences early-stage tauopathy caused by mechanical injury. Using microfabricated cell culture constructs to control the growth of neurites and an in vitro simulated TBI device to apply controlled mechanical deformation, we found that neuronal orientation with respect to the direction of a uniaxial high-strain-rate stretch injury influences the degree of tau pathology in injured neurons. We found that a mechanical stretch applied parallel to the neurite alignment induces greater mislocalization of tau proteins to dendritic spines than does a stretch with the same strain applied perpendicular to the neurites. Synaptic function, characterized by the amplitude of miniature excitatory postsynaptic currents, was similarly decreased in neurons with neurites aligned parallel to stretch, whereas in neurons aligned perpendicular to stretch, it had little to no functional loss. Experimental injury parameters (strain, strain rate, direction of stretch) were combined with a standard viscoelastic solid model to show that in our in vitro model, neurite work density during stretch correlates with tau mislocalization. These findings suggest that in a TBI, the magnitude of brain deformation is not wholly predictive of neurodegenerative consequences of TBI but that deformation relative to local neuronal architecture and the neurite mechanical energy during injury are better metrics for predicting trauma-induced tauopathy.
Collapse
Affiliation(s)
| | - Dezhi Liao
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota.
| | | |
Collapse
|
44
|
Zhou H, Hu L, Li J, Ruan W, Cao Y, Zhuang J, Xu H, Peng Y, Zhang Z, Xu C, Yu Q, Li Y, Dou Z, Hu J, Wu X, Yu X, Gu C, Cao S, Yan F, Chen G. AXL kinase-mediated astrocytic phagocytosis modulates outcomes of traumatic brain injury. J Neuroinflammation 2021; 18:154. [PMID: 34233703 PMCID: PMC8264993 DOI: 10.1186/s12974-021-02201-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 06/22/2021] [Indexed: 11/23/2022] Open
Abstract
Background Complex changes in the brain microenvironment following traumatic brain injury (TBI) can cause neurological impairments for which there are few efficacious therapeutic interventions. The reactivity of astrocytes is one of the keys to microenvironmental changes, such as neuroinflammation, but its role and the molecular mechanisms that underpin it remain unclear. Methods Male C57BL/6J mice were subjected to the controlled cortical impact (CCI) to develop a TBI model. The specific ligand of AXL receptor tyrosine kinase (AXL), recombinant mouse growth arrest-specific 6 (rmGas6) was intracerebroventricularly administered, and selective AXL antagonist R428 was intraperitoneally applied at 30 min post-modeling separately. Post-TBI assessments included neurobehavioral assessments, transmission electron microscopy, immunohistochemistry, and western blotting. Real-time polymerase chain reaction (RT-PCR), siRNA transfection, and flow cytometry were performed for mechanism assessments in primary cultured astrocytes. Results AXL is upregulated mainly in astrocytes after TBI and promotes astrocytes switching to a phenotype that exhibits the capability of ingesting degenerated neurons or debris. As a result, this astrocytic transformation promotes the limitation of neuroinflammation and recovery of neurological dysfunction. Pharmacological inhibition of AXL in astrocytes significantly decreased astrocytic phagocytosis both in vivo and in primary astrocyte cultures, in contrast to the effect of treatment with the rmGas6. AXL activates the signal transducer and activator of the transcription 1 (STAT1) pathway thereby further upregulating ATP-binding cassette transporter 1 (ABCA1). Moreover, the supernatant from GAS6-depleted BV2 cells induced limited enhancement of astrocytic phagocytosis in vitro. Conclusion Our work establishes the role of AXL in the transformation of astrocytes to a phagocytic phenotype via the AXL/STAT1/ABCA1 pathway which contributes to the separation of healthy brain tissue from injury-induced cell debris, further ameliorating neuroinflammation and neurological impairments after TBI. Collectively, our findings provide a potential therapeutic target for TBI. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02201-3.
Collapse
Affiliation(s)
- Hang Zhou
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road88th, Hangzhou, 310016, China
| | - Libin Hu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road88th, Hangzhou, 310016, China
| | - Jianru Li
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road88th, Hangzhou, 310016, China
| | - Wu Ruan
- Department of Burn and Plastic Surgery, Children's Hospital, Zhejiang University School of Medicine, No. 3333 Binsheng Road, Zhejiang, 310052, Hangzhou, China
| | - Yang Cao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road88th, Hangzhou, 310016, China
| | - Jianfeng Zhuang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road88th, Hangzhou, 310016, China
| | - Hangzhe Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road88th, Hangzhou, 310016, China
| | - Yucong Peng
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road88th, Hangzhou, 310016, China
| | - Zhongyuan Zhang
- Department of Neurosurgery, Children's Hospital, Zhejiang University School of Medicine, No. 3333 Binsheng Road, Zhejiang, 310052, Hangzhou, China
| | - Chaoran Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road88th, Hangzhou, 310016, China
| | - Qian Yu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road88th, Hangzhou, 310016, China
| | - Yin Li
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road88th, Hangzhou, 310016, China
| | - Zhangqi Dou
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road88th, Hangzhou, 310016, China
| | - Junwen Hu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road88th, Hangzhou, 310016, China
| | - Xinyan Wu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road88th, Hangzhou, 310016, China
| | - Xiaobo Yu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road88th, Hangzhou, 310016, China
| | - Chi Gu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road88th, Hangzhou, 310016, China
| | - Shenglong Cao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road88th, Hangzhou, 310016, China
| | - Feng Yan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road88th, Hangzhou, 310016, China.
| | - Gao Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road88th, Hangzhou, 310016, China.
| |
Collapse
|
45
|
Shi W, Dong P, Kuss MA, Gu L, Kievit F, Kim HJ, Duan B. Design and Evaluation of an In Vitro Mild Traumatic Brain Injury Modeling System Using 3D Printed Mini Impact Device on the 3D Cultured Human iPSC Derived Neural Progenitor Cells. Adv Healthc Mater 2021; 10:e2100180. [PMID: 33890428 PMCID: PMC8222191 DOI: 10.1002/adhm.202100180] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/15/2021] [Indexed: 12/13/2022]
Abstract
Despite significant progress in understanding the disease mechanism of traumatic brain injury (TBI), promising preclinical therapeutics have seldom been translated into successful clinical outcomes, partially because the model animals have physiological and functional differences in the central nervous system (CNS) compared to humans. Human relevant models are thus urgently required. Here, an in vitro mild TBI (mTBI) modeling system is reported based on 3D cultured human induced pluripotent stem cells (iPSC) derived neural progenitor cells (iPSC-NPCs) to evaluate consequences of single and repetitive mTBI using a 3D printed mini weight-drop impact device. Computational simulation is performed to understand the single/cumulative effects of weight-drop impact on the NPC differentiated neurospheres. Experimental results reveal that neurospheres show reactive astrogliosis and glial scar formation after repetitive (10 hits) mild impacts, while no astrocyte activation is found after one or two mild impacts. A 3D co-culture model of human microglia cells with neurospheres is further developed. It is found that astrocyte response is promoted even after two mild impacts, possibly caused by the chronic neuroinflammation after microglia activation. The in vitro mTBI modeling system recapitulates several hallmarks of the brain impact injury and might serve as a good platform for future drug screening.
Collapse
Affiliation(s)
- Wen Shi
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska, Medical Center, Omaha, NE, 68198, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska, Medical Center, Omaha, NE, 68198, USA
| | - Pengfei Dong
- Department of Biomedical and Chemical Engineering and Science, Florida Institute of Technology, Melbourne, FL, 32901, USA
| | - Mitchell A Kuss
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska, Medical Center, Omaha, NE, 68198, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska, Medical Center, Omaha, NE, 68198, USA
| | - Linxia Gu
- Department of Biomedical and Chemical Engineering and Science, Florida Institute of Technology, Melbourne, FL, 32901, USA
| | - Forrest Kievit
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA
| | - Hyung Joon Kim
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska, Medical Center, Omaha, NE, 68198, USA
- Eppley Institute, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Bin Duan
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska, Medical Center, Omaha, NE, 68198, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska, Medical Center, Omaha, NE, 68198, USA
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Department of Mechanical Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| |
Collapse
|
46
|
Meyer LJ, Riess ML. Evaluation of In Vitro Neuronal Protection by Postconditioning with Poloxamer 188 Following Simulated Traumatic Brain Injury. Life (Basel) 2021; 11:316. [PMID: 33917288 PMCID: PMC8067401 DOI: 10.3390/life11040316] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/16/2021] [Accepted: 03/29/2021] [Indexed: 12/16/2022] Open
Abstract
Traumatic brain injury (TBI) leads to morbidity and mortality worldwide. Reperfusion after ischemia adds detrimental injury to cells. Ischemia/reperfusion (I/R) injures cells in a variety of ways including cell membrane disruption. Hence, methods to improve endogenous membrane resealing capacity are crucial. Poloxamer (P) 188, an amphiphilic triblock copolymer, was found to be effective against I/R and mechanical injury in various experimental settings. The aim of this study was to establish an in vitro mouse neuronal TBI model and, further, to investigate if postconditioning with P188 directly interacts with neurons after compression and simulated I/R injury, when administered at the start of reoxygenation. Cellular function was assessed by cell number/viability, mitochondrial viability, membrane damage by lactated dehydrogenase (LDH) release and FM1-43 incorporation as well as apoptosis-activation by Caspase 3. Five hours hypoxia ± compression with 2 h reoxygenation proved to be a suitable model for TBI. Compared to normoxic cells not exposed to compression, cell number and mitochondrial viability decreased, whereas membrane injury by LDH release/FM1-43 dye incorporation and Caspase 3 activity increased in cells exposed to hypoxic conditions with compression followed by reoxygenation. P188 did not protect neurons from simulated I/R and/or compression injury. Future research is indicated.
Collapse
Affiliation(s)
- Luise J. Meyer
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Anesthesiology, University Medicine Greifswald, 17475 Greifswald, Germany;
| | - Matthias L. Riess
- Anesthesiology, TVHS VA Medical Center, Nashville, TN 37212, USA; Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
47
|
The Role of BDNF in Experimental and Clinical Traumatic Brain Injury. Int J Mol Sci 2021; 22:ijms22073582. [PMID: 33808272 PMCID: PMC8037220 DOI: 10.3390/ijms22073582] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/23/2021] [Accepted: 03/26/2021] [Indexed: 02/07/2023] Open
Abstract
Traumatic brain injury is one of the leading causes of mortality and morbidity in the world with no current pharmacological treatment. The role of BDNF in neural repair and regeneration is well established and has also been the focus of TBI research. Here, we review experimental animal models assessing BDNF expression following injury as well as clinical studies in humans including the role of BDNF polymorphism in TBI. There is a large heterogeneity in experimental setups and hence the results with different regional and temporal changes in BDNF expression. Several studies have also assessed different interventions to affect the BDNF expression following injury. Clinical studies highlight the importance of BDNF polymorphism in the outcome and indicate a protective role of BDNF polymorphism following injury. Considering the possibility of affecting the BDNF pathway with available substances, we discuss future studies using transgenic mice as well as iPSC in order to understand the underlying mechanism of BDNF polymorphism in TBI and develop a possible pharmacological treatment.
Collapse
|
48
|
Zhou Z, Domel AG, Li X, Grant G, Kleiven S, Camarillo D, Zeineh M. White Matter Tract-Oriented Deformation Is Dependent on Real-Time Axonal Fiber Orientation. J Neurotrauma 2021; 38:1730-1745. [PMID: 33446060 DOI: 10.1089/neu.2020.7412] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Traumatic axonal injury (TAI) is a critical public health issue with its pathogenesis remaining largely elusive. Finite element (FE) head models are promising tools to bridge the gap between mechanical insult, localized brain response, and resultant injury. In particular, the FE-derived deformation along the direction of white matter (WM) tracts (i.e., tract-oriented strain) has been shown to be an appropriate predictor for TAI. The evolution of fiber orientation in time during the impact and its potential influence on the tract-oriented strain remains unknown, however. To address this question, the present study leveraged an embedded element approach to track real-time fiber orientation during impacts. A new scheme to calculate the tract-oriented strain was proposed by projecting the strain tensors from pre-computed simulations along the temporal fiber direction instead of its static counterpart directly obtained from diffuse tensor imaging. The results revealed that incorporating the real-time fiber orientation not only altered the direction but also amplified the magnitude of the tract-oriented strain, resulting in a generally more extended distribution and a larger volume ratio of WM exposed to high deformation along fiber tracts. These effects were exacerbated with the impact severities characterized by the acceleration magnitudes. Results of this study provide insights into how best to incorporate fiber orientation in head injury models and derive the WM tract-oriented deformation from computational simulations, which is important for furthering our understanding of the underlying mechanisms of TAI.
Collapse
Affiliation(s)
- Zhou Zhou
- Department of Bioengineering, Stanford University, Stanford, California, USA
| | - August G Domel
- Department of Bioengineering, Stanford University, Stanford, California, USA
| | - Xiaogai Li
- Neuronic Engineering, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Gerald Grant
- Department of Neurosurgery, Stanford University, Stanford, California, USA.,Department of Neurology, Stanford University, Stanford, California, USA
| | - Svein Kleiven
- Neuronic Engineering, KTH Royal Institute of Technology, Stockholm, Sweden
| | - David Camarillo
- Department of Bioengineering, Stanford University, Stanford, California, USA.,Department of Neurosurgery, Stanford University, Stanford, California, USA.,Department of Mechanical Engineering, Stanford University, Stanford, California, USA
| | - Michael Zeineh
- Department of Radiology, Stanford University, Stanford, California, USA
| |
Collapse
|
49
|
Estrada JB, Cramer HC, Scimone MT, Buyukozturk S, Franck C. Neural cell injury pathology due to high-rate mechanical loading. BRAIN MULTIPHYSICS 2021. [DOI: 10.1016/j.brain.2021.100034] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
50
|
Nikolakopoulou P, Rauti R, Voulgaris D, Shlomy I, Maoz BM, Herland A. Recent progress in translational engineered in vitro models of the central nervous system. Brain 2020; 143:3181-3213. [PMID: 33020798 PMCID: PMC7719033 DOI: 10.1093/brain/awaa268] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 06/17/2020] [Accepted: 06/21/2020] [Indexed: 02/07/2023] Open
Abstract
The complexity of the human brain poses a substantial challenge for the development of models of the CNS. Current animal models lack many essential human characteristics (in addition to raising operational challenges and ethical concerns), and conventional in vitro models, in turn, are limited in their capacity to provide information regarding many functional and systemic responses. Indeed, these challenges may underlie the notoriously low success rates of CNS drug development efforts. During the past 5 years, there has been a leap in the complexity and functionality of in vitro systems of the CNS, which have the potential to overcome many of the limitations of traditional model systems. The availability of human-derived induced pluripotent stem cell technology has further increased the translational potential of these systems. Yet, the adoption of state-of-the-art in vitro platforms within the CNS research community is limited. This may be attributable to the high costs or the immaturity of the systems. Nevertheless, the costs of fabrication have decreased, and there are tremendous ongoing efforts to improve the quality of cell differentiation. Herein, we aim to raise awareness of the capabilities and accessibility of advanced in vitro CNS technologies. We provide an overview of some of the main recent developments (since 2015) in in vitro CNS models. In particular, we focus on engineered in vitro models based on cell culture systems combined with microfluidic platforms (e.g. 'organ-on-a-chip' systems). We delve into the fundamental principles underlying these systems and review several applications of these platforms for the study of the CNS in health and disease. Our discussion further addresses the challenges that hinder the implementation of advanced in vitro platforms in personalized medicine or in large-scale industrial settings, and outlines the existing differentiation protocols and industrial cell sources. We conclude by providing practical guidelines for laboratories that are considering adopting organ-on-a-chip technologies.
Collapse
Affiliation(s)
- Polyxeni Nikolakopoulou
- AIMES, Center for the Advancement of Integrated Medical and Engineering Sciences, Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Rossana Rauti
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Dimitrios Voulgaris
- Division of Micro and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Iftach Shlomy
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Ben M Maoz
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel
| | - Anna Herland
- AIMES, Center for the Advancement of Integrated Medical and Engineering Sciences, Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
- Division of Micro and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden
| |
Collapse
|