1
|
Dwyer MKR, Amelinez-Robles N, Polsfuss I, Herbert K, Kim C, Varghese N, Parry TJ, Buller B, Verdoorn TA, Billing CB, Morrison B. NTS-105 decreased cell death and preserved long-term potentiation in an in vitro model of moderate traumatic brain injury. Exp Neurol 2024; 371:114608. [PMID: 37949202 DOI: 10.1016/j.expneurol.2023.114608] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/27/2023] [Accepted: 11/05/2023] [Indexed: 11/12/2023]
Abstract
Traumatic brain injury (TBI) is a major cause of hospitalization and death. To mitigate these human costs, the search for effective drugs to treat TBI continues. In the current study, we evaluated the efficacy of the novel neurosteroid, NTS-105, to reduce post-traumatic pathobiology in an in vitro model of moderate TBI that utilizes an organotypic hippocampal slice culture. NTS-105 inhibited activation of the androgen receptor and the mineralocorticoid receptor, partially activated the progesterone B receptor and was not active at the glucocorticoid receptor. Treatment with NTS-105 starting one hour after injury decreased post-traumatic cell death in a dose-dependent manner, with 10 nM NTS-105 being most effective. Post-traumatic administration of 10 nM NTS-105 also prevented deficits in long-term potentiation (LTP) without adversely affecting neuronal activity in naïve cultures. We propose that the high potency pleiotropic action of NTS-105 beneficial effects at multiple receptors (e.g. androgen, mineralocorticoid and progesterone) provides significant mechanistic advantages over native neurosteroids such as progesterone, which lacked clinical success for the treatment of TBI. Our results suggest that this pleiotropic pharmacology may be a promising strategy for the effective treatment of TBI, and future studies should test its efficacy in pre-clinical animal models of TBI.
Collapse
Affiliation(s)
- Mary Kate R Dwyer
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States of America
| | - Nicolas Amelinez-Robles
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States of America
| | - Isabella Polsfuss
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States of America
| | - Keondre Herbert
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States of America
| | - Carolyn Kim
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States of America
| | - Nevin Varghese
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States of America
| | - Tom J Parry
- NeuroTrauma Sciences, LLC, Alpharetta, GA 30009, United States of America
| | - Benjamin Buller
- NeuroTrauma Sciences, LLC, Alpharetta, GA 30009, United States of America
| | - Todd A Verdoorn
- NeuroTrauma Sciences, LLC, Alpharetta, GA 30009, United States of America
| | - Clare B Billing
- BioPharmaWorks, LLC, Groton, CT 06340, United States of America
| | - Barclay Morrison
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States of America.
| |
Collapse
|
2
|
Pischiutta F, Cavaleiro H, Caruso E, Tribuzio F, Di Marzo N, Moro F, Kobeissy F, Wang KK, Salgado AJ, Zanier ER. A novel organotypic cortical slice culture model for traumatic brain injury: molecular changes induced by injury and mesenchymal stromal cell secretome treatment. Front Cell Neurosci 2023; 17:1217987. [PMID: 37534042 PMCID: PMC10390737 DOI: 10.3389/fncel.2023.1217987] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 06/23/2023] [Indexed: 08/04/2023] Open
Abstract
Traumatic brain injury (TBI) is a major worldwide neurological disorder with no neuroprotective treatment available. Three-dimensional (3D) in vitro models of brain contusion serving as a screening platform for drug testing are lacking. Here we developed a new in vitro model of brain contusion on organotypic cortical brain slices and tested its responsiveness to mesenchymal stromal cell (MSC) derived secretome. A focal TBI was induced on organotypic slices by an electromagnetic impactor. Compared to control condition, a temporal increase in cell death was observed after TBI by propidium iodide incorporation and lactate dehydrogenase release assays up to 48 h post-injury. TBI induced gross neuronal loss in the lesion core, with disruption of neuronal arborizations measured by microtubule-associated protein-2 (MAP-2) immunostaining and associated with MAP-2 gene down-regulation. Neuronal damage was confirmed by increased levels of neurofilament light chain (NfL), microtubule associated protein (Tau) and ubiquitin C-terminal hydrolase L1 (UCH-L1) released into the culture medium 48 h after TBI. We detected glial activation with microglia cells acquiring an amoeboid shape with less ramified morphology in the contusion core. MSC-secretome treatment, delivered 1 h post-injury, reduced cell death in the contusion core, decreased NfL release in the culture media, promoted neuronal reorganization and improved microglia survival/activation. Our 3D in vitro model of brain contusion recapitulates key features of TBI pathology. We showed protective effects of MSC-secretome, suggesting the model stands as a tractable medium/high throughput, ethically viable, and pathomimetic biological asset for testing new cell-based therapies.
Collapse
Affiliation(s)
- Francesca Pischiutta
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Helena Cavaleiro
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s–PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Enrico Caruso
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
- Neuroscience Intensive Care Unit, Department of Anesthesia and Critical Care, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Francesca Tribuzio
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Noemi Di Marzo
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
- Centro Ricerca Tettamanti, Clinica Pediatrica, Università di Milano-Bicocca, Fondazione MBBM, Monza, Italy
| | - Federico Moro
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Firas Kobeissy
- Program for Neurotrauma, Neuroproteomics and Biomarkers Research, Departments of Emergency Medicine, Psychiatry, Neuroscience and Chemistry, University of Florida, Gainesville, FL, United States
- Department of Neurobiology, Center for Neurotrauma, Multiomics and Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, United States
| | - Kevin K. Wang
- Program for Neurotrauma, Neuroproteomics and Biomarkers Research, Departments of Emergency Medicine, Psychiatry, Neuroscience and Chemistry, University of Florida, Gainesville, FL, United States
- Department of Neurobiology, Center for Neurotrauma, Multiomics and Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, United States
| | - António J. Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s–PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Elisa R. Zanier
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| |
Collapse
|
3
|
Varghese N, Morrison B. Partial Depletion of Microglia Attenuates Long-Term Potentiation Deficits following Repeated Blast Traumatic Brain Injury in Organotypic Hippocampal Slice Cultures. J Neurotrauma 2023; 40:547-560. [PMID: 36508265 PMCID: PMC10081725 DOI: 10.1089/neu.2022.0284] [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] [Indexed: 12/14/2022] Open
Abstract
Blast-induced traumatic brain injury (bTBI) has been a health concern in both military and civilian populations due to recent military and geopolitical conflicts. Military service members are frequently exposed to repeated bTBI throughout their training and deployment. Our group has previously reported compounding functional deficits as a result of increased number of blast exposures. In this study, we further characterized the decrease in long-term potentiation (LTP) by varying the blast injury severity and the inter-blast interval between two blast exposures. LTP deficits were attenuated with increasing inter-blast intervals. We also investigated changes in microglial activation; expression of CD68 was increased and expression of CD206 was decreased after multiple blast exposures. Expression of macrophage inflammatory protein (MIP)-1α, interleukin (IL)-1β, monocyte chemoattractant protein (MCP)-1, interferon gamma-inducible protein (IP)-10, and regulated on activation, normal T cell expressed and secreted (RANTES) increased, while expression of IL-10 decreased in the acute period after both single and repeated bTBI. By partially depleting microglia prior to injury, LTP deficits after injury were significantly reduced. Treatment with the novel drug, MW-189, prevented LTP deficits when administered immediately following a repeated bTBI and even when administered only for an acute period (24 h) between two blast injuries. These findings could inform the development of therapeutic strategies to treat the neurological deficits of repeated bTBI suggesting that microglia play a major role in functional neuronal deficits and may be a viable therapeutic target to lessen the neurophysiological deficits after bTBI.
Collapse
Affiliation(s)
- Nevin Varghese
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Barclay Morrison
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| |
Collapse
|
4
|
Varghese N, Morrison B. Inhibition of cyclooxygenase and EP3 receptor improved long term potentiation in a rat organotypic hippocampal model of repeated blast traumatic brain injury. Neurochem Int 2023; 163:105472. [PMID: 36599378 DOI: 10.1016/j.neuint.2022.105472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/09/2022] [Accepted: 12/30/2022] [Indexed: 01/03/2023]
Abstract
Blast-induced traumatic brain injury (bTBI) is a health concern in military service members who are exposed to multiple blasts throughout their training and deployment. Our group has previously reported decreased long term potentiation (LTP) following repeated bTBI in a rat organotypic hippocampal slice culture (OHSC) model. In this study, we investigated changes in inflammatory markers like cyclooxygenase (COX) and tested the efficacy of COX or prostaglandin EP3 receptor (EP3R) inhibitors in attenuating LTP deficits. Expression of COX-2 was increased 48 h following repeated injury, whereas COX-1 expression was unchanged. EP3R expression was upregulated, and cyclic adenosine monophosphate (cAMP) concentration was decreased after repeated blast exposure. Post-traumatic LTP deficits improved after treatment with a COX-1 specific inhibitor, SC-560, a COX-2 specific inhibitor, rofecoxib, a pan-COX inhibitor, ibuprofen, or an EP3R inhibitor, L-798,106. Delayed treatment with ibuprofen and L-798,106 also prevented LTP deficits. These findings suggest that bTBI induced neuroinflammation may be responsible for some functional deficits that we have observed in injured OHSCs. Additionally, COX and EP3R inhibition may be viable therapeutic strategies to reduce neurophysiological deficits after repeated bTBI.
Collapse
Affiliation(s)
- Nevin Varghese
- Department of Biomedical Engineering, Columbia University, 1210 Amsterdam Avenue, New York, NY, 10027, USA.
| | - Barclay Morrison
- Department of Biomedical Engineering, Columbia University, 1210 Amsterdam Avenue, New York, NY, 10027, USA.
| |
Collapse
|
5
|
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: 1.7] [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
|
6
|
Siedhoff HR, Chen S, Song H, Cui J, Cernak I, Cifu DX, DePalma RG, Gu Z. Perspectives on Primary Blast Injury of the Brain: Translational Insights Into Non-inertial Low-Intensity Blast Injury. Front Neurol 2022; 12:818169. [PMID: 35095749 PMCID: PMC8794583 DOI: 10.3389/fneur.2021.818169] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 12/20/2021] [Indexed: 12/18/2022] Open
Abstract
Most traumatic brain injuries (TBIs) during military deployment or training are clinically "mild" and frequently caused by non-impact blast exposures. Experimental models were developed to reproduce the biological consequences of high-intensity blasts causing moderate to severe brain injuries. However, the pathophysiological mechanisms of low-intensity blast (LIB)-induced neurological deficits have been understudied. This review provides perspectives on primary blast-induced mild TBI models and discusses translational aspects of LIB exposures as defined by standardized physical parameters including overpressure, impulse, and shock wave velocity. Our mouse LIB-exposure model, which reproduces deployment-related scenarios of open-field blast (OFB), caused neurobehavioral changes, including reduced exploratory activities, elevated anxiety-like levels, impaired nesting behavior, and compromised spatial reference learning and memory. These functional impairments associate with subcellular and ultrastructural neuropathological changes, such as myelinated axonal damage, synaptic alterations, and mitochondrial abnormalities occurring in the absence of gross- or cellular damage. Biochemically, we observed dysfunctional mitochondrial pathways that led to elevated oxidative stress, impaired fission-fusion dynamics, diminished mitophagy, decreased oxidative phosphorylation, and compensated cell respiration-relevant enzyme activity. LIB also induced increased levels of total tau, phosphorylated tau, and amyloid β peptide, suggesting initiation of signaling cascades leading to neurodegeneration. We also compare translational aspects of OFB findings to alternative blast injury models. By scoping relevant recent research findings, we provide recommendations for future preclinical studies to better reflect military-operational and clinical realities. Overall, better alignment of preclinical models with clinical observations and experience related to military injuries will facilitate development of more precise diagnosis, clinical evaluation, treatment, and rehabilitation.
Collapse
Affiliation(s)
- Heather R. Siedhoff
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO, United States
- Harry S. Truman Memorial Veterans' Hospital Research Service, Columbia, MO, United States
| | - Shanyan Chen
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO, United States
- Harry S. Truman Memorial Veterans' Hospital Research Service, Columbia, MO, United States
| | - Hailong Song
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO, United States
- Harry S. Truman Memorial Veterans' Hospital Research Service, Columbia, MO, United States
| | - Jiankun Cui
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO, United States
- Harry S. Truman Memorial Veterans' Hospital Research Service, Columbia, MO, United States
| | - Ibolja Cernak
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA, United States
| | - David X. Cifu
- Department of Physical Medicine and Rehabilitation, Virginia Commonwealth University School of Medicine, Richmond, VA, United States
| | - Ralph G. DePalma
- Office of Research and Development, Department of Veterans Affairs, Washington, DC, United States
- Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Zezong Gu
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO, United States
- Harry S. Truman Memorial Veterans' Hospital Research Service, Columbia, MO, United States
| |
Collapse
|
7
|
Schumm SN, Gabrieli D, Meaney DF. Plasticity impairment exposes CA3 vulnerability in a hippocampal network model of mild traumatic brain injury. Hippocampus 2022; 32:231-250. [PMID: 34978378 DOI: 10.1002/hipo.23402] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/08/2021] [Accepted: 11/18/2021] [Indexed: 11/10/2022]
Abstract
Proper function of the hippocampus is critical for executing cognitive tasks such as learning and memory. Traumatic brain injury (TBI) and other neurological disorders are commonly associated with cognitive deficits and hippocampal dysfunction. Although there are many existing models of individual subregions of the hippocampus, few models attempt to integrate the primary areas into one system. In this work, we developed a computational model of the hippocampus, including the dentate gyrus, CA3, and CA1. The subregions are represented as an interconnected neuronal network, incorporating well-characterized ex vivo slice electrophysiology into the functional neuron models and well-documented anatomical connections into the network structure. In addition, since plasticity is foundational to the role of the hippocampus in learning and memory as well as necessary for studying adaptation to injury, we implemented spike-timing-dependent plasticity among the synaptic connections. Our model mimics key features of hippocampal activity, including signal frequencies in the theta and gamma bands and phase-amplitude coupling in area CA1. We also studied the effects of spike-timing-dependent plasticity impairment, a potential consequence of TBI, in our model and found that impairment decreases broadband power in CA3 and CA1 and reduces phase coherence between these two subregions, yet phase-amplitude coupling in CA1 remains intact. Altogether, our work demonstrates characteristic hippocampal activity with a scaled network model of spiking neurons and reveals the sensitive balance of plasticity mechanisms in the circuit through one manifestation of mild traumatic injury.
Collapse
Affiliation(s)
- Samantha N Schumm
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David Gabrieli
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David F Meaney
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Neurosurgery, Penn Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
8
|
Zhao L, Zhang L, Zhu W, Chen H, Ding Y, Cui G. Inhibition of microRNA-203 protects against traumatic brain injury induced neural damages via suppressing neuronal apoptosis and dementia-related molecues. Physiol Behav 2021; 228:113190. [PMID: 33002497 DOI: 10.1016/j.physbeh.2020.113190] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/30/2020] [Accepted: 09/26/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND Traumatic brain injury (TBI) can lead to cognitive dysfunction and motor dysfunction. TBI is a potential risk factor for subsequent dementia. Hyperphosphorylation of Tau and ApoE4 has been found in patients with TBI. A significant increase in miR-203 was also found in the peripheral blood of TBI mice. Thus, we hypothesize that miR-203 inhibitor protects against neuronal damage and behavioral deficits by inhibition of Tau phosphorylation, ApoE4 expression and apoptosis. METHODS TBI mice were induced and treated with miR-203 inhibitor. Tau phosphorylation and ApoE4, hippocampal long-term potentiation (LTP), learning and memory, and motor function were separately detected by Western blot analysis, electrophysiology recording and behavioral assessments including Morris water maze test, beam-balance test, beam-walk test and rotarod test. Caspase-3 activity and bcl-2 expression were detected by ELISA. RESULTS TBI induction led to increased phosphorylation of Tau and ApoE4 expression. Administration of miR-203 inhibitor suppressed TBI induced ApoE4 expression and Tau hyperphosphorylation, rescued TBI mediated hippocampal LTP deficits and hippocampus dependent learning and memory dysfunction. miR-203 inhibitor treatment also improved motor function. In addition, miR-203 inhibitor treatment inhibited neuronal apoptosis by inhibiting caspase-3 activity and increasing bcl-2 expression. CONCLUSION miR-203 inhibitor treatment can rescue TBI-induced neural damage by inhibiting neuronal apoptosis and dementia markers like ApoE4 expression and Tau phosphorylation.
Collapse
Affiliation(s)
- Li Zhao
- Department of Pharmacy, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, No. 20 Yuhuangding East Road, Yantai 264000, Shangdong, China
| | - Lei Zhang
- Department of Pharmacy, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, No. 20 Yuhuangding East Road, Yantai 264000, Shangdong, China
| | - Wei Zhu
- Department of Neurosurgery, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, No. 20 Yuhuangding East Road, Yantai 264000, Shangdong, China
| | - Hongguang Chen
- Department of Neurosurgery, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, No. 20 Yuhuangding East Road, Yantai 264000, Shangdong, China
| | - Yuexia Ding
- Department of Pharmacy, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, No. 20 Yuhuangding East Road, Yantai 264000, Shangdong, China
| | - Guangqiang Cui
- Department of Neurosurgery, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, No. 20 Yuhuangding East Road, Yantai 264000, Shangdong, China.
| |
Collapse
|
9
|
Zhu W, Zhao L, Li T, Xu H, Ding Y, Cui G. Docosahexaenoic acid ameliorates traumatic brain injury involving JNK-mediated Tau phosphorylation signaling. Neurosci Res 2020; 157:44-50. [DOI: 10.1016/j.neures.2019.07.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 07/14/2019] [Accepted: 07/22/2019] [Indexed: 10/26/2022]
|
10
|
Grüßer L, Blaumeiser-Debarry R, Rossaint R, Krings M, Kremer B, Höllig A, Coburn M. A 6-Step Approach to Gain Higher Quality Results From Organotypic Hippocampal Brain Slices in a Traumatic Brain Injury Model. Basic Clin Neurosci 2020; 10:485-498. [PMID: 32284838 PMCID: PMC7149959 DOI: 10.32598/bcn.9.10.235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 09/27/2018] [Accepted: 03/22/2019] [Indexed: 01/20/2023] Open
Abstract
Introduction Organotypic Hippocampal Brain Slices (OHBS) provide an advantageous alternative to in vivo models to scrutinize Traumatic Brain Injury (TBI). We followed a well-established TBI protocol, but noticed that several factors may influence the results in such a setup. Here, we describe a structured approach to generate more comparable results and discuss why specific eligibility criteria should be applied. Methods We defined necessary checkpoints and developed inclusion and exclusion criteria that take the observed variation in such a model into consideration. Objective measures include the identification and exclusion of pre-damaged slices and outliers. Six steps were outlined in this study. Results A six-step approach to enhance comparability is proposed and summarized in a flowchart. We applied the suggested measures to data derived from our TBI-experiments examining the impact of three different interventions in 1459 OHBS. Our exemplary results show that through equal requirements set for all slices more precise findings are ensured. Conclusion Results in a TBI experiment on OHBS should be analyzed critically as inhomogeneities may occur. In order to ensure more precise findings, a structured approach of comparing the results should be followed. Further research is recommended to confirm and further develop this framework.
Collapse
Affiliation(s)
- Linda Grüßer
- Department of Anesthesiology, RWTH Aachen University Hospital, Aachen, Germany
| | | | - Rolf Rossaint
- Department of Anesthesiology, RWTH Aachen University Hospital, Aachen, Germany
| | - Matthias Krings
- Department of Anesthesiology and Intensive Care, Medizinisches Zentrum StaedteRegion Aachen, Aachen, Germany
| | - Benedikt Kremer
- Department of Neurosurgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Anke Höllig
- Department of Neurosurgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Mark Coburn
- Department of Anesthesiology, RWTH Aachen University Hospital, Aachen, Germany
| |
Collapse
|
11
|
Direct Observation of Low Strain, High Rate Deformation of Cultured Brain Tissue During Primary Blast. Ann Biomed Eng 2019; 48:1196-1206. [DOI: 10.1007/s10439-019-02437-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 12/08/2019] [Indexed: 10/25/2022]
|
12
|
Bugay V, Bozdemir E, Vigil FA, Chun SH, Holstein DM, Elliott WR, Sprague CJ, Cavazos JE, Zamora DO, Rule G, Shapiro MS, Lechleiter JD, Brenner R. A Mouse Model of Repetitive Blast Traumatic Brain Injury Reveals Post-Trauma Seizures and Increased Neuronal Excitability. J Neurotrauma 2019; 37:248-261. [PMID: 31025597 DOI: 10.1089/neu.2018.6333] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Repetitive blast traumatic brain injury (TBI) affects numerous soldiers on the battlefield. Mild TBI has been shown to have long-lasting effects with repeated injury. We have investigated effects on neuronal excitability after repetitive, mild TBI in a mouse model of blast-induced brain injury. We exposed mice to mild blast trauma of an average peak overpressure of 14.6 psi, repeated across three consecutive days. While a single exposure did not reveal trauma as indicated by the glial fibrillary acidic protein indicator, three repetitive blasts did show significant increases. As well, mice had an increased indicator of inflammation (Iba-1) and increased tau, tau phosphorylation, and altered cytokine levels in the spleen. Video-electroencephalographic monitoring 48 h after the final blast exposure demonstrated seizures in 50% (12/24) of the mice, most of which were non-convulsive seizures. Long-term monitoring revealed that spontaneous seizures developed in at least 46% (6/13) of the mice. Patch clamp recording of dentate gyrus hippocampus neurons 48 h post-blast TBI demonstrated a shortened latency to the first spike and hyperpolarization of action potential threshold. We also found that evoked excitatory postsynaptic current amplitudes were significantly increased. These findings indicate that mild, repetitive blast exposures cause increases in neuronal excitability and seizures and eventual epilepsy development in some animals. The non-convulsive nature of the seizures suggests that subclinical seizures may occur in individuals experiencing even mild blast events, if repeated.
Collapse
Affiliation(s)
- Vladislav Bugay
- Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas
| | - Eda Bozdemir
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, Texas
| | - Fabio A Vigil
- Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas
| | - Sang H Chun
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, Texas
| | - Deborah M Holstein
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, Texas
| | - William R Elliott
- Sensory Trauma, United States Army Institute of Surgical Research, Fort Sam Houston San Antonio, Texas
| | - Cassie J Sprague
- Sensory Trauma, United States Army Institute of Surgical Research, Fort Sam Houston San Antonio, Texas
| | - Jose E Cavazos
- Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas.,Department of Neurology, University of Texas Health San Antonio, San Antonio, Texas
| | - David O Zamora
- Sensory Trauma, United States Army Institute of Surgical Research, Fort Sam Houston San Antonio, Texas
| | | | - Mark S Shapiro
- Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas
| | - James D Lechleiter
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, Texas
| | - Robert Brenner
- Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas
| |
Collapse
|
13
|
Glotfelty EJ, Delgado TE, Tovar-y-Romo LB, Luo Y, Hoffer BJ, Olson L, Karlsson TE, Mattson MP, Harvey BK, Tweedie D, Li Y, Greig NH. Incretin Mimetics as Rational Candidates for the Treatment of Traumatic Brain Injury. ACS Pharmacol Transl Sci 2019; 2:66-91. [PMID: 31396586 PMCID: PMC6687335 DOI: 10.1021/acsptsci.9b00003] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Indexed: 12/17/2022]
Abstract
Traumatic brain injury (TBI) is becoming an increasing public health issue. With an annually estimated 1.7 million TBIs in the United States (U.S) and nearly 70 million worldwide, the injury, isolated or compounded with others, is a major cause of short- and long-term disability and mortality. This, along with no specific treatment, has made exploration of TBI therapies a priority of the health system. Age and sex differences create a spectrum of vulnerability to TBI, with highest prevalence among younger and older populations. Increased public interest in the long-term effects and prevention of TBI have recently reached peaks, with media attention bringing heightened awareness to sport and war related head injuries. Along with short-term issues, TBI can increase the likelihood for development of long-term neurodegenerative disorders. A growing body of literature supports the use of glucagon-like peptide-1 (GLP-1), glucose-dependent insulinotropic peptide (GIP), and glucagon (Gcg) receptor (R) agonists, along with unimolecular combinations of these therapies, for their potent neurotrophic/neuroprotective activities across a variety of cellular and animal models of chronic neurodegenerative diseases (Alzheimer's and Parkinson's diseases) and acute cerebrovascular disorders (stroke). Mild or moderate TBI shares many of the hallmarks of these conditions; recent work provides evidence that use of these compounds is an effective strategy for its treatment. Safety and efficacy of many incretin-based therapies (GLP-1 and GIP) have been demonstrated in humans for the treatment of type 2 diabetes mellitus (T2DM), making these compounds ideal for rapid evaluation in clinical trials of mild and moderate TBI.
Collapse
Affiliation(s)
- Elliot J. Glotfelty
- Translational
Gerontology Branch, and Laboratory of Neurosciences, Intramural
Research Program, National Institute on
Aging, National Institutes of Health, Baltimore, Maryland 21224, United States
- Department
of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Thomas E. Delgado
- Translational
Gerontology Branch, and Laboratory of Neurosciences, Intramural
Research Program, National Institute on
Aging, National Institutes of Health, Baltimore, Maryland 21224, United States
| | - Luis B. Tovar-y-Romo
- Division
of Neuroscience, Institute of Cellular Physiology, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Yu Luo
- Department
of Molecular Genetics, University of Cincinnati, Cincinnati, Ohio 45221, United States
| | - Barry J. Hoffer
- Department
of Neurosurgery, Case Western Reserve University
School of Medicine, Cleveland, Ohio 44106, United States
| | - Lars Olson
- Department
of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - Mark P. Mattson
- Translational
Gerontology Branch, and Laboratory of Neurosciences, Intramural
Research Program, National Institute on
Aging, National Institutes of Health, Baltimore, Maryland 21224, United States
| | - Brandon K. Harvey
- Molecular
Mechanisms of Cellular Stress and Inflammation Unit, Integrative Neuroscience
Department, National Institute on Drug Abuse,
National Institutes of Health, Baltimore, Maryland 21224, United States
| | - David Tweedie
- Translational
Gerontology Branch, and Laboratory of Neurosciences, Intramural
Research Program, National Institute on
Aging, National Institutes of Health, Baltimore, Maryland 21224, United States
| | - Yazhou Li
- Translational
Gerontology Branch, and Laboratory of Neurosciences, Intramural
Research Program, National Institute on
Aging, National Institutes of Health, Baltimore, Maryland 21224, United States
| | - Nigel H. Greig
- Translational
Gerontology Branch, and Laboratory of Neurosciences, Intramural
Research Program, National Institute on
Aging, National Institutes of Health, Baltimore, Maryland 21224, United States
| |
Collapse
|
14
|
Swiatkowski P, Sewell E, Sweet ES, Dickson S, Swanson RA, McEwan SA, Cuccolo N, McDonnell ME, Patel MV, Varghese N, Morrison B, Reitz AB, Meaney DF, Firestein BL. Cypin: A novel target for traumatic brain injury. Neurobiol Dis 2018; 119:13-25. [PMID: 30031156 DOI: 10.1016/j.nbd.2018.07.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 07/06/2018] [Accepted: 07/17/2018] [Indexed: 12/13/2022] Open
Abstract
Cytosolic PSD-95 interactor (cypin), the primary guanine deaminase in the brain, plays key roles in shaping neuronal circuits and regulating neuronal survival. Despite this pervasive role in neuronal function, the ability for cypin activity to affect recovery from acute brain injury is unknown. A key barrier in identifying the role of cypin in neurological recovery is the absence of pharmacological tools to manipulate cypin activity in vivo. Here, we use a small molecule screen to identify two activators and one inhibitor of cypin's guanine deaminase activity. The primary screen identified compounds that change the initial rate of guanine deamination using a colorimetric assay, and secondary screens included the ability of the compounds to protect neurons from NMDA-induced injury and NMDA-induced decreases in frequency and amplitude of miniature excitatory postsynaptic currents. Hippocampal neurons pretreated with activators preserved electrophysiological function and survival after NMDA-induced injury in vitro, while pretreatment with the inhibitor did not. The effects of the activators were abolished when cypin was knocked down. Administering either cypin activator directly into the brain one hour after traumatic brain injury significantly reduced fear conditioning deficits 5 days after injury, while delivering the cypin inhibitor did not improve outcome after TBI. Together, these data demonstrate that cypin activation is a novel approach for improving outcome after TBI and may provide a new pathway for reducing the deficits associated with TBI in patients.
Collapse
Affiliation(s)
- Przemyslaw Swiatkowski
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854-8082, USA; Graduate Program in Molecular Biosciences, Rutgers University, 604 Allison Road, Piscataway, NJ 08854-8082, USA
| | - Emily Sewell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104-6391, USA
| | - Eric S Sweet
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854-8082, USA; Graduate Program in Neurosciences, Rutgers University, 604 Allison Road, Piscataway, NJ 08854-8082, USA
| | - Samantha Dickson
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104-6391, USA
| | - Rachel A Swanson
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854-8082, USA
| | - Sara A McEwan
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854-8082, USA; Graduate Program in Neurosciences, Rutgers University, 604 Allison Road, Piscataway, NJ 08854-8082, USA
| | - Nicholas Cuccolo
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854-8082, USA
| | - Mark E McDonnell
- Fox Chase Chemical Diversity Center, Inc., Doylestown, PA 18902, USA
| | - Mihir V Patel
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854-8082, USA; Graduate Program in Neurosciences, Rutgers University, 604 Allison Road, Piscataway, NJ 08854-8082, USA
| | - Nevin Varghese
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Barclay Morrison
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Allen B Reitz
- Fox Chase Chemical Diversity Center, Inc., Doylestown, PA 18902, USA
| | - David F Meaney
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104-6391, USA
| | - Bonnie L Firestein
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854-8082, USA.
| |
Collapse
|
15
|
Campos-Pires R, Koziakova M, Yonis A, Pau A, Macdonald W, Harris K, Edge CJ, Franks NP, Mahoney PF, Dickinson R. Xenon Protects against Blast-Induced Traumatic Brain Injury in an In Vitro Model. J Neurotrauma 2018; 35:1037-1044. [PMID: 29285980 PMCID: PMC5899289 DOI: 10.1089/neu.2017.5360] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The aim of this study was to evaluate the neuroprotective efficacy of the inert gas xenon as a treatment for patients with blast-induced traumatic brain injury in an in vitro laboratory model. We developed a novel blast traumatic brain injury model using C57BL/6N mouse organotypic hippocampal brain-slice cultures exposed to a single shockwave, with the resulting injury quantified using propidium iodide fluorescence. A shock tube blast generator was used to simulate open field explosive blast shockwaves, modeled by the Friedlander waveform. Exposure to blast shockwave resulted in significant (p < 0.01) injury that increased with peak-overpressure and impulse of the shockwave, and which exhibited a secondary injury development up to 72 h after trauma. Blast-induced propidium iodide fluorescence overlapped with cleaved caspase-3 immunofluorescence, indicating that shock-wave–induced cell death involves apoptosis. Xenon (50% atm) applied 1 h after blast exposure reduced injury 24 h (p < 0.01), 48 h (p < 0.05), and 72 h (p < 0.001) later, compared with untreated control injury. Xenon-treated injured slices were not significantly different from uninjured sham slices at 24 h and 72 h. We demonstrate for the first time that xenon treatment after blast traumatic brain injury reduces initial injury and prevents subsequent injury development in vitro. Our findings support the idea that xenon may be a potential first-line treatment for those with blast-induced traumatic brain injury.
Collapse
Affiliation(s)
- Rita Campos-Pires
- 1 Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London , London, United Kingdom .,2 Royal British Legion Centre for Blast Injury Studies, Department of Bioengineering, Imperial College London , London, United Kingdom
| | - Mariia Koziakova
- 1 Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London , London, United Kingdom .,2 Royal British Legion Centre for Blast Injury Studies, Department of Bioengineering, Imperial College London , London, United Kingdom
| | - Amina Yonis
- 1 Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London , London, United Kingdom
| | - Ashni Pau
- 1 Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London , London, United Kingdom
| | - Warren Macdonald
- 2 Royal British Legion Centre for Blast Injury Studies, Department of Bioengineering, Imperial College London , London, United Kingdom .,3 Department of Bioengineering, Imperial College London , London, United Kingdom
| | - Katie Harris
- 1 Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London , London, United Kingdom
| | - Christopher J Edge
- 4 Department of Life Sciences, Imperial College London , London, United Kingdom .,5 Department of Anaesthetics, Royal Berkshire Hospital NHS Foundation Trust , Reading, United Kingdom
| | - Nicholas P Franks
- 4 Department of Life Sciences, Imperial College London , London, United Kingdom
| | - Peter F Mahoney
- 6 Royal Centre for Defence Medicine , Medical Directorate Joint Force Command, ICT Centre, Birmingham, United Kingdom
| | - Robert Dickinson
- 1 Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London , London, United Kingdom .,2 Royal British Legion Centre for Blast Injury Studies, Department of Bioengineering, Imperial College London , London, United Kingdom
| |
Collapse
|
16
|
Sawyer TW, Ritzel DV, Wang Y, Josey T, Villanueva M, Nelson P, Song Y, Shei Y, Hennes G, Vair C, Parks S, Fan C, McLaws L. Primary Blast Causes Delayed Effects without Cell Death in Shell-Encased Brain Cell Aggregates. J Neurotrauma 2017; 35:174-186. [PMID: 28726571 DOI: 10.1089/neu.2016.4961] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Previous work in this laboratory used underwater explosive exposures to isolate the effects of shock-induced principle stress without shear on rat brain aggregate cultures. The current study has utilized simulated air blast to expose aggregates in suspension and enclosed within a spherical shell, enabling the examination of a much more complex biomechanical insult. Culture medium-filled spheres were exposed to single pulse overpressures of 15-30 psi (∼6-7 msec duration) and measurements within the sphere at defined sites showed complex and spatially dependent pressure changes. When brain aggregates were exposed to similar conditions, no cell death was observed and no changes in several commonly used biomarkers of traumatic brain injury (TBI) were noted. However, similarly to underwater blast, immediate and transient increases in the protein kinase B signaling pathway were observed at early time-points (3 days). In contrast, the oligodendrocyte marker 2',3'-cyclic nucleotide 3'-phosphodiesterase, as well as vascular endothelial growth factor, both displayed markedly delayed (14-28 days) and pressure-dependent responses. The imposition of a spherical shell between the single pulse shock wave and the target brain tissue introduces greatly increased complexity to the insult. This work shows that brain tissue can not only discriminate the nature of the pressure changes it experiences, but that a portion of its response is significantly delayed. These results have mechanistic implications for the study of primary blast-induced TBI and also highlight the importance of rigorously characterizing the actual pressure variations experienced by target tissue in primary blast studies.
Collapse
Affiliation(s)
- Thomas W Sawyer
- 1 Defence Research and Development Canada, Suffield Research Center , Medicine Hat, Alberta, Canada
| | | | - Yushan Wang
- 1 Defence Research and Development Canada, Suffield Research Center , Medicine Hat, Alberta, Canada
| | - Tyson Josey
- 1 Defence Research and Development Canada, Suffield Research Center , Medicine Hat, Alberta, Canada
| | - Mercy Villanueva
- 1 Defence Research and Development Canada, Suffield Research Center , Medicine Hat, Alberta, Canada
| | - Peggy Nelson
- 1 Defence Research and Development Canada, Suffield Research Center , Medicine Hat, Alberta, Canada
| | - Yanfeng Song
- 1 Defence Research and Development Canada, Suffield Research Center , Medicine Hat, Alberta, Canada
| | - Yimin Shei
- 1 Defence Research and Development Canada, Suffield Research Center , Medicine Hat, Alberta, Canada
| | - Grant Hennes
- 1 Defence Research and Development Canada, Suffield Research Center , Medicine Hat, Alberta, Canada
| | - Cory Vair
- 1 Defence Research and Development Canada, Suffield Research Center , Medicine Hat, Alberta, Canada
| | | | - Changyang Fan
- 4 Canada West Biosciences , Camrose, Alberta, Canada
| | - Lori McLaws
- 1 Defence Research and Development Canada, Suffield Research Center , Medicine Hat, Alberta, Canada
| |
Collapse
|
17
|
Wang Y, Sawyer TW, Tse YC, Fan C, Hennes G, Barnes J, Josey T, Weiss T, Nelson P, Wong TP. Primary Blast-Induced Changes in Akt and GSK 3β Phosphorylation in Rat Hippocampus. Front Neurol 2017; 8:413. [PMID: 28868045 PMCID: PMC5563325 DOI: 10.3389/fneur.2017.00413] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 07/31/2017] [Indexed: 12/30/2022] Open
Abstract
Traumatic brain injury (TBI) due to blast from improvised explosive devices has been a leading cause of morbidity and mortality in recent conflicts in Iraq and Afghanistan. However, the mechanisms of primary blast-induced TBI are not well understood. The Akt signal transduction pathway has been implicated in various brain pathologies including TBI. In the present study, the effects of simulated primary blast waves on the phosphorylation status of Akt and its downstream effector kinase, glycogen synthase kinase 3β (GSK3β), in rat hippocampus, were investigated. Male Sprague-Dawley (SD) rats (350–400 g) were exposed to a single pulse shock wave (25 psi; ~7 ms duration) and sacrificed 1 day, 1 week, or 6 weeks after exposure. Total and phosphorylated Akt, as well as phosphorylation of its downstream effector kinase GSK3β (at serine 9), were detected with western blot analysis and immunohistochemistry. Results showed that Akt phosphorylation at both serine 473 and threonine 308 was increased 1 day after blast on the ipsilateral side of the hippocampus, and this elevation persisted until at least 6 weeks postexposure. Similarly, phosphorylation of GSK3β at serine 9, which inhibits GSK3β activity, was also increased starting at 1 day and persisted until at least 6 weeks after primary blast on the ipsilateral side. In contrast, p-Akt was increased at 1 and 6 weeks on the contralateral side, while p-GSK3β was increased 1 day and 1 week after primary blast exposure. No significant changes in total protein levels of Akt and GSK were observed on either side of the hippocampus at any time points. Immunohistochemical results showed that increased p-Akt was mainly of neuronal origin in the CA1 region of the hippocampus and once phosphorylated, the majority was translocated to the dendritic and plasma membranes. Finally, electrophysiological data showed that evoked synaptic N-methyl-d-aspartate (NMDA) receptor activity was significantly increased 6 weeks after primary blast, suggesting that increased Akt phosphorylation may enhance synaptic NMDA receptor activation, or that enhanced synaptic NMDA receptor activation may increase Akt phosphorylation.
Collapse
Affiliation(s)
- Yushan Wang
- Defence Research and Development Canada, Suffield Research Centre, Medicine Hat, AB, Canada
| | - Thomas W Sawyer
- Defence Research and Development Canada, Suffield Research Centre, Medicine Hat, AB, Canada
| | - Yiu Chung Tse
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Changyang Fan
- Defence Research and Development Canada, Suffield Research Centre, Medicine Hat, AB, Canada
| | - Grant Hennes
- Defence Research and Development Canada, Suffield Research Centre, Medicine Hat, AB, Canada
| | - Julia Barnes
- Defence Research and Development Canada, Suffield Research Centre, Medicine Hat, AB, Canada
| | - Tyson Josey
- Defence Research and Development Canada, Suffield Research Centre, Medicine Hat, AB, Canada
| | - Tracy Weiss
- Defence Research and Development Canada, Suffield Research Centre, Medicine Hat, AB, Canada
| | - Peggy Nelson
- Defence Research and Development Canada, Suffield Research Centre, Medicine Hat, AB, Canada
| | - Tak Pan Wong
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| |
Collapse
|
18
|
Vogel EW, Morales FN, Meaney DF, Bass CR, Morrison B. Phosphodiesterase-4 inhibition restored hippocampal long term potentiation after primary blast. Exp Neurol 2017; 293:91-100. [PMID: 28366471 PMCID: PMC6016024 DOI: 10.1016/j.expneurol.2017.03.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 03/08/2017] [Accepted: 03/30/2017] [Indexed: 01/03/2023]
Abstract
Due to recent military conflicts and terrorist attacks, blast-induced traumatic brain injury (bTBI) presents a health concern for military and civilian personnel alike. Although secondary blast (penetrating injury) and tertiary blast (inertia-driven brain deformation) are known to be injurious, the effects of primary blast caused by the supersonic shock wave interacting with the skull and brain remain debated. Our group previously reported that in vitro primary blast exposure reduced long-term potentiation (LTP), the electrophysiological correlate of learning and memory, in rat organotypic hippocampal slice cultures (OHSCs) and that primary blast affects key proteins governing LTP. Recent studies have investigated phosphodiesterase-4 (PDE4) inhibition as a therapeutic strategy for reducing LTP deficits following inertia-driven TBI. We investigated the therapeutic potential of PDE4 inhibitors, specifically roflumilast, to ameliorate primary blast-induced deficits in LTP. We found that roflumilast at concentrations of 1nM or greater prevented deficits in neuronal plasticity measured 24h post-injury. We also observed a therapeutic window of at least 6h, but <23h. Additionally, we investigated molecular mechanisms that could elucidate this therapeutic effect. Roflumilast treatment (1nM delivered 6h post-injury) significantly increased total AMPA glutamate receptor 1 (GluR1) subunit expression, phosphorylation of the GluR1 subunit at the serine-831 site, and phosphorylation of stargazin at the serine-239/240 site upon LTP induction, measured 24h following injury. Roflumilast treatment significantly increased PSD-95 regardless of LTP induction. These findings indicate that further investigation into the translation of PDE4 inhibition as a therapy following bTBI is warranted.
Collapse
Affiliation(s)
- Edward W Vogel
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Fatima N Morales
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - David F Meaney
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Cameron R Bass
- Department of Biomedical Engineering, Duke University, Durham, NC 27705, USA
| | - Barclay Morrison
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA.
| |
Collapse
|
19
|
Exendin-4 attenuates blast traumatic brain injury induced cognitive impairments, losses of synaptophysin and in vitro TBI-induced hippocampal cellular degeneration. Sci Rep 2017. [PMID: 28623327 PMCID: PMC5473835 DOI: 10.1038/s41598-017-03792-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Mild blast traumatic brain injury (B-TBI) induced lasting cognitive impairments in novel object recognition and less severe deficits in Y-maze behaviors. B-TBI significantly reduced the levels of synaptophysin (SYP) protein staining in cortical (CTX) and hippocampal (HIPP) tissues. Treatment with exendin-4 (Ex-4) delivered by subcutaneous micro-osmotic pumps 48 hours prior to or 2 hours immediately after B-TBI prevented the induction of both cognitive deficits and B-TBI induced changes in SYP staining. The effects of a series of biaxial stretch injuries (BSI) on a neuronal derived cell line, HT22 cells, were assessed in an in vitro model of TBI. Biaxial stretch damage induced shrunken neurites and cell death. Treatment of HT22 cultures with Ex-4 (25 to 100 nM), prior to injury, attenuated the cytotoxic effects of BSI and preserved neurite length similar to sham treated cells. These data imply that treatment with Ex-4 may represent a viable option for the management of secondary events triggered by blast-induced, mild traumatic brain injury that is commonly observed in militarized zones.
Collapse
|
20
|
Miller AP, Shah AS, Aperi BV, Kurpad SN, Stemper BD, Glavaski-Joksimovic A. Acute death of astrocytes in blast-exposed rat organotypic hippocampal slice cultures. PLoS One 2017; 12:e0173167. [PMID: 28264063 PMCID: PMC5338800 DOI: 10.1371/journal.pone.0173167] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 02/16/2017] [Indexed: 01/06/2023] Open
Abstract
Blast traumatic brain injury (bTBI) affects civilians, soldiers, and veterans worldwide and presents significant health concerns. The mechanisms of neurodegeneration following bTBI remain elusive and current therapies are largely ineffective. It is important to better characterize blast-evoked cellular changes and underlying mechanisms in order to develop more effective therapies. In the present study, our group utilized rat organotypic hippocampal slice cultures (OHCs) as an in vitro system to model bTBI. OHCs were exposed to either 138 ± 22 kPa (low) or 273 ± 23 kPa (high) overpressures using an open-ended helium-driven shock tube, or were assigned to sham control group. At 2 hours (h) following injury, we have characterized the astrocytic response to a blast overpressure. Immunostaining against the astrocytic marker glial fibrillary acidic protein (GFAP) revealed acute shearing and morphological changes in astrocytes, including clasmatodendrosis. Moreover, overlap of GFAP immunostaining and propidium iodide (PI) indicated astrocytic death. Quantification of the number of dead astrocytes per counting area in the hippocampal cornu Ammonis 1 region (CA1), demonstrated a significant increase in dead astrocytes in the low- and high-blast, compared to sham control OHCs. However only a small number of GFAP-expressing astrocytes were co-labeled with the apoptotic marker Annexin V, suggesting necrosis as the primary type of cell death in the acute phase following blast exposure. Moreover, western blot analyses revealed calpain mediated breakdown of GFAP. The dextran exclusion additionally indicated membrane disruption as a potential mechanism of acute astrocytic death. Furthermore, although blast exposure did not evoke significant changes in glutamate transporter 1 (GLT-1) expression, loss of GLT-1-expressing astrocytes suggests dysregulation of glutamate uptake following injury. Our data illustrate the profound effect of blast overpressure on astrocytes in OHCs at 2 h following injury and suggest increased calpain activity and membrane disruption as potential underlying mechanisms.
Collapse
Affiliation(s)
- Anna P. Miller
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin, United States of America
| | - Alok S. Shah
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin, United States of America
| | - Brandy V. Aperi
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin, United States of America
| | - Shekar N. Kurpad
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin, United States of America
| | - Brian D. Stemper
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin, United States of America
| | - Aleksandra Glavaski-Joksimovic
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin, United States of America
| |
Collapse
|
21
|
Bioorthogonal chemical imaging of metabolic activities in live mammalian hippocampal tissues with stimulated Raman scattering. Sci Rep 2016; 6:39660. [PMID: 28000773 PMCID: PMC5175176 DOI: 10.1038/srep39660] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 11/25/2016] [Indexed: 12/22/2022] Open
Abstract
Brain is an immensely complex system displaying dynamic and heterogeneous metabolic activities. Visualizing cellular metabolism of nucleic acids, proteins, and lipids in brain with chemical specificity has been a long-standing challenge. Recent development in metabolic labeling of small biomolecules allows the study of these metabolisms at the global level. However, these techniques generally require nonphysiological sample preparation for either destructive mass spectrometry imaging or secondary labeling with relatively bulky fluorescent labels. In this study, we have demonstrated bioorthogonal chemical imaging of DNA, RNA, protein and lipid metabolism in live rat brain hippocampal tissues by coupling stimulated Raman scattering microscopy with integrated deuterium and alkyne labeling. Heterogeneous metabolic incorporations for different molecular species and neurogenesis with newly-incorporated DNA were observed in the dentate gyrus of hippocampus at the single cell level. We further applied this platform to study metabolic responses to traumatic brain injury in hippocampal slice cultures, and observed marked upregulation of protein and lipid metabolism particularly in the hilus region of the hippocampus within days of mechanical injury. Thus, our method paves the way for the study of complex metabolic profiles in live brain tissue under both physiological and pathological conditions with single-cell resolution and minimal perturbation.
Collapse
|
22
|
Vogel EW, Rwema SH, Meaney DF, Bass CRD, Morrison B. Primary Blast Injury Depressed Hippocampal Long-Term Potentiation through Disruption of Synaptic Proteins. J Neurotrauma 2016; 34:1063-1073. [PMID: 27573357 DOI: 10.1089/neu.2016.4578] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Blast-induced traumatic brain injury (bTBI) is a major threat to United States service members in military conflicts worldwide. The effects of primary blast, caused by the supersonic shockwave interacting with the skull and brain, remain unclear. Our group has previously reported that in vitro primary blast exposure can reduce long-term potentiation (LTP), the electrophysiological correlate of learning and memory, in rat organotypic hippocampal slice cultures (OHSCs) without significant changes to cell viability or basal, evoked neuronal function. We investigated the time course of primary blast-induced deficits in LTP and the molecular mechanisms that could underlie these deficits. We found that pure primary blast exposure induced LTP deficits in a delayed manner, requiring longer than 1 hour to develop, and that these deficits spontaneously recovered by 10 days following exposure depending on blast intensity. Additionally, we observed that primary blast exposure reduced total α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) glutamate receptor 1 (GluR1) subunit expression and phosphorylation of the GluR1 subunit at the serine-831 site. Blast also reduced the expression of postsynaptic density protein-95 (PSD-95) and phosphorylation of stargazin protein at the serine-239/240 site. Finally, we found that modulation of the cyclic adenosine monophosphate (cAMP) pathway ameliorated electrophysiological and protein-expression changes caused by blast. These findings could inform the development of novel therapies to treat blast-induced loss of neuronal function.
Collapse
Affiliation(s)
- Edward W Vogel
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - Steve H Rwema
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - David F Meaney
- 2 Department of Bioengineering, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Cameron R Dale Bass
- 3 Department of Biomedical Engineering, Duke University , Durham, North Carolina
| | - Barclay Morrison
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| |
Collapse
|
23
|
Beamer M, Tummala SR, Gullotti D, Kopil C, Gorka S, Bass CRD, Morrison B, Cohen AS, Meaney DF. Primary blast injury causes cognitive impairments and hippocampal circuit alterations. Exp Neurol 2016. [PMID: 27246999 DOI: 10.1016/j.expneurol.2016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Abstract
Blast-induced traumatic brain injury (bTBI) and its long term consequences are a major health concern among veterans. Despite recent work enhancing our knowledge about bTBI, very little is known about the contribution of the blast wave alone to the observed sequelae. Herein, we isolated its contribution in a mouse model by constraining the animals' heads during exposure to a shockwave (primary blast). Our results show that exposure to primary blast alone results in changes in hippocampus-dependent behaviors that correspond with electrophysiological changes in area CA1 and are accompanied by reactive gliosis. Specifically, five days after exposure, behavior in an open field and performance in a spatial object recognition (SOR) task were significantly different from sham. Network electrophysiology, also performed five days after injury, demonstrated a significant decrease in excitability and increase in inhibitory tone. Immunohistochemistry for GFAP and Iba1 performed ten days after injury showed a significant increase in staining. Interestingly, a threefold increase in the impulse of the primary blast wave did not exacerbate these measures. However, we observed a significant reduction in the contribution of the NMDA receptors to the field EPSP at the highest blast exposure level. Our results emphasize the need to account for the effects of primary blast loading when studying the sequelae of bTBI.
Collapse
Affiliation(s)
- Matthew Beamer
- Department of Bioengineering(1), University of Pennsylvania, Philadelphia, PA, USA
| | - Shanti R Tummala
- Department of Bioengineering(1), University of Pennsylvania, Philadelphia, PA, USA
| | - David Gullotti
- Department of Bioengineering(1), University of Pennsylvania, Philadelphia, PA, USA
| | - Catherine Kopil
- Department of Bioengineering(1), University of Pennsylvania, Philadelphia, PA, USA
| | - Samuel Gorka
- Department of Bioengineering(1), University of Pennsylvania, Philadelphia, PA, USA
| | | | - Barclay Morrison
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Akiva S Cohen
- Department of Anesthesiology and Critical Care Medicine, University of Pennsylvania, Philadelphia, PA, USA; Division of Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - David F Meaney
- Department of Bioengineering(1), University of Pennsylvania, Philadelphia, PA, USA; Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
24
|
Effgen GB, Morrison B. Memantine Reduced Cell Death, Astrogliosis, and Functional Deficits in an in vitro Model of Repetitive Mild Traumatic Brain Injury. J Neurotrauma 2016; 34:934-942. [PMID: 27450515 DOI: 10.1089/neu.2016.4528] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Clinical studies suggest that athletes with a history of concussion may be at risk for additional mild traumatic brain injury (mTBI), and repetitive exposure to mTBI acutely increases risk for more significant and persistent symptoms and increases future risk for developing neurodegenerative diseases. Currently, symptoms of mTBI are managed with rest and pain medication; there are no drugs approved by the Food and Drug Administration (FDA) that target the biochemical pathology underlying mTBI to treat or prevent acute and long-term effects of repetitive mTBI. Memantine is an FDA-approved drug for treating Alzheimer's disease, and also was shown to be neuroprotective in rodents following a single, moderate to severe TBI. Therefore, we investigated the potential for memantine to mitigate negative outcomes from repetitive mild stretch injury in organotypical hippocampal slice cultures. Samples received two injuries 24 h apart; injury resulted in significant cell death, loss of long-term potentiation (LTP), and astrogliosis compared with naïve, uninjured samples. Delivery of 1.5 μM memantine 1 h following each stretch significantly reduced the effect of injury for all outcome measures, and did not alter those outcome measures that were unaffected by the injury. Therefore, memantine warrants further pre-clinical and clinical investigation for its therapeutic efficacy to prevent cognitive deficits and neuropathology from multiple mTBIs.
Collapse
Affiliation(s)
- Gwen B Effgen
- Department of Biomedical Engineering, Columbia University , New York, New York
| | - Barclay Morrison
- Department of Biomedical Engineering, Columbia University , New York, New York
| |
Collapse
|
25
|
Beamer M, Tummala SR, Gullotti D, Kopil C, Gorka S, Bass CRD, Morrison B, Cohen AS, Meaney DF. Primary blast injury causes cognitive impairments and hippocampal circuit alterations. Exp Neurol 2016; 283:16-28. [PMID: 27246999 DOI: 10.1016/j.expneurol.2016.05.025] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 05/14/2016] [Accepted: 05/20/2016] [Indexed: 11/17/2022]
Abstract
Blast-induced traumatic brain injury (bTBI) and its long term consequences are a major health concern among veterans. Despite recent work enhancing our knowledge about bTBI, very little is known about the contribution of the blast wave alone to the observed sequelae. Herein, we isolated its contribution in a mouse model by constraining the animals' heads during exposure to a shockwave (primary blast). Our results show that exposure to primary blast alone results in changes in hippocampus-dependent behaviors that correspond with electrophysiological changes in area CA1 and are accompanied by reactive gliosis. Specifically, five days after exposure, behavior in an open field and performance in a spatial object recognition (SOR) task were significantly different from sham. Network electrophysiology, also performed five days after injury, demonstrated a significant decrease in excitability and increase in inhibitory tone. Immunohistochemistry for GFAP and Iba1 performed ten days after injury showed a significant increase in staining. Interestingly, a threefold increase in the impulse of the primary blast wave did not exacerbate these measures. However, we observed a significant reduction in the contribution of the NMDA receptors to the field EPSP at the highest blast exposure level. Our results emphasize the need to account for the effects of primary blast loading when studying the sequelae of bTBI.
Collapse
Affiliation(s)
- Matthew Beamer
- Department of Bioengineering(1), University of Pennsylvania, Philadelphia, PA, USA
| | - Shanti R Tummala
- Department of Bioengineering(1), University of Pennsylvania, Philadelphia, PA, USA
| | - David Gullotti
- Department of Bioengineering(1), University of Pennsylvania, Philadelphia, PA, USA
| | - Catherine Kopil
- Department of Bioengineering(1), University of Pennsylvania, Philadelphia, PA, USA
| | - Samuel Gorka
- Department of Bioengineering(1), University of Pennsylvania, Philadelphia, PA, USA
| | | | - Barclay Morrison
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Akiva S Cohen
- Department of Anesthesiology and Critical Care Medicine, University of Pennsylvania, Philadelphia, PA, USA; Division of Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - David F Meaney
- Department of Bioengineering(1), University of Pennsylvania, Philadelphia, PA, USA; Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
26
|
Effgen GB, Ong T, Nammalwar S, Ortuño AI, Meaney DF, 'Dale' Bass CR, Morrison B. Primary Blast Exposure Increases Hippocampal Vulnerability to Subsequent Exposure: Reducing Long-Term Potentiation. J Neurotrauma 2016; 33:1901-1912. [PMID: 26699926 DOI: 10.1089/neu.2015.4327] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Up to 80% of injuries sustained by U.S. soldiers in Operation Enduring Freedom and Operation Iraqi Freedom were the result of blast exposure from improvised explosive devices. Some soldiers experience multiple blasts while on duty, and it has been suggested that symptoms of repetitive blast are similar to those that follow multiple non-blast concussions, such as sport-related concussion. Despite the interest in the effects of repetitive blast exposure, it remains unknown whether an initial blast renders the brain more vulnerable to subsequent exposure, resulting in a synergistic injury response. To investigate the effect of multiple primary blasts on the brain, organotypic hippocampal slice cultures were exposed to single or repetitive (two or three total) primary blasts of varying intensities. Long-term potentiation was significantly reduced following two Level 2 (92.7 kPa, 1.4 msec, 38.5 kPa·msec) blasts delivered 24 h apart without altering basal evoked response. This deficit persisted when the interval between injuries was increased to 72 h but not when the interval was extended to 144 h. The repeated blast exposure with a 24 h interval increased microglia staining and activation significantly but did not significantly increase cell death or damage axons, dendrites, or principal cell layers. Lack of overt structural damage and change in basal stimulated neuron response suggest that injury from repetitive primary blast exposure may specifically affect long-term potentiation. Our studies suggest repetitive primary blasts can exacerbate injury dependent on the injury severity and interval between exposures.
Collapse
Affiliation(s)
- Gwen B Effgen
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - Tiffany Ong
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - Shruthi Nammalwar
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - Andrea I Ortuño
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - David F Meaney
- 2 Department of Bioengineering, University of Pennsylvania , Philadelphia, Pennsylvania
| | | | - Barclay Morrison
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| |
Collapse
|
27
|
Stemper BD, Shah AS, Budde MD, Olsen CM, Glavaski-Joksimovic A, Kurpad SN, McCrea M, Pintar FA. Behavioral Outcomes Differ between Rotational Acceleration and Blast Mechanisms of Mild Traumatic Brain Injury. Front Neurol 2016; 7:31. [PMID: 27014184 PMCID: PMC4789366 DOI: 10.3389/fneur.2016.00031] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 02/29/2016] [Indexed: 11/20/2022] Open
Abstract
Mild traumatic brain injury (mTBI) can result from a number of mechanisms, including blunt impact, head rotational acceleration, exposure to blast, and penetration of projectiles. Mechanism is likely to influence the type, severity, and chronicity of outcomes. The objective of this study was to determine differences in the severity and time course of behavioral outcomes following blast and rotational mTBI. The Medical College of Wisconsin (MCW) Rotational Injury model and a shock tube model of primary blast injury were used to induce mTBI in rats and behavioral assessments were conducted within the first week, as well as 30 and 60 days following injury. Acute recovery time demonstrated similar increases over protocol-matched shams, indicating acute injury severity equivalence between the two mechanisms. Post-injury behavior in the elevated plus maze demonstrated differing trends, with rotationally injured rats acutely demonstrating greater activity, whereas blast-injured rats had decreased activity that developed at chronic time points. Similarly, blast-injured rats demonstrated trends associated with cognitive deficits that were not apparent following rotational injuries. These findings demonstrate that rotational and blast injury result in behavioral changes with different qualitative and temporal manifestations. Whereas rotational injury was characterized by a rapidly emerging phenotype consistent with behavioral disinhibition, blast injury was associated with emotional and cognitive differences that were not evident acutely, but developed later, with an anxiety-like phenotype still present in injured animals at our most chronic measurements.
Collapse
Affiliation(s)
- Brian D. Stemper
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, USA
- Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, WI, USA
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Alok S. Shah
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Matthew D. Budde
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, USA
- Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, WI, USA
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Christopher M. Olsen
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Shekar N. Kurpad
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Michael McCrea
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Frank A. Pintar
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, USA
- Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, WI, USA
| |
Collapse
|