1
|
Tseitlin L, Schreiber S, Richmond-Hacham B, Bikovski L, Pick CG. Enhancing cognitive function after traumatic brain injury in male mice: The benefits of running regardless of intervention timing. Exp Neurol 2025; 384:115069. [PMID: 39577609 DOI: 10.1016/j.expneurol.2024.115069] [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: 08/14/2024] [Revised: 11/14/2024] [Accepted: 11/17/2024] [Indexed: 11/24/2024]
Abstract
The significant benefits of physical activity are well-documented in academic literature, with growing evidence highlighting its positive effects (among others) on memory and cognitive function. Exercise, particularly aerobic activities, has been shown to mitigate neuroinflammatory processes, promote neuronal regeneration, facilitate recovery from cerebral trauma, and reduce the risk of neurodegenerative diseases. Among neurological conditions, traumatic brain injury (TBI) is the most common in individuals under 50, with 80-90 % of cases categorized as mild traumatic brain injury (mTBI). This study investigates the impact of exercise on visual and spatial memory deficits in mice following mTBI. ICR mice were subjected to a seventeen-day treadmill training protocol initiated at four different time intervals post-mTBI (2, 7, 13, and 30 days). A battery of specific behavioral tests was used to assess anxiety-like behaviors, motor skills, and visual and spatial memory. Our results indicate that running positively affected mTBI in both novel object recognition (p < 0.001) and Y-maze (p < 0.001) regardless of the running protocol's initiation time, demonstrating that aerobic exercise significantly alleviates cognitive deficits associated with mTBI. Importantly, mTBI did not appear to impact motor abilities or anxiety-like behaviors based on the assessment paradigms utilized. In conclusion, aerobic exercise effectively enhances visual and spatial memory post-mTBI, with promising results observed even when the running protocol is initiated up to one-month post-injury.
Collapse
Affiliation(s)
- Liron Tseitlin
- Department of Anatomy and Anthropology, Tel Aviv University Faculty of Medicine, Tel Aviv, Israel
| | - Shaul Schreiber
- Department of Psychiatry, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Dr. Miriam and Sheldon G. Adelson Clinic for Drug Abuse Treatment and Research, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Bar Richmond-Hacham
- Department of Anatomy and Anthropology, Tel Aviv University Faculty of Medicine, Tel Aviv, Israel
| | - Lior Bikovski
- Myers Neuro-Behavioral Core Facility, Tel Aviv University Faculty of Medicine, Tel Aviv, Israel; School of Behavioral Sciences, Netanya Academic College, Netanya, Israel
| | - Chaim G Pick
- Department of Anatomy and Anthropology, Tel Aviv University Faculty of Medicine, Tel Aviv, Israel; School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel; Sylvan Adams Sports Institute, Tel Aviv University, Tel Aviv, Israel; Dr. Miriam and Sheldon G. Adelson Chair and Center for the Biology of Addictive Diseases, Tel-Aviv University, Tel-Aviv, Israel.
| |
Collapse
|
2
|
Richmond-Hacham B, Tseitlin L, Bikovski L, Pick CG. Investigation of Mild Traumatic Brain Injury Home Cage Behavior: The Home Cage Assay Advantages. J Neurotrauma 2024; 41:e1780-e1792. [PMID: 38517091 DOI: 10.1089/neu.2023.0459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024] Open
Abstract
This study utilized the Noldus PhenoTyper Home Cage Monitoring system (HCM) to assess the behavioral and cognitive changes of experimental closed-head mild traumatic brain injury (mTBI). Seventy-nine adult male Institute of Cancer Research (ICR) mice were subjected to either a sham procedure or closed-head mTBI using the weight-drop model. Seven days post-injury, separate cohorts of mice underwent either a non-cognitive or a cognitive home cage assessment, a treadmill fatigue test, or the Open Field Test. mTBI significantly influenced habituation behavior and circadian wheel-running activity. Notably, mTBI mice exhibited an increased frequency of visits to the running wheel, but each visit was shorter than those of controls. No significant differences between the groups in discrimination or reversal learning performance were observed. However, during the reversal learning stage, mTBI mice performed similarly to their initial discrimination learning levels, suggesting an abnormally faster rate of reversal learning. Home cage monitoring is a valuable tool for studying the subtle effects of mTBI, complementing traditional assays. The automated evaluation of habituation to novel stimuli (e.g., novel environment) could serve as a potentially sensitive tool for assessing mTBI-associated behavioral deficits.
Collapse
Affiliation(s)
- Bar Richmond-Hacham
- Department of Anatomy and Anthropology, Tel Aviv University Faculty of Medicine, Tel Aviv, Israel
| | - Liron Tseitlin
- Department of Anatomy and Anthropology, Tel Aviv University Faculty of Medicine, Tel Aviv, Israel
| | - Lior Bikovski
- Myers Neuro-Behavioral Core Facility, Tel Aviv University Faculty of Medicine, Tel Aviv, Israel
- School of Behavioral Sciences, Netanya Academic College, Netanya, Israel
| | - Chaim G Pick
- Department of Anatomy and Anthropology, Tel Aviv University Faculty of Medicine, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Sylvan Adams Sports Institute, Tel Aviv University, Tel Aviv, Israel
- Dr. Miriam and Sheldon G. Adelson Chair and Center for the Biology of Addictive Diseases, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
3
|
Huerta de la Cruz S, Santiago-Castañeda C, Rodríguez-Palma EJ, Rocha L, Sancho M. Lateral fluid percussion injury: A rat model of experimental traumatic brain injury. Methods Cell Biol 2024; 185:197-224. [PMID: 38556449 DOI: 10.1016/bs.mcb.2024.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
Traumatic brain injury (TBI) represents one of the leading causes of disability and death worldwide. The annual economic impact of TBI-including direct and indirect costs-is high, particularly impacting low- and middle-income countries. Despite extensive research, a comprehensive understanding of the primary and secondary TBI pathophysiology, followed by the development of promising therapeutic approaches, remains limited. These fundamental caveats in knowledge have motivated the development of various experimental models to explore the molecular mechanisms underpinning the pathogenesis of TBI. In this context, the Lateral Fluid Percussion Injury (LFPI) model produces a brain injury that mimics most of the neurological and systemic aspects observed in human TBI. Moreover, its high reproducibility makes the LFPI model one of the most widely used rodent-based TBI models. In this chapter, we provide a detailed surgical protocol of the LFPI model used to induce TBI in adult Wistar rats. We further highlight the neuroscore test as a valuable tool for the evaluation of TBI-induced sensorimotor consequences and their severity in rats. Lastly, we briefly summarize the current knowledge on the pathological aspects and functional outcomes observed in the LFPI-induced TBI model in rodents.
Collapse
Affiliation(s)
- Saúl Huerta de la Cruz
- Department of Pharmacology, University of Vermont, Burlington, VT, United States; Departamento de Farmacobiología, Cinvestav Sede Sur, Ciudad de México, México.
| | | | - Erick J Rodríguez-Palma
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, Sede Sur, Mexico City, Mexico
| | - Luisa Rocha
- Departamento de Farmacobiología, Cinvestav Sede Sur, Ciudad de México, México
| | - Maria Sancho
- Department of Pharmacology, University of Vermont, Burlington, VT, United States; Department of Physiology, Faculty of Medicine, Universidad Complutense de Madrid, Madrid, Spain.
| |
Collapse
|
4
|
Bharadwaj VN, Sahbaie P, Shi X, Irvine KA, Yeomans DC, Clark JD. Effect of Voluntary Exercise on Endogenous Pain Control Systems and Post-traumatic Headache in Mice. THE JOURNAL OF PAIN 2023; 24:1859-1874. [PMID: 37271350 PMCID: PMC11806938 DOI: 10.1016/j.jpain.2023.05.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 05/16/2023] [Accepted: 05/30/2023] [Indexed: 06/06/2023]
Abstract
Traumatic brain injury (TBI) can cause acute and chronic pain along with motor, cognitive, and emotional problems. Although the mechanisms are poorly understood, previous studies suggest disruptions in endogenous pain modulation may be involved. Voluntary exercise after a TBI has been shown to reduce some consequences of injury including cognitive impairment. We hypothesized, therefore, that voluntary exercise could augment endogenous pain control systems in a rodent model of TBI. For these studies, we used a closed-head impact procedure in male mice modeling mild TBI. We investigated the effect of voluntary exercise on TBI-induced hindpaw nociceptive sensitization, diffuse noxious inhibitory control failure, and periorbital sensitization after bright light stress, a model of post-traumatic headache. Furthermore, we investigated the effects of exercise on memory, circulating markers of brain injury, neuroinflammation, and spinal cord gene expression. We observed that exercise significantly reduced TBI-induced hindpaw allodynia and periorbital allodynia in the first week following TBI. We also showed that exercise improved the deficits associated with diffuse noxious inhibitory control and reduced bright light stress-induced allodynia up to 2 months after TBI. In addition, exercise preserved memory and reduced TBI-induced increases in spinal BDNF, CXCL1, CXCL2, and prodynorphin expression, all genes previously linked to TBI-induced nociceptive sensitization. Taken together, our observations suggest that voluntary exercise may reduce pain after TBI by reducing TBI-induced changes in nociceptive signaling and preserving endogenous pain control systems. PERSPECTIVE: This article evaluates the effects of exercise on pain-related behaviors in a preclinical model of traumatic brain injury (TBI). The findings show that exercise reduces nociceptive sensitization, loss of diffuse noxious inhibitory control, memory deficits, and spinal nociception-related gene expression after TBI. Exercise may reduce or prevent pain after TBI.
Collapse
Affiliation(s)
- Vimala N Bharadwaj
- Department of Anesthesia, Perioperative and Pain Medicine, Stanford University, School of Medicine, Stanford, California.
| | - Peyman Sahbaie
- Department of Anesthesia, Perioperative and Pain Medicine, Stanford University, School of Medicine, Stanford, California; Anesthesiology Service, Veterans Affairs Palo Alto Health Care System, Palo Alto, California
| | - Xiaoyou Shi
- Department of Anesthesia, Perioperative and Pain Medicine, Stanford University, School of Medicine, Stanford, California; Anesthesiology Service, Veterans Affairs Palo Alto Health Care System, Palo Alto, California
| | - Karen-Amanda Irvine
- Department of Anesthesia, Perioperative and Pain Medicine, Stanford University, School of Medicine, Stanford, California; Anesthesiology Service, Veterans Affairs Palo Alto Health Care System, Palo Alto, California
| | - David C Yeomans
- Department of Anesthesia, Perioperative and Pain Medicine, Stanford University, School of Medicine, Stanford, California
| | - J David Clark
- Department of Anesthesia, Perioperative and Pain Medicine, Stanford University, School of Medicine, Stanford, California; Anesthesiology Service, Veterans Affairs Palo Alto Health Care System, Palo Alto, California
| |
Collapse
|
5
|
Deshetty UM, Periyasamy P. Potential Biomarkers in Experimental Animal Models for Traumatic Brain Injury. J Clin Med 2023; 12:3923. [PMID: 37373618 DOI: 10.3390/jcm12123923] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/05/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Traumatic brain injury (TBI) is a complex and multifaceted disorder that has become a significant public health concern worldwide due to its contribution to mortality and morbidity. This condition encompasses a spectrum of injuries, including axonal damage, contusions, edema, and hemorrhage. Unfortunately, specific effective therapeutic interventions to improve patient outcomes following TBI are currently lacking. Various experimental animal models have been developed to mimic TBI and evaluate potential therapeutic agents to address this issue. These models are designed to recapitulate different biomarkers and mechanisms involved in TBI. However, due to the heterogeneous nature of clinical TBI, no single experimental animal model can effectively mimic all aspects of human TBI. Accurate emulation of clinical TBI mechanisms is also tricky due to ethical considerations. Therefore, the continued study of TBI mechanisms and biomarkers, of the duration and severity of brain injury, treatment strategies, and animal model optimization is necessary. This review focuses on the pathophysiology of TBI, available experimental TBI animal models, and the range of biomarkers and detection methods for TBI. Overall, this review highlights the need for further research to improve patient outcomes and reduce the global burden of TBI.
Collapse
Affiliation(s)
- Uma Maheswari Deshetty
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Palsamy Periyasamy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
6
|
Freidin D, Har-Even M, Rubovitch V, Murray KE, Maggio N, Shavit-Stein E, Keidan L, Citron BA, Pick CG. Cognitive and Cellular Effects of Combined Organophosphate Toxicity and Mild Traumatic Brain Injury. Biomedicines 2023; 11:1481. [PMID: 37239152 PMCID: PMC10216664 DOI: 10.3390/biomedicines11051481] [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: 03/13/2023] [Revised: 05/12/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
Traumatic brain injury (TBI) is considered the most common neurological disorder among people under the age of 50. In modern combat zones, a combination of TBI and organophosphates (OP) can cause both fatal and long-term effects on the brain. We utilized a mouse closed-head TBI model induced by a weight drop device, along with OP exposure to paraoxon. Spatial and visual memory as well as neuron loss and reactive astrocytosis were measured 30 days after exposure to mild TBI (mTBI) and/or paraoxon. Molecular and cellular changes were assessed in the temporal cortex and hippocampus. Cognitive and behavioral deficits were most pronounced in animals that received a combination of paraoxon exposure and mTBI, suggesting an additive effect of the insults. Neuron survival was reduced in proximity to the injury site after exposure to paraoxon with or without mTBI, whereas in the dentate gyrus hilus, cell survival was only reduced in mice exposed to paraoxon prior to sustaining a mTBI. Neuroinflammation was increased in the dentate gyrus in all groups exposed to mTBI and/or to paraoxon. Astrocyte morphology was significantly changed in mice exposed to paraoxon prior to sustaining an mTBI. These results provide further support for assumptions concerning the effects of OP exposure following the Gulf War. This study reveals additional insights into the potentially additive effects of OP exposure and mTBI, which may result in more severe brain damage on the modern battlefield.
Collapse
Affiliation(s)
- Dor Freidin
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; (D.F.); (M.H.-E.)
| | - Meirav Har-Even
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; (D.F.); (M.H.-E.)
| | - Vardit Rubovitch
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; (D.F.); (M.H.-E.)
| | - Kathleen E. Murray
- Laboratory of Molecular Biology, VA New Jersey Health Care System, Research & Development, East Orange, NJ 07018, USA
- Rutgers School of Graduate Studies, Newark, NJ 07103, USA
| | - Nicola Maggio
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan 52626202, Israel
- Department of Neurology and Neurosurgery, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Efrat Shavit-Stein
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan 52626202, Israel
- Department of Neurology and Neurosurgery, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Lee Keidan
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; (D.F.); (M.H.-E.)
| | - Bruce A. Citron
- Laboratory of Molecular Biology, VA New Jersey Health Care System, Research & Development, East Orange, NJ 07018, USA
- Rutgers School of Graduate Studies, Newark, NJ 07103, USA
- Department of Pharmacology, Physiology & Neuroscience, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Chaim G. Pick
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; (D.F.); (M.H.-E.)
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
- Sylvan Adams Sports Institute, Tel Aviv University, Tel Aviv 6997801, Israel
- The Dr. Miriam and Sheldon G. Adelson Chair and Center for the Biology of Addictive Diseases, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
7
|
Danis A, Baranoglu Kilinc Y, Torun IE, Hanci F, Kilinc E, Ankarali H. Esculetin alleviates pentylenetetrazole-induced seizures, cognitive impairment and pro-inflammatory cytokines and suppresses penicillin-induced epileptiform activity in rats. Life Sci 2023; 313:121300. [PMID: 36535399 DOI: 10.1016/j.lfs.2022.121300] [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: 09/21/2022] [Revised: 12/07/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
AIMS To investigate the effects of different doses of esculetin on epileptiform activity, behavioral seizures, memory impairment, and cortical and hippocampal NF-κB, as a mediator of pro-inflammatory gene induction, and pro-inflammatory cytokines in penicillin- and pentylenetetrazole(PTZ)-induced seizure models in rats. MAIN METHODS Different doses of esculetin (5, 10, and 20 mg/kg), and diazepam (5 mg/kg) as a positive control, were tested in penicillin- and pentylenetetrazole(PTZ)-induced seizure models. In the PTZ model, cognitive function, behavioral seizures, and cortical and hippocampal pro-inflammatory biomarkers and survival factor was evaluated. In the penicillin model, the frequency and amplitude of electrophysiological epileptiform activity were assessed. KEY FINDINGS In the PTZ model, the 10 mg/kg esculetin displayed anticonvulsant effects by extending onset-times of myoclonic-jerk and generalized tonic-clonic seizure, and by diminishing seizure severity and duration of generalized tonic-clonic seizure. It also ameliorated PTZ-induced cognitive impairment, and cortical and hippocampal activin-A, IL-1β, IL-6 and NF-κB levels. In the penicillin model, the 10 mg/kg esculetin decreased the frequency of spikes without changing the amplitude of spikes. As a positive-control, diazepam reversed all changes induced by both PTZ and penicillin. SIGNIFICANCE Esculetin exhibits anticonvulsant and memory-improving effects by alleviating the behavioral and electrophysiological characteristics of epileptic seizures. Additionally, esculetin exerts anti-neuroinflammatory actions in the PTZ-induced seizures model. Thus, esculetin may be a multi-targeted promising agent with anticonvulsant and anti-neuroinflammatory effects in the treatment of epilepsy.
Collapse
Affiliation(s)
- Aysegul Danis
- Department of Child Neurology, Bolu Abant Izzet Baysal University, Bolu, Turkey
| | | | - Ibrahim Ethem Torun
- Department of Physiology, Bolu Abant Izzet Baysal University, Medical Faculty, Bolu, Turkey
| | - Fatma Hanci
- Department of Child Neurology, Bolu Abant Izzet Baysal University, Bolu, Turkey
| | - Erkan Kilinc
- Department of Physiology, Bolu Abant Izzet Baysal University, Medical Faculty, Bolu, Turkey.
| | - Handan Ankarali
- Department of Biostatistics and Medical Informatics, Istanbul Medeniyet University, Istanbul, Turkey
| |
Collapse
|
8
|
Zhao Y, Mu H, Huang Y, Li S, Wang Y, Stetler RA, Bennett MVL, Dixon CE, Chen J, Shi Y. Microglia-specific deletion of histone deacetylase 3 promotes inflammation resolution, white matter integrity, and functional recovery in a mouse model of traumatic brain injury. J Neuroinflammation 2022; 19:201. [PMID: 35933343 PMCID: PMC9357327 DOI: 10.1186/s12974-022-02563-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 07/29/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Histone deacetylases (HDACs) are believed to exacerbate traumatic brain injury (TBI) based on studies using pan-HDAC inhibitors. However, the HDAC isoform responsible for the detrimental effects and the cell types involved remain unknown, which may hinder the development of specific targeting strategies that boost therapeutic efficacy while minimizing side effects. Microglia are important mediators of post-TBI neuroinflammation and critically impact TBI outcome. HDAC3 was reported to be essential to the inflammatory program of in vitro cultured macrophages, but its role in microglia and in the post-TBI brain has not been investigated in vivo. METHODS We generated HDAC3LoxP mice and crossed them with CX3CR1CreER mice, enabling in vivo conditional deletion of HDAC3. Microglia-specific HDAC3 knockout (HDAC3 miKO) was induced in CX3CR1CreER:HDAC3LoxP mice with 5 days of tamoxifen treatment followed by a 30-day development interval. The effects of HDAC3 miKO on microglial phenotype and neuroinflammation were examined 3-5 days after TBI induced by controlled cortical impact. Neurological deficits and the integrity of white matter were assessed for 6 weeks after TBI by neurobehavioral tests, immunohistochemistry, electron microscopy, and electrophysiology. RESULTS HDAC3 miKO mice harbored specific deletion of HDAC3 in microglia but not in peripheral monocytes. HDAC3 miKO reduced the number of microglia by 26%, but did not alter the inflammation level in the homeostatic brain. After TBI, proinflammatory microglial responses and brain inflammation were markedly alleviated by HDAC3 miKO, whereas the infiltration of blood immune cells was unchanged, suggesting a primary effect of HDAC3 miKO on modulating microglial phenotype. Importantly, HDAC3 miKO was sufficient to facilitate functional recovery for 6 weeks after TBI. TBI-induced injury to axons and myelin was ameliorated, and signal conduction by white matter fiber tracts was significantly enhanced in HDAC3 miKO mice. CONCLUSION Using a novel microglia-specific conditional knockout mouse model, we delineated for the first time the role of microglial HDAC3 after TBI in vivo. HDAC3 miKO not only reduced proinflammatory microglial responses, but also elicited long-lasting improvement of white matter integrity and functional recovery after TBI. Microglial HDAC3 is therefore a promising therapeutic target to improve long-term outcomes after TBI.
Collapse
Affiliation(s)
- Yongfang Zhao
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Hongfeng Mu
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Yichen Huang
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Sicheng Li
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Yangfan Wang
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - R Anne Stetler
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15261, USA
| | - Michael V L Bennett
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - C Edward Dixon
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15261, USA
| | - Jun Chen
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15261, USA.
| | - Yejie Shi
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
9
|
Ratliff WA, Saykally JN, Keeley KL, Driscoll DC, Murray KE, Okuka M, Mervis RF, Delic V, Citron BA. Sidestream Smoke Affects Dendritic Complexity and Astrocytes After Model Mild Closed Head Traumatic Brain Injury. Cell Mol Neurobiol 2022; 42:1453-1463. [PMID: 33417143 PMCID: PMC8263792 DOI: 10.1007/s10571-020-01036-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 12/28/2020] [Indexed: 11/26/2022]
Abstract
Mild traumatic brain injuries can have long-term consequences that interfere with the life of the patient and impose a burden on our health care system. Oxidative stress has been identified as a contributing factor for the progression of neurodegeneration following TBI. A major source of oxidative stress for many veterans is cigarette smoking and second-hand smoke, which has been shown to have an effect on TBI recovery. To examine the potential influences of second-hand smoke during recovery from TBI, we utilized a mouse model of closed head injury, followed by repeated exposure to cigarette smoke and treatment with a neuroprotective antioxidant. We found that neither the mild injuries nor the smoke exposure produced axonal damage detectable with amino cupric silver staining. However, complexity in the dendritic arbors was significantly reduced after mild TBI plus smoke exposure. In the hippocampus, there were astrocytic responses, including Cyp2e1 upregulation, after the injury and tobacco smoke insult. This study provides useful context for the importance of lifestyle changes, such as reducing or eliminating cigarette smoking, during recovery from TBI.
Collapse
Affiliation(s)
- Whitney A Ratliff
- Laboratory of Molecular Biology, Research and Development 151, Bay Pines VA Healthcare System, Bay Pines, FL, 33744, USA
- Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA
| | - Jessica N Saykally
- Laboratory of Molecular Biology, Research and Development 151, Bay Pines VA Healthcare System, Bay Pines, FL, 33744, USA
- Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA
| | - Kristen L Keeley
- Laboratory of Molecular Biology, Research and Development 151, Bay Pines VA Healthcare System, Bay Pines, FL, 33744, USA
- Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA
| | - David C Driscoll
- Laboratory of Molecular Biology, Research and Development 151, Bay Pines VA Healthcare System, Bay Pines, FL, 33744, USA
| | - Kathleen E Murray
- VA New Jersey Health Care System, Research & Development, East Orange, NJ, 07018, USA
- Department of Pharmacology, Physiology, & Neuroscience, Rutgers - New Jersey Medical School, Newark, NJ, 07103, USA
| | - Maja Okuka
- Department of Obstetrics and Gynecology, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA
| | | | - Vedad Delic
- VA New Jersey Health Care System, Research & Development, East Orange, NJ, 07018, USA
- Department of Pharmacology, Physiology, & Neuroscience, Rutgers - New Jersey Medical School, Newark, NJ, 07103, USA
| | - Bruce A Citron
- Laboratory of Molecular Biology, Research and Development 151, Bay Pines VA Healthcare System, Bay Pines, FL, 33744, USA.
- Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA.
- VA New Jersey Health Care System, Research & Development, East Orange, NJ, 07018, USA.
- Department of Pharmacology, Physiology, & Neuroscience, Rutgers - New Jersey Medical School, Newark, NJ, 07103, USA.
| |
Collapse
|
10
|
Qubty D, Frid K, Har-Even M, Rubovitch V, Gabizon R, Pick CG. Nano-PSO Administration Attenuates Cognitive and Neuronal Deficits Resulting from Traumatic Brain Injury. Molecules 2022; 27:molecules27092725. [PMID: 35566074 PMCID: PMC9105273 DOI: 10.3390/molecules27092725] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 11/17/2022] Open
Abstract
Traumatic Brain Injury (TBI), is one of the most common causes of neurological damage in young populations. It is widely considered as a risk factor for neurodegenerative diseases, such as Alzheimer’s disease (AD) and Parkinson’s (PD) disease. These diseases are characterized in part by the accumulation of disease-specific misfolded proteins and share common pathological features, such as neuronal death, as well as inflammatory and oxidative damage. Nano formulation of Pomegranate seed oil [Nano-PSO (Granagard TM)] has been shown to target its active ingredient to the brain and thereafter inhibit memory decline and neuronal death in mice models of AD and genetic Creutzfeldt Jacob disease. In this study, we show that administration of Nano-PSO to mice before or after TBI application prevents cognitive and behavioral decline. In addition, immuno-histochemical staining of the brain indicates that preventive Nano-PSO treatment significantly decreased neuronal death, reduced gliosis and prevented mitochondrial damage in the affected cells. Finally, we examined levels of Sirtuin1 (SIRT1) and Synaptophysin (SYP) in the cortex using Western blotting. Nano-PSO consumption led to higher levels of SIRT1 and SYP protein postinjury. Taken together, our results indicate that Nano-PSO, as a natural brain-targeted antioxidant, can prevent part of TBI-induced damage.
Collapse
Affiliation(s)
- Doaa Qubty
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; (D.Q.); (M.H.-E.); (V.R.)
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Kati Frid
- The Agnes Ginges Center for Human Neurogenetics, Department of Neurology, Hadassah University Hospital, Medical School, The Hebrew University, Jerusalem 91120, Israel; (K.F.); (R.G.)
| | - Meirav Har-Even
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; (D.Q.); (M.H.-E.); (V.R.)
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
- Sylvan Adams Sports Institute, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Vardit Rubovitch
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; (D.Q.); (M.H.-E.); (V.R.)
| | - Ruth Gabizon
- The Agnes Ginges Center for Human Neurogenetics, Department of Neurology, Hadassah University Hospital, Medical School, The Hebrew University, Jerusalem 91120, Israel; (K.F.); (R.G.)
| | - Chaim G Pick
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; (D.Q.); (M.H.-E.); (V.R.)
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
- Sylvan Adams Sports Institute, Tel Aviv University, Tel Aviv 6997801, Israel
- The Dr. Miriam and Sheldon G. Adelson Chair and Center for the Biology of Addictive Diseases, Tel Aviv University, Tel Aviv 6997801, Israel
- Correspondence:
| |
Collapse
|
11
|
Fan L, Xu H, Su J, Qin J, Gao K, Ou M, Peng S, Shen H, Li N. Discriminating mild traumatic brain injury using sparse dictionary learning of functional network dynamics. Brain Behav 2021; 11:e2414. [PMID: 34775693 PMCID: PMC8671791 DOI: 10.1002/brb3.2414] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 09/23/2021] [Accepted: 10/13/2021] [Indexed: 11/06/2022] Open
Abstract
Mild traumatic brain injury (mTBI) is usually caused by a bump, blow, or jolt to the head or penetrating head injury, and carries the risk of inducing cognitive disorders. However, identifying the biomarkers for the diagnosis of mTBI is challenging as evident abnormalities in brain anatomy are rarely found in patients with mTBI. In this study, we tested whether the alteration of functional network dynamics could be used as potential biomarkers to better diagnose mTBI. We propose a sparse dictionary learning framework to delineate spontaneous fluctuation of functional connectivity into the subject-specific time-varying evolution of a set of overlapping group-level sparse connectivity components (SCCs) based on the resting-state functional magnetic resonance imaging (fMRI) data from 31 mTBI patients in the early acute phase (<3 days postinjury) and 31 healthy controls (HCs). The identified SCCs were consistently distributed in the cohort of subjects without significant inter-group differences in connectivity patterns. Nevertheless, subject-specific temporal expression of these SCCs could be used to discriminate patients with mTBI from HCs with a classification accuracy of 74.2% (specificity 64.5% and sensitivity 83.9%) using leave-one-out cross-validation. Taken together, our findings indicate neuroimaging biomarkers for mTBI individual diagnosis based on the temporal expression of SCCs underlying time-resolved functional connectivity.
Collapse
Affiliation(s)
- Liangwei Fan
- College of Intelligence Science and Technology, National University of Defense Technology, Changsha, China
| | - Huaze Xu
- College of Intelligence Science and Technology, National University of Defense Technology, Changsha, China
| | - Jianpo Su
- College of Intelligence Science and Technology, National University of Defense Technology, Changsha, China
| | - Jian Qin
- College of Intelligence Science and Technology, National University of Defense Technology, Changsha, China
| | - Kai Gao
- College of Intelligence Science and Technology, National University of Defense Technology, Changsha, China
| | - Min Ou
- College of Intelligence Science and Technology, National University of Defense Technology, Changsha, China
| | - Song Peng
- Radiology Department, Xiangya 3rd Hospital, Central South University, Changsha, China
| | - Hui Shen
- College of Intelligence Science and Technology, National University of Defense Technology, Changsha, China
| | - Na Li
- Radiology Department, Xiangya 3rd Hospital, Central South University, Changsha, China
| |
Collapse
|
12
|
Qubty D, Schreiber S, Rubovitch V, Boag A, Pick CG. No Significant Effects of Cellphone Electromagnetic Radiation on Mice Memory or Anxiety: Some Mixed Effects on Traumatic Brain Injured Mice. Neurotrauma Rep 2021; 2:381-390. [PMID: 34723249 PMCID: PMC8550818 DOI: 10.1089/neur.2021.0009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Current literature details an array of contradictory results regarding the effect of radiofrequency electromagnetic radiation (RF-EMR) on health, both in humans and in animal models. The present study was designed to ascertain the conflicting data published regarding the possible impact of cellular exposure (radiation) on male and female mice as far as spatial memory, anxiety, and general well-being is concerned. To increase the likelihood of identifying possible "subtle" effects, we chose to test it in already cognitively impaired (following mild traumatic brain injury; mTBI) mice. Exposure to cellular radiation by itself had no significant impact on anxiety levels or spatial/visual memory in mice. When examining the dual impact of mTBI and cellular radiation on anxiety, no differences were found in the anxiety-like behavior as seen at the elevated plus maze (EPM). When exposed to both mTBI and cellular radiation, our results show improvement of visual memory impairment in both female and male mice, but worsening of the spatial memory of female mice. These results do not allow for a decisive conclusion regarding the possible hazards of cellular radiation on brain function in mice, and the mTBI did not facilitate identification of subtle effects by augmenting them.
Collapse
Affiliation(s)
- Doaa Qubty
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shaul Schreiber
- Department of Psychiatry, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Vardit Rubovitch
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Amir Boag
- School of Electrical Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Chaim G Pick
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Psychiatry, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,The Dr. Miriam and Sheldon G. Adelson Center for the Biology of Addictive Diseases, Tel Aviv University, Tel Aviv, Israel.,Sylvan Adams Sports Institute, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
13
|
Traumatic Brain Injury: An Age-Dependent View of Post-Traumatic Neuroinflammation and Its Treatment. Pharmaceutics 2021; 13:pharmaceutics13101624. [PMID: 34683918 PMCID: PMC8537402 DOI: 10.3390/pharmaceutics13101624] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/24/2021] [Accepted: 09/26/2021] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability all over the world. TBI leads to (1) an inflammatory response, (2) white matter injuries and (3) neurodegenerative pathologies in the long term. In humans, TBI occurs most often in children and adolescents or in the elderly, and it is well known that immune responses and the neuroregenerative capacities of the brain, among other factors, vary over a lifetime. Thus, age-at-injury can influence the consequences of TBI. Furthermore, age-at-injury also influences the pharmacological effects of drugs. However, the post-TBI inflammatory, neuronal and functional consequences have been mostly studied in experimental young adult animal models. The specificity and the mechanisms underlying the consequences of TBI and pharmacological responses are poorly understood in extreme ages. In this review, we detail the variations of these age-dependent inflammatory responses and consequences after TBI, from an experimental point of view. We investigate the evolution of microglial, astrocyte and other immune cells responses, and the consequences in terms of neuronal death and functional deficits in neonates, juvenile, adolescent and aged male animals, following a single TBI. We also describe the pharmacological responses to anti-inflammatory or neuroprotective agents, highlighting the need for an age-specific approach to the development of therapies of TBI.
Collapse
|
14
|
Faillot M, Chaillet A, Palfi S, Senova S. Rodent models used in preclinical studies of deep brain stimulation to rescue memory deficits. Neurosci Biobehav Rev 2021; 130:410-432. [PMID: 34437937 DOI: 10.1016/j.neubiorev.2021.08.012] [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: 02/08/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 11/28/2022]
Abstract
Deep brain stimulation paradigms might be used to treat memory disorders in patients with stroke or traumatic brain injury. However, proof of concept studies in animal models are needed before clinical translation. We propose here a comprehensive review of rodent models for Traumatic Brain Injury and Stroke. We systematically review the histological, behavioral and electrophysiological features of each model and identify those that are the most relevant for translational research.
Collapse
Affiliation(s)
- Matthieu Faillot
- Neurosurgery department, Henri Mondor University Hospital, APHP, DMU CARE, Université Paris Est Créteil, Mondor Institute for Biomedical Research, INSERM U955, Team 15, Translational Neuropsychiatry, France
| | - Antoine Chaillet
- Laboratoire des Signaux et Systèmes (L2S-UMR8506) - CentraleSupélec, Université Paris Saclay, Institut Universitaire de France, France
| | - Stéphane Palfi
- Neurosurgery department, Henri Mondor University Hospital, APHP, DMU CARE, Université Paris Est Créteil, Mondor Institute for Biomedical Research, INSERM U955, Team 15, Translational Neuropsychiatry, France
| | - Suhan Senova
- Neurosurgery department, Henri Mondor University Hospital, APHP, DMU CARE, Université Paris Est Créteil, Mondor Institute for Biomedical Research, INSERM U955, Team 15, Translational Neuropsychiatry, France.
| |
Collapse
|
15
|
Santana-Gomez CE, Medel-Matus JS, Rundle BK. Animal models of post-traumatic epilepsy and their neurobehavioral comorbidities. Seizure 2021; 90:9-16. [DOI: 10.1016/j.seizure.2021.05.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 05/07/2021] [Accepted: 05/09/2021] [Indexed: 12/30/2022] Open
|
16
|
Yang Q, Yu J, Qin H, Liu L, Di C, Zhuang Q, Yin H. Irbesartan suppresses lipopolysaccharide (LPS)-induced blood-brain barrier (BBB) dysfunction by inhibiting the activation of MLCK/MLC. Int Immunopharmacol 2021; 98:107834. [PMID: 34174702 DOI: 10.1016/j.intimp.2021.107834] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 12/26/2022]
Abstract
The basic function of the blood-brain barrier (BBB) is to selectively regulate the infiltration of solutes from the circulating blood into the central nervous system (CNS). Impaired BBB activity is related to brain damage caused by stroke, traumatic injury, neurodegenerative diseases, etc. Comprised of a monolayer of endothelial cells, the integrity of the BBB is determined by the expression of tight junction proteins and the contractile activity of the perijunctional apical actomyosin ring. Irbesartan, an AT1R antagonist, has been widely used for the treatment of hypertension. However, the pharmacological function of Irbesartan in the balance of the BBB is still unknown. In the present study, we performed both in-vivo and in-vitro experiments using lipopolysaccharide (LPS) to explore the mechanism behind the protective effects of Irbesartan against the BBB impairment. The results of our mouse model study revealed that Irbesartan could reduce BBB permeability, restore the expression of Occludin, and suppress the expression of inflammatory mediators, including interleukin-6, monocyte chemoattractant protein-1, and intercellular adhesion molecule-1. Additionally, Irbesartan improved LPS-induced depressive-like behavior. In our in vitro experiments, human brain microvascular endothelial cells (HBMVECs) stimulated with LPS demonstrated decreased endothelial permeability and increased occludin expression in response to Irbesartan treatment. Importantly, we found that the protective effects of Irbesartan were mediated through the NF-κB/MLC/MLCK signaling pathway, as blockage of NF-κB abolished the effects of Irbesartan. Our findings provide a basis for further research into the neuroprotective mechanism of Irbesartan.
Collapse
Affiliation(s)
- Qixia Yang
- Department of Pharmacy, Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277100, China
| | - Juanjuan Yu
- Department of Pharmacy, Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277100, China
| | - Hao Qin
- Department of Neurosurgery, Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277100, China
| | - Long Liu
- Department of Neurosurgery, Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277100, China
| | - Chao Di
- Department of Neurosurgery, Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277100, China
| | - Qiang Zhuang
- Department of Neurosurgery, Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277100, China
| | - Hang Yin
- Department of Neurosurgery, Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277100, China.
| |
Collapse
|
17
|
Fakhoury M, Shakkour Z, Kobeissy F, Lawand N. Depression following traumatic brain injury: a comprehensive overview. Rev Neurosci 2020; 32:289-303. [PMID: 33661587 DOI: 10.1515/revneuro-2020-0037] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 10/21/2020] [Indexed: 12/19/2022]
Abstract
Traumatic brain injury (TBI) represents a major health concern affecting the neuropsychological health; TBI is accompanied by drastic long-term adverse complications that can influence many aspects of the life of affected individuals. A substantial number of studies have shown that mood disorders, particularly depression, are the most frequent complications encountered in individuals with TBI. Post-traumatic depression (P-TD) is present in approximately 30% of individuals with TBI, with the majority of individuals experiencing symptoms of depression during the first year following head injury. To date, the mechanisms of P-TD are far from being fully understood, and effective treatments that completely halt this condition are still lacking. The aim of this review is to outline the current state of knowledge on the prevalence and risk factors of P-TD, to discuss the accompanying brain changes at the anatomical, molecular and functional levels, and to discuss current approaches used for the treatment of P-TD.
Collapse
Affiliation(s)
- Marc Fakhoury
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Zaynab Shakkour
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Nada Lawand
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,Department of Neurology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
18
|
Hoffman J, Yu J, Kirstein C, Kindy MS. Combined Effects of Repetitive Mild Traumatic Brain Injury and Alcohol Drinking on the Neuroinflammatory Cytokine Response and Cognitive Behavioral Outcomes. Brain Sci 2020; 10:brainsci10110876. [PMID: 33228251 PMCID: PMC7699568 DOI: 10.3390/brainsci10110876] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/09/2020] [Accepted: 11/13/2020] [Indexed: 12/22/2022] Open
Abstract
The relationship between alcohol consumption and traumatic brain injury (TBI) often focuses on alcohol consumption increasing the likelihood of incurring a TBI, rather than alcohol use outcomes after TBI. However, patients without a history of an alcohol use disorder can also show increased problem drinking after single or multiple TBIs. Alcohol and mild TBI share diffuse deleterious neurological impacts and cognitive impairments; therefore, the purpose of these studies was to determine if an interaction on brain and behavior outcomes occurs when alcohol is consumed longitudinally after TBI. To examine the impact of mild repetitive TBI (rmTBI) on voluntary alcohol consumption, mice were subjected to four mild TBI or sham procedures over a 2 week period, then offered alcohol (20% v/v) for 2 weeks using the two-bottle choice, drinking in the dark protocol. Following the drinking period, mice were evaluated for neuroinflammatory cytokine response or tested for cognitive and behavioral deficits. Results indicate no difference in alcohol consumption or preference following rmTBI as compared to sham; however, increases in the neuroinflammatory cytokine response due to alcohol consumption and some mild cognitive behavioral deficits after rmTBI and alcohol consumption were observed. These data suggest that the cytokine response to alcohol drinking and rmTBI + alcohol drinking is not necessarily aggregate, but the combination does result in an exacerbation of cognitive behavioral outcomes.
Collapse
Affiliation(s)
- Jessica Hoffman
- Department of Psychiatry, Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Correspondence: (J.H.); (M.S.K.); Tel.: +1-919-843-4389 (J.H.)
| | - Jin Yu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA;
| | - Cheryl Kirstein
- Department of Psychology, College of Arts and Sciences, University of South Florida, Tampa, FL 33612, USA;
| | - Mark S. Kindy
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA;
- James A. Haley VA Medical Center, Tampa, FL 33612, USA
- Shriners Hospital for Children, Tampa, FL 33612, USA
- Correspondence: (J.H.); (M.S.K.); Tel.: +1-919-843-4389 (J.H.)
| |
Collapse
|
19
|
Kempuraj D, Ahmed ME, Selvakumar GP, Thangavel R, Raikwar SP, Zaheer SA, Iyer SS, Govindarajan R, Nattanmai Chandrasekaran P, Burton C, James D, Zaheer A. Acute Traumatic Brain Injury-Induced Neuroinflammatory Response and Neurovascular Disorders in the Brain. Neurotox Res 2020; 39:359-368. [PMID: 32955722 PMCID: PMC7502806 DOI: 10.1007/s12640-020-00288-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 09/12/2020] [Accepted: 09/14/2020] [Indexed: 02/07/2023]
Abstract
Acute traumatic brain injury (TBI) leads to neuroinflammation, neurodegeneration, cognitive decline, psychological disorders, increased blood-brain barrier (BBB) permeability, and microvascular damage in the brain. Inflammatory mediators secreted from activated glial cells, neurons, and mast cells are implicated in the pathogenesis of TBI through secondary brain damage. Abnormalities or damage to the neurovascular unit is the indication of secondary injuries in the brain after TBI. However, the precise mechanisms of molecular and ultrastructural neurovascular alterations involved in the pathogenesis of acute TBI are not yet clearly understood. Moreover, currently, there are no precision-targeted effective treatment options to prevent the sequelae of TBI. In this study, mice were subjected to closed head weight-drop-induced acute TBI and evaluated neuroinflammatory and neurovascular alterations in the brain by immunofluorescence staining or quantitation by enzyme-linked immunosorbent assay (ELISA) procedure. Mast cell stabilizer drug cromolyn was administered to inhibit the neuroinflammatory response of TBI. Results indicate decreased level of pericyte marker platelet-derived growth factor receptor-beta (PDGFR-β) and BBB-associated tight junction proteins junctional adhesion molecule-A (JAM-A) and zonula occludens-1 (ZO-1) in the brains 7 days after weight-drop-induced acute TBI as compared with the brains from sham control mice indicating acute TBI-associated BBB/tight junction protein disruption. Further, the administration of cromolyn drug significantly inhibited acute TBI-associated decrease of PDGFR-β, JAM-A, and ZO-1 in the brain. These findings suggest that acute TBI causes BBB/tight junction damage and that cromolyn administration could protect this acute TBI-induced brain damage as well as its long-time consequences.
Collapse
Affiliation(s)
- Duraisamy Kempuraj
- Department of Neurology, School of Medicine, University of Missouri, 1 Hospital Drive, Columbia, MO, USA. .,The Center for Translational Neuroscience, School of Medicine, University of Missouri, 1 Hospital Drive, Columbia, MO, USA. .,Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, MO, USA.
| | - Mohammad Ejaz Ahmed
- Department of Neurology, School of Medicine, University of Missouri, 1 Hospital Drive, Columbia, MO, USA.,The Center for Translational Neuroscience, School of Medicine, University of Missouri, 1 Hospital Drive, Columbia, MO, USA.,Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, MO, USA
| | - Govindhasamy Pushpavathi Selvakumar
- Department of Neurology, School of Medicine, University of Missouri, 1 Hospital Drive, Columbia, MO, USA.,The Center for Translational Neuroscience, School of Medicine, University of Missouri, 1 Hospital Drive, Columbia, MO, USA.,Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, MO, USA
| | - Ramasamy Thangavel
- Department of Neurology, School of Medicine, University of Missouri, 1 Hospital Drive, Columbia, MO, USA.,The Center for Translational Neuroscience, School of Medicine, University of Missouri, 1 Hospital Drive, Columbia, MO, USA.,Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, MO, USA
| | - Sudhanshu P Raikwar
- Department of Neurology, School of Medicine, University of Missouri, 1 Hospital Drive, Columbia, MO, USA.,The Center for Translational Neuroscience, School of Medicine, University of Missouri, 1 Hospital Drive, Columbia, MO, USA.,Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, MO, USA
| | - Smita A Zaheer
- Department of Neurology, School of Medicine, University of Missouri, 1 Hospital Drive, Columbia, MO, USA.,The Center for Translational Neuroscience, School of Medicine, University of Missouri, 1 Hospital Drive, Columbia, MO, USA
| | - Shankar S Iyer
- Department of Neurology, School of Medicine, University of Missouri, 1 Hospital Drive, Columbia, MO, USA.,The Center for Translational Neuroscience, School of Medicine, University of Missouri, 1 Hospital Drive, Columbia, MO, USA.,Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, MO, USA
| | - Raghav Govindarajan
- Department of Neurology, School of Medicine, University of Missouri, 1 Hospital Drive, Columbia, MO, USA
| | | | | | | | - Asgar Zaheer
- Department of Neurology, School of Medicine, University of Missouri, 1 Hospital Drive, Columbia, MO, USA. .,The Center for Translational Neuroscience, School of Medicine, University of Missouri, 1 Hospital Drive, Columbia, MO, USA. .,Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, MO, USA.
| |
Collapse
|
20
|
Qubty D, Rubovitch V, Benromano T, Ovadia M, Pick CG. Orally Administered Cinnamon Extract Attenuates Cognitive and Neuronal Deficits Following Traumatic Brain Injury. J Mol Neurosci 2020; 71:178-186. [PMID: 32901372 DOI: 10.1007/s12031-020-01688-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 08/14/2020] [Indexed: 12/27/2022]
Abstract
The present paper shows how cinnamon extract (CE) consumption mitigates neuronal loss and memory impairment following traumatic brain injury (TBI), one of the world's most common neurodegenerative diseases. TBI patients suffer short- and long-term behavioral, cognitive, and emotional impairments, including difficulties in concentration, memory loss, and depression. Research shows that CE application can mitigate cognitive and behavioral impairments in animal models for Alzheimer's and Parkinson's disease, whose pathophysiology is similar to that of TBI. This study builds on prior research by showing similar results in TBI mice models. After drinking CE for a week, mice were injured using our 70-g weight drop TBI device. For 2 weeks thereafter, the mice continued drinking CE alongside standard lab nutrition. Subsequently, the mice underwent behavioral tests to assess their memory, motor activity, and anxiety. The mice brains were harvested for immunohistochemistry staining to evaluate overall neuronal survival. Our results show that CE consumption almost completely mitigates memory impairment and decreases neuronal loss after TBI. Mice that did not consume CE demonstrated impaired memory. Our results also show that CE consumption attenuated neuronal loss in the temporal cortex and the dentate gyrus. Mice that did not consume CE suffered a significant neuronal loss. There were no significant differences in anxiety levels and motor activity between all groups. These findings show a new therapeutic approach to improve cognitive function and decrease memory loss after TBI.
Collapse
Affiliation(s)
- Doaa Qubty
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Vardit Rubovitch
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Tali Benromano
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Michael Ovadia
- Department of Zoology, Tel Aviv University, Tel Aviv, Israel
| | - Chaim G Pick
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel. .,The Dr. Miriam and Sheldon G. Adelson Chair and Center for the Biology of Addictive Diseases, Tel-Aviv University, Tel-Aviv, Israel. .,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel. .,Sylvan Adams Sports Institute, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
21
|
Silymarin sex-dependently improves cognitive functions and alters TNF-α, BDNF, and glutamate in the hippocampus of mice with mild traumatic brain injury. Life Sci 2020; 257:118049. [DOI: 10.1016/j.lfs.2020.118049] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 06/30/2020] [Accepted: 07/01/2020] [Indexed: 02/07/2023]
|
22
|
Tweedie D, Karnati HK, Mullins R, Pick CG, Hoffer BJ, Goetzl EJ, Kapogiannis D, Greig NH. Time-dependent cytokine and chemokine changes in mouse cerebral cortex following a mild traumatic brain injury. eLife 2020; 9:55827. [PMID: 32804078 PMCID: PMC7473773 DOI: 10.7554/elife.55827] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 08/14/2020] [Indexed: 02/07/2023] Open
Abstract
Traumatic brain injury (TBI) is a serious global health problem, many individuals live with TBI-related neurological dysfunction. A lack of biomarkers of TBI has impeded medication development. To identify new potential biomarkers, we time-dependently evaluated mouse brain tissue and neuronally derived plasma extracellular vesicle proteins in a mild model of TBI with parallels to concussive head injury. Mice (CD-1, 30–40 g) received a sham procedure or 30 g weight-drop and were euthanized 8, 24, 48, 72, 96 hr, 7, 14 and 30 days later. We quantified ipsilateral cortical proteins, many of which differed from sham by 8 hours post-mTBI, particularly GAS-1 and VEGF-B were increased while CXCL16 reduced, 23 proteins changed in 4 or more of the time points. Gene ontology pathways mapped from altered proteins over time related to pathological and physiological processes. Validation of proteins identified in this study may provide utility as treatment response biomarkers.
Collapse
Affiliation(s)
- David Tweedie
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, NIH, Baltimore, United States
| | - Hanuma Kumar Karnati
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, NIH, Baltimore, United States
| | - Roger Mullins
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute on Aging, NIH, Baltimore, United States
| | - Chaim G Pick
- Department of Anatomy and Anthropology, Sackler School of Medicine, Sylvan Adams Sports Institute, and Dr. Miriam and SheldonG. Adelson Chair and Center for the Biology of Addictive Diseases, Tel Aviv University, Tel Aviv, Israel
| | - Barry J Hoffer
- Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, United States
| | - Edward J Goetzl
- Department of Medicine, University of California Medical Center, San Francisco, San Francisco, United States
| | - Dimitrios Kapogiannis
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute on Aging, NIH, Baltimore, United States
| | - Nigel H Greig
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, NIH, Baltimore, United States
| |
Collapse
|
23
|
Ahmed ME, Selvakumar GP, Kempuraj D, Raikwar SP, Thangavel R, Bazley K, Wu K, Khan O, Khan A, Zaheer S, Iyer S, Burton C, James D, Zaheer A. Glia Maturation Factor (GMF) Regulates Microglial Expression Phenotypes and the Associated Neurological Deficits in a Mouse Model of Traumatic Brain Injury. Mol Neurobiol 2020; 57:4438-4450. [PMID: 32737763 DOI: 10.1007/s12035-020-02040-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 07/24/2020] [Indexed: 12/20/2022]
Abstract
Traumatic brain injury (TBI) induces inflammatory responses through microglial activation and polarization towards a more inflammatory state that contributes to the deleterious secondary brain injury. Glia maturation factor (GMF) is a pro-inflammatory protein that is responsible for neuroinflammation following insult to the brain, such as in TBI. We hypothesized that the absence of GMF in GMF-knockout (GMF-KO) mice would regulate microglial activation state and the M1/M2 phenotypes following TBI. We used the weight drop model of TBI in C57BL/6 mice wild-type (WT) and GMF-KO mice. Immunofluorescence staining, Western blot, and ELISA assays were performed to confirm TBI-induced histopathological and neuroinflammatory changes. Behavioral analysis was done to check motor coordination ability and cognitive function. We demonstrated that the deletion of GMF in GMF-KO mice significantly limited lesion volume, attenuated neuronal loss, inhibited gliosis, and activated microglia adopted predominantly anti-inflammatory (M2) phenotypes. Using an ELISA method, we found a gradual decrease in pro-inflammatory cytokines (TNF-α and IL-6) and upregulation of anti-inflammatory cytokines (IL-4 and IL-10) in GMF-KO mice compared with WT mice, thus, promoting the transition of microglia towards a more predominantly anti-inflammatory (M2) phenotype. GMF-KO mice showed significant improvement in motor ability, memory, and cognition. Overall, our results demonstrate that GMF deficiency regulates microglial polarization, which ameliorates neuronal injury and behavioral impairments following TBI in mice and concludes that GMF is a regulator of neuroinflammation and an ideal therapeutic target for the treatment of TBI.
Collapse
Affiliation(s)
- Mohammad Ejaz Ahmed
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA
| | - Govindhasamy Pushpavathi Selvakumar
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA
| | - Duraisamy Kempuraj
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA
| | - Sudhanshu P Raikwar
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA
| | - Ramasamy Thangavel
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA
| | - Kieran Bazley
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Kristopher Wu
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Osaid Khan
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Asher Khan
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Smita Zaheer
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Shankar Iyer
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA
| | | | | | - Asgar Zaheer
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.
| |
Collapse
|
24
|
Ratliff WA, Delic V, Pick CG, Citron BA. Dendritic arbor complexity and spine density changes after repetitive mild traumatic brain injury and neuroprotective treatments. Brain Res 2020; 1746:147019. [PMID: 32681835 DOI: 10.1016/j.brainres.2020.147019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 06/25/2020] [Accepted: 07/10/2020] [Indexed: 12/17/2022]
Abstract
Traumatic brain injury has been described as the signature affliction of recent military conflicts and repetitive TBIs, particularly associated with military and athletic activities, typically result in more severe clinical effects. The majority of TBIs are mild, but they can result in long term cognitive deficits for which there is no effective treatment. One of the most significant deficits observed in TBI patients is memory loss, which suggests that TBI can induce pathological changes within the hippocampus. tert-butylhydroquinone (tBHQ) and pioglitazone activate the Nrf2 and PPAR-γ transcription factors, respectively, and both have been shown to be neuroprotective in model systems. We examined the morphological changes within the hippocampus following repetitive mild TBI and simultaneous treatment with both factors. We utilized a closed head injury mouse model with five injuries over 5 weeks. Our results showed marked morphological changes among the dendrites and dendritic spines of the neurons of the dentate gyrus of the hippocampus. We observed decreases in overall dendritic length, as well as in the quantity and density of dendritic spines. Our treatment partially ameliorated these effects, suggesting that the Nrf2 and PPAR-γ transcription factors may be important targets for future drug development in the treatment of TBI in humans.
Collapse
Affiliation(s)
- Whitney A Ratliff
- Laboratory of Molecular Biology, Bay Pines VA Healthcare System, Research and Development 151, Bldg. 22 Rm. 123, 10000 Bay Pines Blvd, Bay Pines, FL 33744, United States
| | - Vedad Delic
- Laboratory of Molecular Biology, VA New Jersey Health Care System, Research & Development (15), Bldg. 16, Rm 16-176 385 Tremont Ave, East Orange, NJ 07018, United States
| | - Chaim G Pick
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel; Dr. Miriam and Sheldon G. Adelson Chair and Center for the Biology of Addictive Diseases, Tel Aviv University, Tel Aviv, Israel
| | - Bruce A Citron
- Laboratory of Molecular Biology, VA New Jersey Health Care System, Research & Development (15), Bldg. 16, Rm 16-176 385 Tremont Ave, East Orange, NJ 07018, United States; Laboratory of Molecular Biology, Bay Pines VA Healthcare System, Research and Development 151, Bldg. 22 Rm. 123, 10000 Bay Pines Blvd, Bay Pines, FL 33744, United States; Department of Pharmacology, Physiology & Neuroscience, Rutgers-New Jersey Medical School, Newark, NJ 07103, United States.
| |
Collapse
|
25
|
Selvakumar GP, Ahmed ME, Iyer SS, Thangavel R, Kempuraj D, Raikwar SP, Bazley K, Wu K, Khan A, Kukulka K, Bussinger B, Zaheer S, Burton C, James D, Zaheer A. Absence of Glia Maturation Factor Protects from Axonal Injury and Motor Behavioral Impairments after Traumatic Brain Injury. Exp Neurobiol 2020; 29:230-248. [PMID: 32565489 PMCID: PMC7344375 DOI: 10.5607/en20017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 06/09/2020] [Accepted: 06/09/2020] [Indexed: 12/11/2022] Open
Abstract
Traumatic brain injury (TBI) causes disability and death, accelerating the progression towards Alzheimer's disease and Parkinson's disease (PD). TBI causes serious motor and cognitive impairments, as seen in PD that arise during the period of the initial insult. However, this has been understudied relative to TBI induced neuroinflammation, motor and cognitive decline that progress towards PD. Neuronal ubiquitin-C-terminal hydrolase- L1 (UCHL1) is a thiol protease that breaks down ubiquitinated proteins and its level represents the severity of TBI. Previously, we demonstrated the molecular action of glia maturation factor (GMF); a proinflammatory protein in mediating neuroinflammation and neuronal loss. Here, we show that the weight drop method induced TBI neuropathology using behavioral tests, western blotting, and immunofluorescence techniques on sections from wild type (WT) and GMF-deficient (GMF-KO) mice. Results reveal a significant improvement in substantia nigral tyrosine hydroxylase and dopamine transporter expression with motor behavioral performance in GMF-KO mice following TBI. In addition, a significant reduction in neuroinflammation was manifested, as shown by activation of nuclear factor-kB, reduced levels of inducible nitric oxide synthase, and cyclooxygenase- 2 expressions. Likewise, neurotrophins including brain-derived neurotrophic factor and glial-derived neurotrophic factor were significantly improved in GMF-KO mice than WT 72 h post-TBI. Consistently, we found that TBI enhances GFAP and UCHL-1 expression and reduces the number of dopaminergic TH-positive neurons in WT compared to GMF-KO mice 72 h post-TBI. Interestingly, we observed a reduction of THpositive tanycytes in the median eminence of WT than GMF-KO mice. Overall, we found that absence of GMF significantly reversed these neuropathological events and improved behavioral outcome. This study provides evidence that PD-associated pathology progression can be initiated upon induction of TBI.
Collapse
Affiliation(s)
- Govindhasamy Pushpavathi Selvakumar
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Mohammad Ejaz Ahmed
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Shankar S Iyer
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Ramasamy Thangavel
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Duraisamy Kempuraj
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Sudhanshu P Raikwar
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Kieran Bazley
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Kristopher Wu
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Asher Khan
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Klaudia Kukulka
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Bret Bussinger
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Smita Zaheer
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | | | | | - Asgar Zaheer
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| |
Collapse
|
26
|
Mast Cell Activation, Neuroinflammation, and Tight Junction Protein Derangement in Acute Traumatic Brain Injury. Mediators Inflamm 2020; 2020:4243953. [PMID: 32684835 PMCID: PMC7333064 DOI: 10.1155/2020/4243953] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/28/2020] [Accepted: 06/02/2020] [Indexed: 02/06/2023] Open
Abstract
Traumatic brain injury (TBI) is one of the major health problems worldwide that causes death or permanent disability through primary and secondary damages in the brain. TBI causes primary brain damage and activates glial cells and immune and inflammatory cells, including mast cells in the brain associated with neuroinflammatory responses that cause secondary brain damage. Though the survival rate and the neurological deficiencies have shown significant improvement in many TBI patients with newer therapeutic options, the underlying pathophysiology of TBI-mediated neuroinflammation, neurodegeneration, and cognitive dysfunctions is understudied. In this study, we analyzed mast cells and neuroinflammation in weight drop-induced TBI. We analyzed mast cell activation by toluidine blue staining, serum chemokine C-C motif ligand 2 (CCL2) level by enzyme-linked immunosorbent assay (ELISA), and proteinase-activated receptor-2 (PAR-2), a mast cell and inflammation-associated protein, vascular endothelial growth factor receptor 2 (VEGFR2), and blood-brain barrier tight junction-associated claudin 5 and Zonula occludens-1 (ZO-1) protein expression in the brains of TBI mice. Mast cell activation and its numbers increased in the brains of 24 h and 72 h TBI when compared with sham control brains without TBI. Mouse brains after TBI show increased CCL2, PAR-2, and VEGFR2 expression and derangement of claudin 5 and ZO-1 expression as compared with sham control brains. TBI can cause mast cell activation, neuroinflammation, and derangement of tight junction proteins associated with increased BBB permeability. We suggest that inhibition of mast cell activation can suppress neuroimmune responses and glial cell activation-associated neuroinflammation and neurodegeneration in TBI.
Collapse
|
27
|
Niziolek GM, Boudreau RM, Baker J, Friend LA, Makley AT, Edwards MJ, Gulbins E, Goodman MD. Acid Sphingomyelinase Inhibition Mitigates Histopathological and Behavioral Changes in a Murine Model of Traumatic Brain Injury. J Neurotrauma 2020; 37:1902-1909. [PMID: 32138594 DOI: 10.1089/neu.2019.6436] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Traumatic brain injury (TBI) can lead to the development of chronic traumatic encephalopathy as a result of neuronal phosphorylated tau (p-tau) protein aggregation and neuroinflammation. Acid sphingomyelinase (Asm) may also contribute to post-TBI neurodegenerative disorders. We hypothesized that Asm inhibition would ameliorate p-tau aggregation, neuroinflammation, and behavioral changes after TBI in a murine model. TBI was generated using a weight-drop method. Asm inhibition in wild-type mice was achieved with a single injection of amitriptyline 1 h after TBI. Genetic Asm ablation was achieved using Asm-deficient mice (Asm-/-). Thirty days after TBI, mice underwent behavioral testing with the forced swim test for symptoms of depression or were euthanized for neurohistological analysis. Neuroinflammation was quantified using the microglial markers, ionized calcium-binding adaptor molecule 1 and transmembrane protein 119. Compared to sham mice, TBI mice demonstrated increased hippocampal p-tau. Mice that received amitriptyline after TBI demonstrated decreased p-tau compared to mice that received a saline control. Further, post-TBI Asm-/- mice demonstrated lower levels of p-tau compared to wild-type mice. Though a decrease in neuroinflammation was observed at 1 month post-TBI, no change was demonstrated with mice treated with amitriptyline. Similarly, TBI mice were more likely to show depression compared to mice that received amitriptyline after TBI. Utilizing a weight-drop method to induce moderate TBI, we have shown that genetic deficiency or pharmacological inhibition of Asm prevented hippocampal p-tau aggregation 1 month after injury as well as decreased symptoms of depression. These findings highlight an opportunity to potentially reduce the long-term consequences of TBI.
Collapse
Affiliation(s)
- Grace M Niziolek
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio, USA
| | - Ryan M Boudreau
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio, USA
| | - Jennifer Baker
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio, USA
| | - Lou Ann Friend
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio, USA
| | - Amy T Makley
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio, USA
| | - Michael J Edwards
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio, USA
| | - Erich Gulbins
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio, USA.,Department of Molecular Biology, University of Duisburg-Essen, Essen, Germany
| | - Michael D Goodman
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
28
|
Ratliff WA, Qubty D, Delic V, Pick CG, Citron BA. Repetitive Mild Traumatic Brain Injury and Transcription Factor Modulation. J Neurotrauma 2020; 37:1910-1917. [PMID: 32292111 DOI: 10.1089/neu.2020.7005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The worldwide incidence of traumatic brain injury (TBI) is ∼0.5% per year and the frequency is significantly higher among military personnel and athletes. Repetitive TBIs are associated with military and athletic activities, and typically involve more severe consequences. The majority of TBIs are mild; however, these still can result in long-term cognitive deficits, and there is currently no effective treatment. tert-Butylhydroquinone (tBHQ) and pioglitazone can activate the nuclear factor (erythroid-derived 2)-like 2 (Nrf2) and peroxisome proliferator-activated receptor-gamma (PPAR-γ) transcription factors, respectively, and each has been shown to be neuroprotective in various model systems. We examined behavioral and gene expression changes after repetitive mild TBI followed by simultaneous treatment with both factors. We used a repetitive closed head injury of mice involving five injuries with a 1-week interval between each TBI. We found that memory performance was significantly reduced by the injuries, unless the TBIs were followed by the tBHQ and pioglitazone administrations. Certain genes; for example, growth hormone and osteopontin, were downregulated by the injury, and this was reversed by the treatment, whereas other genes; for example, a tumor necrosis factor receptor, were upregulated by the injury and restored if the post-injury treatment was administered. Analysis of gene expression levels affected by the injury and/or the treatment point to potential mechanisms that could be exploited therapeutically.
Collapse
Affiliation(s)
- Whitney A Ratliff
- Laboratory of Molecular Biology, Bay Pines VA Healthcare System, Research and Development, Bay Pines, Florida, USA
| | - Doaa Qubty
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Vedad Delic
- Laboratory of Molecular Biology, VA New Jersey Health Care System, Research and Development, East Orange, New Jersey, USA
| | - Chaim G Pick
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.,Dr. Miriam and Sheldon G. Adelson Chair and Center for the Biology of Addictive Diseases, Tel Aviv University, Tel Aviv, Israel
| | - Bruce A Citron
- Laboratory of Molecular Biology, Bay Pines VA Healthcare System, Research and Development, Bay Pines, Florida, USA.,Laboratory of Molecular Biology, VA New Jersey Health Care System, Research and Development, East Orange, New Jersey, USA.,Department of Pharmacology, Physiology, and Neuroscience, Rutgers-New Jersey Medical School, Newark, New Jersey, USA
| |
Collapse
|
29
|
Bader M, Li Y, Tweedie D, Shlobin NA, Bernstein A, Rubovitch V, Tovar-y-Romo LB, DiMarchi RD, Hoffer BJ, Greig NH, Pick CG. Neuroprotective Effects and Treatment Potential of Incretin Mimetics in a Murine Model of Mild Traumatic Brain Injury. Front Cell Dev Biol 2020; 7:356. [PMID: 31998717 PMCID: PMC6965031 DOI: 10.3389/fcell.2019.00356] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 12/10/2019] [Indexed: 12/16/2022] Open
Abstract
Traumatic brain injury (TBI) is a commonly occurring injury in sports, victims of motor vehicle accidents, and falls. TBI has become a pressing public health concern with no specific therapeutic treatment. Mild TBI (mTBI), which accounts for approximately 90% of all TBI cases, may frequently lead to long-lasting cognitive, behavioral, and emotional impairments. The incretins glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are gastrointestinal hormones that induce glucose-dependent insulin secretion, promote β-cell proliferation, and enhance resistance to apoptosis. GLP-1 mimetics are marketed as treatments for type 2 diabetes mellitus (T2DM) and are well tolerated. Both GLP-1 and GIP mimetics have shown neuroprotective properties in animal models of Parkinson's and Alzheimer's disease. The aim of this study is to evaluate the potential neuroprotective effects of liraglutide, a GLP-1 analog, and twincretin, a dual GLP-1R/GIPR agonist, in a murine mTBI model. First, we subjected mice to mTBI using a weight-drop device and, thereafter, administered liraglutide or twincretin as a 7-day regimen of subcutaneous (s.c.) injections. We then investigated the effects of these drugs on mTBI-induced cognitive impairments, neurodegeneration, and neuroinflammation. Finally, we assessed their effects on neuroprotective proteins expression that are downstream to GLP-1R/GIPR activation; specifically, PI3K and PKA phosphorylation. Both drugs ameliorated mTBI-induced cognitive impairments evaluated by the novel object recognition (NOR) and the Y-maze paradigms in which neither anxiety nor locomotor activity were confounds, as the latter were unaffected by either mTBI or drugs. Additionally, both drugs significantly mitigated mTBI-induced neurodegeneration and neuroinflammation, as quantified by immunohistochemical staining with Fluoro-Jade/anti-NeuN and anti-Iba-1 antibodies, respectively. mTBI challenge significantly decreased PKA phosphorylation levels in ipsilateral cortex, which was mitigated by both drugs. However, PI3K phosphorylation was not affected by mTBI. These findings offer a new potential therapeutic approach to treat mTBI, and support further investigation of the neuroprotective effects and mechanism of action of incretin-based therapies for neurological disorders.
Collapse
Affiliation(s)
- Miaad Bader
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yazhou Li
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - David Tweedie
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Nathan A. Shlobin
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Adi Bernstein
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Vardit Rubovitch
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Luis B. Tovar-y-Romo
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
- Division of Neuroscience, Institute of Cellular Physiology, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | | - Barry J. Hoffer
- Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Nigel H. Greig
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Chaim G. Pick
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Center for the Biology of Addictive Diseases, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
30
|
Kim M, Song M, Oh HJ, Hui J, Bae W, Shin J, Ji SD, Koh YH, Suh JW, Park H, Maeng S. Evaluating the Memory Enhancing Effects of Angelica gigas in Mouse Models of Mild Cognitive Impairments. Nutrients 2019; 12:nu12010097. [PMID: 31905851 PMCID: PMC7019643 DOI: 10.3390/nu12010097] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/01/2019] [Accepted: 11/08/2019] [Indexed: 12/15/2022] Open
Abstract
(1) Background: By 2050, it is estimated that 130 million people will be diagnosed with dementia, and currently approved medicines only slow the progression. So preventive intervention is important to treat dementia. Mild cognitive impairment is a condition characterized by some deterioration in cognitive function and increased risk of progressing to dementia. Therefore, the treatment of mild cognitive impairment (MCI) is a possible way to prevent dementia. Angelica gigas reduces neuroinflammation, improves circulation, and inhibits cholinesterase, which can be effective in the prevention of Alzheimer’s disease and vascular dementia and the progression of mild cognitive impairment. (2) Methods: Angelica gigas (AG) extract 1 mg/kg was administered to mildly cognitive impaired mice, models based on mild traumatic brain injury and chronic mild stress. Then, spatial, working, and object recognition and fear memory were measured. (3) Result: Angelica gigas improved spatial learning, working memory, and suppressed fear memory in the mild traumatic brain injury model. It also improved spatial learning and suppressed cued fear memory in the chronic mild stress model animals. (4) Conclusions: Angelica gigas can improve cognitive symptoms in mild cognitive impairment model mice.
Collapse
Affiliation(s)
- Minsang Kim
- Graduate School of Interdisciplinary Program of Biomodulation Collage of Natural Science, Myongji University, Yongin 17058, Korea;
| | - Minah Song
- Graduate School of East-West Medical Science, Kyung Hee University, Yongin 17104, Korea; (M.S.); (H.-J.O.); (W.B.); (J.S.)
| | - Hee-Jin Oh
- Graduate School of East-West Medical Science, Kyung Hee University, Yongin 17104, Korea; (M.S.); (H.-J.O.); (W.B.); (J.S.)
| | - Jin Hui
- Center for Nutraceutical and Pharmaceutical Materials, Myongji University, Yongin 17058, Korea; (J.H.); (J.W.S.)
| | - Woori Bae
- Graduate School of East-West Medical Science, Kyung Hee University, Yongin 17104, Korea; (M.S.); (H.-J.O.); (W.B.); (J.S.)
| | - Jihwan Shin
- Graduate School of East-West Medical Science, Kyung Hee University, Yongin 17104, Korea; (M.S.); (H.-J.O.); (W.B.); (J.S.)
| | - Sang-Dock Ji
- Department of Agricultural Biology, National Academy of Agricultural Science, Rural Development Administration, Wanju-gun, Jeollabuk-do 55365, Korea;
| | - Young Ho Koh
- ILSONG Institute of Life Science, Hallym University, Anyang 14066, Korea;
- Department of Bio-Medical Gerontology, Hallym University Graduate School, Chuncheon 24252, Korea
| | - Joo Won Suh
- Center for Nutraceutical and Pharmaceutical Materials, Myongji University, Yongin 17058, Korea; (J.H.); (J.W.S.)
| | - Hyunwoo Park
- Health Park Co., Ltd., #2502, Gangnam-dae-Ro 305, Sucho-gu, Seoul 06628, Korea
- Correspondence: (H.P.); (S.M.); Tel.: +82-10-5440-0169 (H.P.); +82-10-5554-0155 (S.M.)
| | - Sungho Maeng
- Graduate School of East-West Medical Science, Kyung Hee University, Yongin 17104, Korea; (M.S.); (H.-J.O.); (W.B.); (J.S.)
- Correspondence: (H.P.); (S.M.); Tel.: +82-10-5440-0169 (H.P.); +82-10-5554-0155 (S.M.)
| |
Collapse
|
31
|
Motor Effects of Minimal Traumatic Brain Injury in Mice. J Mol Neurosci 2019; 70:365-377. [PMID: 31820347 DOI: 10.1007/s12031-019-01422-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 10/14/2019] [Indexed: 12/13/2022]
Abstract
Traumatic brain injury (TBI) is considered to be the leading cause of disability and death among young people. Up to 30% of mTBI patients report motor impairments, such as altered coordination and impaired balance and gait. The objective of the present study was to characterize motor performance and motor learning changes, in order to achieve a more thorough understanding of the possible motor consequences of mTBI in humans. Mice were exposed to traumatic brain injury using the weight-drop model and subsequently subjected to a battery of behavioral motor tests. Immunohistochemistry was conducted in order to evaluate neuronal survival and synaptic connectivity. TBI mice showed a different walking pattern on the Erasmus ladder task, without any significant impairment in motor performance and motor learning. In the running wheels, mTBI mice showed reduced activity during the second dark phase and increased activity during the second light phase compared to the control mice. There was no difference in the sum of wheel revolutions throughout the experiment. On the Cat-Walk paradigm, the mice showed a wider frontal base of support post mTBI. The same mice spent a significantly greater percent of time standing on three paws post mTBI compared with controls. mTBI mice also showed a decrease in the number of neurons in the temporal cortex compared with the control group. In summary, mTBI mice suffered from mild motor impairments, minor changes in the circadian clock, and neuronal damage. A more in-depth examination of the mechanisms by which mTBI compensate for motor deficits is necessary.
Collapse
|
32
|
Li Y, Glotfelty EJ, Namdar I, Tweedie D, Olson L, Hoffer BJ, DiMarchi RD, Pick CG, Greig NH. Neurotrophic and neuroprotective effects of a monomeric GLP-1/GIP/Gcg receptor triagonist in cellular and rodent models of mild traumatic brain injury. Exp Neurol 2019; 324:113113. [PMID: 31730763 DOI: 10.1016/j.expneurol.2019.113113] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 11/08/2019] [Accepted: 11/11/2019] [Indexed: 02/08/2023]
Abstract
A synthetic monomeric peptide triple receptor agonist, termed "Triagonist" that incorporates glucagon-like peptide-1 (GLP-1), glucose-dependent insulinotropic polypeptide (GIP) and glucagon (Gcg) actions, was previously developed to improve upon metabolic and glucose regulatory benefits of single and dual receptor agonists in rodent models of diet-induced obesity and type 2 diabetes. In the current study, the neurotrophic and neuroprotective actions of this Triagonist were probed in cellular and mouse models of mild traumatic brain injury (mTBI), a prevalent cause of neurodegeneration in both the young and elderly. Triagonist dose- and time-dependently elevated cyclic AMP levels in cultured human SH-SY5Y neuronal cells, and induced neurotrophic and neuroprotective actions, mitigating oxidative stress and glutamate excitotoxicity. These actions were inhibited only by the co-administration of antagonists for all three receptor types, indicating the balanced co-involvement of GLP-1, GIP and Gcg receptors. To evaluate physiological relevance, a clinically translatable dose of Triagonist was administered subcutaneously, once daily for 7 days, to mice following a 30 g weight drop close head injury. Triagonist fully mitigated mTBI-induced visual and spatial memory deficits, evaluated at 7 and 30 days post injury. These results establish Triagonist as a novel neurotrophic/protective agent worthy of further evaluation as a TBI treatment strategy.
Collapse
Affiliation(s)
- Yazhou Li
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| | - Elliot J Glotfelty
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA; Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Inbar Namdar
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - David Tweedie
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Lars Olson
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Barry J Hoffer
- Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | | | - Chagi G Pick
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Nigel H Greig
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| |
Collapse
|
33
|
Sex differences in cued fear responses and parvalbumin cell density in the hippocampus following repetitive concussive brain injuries in C57BL/6J mice. PLoS One 2019; 14:e0222153. [PMID: 31487322 PMCID: PMC6728068 DOI: 10.1371/journal.pone.0222153] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 08/22/2019] [Indexed: 02/07/2023] Open
Abstract
There is strong evidence to suggest a link between repeated head trauma and cognitive and emotional disorders, and Repetitive concussive brain injuries (rCBI) may also be a risk factor for depression and anxiety disorders. Animal models of brain injury afford the opportunity for controlled study of the effects of injury on functional outcomes. In this study, male and cycling female C57BL/6J mice sustained rCBI (3x) at 24-hr intervals and were tested in a context and cued fear conditioning paradigm, open field (OF), elevated zero maze and tail suspension test. All mice with rCBI showed less freezing behavior than sham control mice during the fear conditioning context test. Injured male, but not female mice also froze less in response to the auditory cue (tone). Injured mice were hyperactive in an OF environment and spent more time in the open quadrants of the elevated zero maze, suggesting decreased anxiety, but there were no differences between injured mice and sham-controls in depressive-like activity on the tail suspension test. Pathologically, injured mice showed increased astrogliosis in the injured cortex and white matter tracts (optic tracts and corpus callosum). There were no changes in the number of parvalbumin-positive interneurons in the cortex or amygdala, but injured male mice had fewer parvalbumin-positive neurons in the hippocampus. Parvalbumin-reactive interneurons of the hippocampus have been previously demonstrated to be involved in hippocampal-cortical interactions required for memory consolidation, and it is possible memory changes in the fear-conditioning paradigm following rCBI are the result of more subtle imbalances in excitation and inhibition both within the amygdala and hippocampus, and between more widespread brain regions that are injured following a diffuse brain injury.
Collapse
|
34
|
Ratliff WA, Saykally JN, Mervis RF, Lin X, Cao C, Citron BA. Behavior, protein, and dendritic changes after model traumatic brain injury and treatment with nanocoffee particles. BMC Neurosci 2019; 20:44. [PMID: 31438853 PMCID: PMC6704525 DOI: 10.1186/s12868-019-0525-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 08/10/2019] [Indexed: 12/14/2022] Open
Abstract
Background Traumatic brain injury (TBI) is a widespread public health problem and a signature injury of our military in modern conflicts. Despite the long-term effects of even mild brain injuries, an effective treatment remains elusive. Coffee and several of its compounds, including caffeine, have been identified as having neuroprotective effects in studies of neurodegenerative disease. Given the molecular similarities between TBI and neurodegenerative disease, we have devised a study to test a nanocoffee extract in the treatment of a mouse model of mild TBI. Results After a single injury and two subsequent injections of nanocoffee, we identified treatment as being associated with improved behavioral outcomes, favorable molecular signaling changes, and dendritic changes suggestive of improved neuronal health. Conclusions We have identified coffee extracts as a potential viable multifaceted treatment approach to target the secondary injury associated with TBI.
Collapse
Affiliation(s)
- Whitney A Ratliff
- Bay Pines VA Healthcare System, Research and Development, 151, Bldg. 22 Rm. 123, 10000 Bay Pines Blvd, Bay Pines, FL, 33744, USA.,Department of Molecular Medicine, USF College of Medicine, 12901 Bruce B. Downs Blvd, MDC 7, Tampa, FL, 33612, USA
| | - Jessica N Saykally
- Bay Pines VA Healthcare System, Research and Development, 151, Bldg. 22 Rm. 123, 10000 Bay Pines Blvd, Bay Pines, FL, 33744, USA.,Department of Molecular Medicine, USF College of Medicine, 12901 Bruce B. Downs Blvd, MDC 7, Tampa, FL, 33612, USA
| | - Ronald F Mervis
- NeuroStructural Analytics, Inc, Columbus, OH, USA.,Center for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 2 Tampa General Circle, Tampa, FL, 33606, USA
| | - Xiaoyang Lin
- The USF-Health Byrd Alzheimer's Institute, College of Pharmacy, University of South Florida, 4001 E. Fletcher Ave, Tampa, FL, 33613, USA.,Department of Pharmaceutical Sciences, USF College of Pharmacy, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA
| | - Chuanhai Cao
- The USF-Health Byrd Alzheimer's Institute, College of Pharmacy, University of South Florida, 4001 E. Fletcher Ave, Tampa, FL, 33613, USA. .,Department of Pharmaceutical Sciences, USF College of Pharmacy, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA.
| | - Bruce A Citron
- Bay Pines VA Healthcare System, Research and Development, 151, Bldg. 22 Rm. 123, 10000 Bay Pines Blvd, Bay Pines, FL, 33744, USA.,Department of Molecular Medicine, USF College of Medicine, 12901 Bruce B. Downs Blvd, MDC 7, Tampa, FL, 33612, USA.,VA New Jersey Health Care System, Research & Development, Bldg. 16, Rm. 16-176, 385 Tremont Ave, Mailstop 15, East Orange, NJ, 07018, USA.,Department of Pharmacology, Physiology & Neuroscience, Rutgers-New Jersey Medical School, 185 South Orange Ave., Newark, NJ, 07101, USA
| |
Collapse
|
35
|
Cognitive and neuropsychiatric impairments vary as a function of injury severity at 12 months post-experimental diffuse traumatic brain injury: Implications for dementia development. Behav Brain Res 2019; 365:66-76. [PMID: 30826298 DOI: 10.1016/j.bbr.2019.02.045] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 02/20/2019] [Accepted: 02/27/2019] [Indexed: 12/14/2022]
Abstract
Traumatic brain injury (TBI) is a common risk factor for later neurodegeneration, which can manifest as dementia. Despite this, little is known about the time-course of development of functional deficits, particularly cognitive and neuropsychiatric impairments, and whether these differ depending on the nature of the initiating insult. Therefore, this study investigated long term functional impairment at 12 months post-injury following diffuse TBI of different severities. Male Sprague-Dawley rats (420-480 g; 10-12 weeks) were either given a sham surgery (n = 14) or subjected to Marmarou's impact acceleration model of diffuse TBI for a single mild TBI (n = 12), repetitive mild TBI (3 mild diffuse injuries at 5 day intervals) (n = 14) or moderate to severe TBI (n = 14). At 12 months after injury, they were tested on a functional battery encompassing motor, neuropsychiatric (anxiety and depressive-like) and cognitive function. Our results showed that moderate to severe TBI animals exhibited significant impairments in cognitive flexibility (p = 0.009) on the Barnes maze when compared to age-matched sham animals. Neither repetitive mild TBI nor single mild TBI animals showed significant functional impairments when compared to shams. Thus, this study provides the first insight into chronic functional impairments associated with different severities of diffuse TBI, with moderate to severe TBI being a higher risk factor for impaired cognitive function at 12 months post-injury. Taken together, this may have implications for risk of dementia development following different severities of injury.
Collapse
|
36
|
Bodnar CN, Roberts KN, Higgins EK, Bachstetter AD. A Systematic Review of Closed Head Injury Models of Mild Traumatic Brain Injury in Mice and Rats. J Neurotrauma 2019; 36:1683-1706. [PMID: 30661454 PMCID: PMC6555186 DOI: 10.1089/neu.2018.6127] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Mild TBI (mTBI) is a significant health concern. Animal models of mTBI are essential for understanding mechanisms, and pathological outcomes, as well as to test therapeutic interventions. A variety of closed head models of mTBI that incorporate different aspects (i.e., biomechanics) of the mTBI have been reported. The aim of the current review was to compile a comprehensive list of the closed head mTBI rodent models, along with the common data elements, and outcomes, with the goal to summarize the current state of the field. Publications were identified from a search of PubMed and Web of Science and screened for eligibility following PRISMA guidelines. Articles were included that were closed head injuries in which the authors classified the injury as mild in rats or mice. Injury model and animal-specific common data elements, as well as behavioral and histological outcomes, were collected and compiled from a total of 402 articles. Our results outline the wide variety of methods used to model mTBI. We also discovered that female rodents and both young and aged animals are under-represented in experimental mTBI studies. Our findings will aid in providing context comparing the injury models and provide a starting point for the selection of the most appropriate model of mTBI to address a specific hypothesis. We believe this review will be a useful starting place for determining what has been done and what knowledge is missing in the field to reduce the burden of mTBI.
Collapse
Affiliation(s)
- Colleen N. Bodnar
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky
| | - Kelly N. Roberts
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky
| | - Emma K. Higgins
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky
| | - Adam D. Bachstetter
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
37
|
Inhibition of Drp1 after traumatic brain injury provides brain protection and improves behavioral performance in rats. Chem Biol Interact 2019; 304:173-185. [DOI: 10.1016/j.cbi.2019.03.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 03/13/2019] [Indexed: 12/31/2022]
|
38
|
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
|
39
|
Bader M, Li Y, Lecca D, Rubovitch V, Tweedie D, Glotfelty E, Rachmany L, Kim HK, Choi HI, Hoffer BJ, Pick CG, Greig NH, Kim DS. Pharmacokinetics and efficacy of PT302, a sustained-release Exenatide formulation, in a murine model of mild traumatic brain injury. Neurobiol Dis 2019; 124:439-453. [PMID: 30471415 PMCID: PMC6710831 DOI: 10.1016/j.nbd.2018.11.023] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 10/29/2018] [Accepted: 11/20/2018] [Indexed: 12/15/2022] Open
Abstract
Traumatic brain injury (TBI) is a neurodegenerative disorder for which no effective pharmacological treatment is available. Glucagon-like peptide 1 (GLP-1) analogues such as Exenatide have previously demonstrated neurotrophic and neuroprotective effects in cellular and animal models of TBI. However, chronic or repeated administration was needed for efficacy. In this study, the pharmacokinetics and efficacy of PT302, a clinically available sustained-release Exenatide formulation (SR-Exenatide) were evaluated in a concussive mild (m)TBI mouse model. A single subcutaneous (s.c.) injection of PT302 (0.6, 0.12, and 0.024 mg/kg) was administered and plasma Exenatide concentrations were time-dependently measured over 3 weeks. An initial rapid regulated release of Exenatide in plasma was followed by a secondary phase of sustained-release in a dose-dependent manner. Short- and longer-term (7 and 30 day) cognitive impairments (visual and spatial deficits) induced by weight drop mTBI were mitigated by a single post-injury treatment with Exenatide delivered by s.c. injection of PT302 in clinically translatable doses. Immunohistochemical evaluation of neuronal cell death and inflammatory markers, likewise, cross-validated the neurotrophic and neuroprotective effects of SR-Exenatide in this mouse mTBI model. Exenatide central nervous system concentrations were 1.5% to 2.0% of concomitant plasma levels under steady-state conditions. These data demonstrate a positive beneficial action of PT302 in mTBI. This convenient single, sustained-release dosing regimen also has application for other neurological disorders, such as Alzheimer's disease, Parkinson's disease, multiple system atrophy and multiple sclerosis where prior preclinical studies, likewise, have demonstrated positive Exenatide actions.
Collapse
Affiliation(s)
- Miaad Bader
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Yazhou Li
- Drug Design and Development Section, Translational Gerontology Branch, Intramural Research Program, National Institutes of Health, National Institute on Aging, Baltimore, MD, USA
| | - Daniela Lecca
- Drug Design and Development Section, Translational Gerontology Branch, Intramural Research Program, National Institutes of Health, National Institute on Aging, Baltimore, MD, USA
| | - Vardit Rubovitch
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - David Tweedie
- Drug Design and Development Section, Translational Gerontology Branch, Intramural Research Program, National Institutes of Health, National Institute on Aging, Baltimore, MD, USA
| | - Elliot Glotfelty
- Drug Design and Development Section, Translational Gerontology Branch, Intramural Research Program, National Institutes of Health, National Institute on Aging, Baltimore, MD, USA; Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Lital Rachmany
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Hee Kyung Kim
- Peptron Inc., Yuseong-gu, Daejeon, Republic of Korea
| | - Ho-Il Choi
- Peptron Inc., Yuseong-gu, Daejeon, Republic of Korea
| | - Barry J Hoffer
- Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Chaim G Pick
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel; Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv 69978, Israel; Center for the Biology of Addictive Diseases, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Nigel H Greig
- Drug Design and Development Section, Translational Gerontology Branch, Intramural Research Program, National Institutes of Health, National Institute on Aging, Baltimore, MD, USA.
| | - Dong Seok Kim
- Drug Design and Development Section, Translational Gerontology Branch, Intramural Research Program, National Institutes of Health, National Institute on Aging, Baltimore, MD, USA; Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
40
|
Koshy Cherian A, Tronson NC, Parikh V, Kucinski A, Blakely RD, Sarter M. Repetitive mild concussion in subjects with a vulnerable cholinergic system: Lasting cholinergic-attentional impairments in CHT+/- mice. Behav Neurosci 2019; 133:448-459. [PMID: 30896190 DOI: 10.1037/bne0000310] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Previous research emphasized the impact of traumatic brain injury on cholinergic systems and associated cognitive functions. Here we addressed the converse question: Because of the available evidence indicating cognitive and neuronal vulnerabilities in humans expressing low-capacity cholinergic systems or with declining cholinergic systems, do injuries cause more severe cognitive decline in such subjects, and what cholinergic mechanisms contribute to such vulnerability? Using mice heterozygous for the choline transporter (CHT+/- mice) as a model for a limited cholinergic capacity, we investigated the cognitive and neuronal consequences of repeated, mild concussion injuries (rmCc). After five rmCc, and compared with wild type (WT) mice, CHT+/- mice exhibited severe and lasting impairments in sustained attention performance, consistent with effects of cholinergic losses on attention. However, rmCc did not affect the integrity of neuronal cell bodies and did not alter the density of cortical synapses. As a cellular mechanism potentially responsible for the attentional impairment in CHT+/- mice, we found that rmCc nearly completely attenuated performance-associated, CHT-mediated choline transport. These results predict that subjects with an already vulnerable cholinergic system will experience severe and lasting cognitive-cholinergic effects after even relatively mild injuries. If confirmed in humans, such subjects may be excluded from, or receive special protection against, activities involving injury risk. Moreover, the treatment and long-term outcome of traumatic brain injuries may benefit from determining the status of cholinergic systems and associated cognitive functions. (PsycINFO Database Record (c) 2019 APA, all rights reserved).
Collapse
Affiliation(s)
| | | | - Vinay Parikh
- Department of Psychology and Neuroscience Program
| | | | | | | |
Collapse
|
41
|
Rubovitch V, Pharayra A, Har-Even M, Dvir O, Mattson MP, Pick CG. Dietary Energy Restriction Ameliorates Cognitive Impairment in a Mouse Model of Traumatic Brain Injury. J Mol Neurosci 2019; 67:613-621. [PMID: 30734244 DOI: 10.1007/s12031-019-01271-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 01/24/2019] [Indexed: 12/12/2022]
Abstract
Traumatic brain injury (TBI) is one of the most common causes of neurological damage in young people. It was previously reported that dietary restriction, by either intermittent fasting (IF) or daily caloric restriction (CR), could protect neurons against dysfunction and degeneration in animal models of stroke and Parkinson's disease. Recently, several studies have shown that the protein Sirtuin 1 (SIRT1) plays a significant role in the induced neuroprotection following dietary restriction. In the present study, we found a significant reduction of SIRT1 levels in the cortex and hippocampus in a mouse model of mild weight-drop closed head TBI. This reduction was prevented in mice maintained on IF (alternate day fasting) and CR initiated after the head trauma. Hippocampus-dependent learning and memory (measured using a novel object recognition test) was impaired 30 days post-injury in mice fed ad libitum, but not in mice in the IF and CR groups. These results suggest a clinical potential for IF and/or CR as an intervention to reduce brain damage and improve functional outcome in TBI patients.
Collapse
Affiliation(s)
- V Rubovitch
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| | - A Pharayra
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - M Har-Even
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - O Dvir
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - M P Mattson
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD, USA
| | - C G Pick
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel.,Sagol School of Neuroscience, Tel-Aviv University, 69978, Tel-Aviv, Israel.,The Dr. Miriam and Sheldon G. Adelson Chair and Center for the Biology of Addictive Diseases, Tel-Aviv University, 69978, Tel-Aviv, Israel
| |
Collapse
|
42
|
GM1 ganglioside prevents axonal regeneration inhibition and cognitive deficits in a mouse model of traumatic brain injury. Sci Rep 2018; 8:13340. [PMID: 30190579 PMCID: PMC6127193 DOI: 10.1038/s41598-018-31623-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 08/13/2018] [Indexed: 11/08/2022] Open
Abstract
Traumatic Brain Injury (TBI) is one of the most common causes of neurological damage in young populations. It has been previously suggested that one of the mechanisms that underlie brain injury is Axonal Outgrowth Inhibition (AOI) that is caused by altered composition of the gangliosides on the axon surface. In the present study, we have found a significant reduction of GM1 ganglioside levels in the cortex in a closed head traumatic brain injury model of a mouse, induced by a weight drop device. In addition, axonal regeneration in the brains of the injured mice was affected as seen by the expression of the axonal marker pNF-H and the growth cones (visualized by F-actin and β-III-tubulin). NeuN immunostaining revealed mTBI-induced damage to neuronal survival. Finally, as expected, spatial and visual memories (measured by the Y-maze and the Novel Object Recognition tests, respectively) were also damaged 7 and 30 days post injury. A single low dose of GM1 shortly after the injury (2 mg/kg; IP) prevented all of the deficits mentioned above. These results reveal additional insights into the neuroprotective characteristics of GM1 in prevention of biochemical, cellular and cognitive changes caused by trauma, and may suggest a potential intervention for mTBI.
Collapse
|
43
|
Arulsamy A, Teng J, Colton H, Corrigan F, Collins-Praino L. Evaluation of early chronic functional outcomes and their relationship to pre-frontal cortex and hippocampal pathology following moderate-severe traumatic brain injury. Behav Brain Res 2018; 348:127-138. [DOI: 10.1016/j.bbr.2018.04.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 03/20/2018] [Accepted: 04/06/2018] [Indexed: 01/02/2023]
|
44
|
Semple BD, Zamani A, Rayner G, Shultz SR, Jones NC. Affective, neurocognitive and psychosocial disorders associated with traumatic brain injury and post-traumatic epilepsy. Neurobiol Dis 2018; 123:27-41. [PMID: 30059725 DOI: 10.1016/j.nbd.2018.07.018] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 07/08/2018] [Accepted: 07/16/2018] [Indexed: 12/13/2022] Open
Abstract
Survivors of traumatic brain injury (TBI) often develop chronic neurological, neurocognitive, psychological, and psychosocial deficits that can have a profound impact on an individual's wellbeing and quality of life. TBI is also a common cause of acquired epilepsy, which is itself associated with significant behavioral morbidity. This review considers the clinical and preclinical evidence that post-traumatic epilepsy (PTE) acts as a 'second-hit' insult to worsen chronic behavioral outcomes for brain-injured patients, across the domains of emotional, cognitive, and psychosocial functioning. Surprisingly, few well-designed studies have specifically examined the relationship between seizures and behavioral outcomes after TBI. The complex mechanisms underlying these comorbidities remain incompletely understood, although many of the biological processes that precipitate seizure occurrence and epileptogenesis may also contribute to the development of chronic behavioral deficits. Further, the relationship between PTE and behavioral dysfunction is increasingly recognized to be a bidirectional one, whereby premorbid conditions are a risk factor for PTE. Clinical studies in this arena are often challenged by the confounding effects of anti-seizure medications, while preclinical studies have rarely examined an adequately extended time course to fully capture the time course of epilepsy development after a TBI. To drive the field forward towards improved treatment strategies, it is imperative that both seizures and neurobehavioral outcomes are assessed in parallel after TBI, both in patient populations and preclinical models.
Collapse
Affiliation(s)
- Bridgette D Semple
- Department of Neuroscience, Monash University, 99 Commercial Road, Melbourne, VIC, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Royal Parade, Parkville, VIC, Australia.
| | - Akram Zamani
- Department of Neuroscience, Monash University, 99 Commercial Road, Melbourne, VIC, Australia.
| | - Genevieve Rayner
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre (Austin Campus), Heidelberg, VIC, Australia; Melbourne School of Psychological Sciences, The University of Melbourne, Parkville, VIC, Australia; Comprehensive Epilepsy Program, Alfred Health, Australia.
| | - Sandy R Shultz
- Department of Neuroscience, Monash University, 99 Commercial Road, Melbourne, VIC, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Royal Parade, Parkville, VIC, Australia.
| | - Nigel C Jones
- Department of Neuroscience, Monash University, 99 Commercial Road, Melbourne, VIC, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Royal Parade, Parkville, VIC, Australia.
| |
Collapse
|
45
|
Effects of Mild Blast Traumatic Brain Injury on Cognitive- and Addiction-Related Behaviors. Sci Rep 2018; 8:9941. [PMID: 29967344 PMCID: PMC6028456 DOI: 10.1038/s41598-018-28062-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 06/07/2018] [Indexed: 12/30/2022] Open
Abstract
Traumatic brain injury (TBI) commonly results in cognitive and psychiatric problems. Cognitive impairments occur in approximately 30% of patients suffering from mild TBI (mTBI), and correlational evidence from clinical studies indicates that substance abuse may be increased following mTBI. However, understanding the lasting cognitive and psychiatric problems stemming from mTBI is difficult in clinical settings where pre-injury assessment may not be possible or accurate. Therefore, we used a previously characterized blast model of mTBI (bTBI) to examine cognitive- and addiction-related outcomes. We previously demonstrated that this model leads to bilateral damage of the medial prefrontal cortex (mPFC), a region critical for cognitive function and addiction. Rats were exposed to bTBI and tested in operant learning tasks several weeks after injury. bTBI rats made more errors during acquisition of a cue discrimination task compared to sham treated rats. Surprisingly, we observed no differences between groups in set shifting and delayed matching to sample, tasks known to require the mPFC. Separate rats performed cocaine self-administration. No group differences were found in intake or extinction, and only subtle differences were observed in drug-primed reinstatement 3-4 months after injury. These findings indicate that bTBI impairs acquisition of a visual discrimination task and that bTBI does not significantly increase the ability of cocaine exposure to trigger drug seeking.
Collapse
|
46
|
Folweiler KA, Samuel S, Metheny HE, Cohen AS. Diminished Dentate Gyrus Filtering of Cortical Input Leads to Enhanced Area Ca3 Excitability after Mild Traumatic Brain Injury. J Neurotrauma 2018; 35:1304-1317. [PMID: 29338620 PMCID: PMC5962932 DOI: 10.1089/neu.2017.5350] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Mild traumatic brain injury (mTBI) disrupts hippocampal function and can lead to long-lasting episodic memory impairments. The encoding of episodic memories relies on spatial information processing within the hippocampus. As the primary entry point for spatial information into the hippocampus, the dentate gyrus is thought to function as a physiological gate, or filter, of afferent excitation before reaching downstream area Cornu Ammonis (CA3). Although injury has previously been shown to alter dentate gyrus network excitability, it is unknown whether mTBI affects dentate gyrus output to area CA3. In this study, we assessed hippocampal function, specifically the interaction between the dentate gyrus and CA3, using behavioral and electrophysiological techniques in ex vivo brain slices 1 week following mild lateral fluid percussion injury (LFPI). Behaviorally, LFPI mice were found to be impaired in an object-place recognition task, indicating that spatial information processing in the hippocampus is disrupted. Extracellular recordings and voltage-sensitive dye imaging demonstrated that perforant path activation leads to the aberrant spread of excitation from the dentate gyrus into area CA3 along the mossy fiber pathway. These results suggest that after mTBI, the dentate gyrus has a diminished capacity to regulate cortical input into the hippocampus, leading to increased CA3 network excitability. The loss of the dentate filtering efficacy reveals a potential mechanism by which hippocampal-dependent spatial information processing is disrupted, and may contribute to memory dysfunction after mTBI.
Collapse
Affiliation(s)
- Kaitlin A. Folweiler
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Anesthesiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Neuroscience Graduate Group, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sandy Samuel
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Anesthesiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hannah E. Metheny
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Anesthesiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Akiva S. Cohen
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Anesthesiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Neuroscience Graduate Group, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
47
|
Arakawa H. Ethological approach to social isolation effects in behavioral studies of laboratory rodents. Behav Brain Res 2018; 341:98-108. [DOI: 10.1016/j.bbr.2017.12.022] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 12/15/2017] [Accepted: 12/17/2017] [Indexed: 12/31/2022]
|
48
|
de la Tremblaye PB, O'Neil DA, LaPorte MJ, Cheng JP, Beitchman JA, Thomas TC, Bondi CO, Kline AE. Elucidating opportunities and pitfalls in the treatment of experimental traumatic brain injury to optimize and facilitate clinical translation. Neurosci Biobehav Rev 2018; 85:160-175. [PMID: 28576511 PMCID: PMC5709241 DOI: 10.1016/j.neubiorev.2017.05.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 05/12/2017] [Indexed: 12/19/2022]
Abstract
The aim of this review is to discuss the research presented in a symposium entitled "Current progress in characterizing therapeutic strategies and challenges in experimental CNS injury" which was presented at the 2016 International Behavioral Neuroscience Society annual meeting. Herein we discuss diffuse and focal traumatic brain injury (TBI) and ensuing chronic behavioral deficits as well as potential rehabilitative approaches. We also discuss the effects of stress on executive function after TBI as well as the response of the endocrine system and regulatory feedback mechanisms. The role of the endocannabinoids after CNS injury is also discussed. Finally, we conclude with a discussion of antipsychotic and antiepileptic drugs, which are provided to control TBI-induced agitation and seizures, respectively. The review consists predominantly of published data.
Collapse
Affiliation(s)
- Patricia B de la Tremblaye
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States
| | - Darik A O'Neil
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States
| | - Megan J LaPorte
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jeffrey P Cheng
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States
| | - Joshua A Beitchman
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States; Department of Child Health, University of Arizona College of Medicine, Phoenix, AZ, United States; Midwestern University, Glendale, AZ, United States
| | - Theresa Currier Thomas
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States; Department of Child Health, University of Arizona College of Medicine, Phoenix, AZ, United States; Phoenix VA Healthcare System, Phoenix, AZ, United States
| | - Corina O Bondi
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States; Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Anthony E Kline
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States; Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, United States; Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States; Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
49
|
Wang G, Zhang YP, Gao Z, Shields LBE, Li F, Chu T, Lv H, Moriarty T, Xu XM, Yang X, Shields CB, Cai J. Pathophysiological and behavioral deficits in developing mice following rotational acceleration-deceleration traumatic brain injury. Dis Model Mech 2018; 11:dmm030387. [PMID: 29208736 PMCID: PMC5818073 DOI: 10.1242/dmm.030387] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 11/16/2017] [Indexed: 01/22/2023] Open
Abstract
Abusive head trauma (AHT) is the leading cause of death from trauma in infants and young children. An AHT animal model was developed on 12-day-old mice subjected to 90° head extension-flexion sagittal shaking repeated 30, 60, 80 and 100 times. The mortality and time until return of consciousness were dependent on the number of repeats and severity of the injury. Following 60 episodes of repeated head shakings, the pups demonstrated apnea and/or bradycardia immediately after injury. Acute oxygen desaturation was observed by pulse oximetry during respiratory and cardiac suppression. The cerebral blood perfusion was assessed by laser speckle contrast analysis (LASCA) using a PeriCam PSI system. There was a severe reduction in cerebral blood perfusion immediately after the trauma that did not significantly improve within 24 h. The injured mice began to experience reversible sensorimotor function at 9 days postinjury (dpi), which had completely recovered at 28 dpi. However, cognitive deficits and anxiety-like behavior remained. Subdural/subarachnoid hemorrhage, damage to the brain-blood barrier and parenchymal edema were found in all pups subjected to 60 insults. Proinflammatory response and reactive gliosis were upregulated at 3 dpi. Degenerated neurons were found in the cerebral cortex and olfactory tubercles at 30 dpi. This mouse model of repetitive brain injury by rotational head acceleration-deceleration partially mimics the major pathophysiological and behavioral events that occur in children with AHT. The resultant hypoxia/ischemia suggests a potential mechanism underlying the secondary rotational acceleration-deceleration-induced brain injury in developing mice.
Collapse
Affiliation(s)
- Guoxiang Wang
- Department of Spine Surgery, Orthopedics Hospital affiliated to the Second Bethune Hospital, Jilin University, Changchun 130041, China
- Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Yi Ping Zhang
- Norton Neuroscience Institute, Norton Healthcare, Louisville, KY 40202, USA
| | - Zhongwen Gao
- Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Lisa B E Shields
- Norton Neuroscience Institute, Norton Healthcare, Louisville, KY 40202, USA
| | - Fang Li
- Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Department of Neurological Surgery, China-Japan Friendship Hospital, Beijing 100029, China
| | - Tianci Chu
- Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Huayi Lv
- Eye Center of the Second Bethune Hospital, Jilin University, Changchun 130041, China
| | - Thomas Moriarty
- Norton Neuroscience Institute, Norton Healthcare, Louisville, KY 40202, USA
| | - Xiao-Ming Xu
- Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Xiaoyu Yang
- Department of Spine Surgery, Orthopedics Hospital affiliated to the Second Bethune Hospital, Jilin University, Changchun 130041, China
| | - Christopher B Shields
- Norton Neuroscience Institute, Norton Healthcare, Louisville, KY 40202, USA
- Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Jun Cai
- Department of Spine Surgery, Orthopedics Hospital affiliated to the Second Bethune Hospital, Jilin University, Changchun 130041, China
- Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
50
|
The effect of an acute systemic inflammatory insult on the chronic effects of a single mild traumatic brain injury. Behav Brain Res 2018; 336:22-31. [DOI: 10.1016/j.bbr.2017.08.035] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 08/18/2017] [Accepted: 08/23/2017] [Indexed: 12/19/2022]
|