101
|
Christie BR, Trivino‐Paredes J, Pinar C, Neale KJ, Meconi A, Reid H, Hutton CP. A Rapid Neurological Assessment Protocol for Repeated Mild Traumatic Brain Injury in Awake Rats. ACTA ACUST UNITED AC 2019; 89:e80. [DOI: 10.1002/cpns.80] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Brian R. Christie
- Division of Medical SciencesUniversity of Victoria Victoria British Columbia Canada
- Island Medical ProgramUniversity of British Columbia Vancouver British Columbia Canada
| | - Juan Trivino‐Paredes
- Division of Medical SciencesUniversity of Victoria Victoria British Columbia Canada
- Island Medical ProgramUniversity of British Columbia Vancouver British Columbia Canada
| | - Cristina Pinar
- Division of Medical SciencesUniversity of Victoria Victoria British Columbia Canada
- Island Medical ProgramUniversity of British Columbia Vancouver British Columbia Canada
| | - Katie J. Neale
- Division of Medical SciencesUniversity of Victoria Victoria British Columbia Canada
- Island Medical ProgramUniversity of British Columbia Vancouver British Columbia Canada
| | - Alicia Meconi
- Division of Medical SciencesUniversity of Victoria Victoria British Columbia Canada
- Island Medical ProgramUniversity of British Columbia Vancouver British Columbia Canada
| | - Hannah Reid
- Division of Medical SciencesUniversity of Victoria Victoria British Columbia Canada
- Island Medical ProgramUniversity of British Columbia Vancouver British Columbia Canada
| | - Craig P. Hutton
- Division of Medical SciencesUniversity of Victoria Victoria British Columbia Canada
- Island Medical ProgramUniversity of British Columbia Vancouver British Columbia Canada
| |
Collapse
|
102
|
The Administration of the New Pyrimidine Derivative-4-{2-[2-(3,4-Dimethoxyphenyl)-Vinyl]-6-Ethyl-4-Oxo-5-Phenyl-4H-Pyrimidine-1-Il}Benzsulfamide Restores the Activity of Brain Cells in Experimental Chronic Traumatic Encephalopathy by Maintaining Mitochondrial Function. ACTA ACUST UNITED AC 2019; 55:medicina55070386. [PMID: 31319603 PMCID: PMC6681389 DOI: 10.3390/medicina55070386] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 07/11/2019] [Accepted: 07/15/2019] [Indexed: 12/14/2022]
Abstract
Background and objectives: To evaluate the effect of a new pyrimidine derivative on the change of mitochondrial function in experimental chronic traumatic encephalopathy. Materials and methods: The study was performed on male mice of the BALB/c line (acute toxicity was assessed) and male rats of the Wistar line, which were modeled chronic traumatic encephalopathy by the method of free fall of the load (weight 150 g from a 50 cm height). The injury to rats was reproduced once a day for 7 days. Further, cognitive functions, changes in sensorimotor deficiency, cerebral blood flow, neuron-specific enolase(NSE), S100β, glial fibrillary acidic protein (GFAP) (in blood serum) and β-amyloid, adenosine triphosphate (ATP) (in brain tissue supernatant) were evaluated. Mitochondrial respiration was also measured. Choline alfoscerate (100 mg/kg) was used as a reference drug. Results: The study found that the use of a new pyrimidine derivative contributed to the preservation of the mitochondrial respirometric function and cognitive functions in rats. In addition, against the administration of test-object marked increase in the concentration of ATP, the velocity of cerebral blood flow was 4.2 times (p < 0.05) and 35.6% (p < 0.05), respectively, as well as reduced concentration and GFAP, NSE, S100β, β-amyloid and sensorimotor deficit at 2.7 (p < 0.05) times; 2 times (p < 0.05); 2.4 times (p < 0.05); of 30.4% (p < 0.05 and 46.5% (p < 0.05), respectively. The LD50 (per os) for the test-object was 4973.56 ± 573.72 mg/kg. Conclusion: Based on the obtained data, high therapeutic efficacy and low systemic toxicity of the application are assumed 4-{2-[2-(3,4-dimethoxyphenyl)-vinyl]-6-ethyl-4-oxo-5-phenyl-4H-pyrimidine-1-Il}benzsulfamide in chronic traumatic encephalopathy.
Collapse
|
103
|
Cannella LA, McGary H, Ramirez SH. Brain interrupted: Early life traumatic brain injury and addiction vulnerability. Exp Neurol 2019; 317:191-201. [PMID: 30862466 PMCID: PMC6544498 DOI: 10.1016/j.expneurol.2019.03.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 02/27/2019] [Accepted: 03/08/2019] [Indexed: 12/20/2022]
Abstract
Recent reports provide evidence for increased risk of substance use disorders (SUD) among patients with a history of early-life traumatic brain injury (TBI). Preclinical research utilizing animal models of TBI have identified injury-induced inflammation, blood-brain barrier permeability, and changes to synapses and neuronal networks within regions of the brain associated with the perception of reward. Importantly, these reward pathway networks are underdeveloped during childhood and adolescence, and early-life TBI pathology may interrupt ongoing maturation. As such, maladaptive changes induced by juvenile brain injury may underlie increased susceptibility to SUD. In this review, we describe the available clinical and preclinical evidence that identifies SUD as a persistent psychiatric consequence of pediatric neurotrauma by discussing (1) the incidence of early-life TBI, (2) how preclinical studies model TBI and SUD, (3) TBI-induced neuropathology and neuroinflammation in the corticostriatal regions of the brain, and (4) the link between childhood or adolescent TBI and addiction in adulthood. In summary, preclinical research utilizes an innovative combination of models of early-life TBI and SUD to recapitulate clinical features and to determine how TBI promotes a risk for the development of SUD. However, causal processes that link TBI and SUD remain unclear. Additional research to identify and therapeutically target underlying mechanisms of aberrant reward pathway development will provide a launching point for TBI and SUD treatment strategies.
Collapse
Affiliation(s)
- Lee Anne Cannella
- Department of Pathology and Laboratory Medicine, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; Center for Substance Abuse Research, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Hannah McGary
- Department of Pathology and Laboratory Medicine, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Servio H Ramirez
- Department of Pathology and Laboratory Medicine, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; Center for Substance Abuse Research, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; Shriners Hospitals Pediatric Research Center, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA.
| |
Collapse
|
104
|
Feldmann LK, Le Prieult F, Felzen V, Thal SC, Engelhard K, Behl C, Mittmann T. Proteasome and Autophagy-Mediated Impairment of Late Long-Term Potentiation (l-LTP) after Traumatic Brain Injury in the Somatosensory Cortex of Mice. Int J Mol Sci 2019; 20:ijms20123048. [PMID: 31234472 PMCID: PMC6627835 DOI: 10.3390/ijms20123048] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 06/17/2019] [Accepted: 06/19/2019] [Indexed: 12/21/2022] Open
Abstract
Traumatic brain injury (TBI) can lead to impaired cognition and memory consolidation. The acute phase (24–48 h) after TBI is often characterized by neural dysfunction in the vicinity of the lesion, but also in remote areas like the contralateral hemisphere. Protein homeostasis is crucial for synaptic long-term plasticity including the protein degradation systems, proteasome and autophagy. Still, little is known about the acute effects of TBI on synaptic long-term plasticity and protein degradation. Thus, we investigated TBI in a controlled cortical impact (CCI) model in the motor and somatosensory cortex of mice ex vivo-in vitro. Late long-term potentiation (l-LTP) was induced by theta-burst stimulation in acute brain slices after survival times of 1–2 days. Protein levels for the plasticity related protein calcium/calmodulin-dependent protein kinase II (CaMKII) was quantified by Western blots, and the protein degradation activity by enzymatical assays. We observed missing maintenance of l-LTP in the ipsilateral hemisphere, however not in the contralateral hemisphere after TBI. Protein levels of CaMKII were not changed but, interestingly, the protein degradation revealed bidirectional changes with a reduced proteasome activity and an increased autophagic flux in the ipsilateral hemisphere. Finally, LTP recordings in the presence of pharmacologically modified protein degradation systems also led to an impaired synaptic plasticity: bath-applied MG132, a proteasome inhibitor, or rapamycin, an activator of autophagy, both administered during theta burst stimulation, blocked the induction of LTP. These data indicate that alterations in protein degradation pathways likely contribute to cognitive deficits in the acute phase after TBI, which could be interesting for future approaches towards neuroprotective treatments early after traumatic brain injury.
Collapse
Affiliation(s)
- Lucia K Feldmann
- Institute for Physiology, UMC of the Johannes Gutenberg University Mainz, Duesbergweg 6, 55128 Mainz, Germany.
| | - Florie Le Prieult
- Institute for Physiology, UMC of the Johannes Gutenberg University Mainz, Duesbergweg 6, 55128 Mainz, Germany.
| | - Vanessa Felzen
- Institute for Pathobiochemistry, UMC of the Johannes Gutenberg University Mainz, Duesbergweg 6, 55128 Mainz, Germany.
| | - Serge C Thal
- Clinics for Anaesthesiology, UMC of the Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.
| | - Kristin Engelhard
- Clinics for Anaesthesiology, UMC of the Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.
| | - Christian Behl
- Institute for Pathobiochemistry, UMC of the Johannes Gutenberg University Mainz, Duesbergweg 6, 55128 Mainz, Germany.
| | - Thomas Mittmann
- Institute for Physiology, UMC of the Johannes Gutenberg University Mainz, Duesbergweg 6, 55128 Mainz, Germany.
| |
Collapse
|
105
|
Kinder HA, Baker EW, Howerth EW, Duberstein KJ, West FD. Controlled Cortical Impact Leads to Cognitive and Motor Function Deficits that Correspond to Cellular Pathology in a Piglet Traumatic Brain Injury Model. J Neurotrauma 2019; 36:2810-2826. [PMID: 31084390 DOI: 10.1089/neu.2019.6405] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability in the United States, with children who sustain a TBI having a greater risk of developing long-lasting cognitive, behavioral, and motor function deficits. This has led to increased interest in utilizing large animal models to study pathophysiologic and functional changes after injury in hopes of identifying novel therapeutic targets. In the present study, a controlled cortical impact (CCI) piglet TBI model was utilized to evaluate cognitive, motor, and histopathologic outcomes. CCI injury (4 m/sec velocity, 9 mm depression, 400 msec dwell time) was induced at the parietal cortex. Compared with normal pigs (n = 5), TBI pigs (n = 5) exhibited appreciable cognitive deficiencies, including significantly impaired spatial memory in spatial T-maze testing and a significant decrease in exploratory behaviors followed by marked hyperactivity in open field testing. Additionally, gait analysis revealed significant increases in cycle time and stance percent, significant decreases in hind reach, and a shift in the total pressure index from the front to the hind limb on the affected side, suggesting TBI impairs gait and balance. Pigs were sacrificed 28 days post-TBI and histological analysis revealed that TBI lead to a significant decrease in neurons and a significant increase in microglia activation and astrogliosis/astrocytosis at the perilesional area, a significant loss in neurons at the dorsal hippocampus, and significantly increased neuroblast proliferation at the subventricular zone. These data demonstrate a strong relationship between TBI-induced cellular changes and functional outcomes in our piglet TBI model that lay the framework for future studies that assess the ability of therapeutic interventions to contribute to functional improvements.
Collapse
Affiliation(s)
- Holly A Kinder
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia.,Department of Animal and Dairy Science, University of Georgia, Athens, Georgia
| | - Emily W Baker
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia.,Department of Animal and Dairy Science, University of Georgia, Athens, Georgia
| | - Elizabeth W Howerth
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia.,Department of Pathology, University of Georgia, Athens, Georgia
| | - Kylee J Duberstein
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia.,Department of Animal and Dairy Science, University of Georgia, Athens, Georgia
| | - Franklin D West
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia.,Department of Animal and Dairy Science, University of Georgia, Athens, Georgia
| |
Collapse
|
106
|
Zhao J, Wang B, Huang T, Guo X, Yang Z, Song J, Zhang M. Glial response in early stages of traumatic brain injury. Neurosci Lett 2019; 708:134335. [PMID: 31207278 DOI: 10.1016/j.neulet.2019.134335] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 06/12/2019] [Accepted: 06/13/2019] [Indexed: 12/13/2022]
Abstract
Traumatic brain in jury affects a number of individuals per year and is a major cause of worldwide death and disability. Yet, its pathophysiological mechanism remains unclear. It is well-known that glial cells, including microglia and astrocytes, are activated and involved in tissue damage and repair in the peri-lesion regions after traumatic brain injury; however, global glial responses are rarely reported. The purpose of this study was to investigate the global activation of microglia and astrocytes 1 day after traumatic brain injury. To test this, we used a weight drop device to inflict traumatic brain injury on left side of the brain and performed hematoxylin-eosin staining to detect tissue damage. We used immunohistochemical staining and western blotting to detect the activation of microglia and astrocytes 1 day after TBI. We found that microglia were significantly activated in ipsilateral regions. Interestingly, we found that astrocytes were also significantly activated in the ipsilateral regions, contralateral cortex, and contralateral corpus callosum. These results suggest that a focal damage can cause a global glial reaction.
Collapse
Affiliation(s)
- Junjie Zhao
- Department of Neurosurgery, the First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi 710061, China
| | - Bo Wang
- Department of Neurosurgery, the First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi 710061, China
| | - Tingqin Huang
- Department of Neurosurgery, the Second Affiliated Hospital of Xi'an Jiaotong University, West fifth Road No.157, Xi'an, Shaanxi 710004, China
| | - Xiaoye Guo
- Department of Neurosurgery, the First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi 710061, China
| | - Zhongbo Yang
- Department of Neurosurgery, the First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi 710061, China
| | - Jinning Song
- Department of Neurosurgery, the First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi 710061, China
| | - Ming Zhang
- Department of Neurosurgery, the Second Affiliated Hospital of Xi'an Jiaotong University, West fifth Road No.157, Xi'an, Shaanxi 710004, China.
| |
Collapse
|
107
|
Lutton EM, Farney SK, Andrews AM, Shuvaev VV, Chuang GY, Muzykantov VR, Ramirez SH. Endothelial Targeted Strategies to Combat Oxidative Stress: Improving Outcomes in Traumatic Brain Injury. Front Neurol 2019; 10:582. [PMID: 31275220 PMCID: PMC6593265 DOI: 10.3389/fneur.2019.00582] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 05/17/2019] [Indexed: 01/29/2023] Open
Abstract
The endothelium is a thin monolayer of specialized cells that lines the luminal wall of blood vessels and constitutes the critical innermost portion of the physical barrier between the blood and the brain termed the blood-brain barrier (BBB). Aberrant changes in the endothelium occur in many neuropathological states, including those with high morbidity and mortality that lack targeted therapeutic interventions, such as traumatic brain injury (TBI). Utilizing ligands of surface determinants expressed on brain endothelium to target and combat injury mechanisms at damaged endothelium offers a new approach to the study of TBI and new avenues for clinical advancement. Many factors influence the targets that are expressed on endothelium. Therefore, the optimization of binding sites and ideal design features of nanocarriers are controllable factors that permit the engineering of nanotherapeutic agents with applicability that is specific to a known disease state. Following TBI, damaged endothelial cells upregulate cell adhesion molecules, including ICAM-1, and are key sites of reactive oxygen species (ROS) generation, including hydrogen peroxide. Reactive oxygen species along with pro-inflammatory mediators are known to contribute to endothelial damage and loss of BBB integrity. The use of targeted endothelial nanomedicine, with conjugates of the antioxidant enzyme catalase linked to anti-ICAM-1 antibodies, has recently been demonstrated to minimize oxidative stress at the BBB and reduce neuropathological outcomes following TBI. Here, we discuss targeted endothelial nanomedicine and its potential to provide benefits in TBI outcomes and future directions of this approach.
Collapse
Affiliation(s)
- Evan M Lutton
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - S Katie Farney
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Allison M Andrews
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Vladimir V Shuvaev
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Gwo-Yu Chuang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Vladimir R Muzykantov
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Servio H Ramirez
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| |
Collapse
|
108
|
Franklin W, Krishnan B, Taglialatela G. Chronic synaptic insulin resistance after traumatic brain injury abolishes insulin protection from amyloid beta and tau oligomer-induced synaptic dysfunction. Sci Rep 2019; 9:8228. [PMID: 31160730 PMCID: PMC6546708 DOI: 10.1038/s41598-019-44635-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 05/14/2019] [Indexed: 02/07/2023] Open
Abstract
Traumatic brain injury (TBI) is a risk factor for Alzheimer's disease (AD), although the mechanisms contributing to this increased risk are unknown. Insulin resistance is an additional risk factor for AD whereby decreased insulin signaling increases synaptic sensitivity to amyloid beta (Aβ) and tau. Considering this, we used rats that underwent a lateral fluid percussion injury at acute and chronic time-points to investigate whether decreased insulin responsiveness in TBI animals is playing a role in synaptic vulnerability to AD pathology. We detected acute and chronic decreases in insulin responsiveness in isolated hippocampal synaptosomes after TBI. In addition to assessing both Aβ and tau binding on synaptosomes, we performed electrophysiology to assess the dysfunctional impact of Aβ and tau oligomers as well as the protective effect of insulin. While we saw no difference in binding or degree of LTP inhibition by either Aβ or tau oligomers between sham and TBI animals, we found that insulin treatment was able to block oligomer-induced LTP inhibition in sham but not in TBI animals. Since insulin treatment has been discussed as a therapy for AD, this gives valuable insight into therapeutic implications of treating AD patients based on one's history of associated risk factors.
Collapse
Affiliation(s)
- Whitney Franklin
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch, Galveston, Texas, 77555, USA
- Department of Neuroscience, Cell Biology & Anatomy, University of Texas Medical Branch, Galveston, Texas, 77555, USA
| | - Balaji Krishnan
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch, Galveston, Texas, 77555, USA
| | - Giulio Taglialatela
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch, Galveston, Texas, 77555, USA.
| |
Collapse
|
109
|
Ren X, Boriero D, Chaiswing L, Bondada S, St Clair DK, Butterfield DA. Plausible biochemical mechanisms of chemotherapy-induced cognitive impairment ("chemobrain"), a condition that significantly impairs the quality of life of many cancer survivors. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1088-1097. [PMID: 30759363 PMCID: PMC6502692 DOI: 10.1016/j.bbadis.2019.02.007] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 02/05/2019] [Accepted: 02/07/2019] [Indexed: 12/13/2022]
Abstract
Increasing numbers of cancer patients survive and live longer than five years after therapy, but very often side effects of cancer treatment arise at same time. One of the side effects, chemotherapy-induced cognitive impairment (CICI), also called "chemobrain" or "chemofog" by patients, brings enormous challenges to cancer survivors following successful chemotherapeutic treatment. Decreased abilities of learning, memory, attention, executive function and processing speed in cancer survivors with CICI, are some of the challenges that greatly impair survivors' quality of life. The molecular mechanisms of CICI involve very complicated processes, which have been the subject of investigation over the past decades. Many mechanistic candidates have been studied including disruption of the blood-brain barrier (BBB), DNA damage, telomere shortening, oxidative stress and associated inflammatory response, gene polymorphism of neural repair, altered neurotransmission, and hormone changes. Oxidative stress is considered as a vital mechanism, since over 50% of FDA-approved anti-cancer drugs can generate reactive oxygen species (ROS) or reactive nitrogen species (RNS), which lead to neuronal death. In this review paper, we discuss these important candidate mechanisms, in particular oxidative stress and the cytokine, TNF-alpha and their potential roles in CICI.
Collapse
Affiliation(s)
- Xiaojia Ren
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA
| | - Diana Boriero
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA; Department of Neurosciences, Biomedicine, and Movement Disorders, Section on Biological Chemistry, University of Verona, 37134 Verona, Italy
| | - Luksana Chaiswing
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Subbarao Bondada
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; Department of Microbiology, Immunology & Molecular Genetics, University of Kentucky, Lexington, KY 40536, USA
| | - Daret K St Clair
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - D Allan Butterfield
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA; Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
110
|
Metabotropic Glutamate Receptor 5 and 8 Modulate the Ameliorative Effect of Ultramicronized Palmitoylethanolamide on Cognitive Decline Associated with Neuropathic Pain. Int J Mol Sci 2019; 20:ijms20071757. [PMID: 30970677 PMCID: PMC6480075 DOI: 10.3390/ijms20071757] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/03/2019] [Accepted: 04/04/2019] [Indexed: 12/23/2022] Open
Abstract
This study investigated whether metabotropic glutamate receptor (mGluR) 5 and 8 are involved in the effect of ultramicronizedpalmitoylethanolamide (um-PEA) on the cognitive behavior and long term potentiation (LTP) at entorhinal cortex (LEC)-dentate gyrus (DG) pathway in mice rendered neuropathic by the spare nerve injury (SNI). SNI reduced discriminative memory and LTP. Um-PEA treatment started after the development of neuropathic pain had no effects in sham mice, whereas it restored cognitive behavior and LTP in SNI mice. 2-Methyl-6-(phenylethynyl) pyridine (MPEP), a selective mGluR5 antagonist, improved cognition in SNI mice and produced a chemical long term depression of the field excitatory postsynaptic potentials (fEPSPs) in sham and SNI mice. After theta burst stimulation (TBS) MPEP restored LTP in SNI mice. In combination with PEA, MPEP antagonized the PEA effect on discriminative memory and decreased LTP in SNI mice. The (RS)-4-(1-amino-1-carboxyethyl)phthalic acid (MDCPG), a selective mGluR8 antagonist, did not affect discriminative memory, but it induced a chemical LTP and prevented the enhancement of fEPSPs after TBS in SNI mice which were treated or not treated with PEA. The effect of PEA on LTP and cognitive behavior was modulated by mGluR5 and mGluR8. In particular in the SNI conditions, the mGluR5 blockade facilitated memory and LTP, but prevented the beneficial effects of PEA on discriminative memory while the mGluR8 blockade, which was ineffective in itself, prevented the favorable action of the PEA on LTP. Thus, although their opposite roles (excitatory/inhibitory of the two receptor subtypes on the glutamatergic system), they appeared to be required for the neuroprotective effect of PEA in conditions of neuropathic pain.
Collapse
|
111
|
Mollayeva T, Mollayeva S, Pacheco N, D'Souza A, Colantonio A. The course and prognostic factors of cognitive outcomes after traumatic brain injury: A systematic review and meta-analysis. Neurosci Biobehav Rev 2019; 99:198-250. [PMID: 30641116 DOI: 10.1016/j.neubiorev.2019.01.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 08/16/2018] [Accepted: 01/10/2019] [Indexed: 12/18/2022]
Abstract
Despite indications that TBI may be a precursor of cognitive decline and subsequent development of Alzheimer's disease, little is known about the time course of this relationship and the factors involved. This systematic review summarizes the evidence pertinent to this subject matter. All English language studies of longitudinal design, and works cited within them, found in six literature databases, were considered, and their quality assessed. Of 65 articles appraised, 44 studies were selected. Results were organized by timing of assessments, injury severity, and cognitive domains assessed. Differences in the course of cognitive performance were observed across injury severity groups and cognitive domains, with differential proportions of reports of improvement, decline, or no change over time. The evidence for genetic, sex-, age-, and injury-related factors as determinants of cognitive outcome was inconsistent. The non-uniform trajectory of cognitive performance post-TBI supports the notion that this construct is non-homogeneous, and that different factors influence its course. Agreement on a core set of predictors and consideration of psychometric properties of outcome measures is needed.
Collapse
Affiliation(s)
- Tatyana Mollayeva
- Rehabilitation Sciences Institute, Faculty of Medicine, University of Toronto, Canada; Toronto Rehabilitation Institute-University Health Network, Ontario, Canada; Acquired Brain Injury Research Lab, University of Toronto, Canada.
| | - Shirin Mollayeva
- Toronto Rehabilitation Institute-University Health Network, Ontario, Canada.
| | - Nicole Pacheco
- Acquired Brain Injury Research Lab, University of Toronto, Canada; Faculty of Health Sciences, McMaster University, Canada.
| | - Andrea D'Souza
- Acquired Brain Injury Research Lab, University of Toronto, Canada.
| | - Angela Colantonio
- Rehabilitation Sciences Institute, Faculty of Medicine, University of Toronto, Canada; Toronto Rehabilitation Institute-University Health Network, Ontario, Canada; Acquired Brain Injury Research Lab, University of Toronto, Canada.
| |
Collapse
|
112
|
Alexander TC, Kiffer F, Groves T, Anderson J, Wang J, Hayar A, Chen MT, Rodriguez A, Allen AR. Effects of thioTEPA chemotherapy on cognition and motor coordination. Synapse 2019; 73:e22085. [PMID: 30586195 DOI: 10.1002/syn.22085] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 12/15/2018] [Accepted: 12/20/2018] [Indexed: 01/10/2023]
Abstract
Cancer survivorship has increased greatly as therapies have become more advanced and effective. Thus, we must now focus on improving the quality of life of patients after treatment. After chemotherapy, many patients experience chemotherapy-induced cognitive decline, indicating a need to investigate pathologies associated with this condition. In this study, we addressed cognitive impairment after thioTEPA treatment by assessing behavior and assaying cytokine production and the structure of dendrites in the hippocampus. Male mice were given three intraperitoneal injections of thioTEPA. Five weeks later, the mice underwent behavior testing, and brains were collected for Golgi staining and cytokine analysis. Behavior tests included y-maze and Morris water maze and licking behavioral task. Cytokines measured include: IL-1α, IL-1β, IL-2, IL-3, IL-4, IL-5, IL-10, IL-12p70, MCP-1, TNF-α, GMCSF, and RANTES. We observed decreased memory retention in behavioral tasks. Also, dendritic arborization and length were decreased after chemotherapy treatment. Finally, thioTEPA decreased cytokine production in animals treated with chemotherapy, compared to saline-treated controls. Here, we used a mouse model to correlate the decreases in dendritic complexity and inflammatory cytokine production with cognitive impairment after chemotherapy.
Collapse
Affiliation(s)
- Tyler C Alexander
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas.,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Frederico Kiffer
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas.,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Thomas Groves
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas.,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas.,Ocala West Veterans Affairs, Ocala, Florida
| | - Julie Anderson
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas.,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Jing Wang
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas.,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Abdallah Hayar
- Department of Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | | | - Analiz Rodriguez
- Department of Neurosurgery, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Antiño R Allen
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas.,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas.,Department of Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| |
Collapse
|
113
|
Senescence-associated-β-galactosidase staining following traumatic brain injury in the mouse cerebrum. PLoS One 2019; 14:e0213673. [PMID: 30856215 PMCID: PMC6411151 DOI: 10.1371/journal.pone.0213673] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 02/26/2019] [Indexed: 01/05/2023] Open
Abstract
Primary and secondary traumatic brain injury (TBI) can cause tissue damage by inducing cell death pathways including apoptosis, necroptosis, and autophagy. However, similar pathways can also lead to senescence. Senescent cells secrete senescence-associated secretory phenotype proteins following persistent DNA damage response signaling, leading to cell disorders. TBI initially activates the cell cycle followed by the subsequent triggering of senescence. This study aims to clarify how the mRNA and protein expression of different markers of cell cycle and senescence are modulated and switched over time after TBI. We performed senescence-associated-β-galactosidase (SA-β-gal) staining, immunohistochemical analysis, and real-time PCR to examine the time-dependent changes in expression levels of proteins and mRNA, related to cell cycle and cellular senescence markers, in the cerebrum during the initial 14 days after TBI using a mouse model of controlled cortical impact (CCI). Within the area adjacent to the cerebral contusion after TBI, the protein and/or mRNA expression levels of cell cycle markers were increased significantly until 4 days after injury and senescence markers were significantly increased at 4, 7, and 14 days after injury. Our findings suggested that TBI initially activated the cell cycle in neurons, astrocytes, and microglia within the area adjacent to the hemicerebrum contusion in TBI, whereas after 4 days, such cells could undergo senescence in a cell-type-dependent manner.
Collapse
|
114
|
Zhuang Z, Shen Z, Chen Y, Dai Z, Zhang X, Mao Y, Zhang B, Zeng H, Chen P, Wu R. Mapping the Changes of Glutamate Using Glutamate Chemical Exchange Saturation Transfer (GluCEST) Technique in a Traumatic Brain Injury Model: A Longitudinal Pilot Study. ACS Chem Neurosci 2019; 10:649-657. [PMID: 30346712 DOI: 10.1021/acschemneuro.8b00482] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Glutamate excitoxicity plays a crucial role in the pathophysiology of traumatic brain injury (TBI) through the initiation of secondary injuries. Glutamate chemical exchange saturation transfer (GluCEST) MRI is a newly developed technique to noninvasively image glutamate in vivo with high sensitivity and spatial resolution. The aim of the present study was to use a rat model of TBI to map changes in brain glutamate distribution and explore the capability of GluCEST imaging for detecting secondary injuries. Sequential GluCEST imaging scans were performed in adult male Sprague-Dawley rats before TBI and at 1, 3, 7, and 14 days after TBI. GluCEST% increased and peaked on day 1 after TBI in the core lesion of injured cortex and peaked on day 3 in the ipsilateral hippocampus, as compared to baseline and controls. GluCEST% gradually declined to baseline by day 14 after TBI. A negative correlation between the GluCEST% of the ipsilateral hippocampus on day 3 and the time in the correct quadrant was observed in injured rats. Immunolabeling for glial fibrillary acidic protein showed significant astrocyte activation in the ipsilateral hippocampus of TBI rats. IL-6 and TNF-α in the core lesion peaked on day 1 postinjury, while those in the ipsilateral hippocampus peaked on day 3. These subsequently gradually declined to sham levels by day 14. It was concluded that GluCEST imaging has potential to be a novel neuroimaging approach for predicting cognitive outcome and to better understand neuroinflammation following TBI.
Collapse
Affiliation(s)
- Zerui Zhuang
- Department of Neurosurgery, The Second Affiliated Hospital, Medical College of Shantou University, Shantou 515041, China
| | - Zhiwei Shen
- Department of Radiology, The Second Affiliated Hospital, Medical College of Shantou University, Shantou 515041, China
| | - Yanzi Chen
- Department of Radiology, The Second Affiliated Hospital, Medical College of Shantou University, Shantou 515041, China
| | - Zhuozhi Dai
- Department of Radiology, The Second Affiliated Hospital, Medical College of Shantou University, Shantou 515041, China
| | - Xiaolei Zhang
- Department of Radiology, The Second Affiliated Hospital, Medical College of Shantou University, Shantou 515041, China
| | - Yifei Mao
- Department of Radiology, The Second Affiliated Hospital, Medical College of Shantou University, Shantou 515041, China
| | - Bingna Zhang
- Translational Medicine, The Second Affiliated Hospital, Medical College of Shantou University, Shantou 515041, China
| | - Haiyan Zeng
- Medical College of Shantou University, Shantou 515041, China
| | - Peidong Chen
- Department of Radiology, The Second Affiliated Hospital, Medical College of Shantou University, Shantou 515041, China
| | - Renhua Wu
- Department of Radiology, The Second Affiliated Hospital, Medical College of Shantou University, Shantou 515041, China
| |
Collapse
|
115
|
Zimering MB, Patel D, Bahn G. Type 2 Diabetes Predicts Increased Risk of Neurodegenerative Complications in Veterans Suffering Traumatic Brain Injury. JOURNAL OF ENDOCRINOLOGY AND DIABETES 2019; 6:137. [PMID: 31828222 PMCID: PMC6905496 DOI: 10.15226/2374-6890/6/3/001137] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
AIMS Obese type 2 diabetes and traumatic brain injury are associated with persistent peripheral and neuro-inflammation, respectively. We tested whether adult type 2 diabetes increased the hazard rate for neurodegeneration complications following traumatic brain injury. METHODS Retrospective chart review of patients treated at the Veterans Affairs New Jersey Healthcare System between 2016-2019 and having a diagnosis of prior traumatic brain injury was performed in adult veterans, age 50 years or older. Cox proportional hazards regression analysis was used to identify risk factors predictive of an increased risk of neurodegeneration, i.e. worsening major depression, dementia or Parkinson's disease following traumatic brain injury. RESULTS Type 2 diabetes predicted a nearly three-fold increased hazard ratio (HR = 2.95, 95% CI 1.15-7.56, P =0.02) for the occurrence of worsening major depression, dementia or Parkinson's disease in eighty adults age 50 years or older who had experienced prior traumatic brain injury. After adjusting for other covariates, hypertension (HR= 4.15, 95% CI 1.21-14.29, P =0.02) was significant and body mass index (HR=1.14, 95% CI 0.99-1.30; P=0.06) modestly significant predictors of the risk for the time to first occurrence of the composite neurodegenerative outcome. CONCLUSION Type 2 diabetes, hypertension and higher body mass index increase the hazard for the occurrence of worsening depression, Parkinson's disease and dementia following traumatic brain injury in middle-aged and older adults.
Collapse
Affiliation(s)
- Mark B. Zimering
- Endocrinology, Veterans Affairs New Jersey Healthcare System, East Orange, NJ
- Rutgers-Robert Wood Johnson Medical School, New Brunswick NJ, USA
| | - Deesha Patel
- Endocrinology, Veterans Affairs New Jersey Healthcare System, East Orange, NJ
| | - Gideon Bahn
- Hines Veterans Affairs Hospital, Hines, Illinois
| |
Collapse
|
116
|
Guignet M, Lein PJ. Neuroinflammation in organophosphate-induced neurotoxicity. ROLE OF INFLAMMATION IN ENVIRONMENTAL NEUROTOXICITY 2019. [DOI: 10.1016/bs.ant.2018.10.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
117
|
Holdnack JA, Iverson GL, Silverberg ND, Tulsky DS, Heinemann AW. NIH toolbox cognition tests following traumatic brain injury: Frequency of low scores. Rehabil Psychol 2018; 62:474-484. [PMID: 29265868 DOI: 10.1037/rep0000145] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
PURPOSE/OBJECTIVE To apply multivariate base rate analyses to the National Institutes of Health Toolbox Cognition Battery (NIHTB-CB) to facilitate the identification of cognitive impairment in individuals with traumatic brain injury (TBI). Research Method/Design: In a multisite cross-sectional design, 158 participants who sustained a complicated mild or moderate TBI (n = 74) or severe TBI (n = 84) at least 1 year earlier were administered the NIHTB-CB. The NIHTB-CB is comprised of 2 crystallized cognition tests (reflecting premorbid ability) and 5 fluid cognition tests, measuring processing speed, memory, and executive functioning. Base rates for obtaining 0 to 5 low fluid cognition scores were calculated across a range of cutoffs for defining a low test score (≤25th to 5th percentiles). Base rates of low scores in the TBI sample were compared to the NIHTB-CB normative sample using diagnostic accuracy statistics. RESULTS The proportion of the TBI sample obtaining low scores decreased as the cutoff for defining a low score decreased. Individuals with lower premorbid cognitive ability, as measured by NIHTB-CB Crystallized Composite score, tended to produce more low scores on the NIHTB-CB fluid cognition tests, even when using fully demographically adjusted scores. Certain patterns of low scores were associated with TBI (defined as likelihood ratio >2.0), whereas others were nonspecific, occurring almost as often in participants without TBI. CONCLUSIONS/IMPLICATIONS Premorbid ability stratified base rate tables provided in this article can guide researchers and clinicians in the interpretation of NIHTB-CB performance in adults with TBI. (PsycINFO Database Record
Collapse
Affiliation(s)
- James A Holdnack
- The Center for Health Assessment Research and Translation, University of Delaware
| | - Grant L Iverson
- Department of Physical Medicine and Rehabilitation, Harvard Medical School
| | - Noah D Silverberg
- Division of Physical Medicine & Rehabilitation, Department of Medicine, University of British Columbia
| | - David S Tulsky
- The Center for Health Assessment Research and Translation, Departments of Physical Therapy, Psychological and Brain Sciences, University of Delaware
| | | |
Collapse
|
118
|
Dobrachinski F, Gerbatin RR, Sartori G, Golombieski RM, Antoniazzi A, Nogueira CW, Royes LF, Fighera MR, Porciúncula LO, Cunha RA, Soares FAA. Guanosine Attenuates Behavioral Deficits After Traumatic Brain Injury by Modulation of Adenosinergic Receptors. Mol Neurobiol 2018; 56:3145-3158. [PMID: 30105669 DOI: 10.1007/s12035-018-1296-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 08/02/2018] [Indexed: 12/16/2022]
Abstract
Traumatic brain injury (TBI) is a leading cause of disability worldwide, triggering chronic neurodegeneration underlying cognitive and mood disorder still without therapeutic prospects. Based on our previous observations that guanosine (GUO) attenuates short-term neurochemical alterations caused by TBI, this study investigated the effects of chronical GUO treatment in behavioral, molecular, and morphological disturbances 21 days after trauma. Rats subject to TBI displayed mood (anxiety-like) and memory dysfunction. This was accompanied by a decreased expression of both synaptic (synaptophysin) and plasticity proteins (BDNF and CREB), a loss of cresyl violet-stained neurons, and increased astrogliosis and microgliosis in the hippocampus. Notably, chronic GUO treatment (7.5 mg/kg i.p. daily starting 1 h after TBI) prevented all these TBI-induced long-term behavioral, neurochemical, and morphological modifications. This neuroprotective effect of GUO was abrogated in the presence of the adenosine A1 receptor antagonist DPCPX (1 mg/kg) but unaltered by the adenosine A2A receptor antagonist SCH58261 (0.05 mg/kg). These findings show that a chronic GUO treatment prevents the long-term mood and memory dysfunction triggered by TBI, which involves adenosinergic receptors.
Collapse
Affiliation(s)
- Fernando Dobrachinski
- Department of Biochemistry and Molecular Biology, Center for Natural and Exact Sciences, Federal University of Santa Maria, UFSM, Santa Maria, RS, 97105-900, Brazil
- CNC - Center for Neurosciences and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Rogério R Gerbatin
- Laboratory of Exercise Biochemistry, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Gláubia Sartori
- Laboratory of Synthesis, Reactivity and Pharmacological Evaluating and Toxicology of Organochalcogens, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Ronaldo M Golombieski
- Department of Biochemistry and Molecular Biology, Center for Natural and Exact Sciences, Federal University of Santa Maria, UFSM, Santa Maria, RS, 97105-900, Brazil
| | - Alfredo Antoniazzi
- Laboratory of Biotechnology and Animal Reproduction - BioRep Veterinary Hospital, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Cristina W Nogueira
- Laboratory of Synthesis, Reactivity and Pharmacological Evaluating and Toxicology of Organochalcogens, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Luiz F Royes
- Laboratory of Exercise Biochemistry, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Michele R Fighera
- Department of Biochemistry and Molecular Biology, Center for Natural and Exact Sciences, Federal University of Santa Maria, UFSM, Santa Maria, RS, 97105-900, Brazil
- Department of Neuropsychiatry, Health Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Lisiane O Porciúncula
- Laboratory of Studies on the Purinergic System, Department of Biochemistry / ICBS, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Rodrigo A Cunha
- CNC - Center for Neurosciences and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Félix A A Soares
- Department of Biochemistry and Molecular Biology, Center for Natural and Exact Sciences, Federal University of Santa Maria, UFSM, Santa Maria, RS, 97105-900, Brazil.
| |
Collapse
|
119
|
Hansen KR, DeWalt GJ, Mohammed AI, Tseng HA, Abdulkerim ME, Bensussen S, Saligrama V, Nazer B, Eldred WD, Han X. Mild Blast Injury Produces Acute Changes in Basal Intracellular Calcium Levels and Activity Patterns in Mouse Hippocampal Neurons. J Neurotrauma 2018; 35:1523-1536. [PMID: 29343209 PMCID: PMC5998839 DOI: 10.1089/neu.2017.5029] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Mild traumatic brain injury (mTBI) represents a serious public health concern. Although much is understood about long-term changes in cell signaling and anatomical pathologies associated with mTBI, little is known about acute changes in neuronal function. Using large scale Ca2+ imaging in vivo, we characterized the intracellular Ca2+ dynamics in thousands of individual hippocampal neurons using a repetitive mild blast injury model in which blasts were directed onto the cranium of unanesthetized mice on two consecutive days. Immediately following each blast event, neurons exhibited two types of changes in Ca2+ dynamics at different time scales. One was a reduction in slow Ca2+ dynamics that corresponded to shifts in basal intracellular Ca2+ levels at a time scale of minutes, suggesting a disruption of biochemical signaling. The second was a reduction in the rates of fast transient Ca2+ fluctuations at the sub-second time scale, which are known to be closely linked to neural activity. Interestingly, the blast-induced changes in basal Ca2+ levels were independent of the changes in the rates of fast Ca2+ transients, suggesting that blasts had heterogeneous effects on different cell populations. Both types of changes recovered after ∼1 h. Together, our results demonstrate that mTBI induced acute, heterogeneous changes in neuronal function, altering intracellular Ca2+ dynamics across different time scales, which may contribute to the initiation of longer-term pathologies.
Collapse
Affiliation(s)
- Kyle R. Hansen
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| | | | - Ali I. Mohammed
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| | - Hua-an Tseng
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| | - Moona E. Abdulkerim
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| | - Seth Bensussen
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| | - Venkatesh Saligrama
- Department of Electrical and Computer Engineering, Boston University, Boston, Massachusetts
| | - Bobak Nazer
- Department of Electrical and Computer Engineering, Boston University, Boston, Massachusetts
| | | | - Xue Han
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| |
Collapse
|
120
|
Meng X, Li N, Zhang Y, Fan D, Yang C, Li H, Guo D, Pan S. Beneficial Effect of β-Elemene Alone and in Combination with Hyperbaric Oxygen in Traumatic Brain Injury by Inflammatory Pathway. Transl Neurosci 2018; 9:33-37. [PMID: 29992051 PMCID: PMC6034101 DOI: 10.1515/tnsci-2018-0007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 03/09/2018] [Indexed: 11/25/2022] Open
Abstract
Background Present study evaluates the neuroprotective effect of β-elemene alone and in combination with hyperbaric oxygen (HO) in traumatic brain injury (TBI). Methodology TBI was induced by dropping a weight from a specific height. All the animals were separated in to five groups (n=20) like control group; TBI group; β-elemene treated group which receives β-elemene (100 mg/kg, i.p.) half an hour after the injury; HO group which receives hyperbaric oxygen therapy and β-elemene + HO group which receives β-elemene (100 mg/kg, i.p.) half an hour after the injury and hyperbaric oxygen therapy. Neurological function was assessed to evaluate the effect of β-elemene in TBI rats. Thereafter level of inflammatory cytokines and expression of protein of inflammatory pathway was assessed in the brain tissues of TBI rats. In addition TUNEL assay was also done for the determination apoptosis in neuronal cells. Result Data of the report reveals that β-elemene alone and in combination with hyperbaric oxygen (HO) significantly decreases the neurological score Compared to TBI group. Moreover level of inflammatory cytokines and expression of LTR4 and casepase 3 significantly decrease and increase in the expression of IkB in β-elemene alone and in combination with hyperbaric oxygen (HO) treated group compared to TBI group. Data of TUNEL assay also reveals that β-elemene treated group shows significant decrease in the TUNEL positive cells and apoptosis index compared to TBI group. Conclusion Thus present study concludes the neuroprotective effect of β-elemene against TBI and it shows synergistic effect on TBI when treated with HO.
Collapse
Affiliation(s)
- Xiangen Meng
- Department of Hyperbric Oxygen, Navy General Hospital, Beijing, 100048, P.R. China
| | - Na Li
- Department of Hyperbric Oxygen, Navy General Hospital, Beijing, 100048, P.R. China
| | - Yu Zhang
- Department of Hyperbric Oxygen, Navy General Hospital, Beijing, 100048, P.R. China
| | - Danfeng Fan
- Department of Hyperbric Oxygen, Navy General Hospital, Beijing, 100048, P.R. China
| | - Chen Yang
- Department of Hyperbric Oxygen, Navy General Hospital, Beijing, 100048, P.R. China
| | - Hang Li
- Department of Hyperbric Oxygen, Navy General Hospital, Beijing, 100048, P.R. China
| | - Dazhi Guo
- Department of Hyperbric Oxygen, Navy General Hospital, Beijing, 100048, P.R. China
| | - Shuyi Pan
- Department of Hyperbric Oxygen, Navy General Hospital, Beijing, 100048, P.R. China
| |
Collapse
|
121
|
Li P, Jiang H, Wu H, Wu D, Li H, Yu J, Lai J. AH6809 decreases production of inflammatory mediators by PGE 2 - EP2 - cAMP signaling pathway in an experimentally induced pure cerebral concussion in rats. Brain Res 2018; 1698:11-28. [PMID: 29792868 DOI: 10.1016/j.brainres.2018.05.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 05/19/2018] [Accepted: 05/20/2018] [Indexed: 12/31/2022]
Abstract
Increasing evidence suggests that PGE2 metabolic pathway is involved in pathological changes of the secondary brain injury after traumatic brain injury. However, the underlying mechanisms, in particular, the correlation between various key enzymes and the brain injury, has remained to be fully explored. More specifically, it remains to be ascertained whether AH6809 (an EP2 receptor antagonist) would interfere with the downstream of the PGE2, regulate the inflammatory mediators and improve neuronal damage in the hippocampus by PGE2 - EP2 - cAMP signaling pathway. The expression and pathological changes of cyclooxygenase-1 (COX-1), cyclooxygenase-2 (COX-2), microsomal prostaglandin-E synthase-1 (mPGES-1), E-prostanoid receptor 2 (EP2), tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and inducible nitricoxide synthase (iNOS) in the CA1 area of hippocampus were evaluated by immunohistochemistry, Western blot and RT-PCR after pure cerebral concussion (PCC) induced by a metal pendulum closed brain injury in rats followed by AH6809 treatment. The morphology and number of neurons in CA1 region were analyzed by cresyl violet staining. The concentration of prostaglandin E2 (PGE2) and cyclic adenosine monophosphate (cAMP) was assayed by ELISA. Many neurons in hippocampal CA1 area appeared to undergo necrosis and the number of neurons was concomitantly reduced after PCC injury. With the passage of time, the protein and mRNA expression of various key enzymes including COX-1, COX-2 and mPGES-1, EP2 receptor, and inflammatory mediators including TNF-α, IL-1β and iNOS was increased; meanwhile, the concentration of PGE2 and cAMP was enhanced. After PCC injury given AH6809 intervention, injury of neurons in hippocampal CA1 area was attenuated. The protein and mRNA expression of COX-1, COX-2, mPGES-1, EP2, TNF-α, IL-1β and iNOS was decreased, this was coupled with reduction of PGE2 and cAMP. The results suggest that PGE2 metabolic pathway is involved in secondary pathological changes of PCC. AH6809 improves the recovery of injured neurons in the hippocampal CA1 area and downregulates the inflammatory mediators by PGE2 - EP2 - cAMP signaling pathway.
Collapse
Affiliation(s)
- Ping Li
- College of Forensic Science, Xi'an Jiaotong University, Key Laboratory of Ministry of Public Health for Forensic Science, Xi'an 710061, Shaanxi, PR China; Department of Anatomy and Histology/Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong District, Kunming 650500, Yunnan, PR China
| | - Hongyan Jiang
- Department of Anatomy and Histology/Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong District, Kunming 650500, Yunnan, PR China
| | - Haiying Wu
- Department of Emergency and Intensive Care Unit, The First Affiliated Hospital, Kunming Medical University, Kunming 650032, Yunnan, PR China
| | - Deye Wu
- Department of Human Anatomy and Histology/Embryology, Qilu Medical University, 246 West Outer Ring Road, Boshan Economic and Technological Development Zone, Zibo 255213, Shandong, PR China
| | - Hengxi Li
- Department of Anatomy and Histology/Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong District, Kunming 650500, Yunnan, PR China
| | - Jianyun Yu
- College of Forensic Science and Key Laboratory of Brain Injury, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong District, Kunming 650500, Yunnan, PR China
| | - Jianghua Lai
- College of Forensic Science, Xi'an Jiaotong University, Key Laboratory of Ministry of Public Health for Forensic Science, Xi'an 710061, Shaanxi, PR China.
| |
Collapse
|
122
|
Allitt BJ, Johnstone VPA, Richards KL, Yan EB, Rajan R. Progesterone Sharpens Temporal Response Profiles of Sensory Cortical Neurons in Animals Exposed to Traumatic Brain Injury. Cell Transplant 2018; 26:1202-1223. [PMID: 28933224 PMCID: PMC5657734 DOI: 10.1177/0963689717714326] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Traumatic brain injury (TBI) initiates a cascade of pathophysiological changes that are both complex and difficult to treat. Progesterone (P4) is a neuroprotective treatment option that has shown excellent preclinical benefits in the treatment of TBI, but these benefits have not translated well in the clinic. We have previously shown that P4 exacerbates the already hypoactive upper cortical responses in the short-term post-TBI and does not reduce upper cortical hyperactivity in the long term, and we concluded that there is no tangible benefit to sensory cortex firing strength. Here we examined the effects of P4 treatment on temporal coding resolution in the rodent sensory cortex in both the short term (4 d) and long term (8 wk) following impact-acceleration–induced TBI. We show that in the short-term postinjury, TBI has no effect on sensory cortex temporal resolution and that P4 also sharpens the response profile in all cortical layers in the uninjured brain and all layers other than layer 2 (L2) in the injured brain. In the long term, TBI broadens the response profile in all cortical layers despite firing rate hyperactivity being localized to upper cortical layers and P4 sharpens the response profile in TBI animals in all layers other than L2 and has no long-term effect in the sham brain. These results indicate that P4 has long-term effects on sensory coding that may translate to beneficial perceptual outcomes. The effects seen here, combined with previous beneficial preclinical data, emphasize that P4 is still a potential treatment option in ameliorating TBI-induced disorders.
Collapse
Affiliation(s)
- Benjamin J Allitt
- 1 Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Victoria P A Johnstone
- 1 Department of Physiology, Monash University, Clayton, Victoria, Australia.,2 School of Anatomy, Physiology and Human Biology, The University of Western Australia, Perth, Western Australia, Australia
| | - Katrina L Richards
- 1 Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Edwin B Yan
- 1 Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Ramesh Rajan
- 1 Department of Physiology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
123
|
Chen YH, Huang EYK, Kuo TT, Miller J, Chiang YH, Hoffer BJ. Impact of Traumatic Brain Injury on Dopaminergic Transmission. Cell Transplant 2018; 26:1156-1168. [PMID: 28933212 PMCID: PMC5657731 DOI: 10.1177/0963689717714105] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Brain trauma is often associated with severe morbidity and is a major public health concern. Even when injury is mild and no obvious anatomic disruption is seen, many individuals suffer disabling neuropsychological impairments such as memory loss, mood dysfunction, substance abuse, and adjustment disorder. These changes may be related to subtle disruption of neural circuits as well as functional changes at the neurotransmitter level. In particular, there is considerable evidence that dopamine (DA) physiology in the nigrostriatal and mesocorticolimbic pathways might be impaired after traumatic brain injury (TBI). Alterations in DA levels can lead to oxidative stress and cellular dysfunction, and DA plays an important role in central nervous system inflammation. Therapeutic targeting of DA pathways may offer benefits for both neuronal survival and functional outcome after TBI. The purpose of this review is to discuss the role of DA pathology in acute TBI and the potential impact of therapies that target these systems for the treatment of TBI.
Collapse
Affiliation(s)
- Yuan-Hao Chen
- Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, Republic of China
- Yuan-Hao Chen, Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, 4F, No. 325, 2nd Sec., Cheng-Kung Road, Nei-Hu District, Taipei City, 114 Taiwan, Republic of China.
| | - Eagle Yi-Kung Huang
- Department of Pharmacology, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Tung-Tai Kuo
- Graduate Institute of Computer and Communication Engineering, National Taipei University of Technology, Taipei, Taiwan, Republic of China
| | - Jonathan Miller
- Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Yung-Hsiao Chiang
- Section of Neurosurgery, Department of Surgery, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan, Republic of China
| | - Barry J. Hoffer
- Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| |
Collapse
|
124
|
Abstract
Mild traumatic brain injury (mTBI) represents a significant public healthcare concern, accounting for the majority of all head injuries. While symptoms are generally transient, some patients go on to experience long-term cognitive impairments and additional mild impacts can result in exacerbated and persisting negative outcomes. To date, studies using a range of experimental models have reported chronic behavioral deficits in the presence of axonal injury and inflammation following repeated mTBI; assessments of oxidative stress and myelin pathology have thus far been limited. However, some models employed induced acute focal damage more suggestive of moderate–severe brain injury and are therefore not relevant to repeated mTBI. Given that the nature of mechanical loading in TBI is implicated in downstream pathophysiological changes, the mechanisms of damage and chronic consequences of single and repeated closed-head mTBI remain to be fully elucidated. This review covers literature on potential mechanisms of damage following repeated mTBI, integrating known mechanisms of pathology underlying moderate–severe TBIs, with recent studies on adult rodent models relevant to direct impact injuries rather than blast-induced damage. Pathology associated with excitotoxicity and cerebral blood flow-metabolism uncoupling, oxidative stress, cell death, blood-brain barrier dysfunction, astrocyte reactivity, microglial activation, diffuse axonal injury, and dysmyelination is discussed, followed by a summary of functional deficits and preclinical assessments of therapeutic strategies. Comprehensive characterization of the pathology underlying delayed and persisting deficits following repeated mTBI is likely to facilitate further development of therapeutic strategies to limit long-term sequelae.
Collapse
Affiliation(s)
- Brooke Fehily
- 1 Experimental and Regenerative Neurosciences, School of Biological sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Melinda Fitzgerald
- 1 Experimental and Regenerative Neurosciences, School of Biological sciences, The University of Western Australia, Perth, Western Australia, Australia.,2 Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia.,3 Perron Institute for Neurological and Translational Science, Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia
| |
Collapse
|
125
|
Becker RE, Kapogiannis D, Greig NH. Does traumatic brain injury hold the key to the Alzheimer's disease puzzle? Alzheimers Dement 2018; 14:431-443. [PMID: 29245000 PMCID: PMC5958613 DOI: 10.1016/j.jalz.2017.11.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 10/10/2017] [Accepted: 11/14/2017] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Neurodegenerative disorders have been a graveyard for hundreds of well-intentioned efforts at drug discovery and development. Concussion and other traumatic brain injuries (TBIs) and Alzheimer's disease (AD) share many overlapping pathologies and possible clinical links. METHODS We searched the literature since 1995 using MEDLINE and Google Scholar for the terms concussion, AD, and shared neuropathologies. We also studied a TBI animal model as a supplement to transgenic (Tg) mouse AD models for evaluating AD drug efficacy by preventing neuronal losses. To evaluate TBI/AD pathologies and neuronal self-induced cell death (apoptosis), we are studying brain extracellular vesicles in plasma and (-)-phenserine pharmacology to probe, in animal models of AD and humans, apoptosis and pathways common to concussion and AD. RESULTS Neuronal cell death and a diverse and significant pathological cascade follow TBIs. Many of the developing pathologies are present in early AD. The use of an animal model of concussion as a supplement to Tg mice provides an indication of an AD drug candidate's potential for preventing apoptosis and resulting progression toward dementia in AD. This weight drop supplementation to Tg mouse models, the experimental drug (-)-phenserine, and plasma-derived extracellular vesicles enriched for neuronal origin to follow biomarkers of neurodegenerative processes, each and in combination, show promise as tools useful for probing the progression of disease in AD, TBI/AD pathologies, apoptosis, and drug effects on rates of apoptosis both preclinically and in humans. (-)-Phenserine both countered many subacute post-TBI pathologies that could initiate clinical AD and, in the concussion and other animal models, showed evidence consistent with direct inhibition of neuronal preprogrammed cell death in the presence of TBI/AD pathologies. DISCUSSION These findings may provide support for expanding preclinical Tg mouse studies in AD with a TBI weight drop model, insights into the progression of pathological targets, their relations to apoptosis, and timing of interventions against these targets and apoptosis. Such studies may demonstrate the potential for drugs to effectively and safely inhibit preprogrammed cell death as a new drug development strategy for use in the fight to defeat AD.
Collapse
Affiliation(s)
- Robert E Becker
- Aristea Translational Medicine Corporation, Park City, UT, USA; Drug Design and Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, Baltimore, MD, USA.
| | - Dimitrios Kapogiannis
- Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging, Baltimore, MD, USA
| | - Nigel H Greig
- Drug Design and Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, Baltimore, MD, USA.
| |
Collapse
|
126
|
Sports-Related Concussion Results in Differential Expression of Nuclear Factor-κB Pathway Genes in Peripheral Blood During the Acute and Subacute Periods. J Head Trauma Rehabil 2018; 31:269-76. [PMID: 26479397 DOI: 10.1097/htr.0000000000000191] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To determine changes in global gene expression in peripheral leukocytes in the acute and subacute periods following a sports-related concussion in athletes. SETTING Samples were collected at 2 universities in Rochester, New York. PARTICIPANTS Fifteen contact sport athletes who experienced a sports-related concussion, and 16 nonconcussed teammates served as controls. DESIGN Blood samples were collected at the start of the season (baseline), within 6 hours of injury (acute), and at 7 days (subacute) postinjury. Differential gene expression was measured using the GeneChip 3' in vitro transcription Expression kit and Affymetrix microarrays, and genes with fold difference of 2 or more were identified using Partek. MAIN MEASURES Whole genome differential gene expression, and cognitive and balance measures to asses for clinical symptoms pre- and postinjury. RESULTS In the concussed athletes, we observed 67 downregulated and 4 upregulated genes in the acute period and 63 downregulated and 2 upregulated genes in the subacute period compared with baseline. Of these, there were 28 genes from both time points involved in the inflammatory response. No significant differences in gene expression were detected in the control group. CONCLUSIONS Our findings suggest that recovery from sports-related concussion relates to modulation of inflammation through cytokine and chemokine gene pathways, which can contribute to future development of personalized therapeutic agents.
Collapse
|
127
|
Karelina K, Nicholson S, Weil ZM. Minocycline blocks traumatic brain injury-induced alcohol consumption and nucleus accumbens inflammation in adolescent male mice. Brain Behav Immun 2018; 69:532-539. [PMID: 29395778 PMCID: PMC6698899 DOI: 10.1016/j.bbi.2018.01.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 01/22/2018] [Accepted: 01/22/2018] [Indexed: 12/26/2022] Open
Abstract
Alcohol use is a well characterized risk factor for traumatic brain injury (TBI); however, emerging clinical and experimental research suggests that TBI may also be an independent risk factor for the development of alcohol use disorders. In particular, TBIs incurred early in life predict the development of problem alcohol use and increase vulnerability to neuroinflammation as a consequence of alcohol use. Critically, the neuroinflammatory response to alcohol, mediated in large part by microglia, may also function as a driver of further alcohol use. Here, we tested the hypothesis that TBI increases alcohol consumption through microglia-mediated neuroinflammation. Mice were injured as juveniles and alcohol consumption and preference were assessed in a free-choice voluntary drinking paradigm in adolescence. TBI increased alcohol consumption; however, treatment with minocycline, an inhibitor of microglial activation, reduced alcohol intake in TBI mice to sham levels. Moreover, a single injection of ethanol (2 g/kg) significantly increased microglial activation in the nucleus accumbens and microglial expression of the proinflammatory cytokine IL-1β in TBI, but not sham or minocycline-treated, mice. Our data implicate TBI-induced microglial activation as a possible mechanism for the development of alcohol use disorders.
Collapse
Affiliation(s)
- Kate Karelina
- Department of Neuroscience, Group in Behavioral Neuroendocrinology, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| | - Samuel Nicholson
- Department of Neuroscience, Group in Behavioral Neuroendocrinology, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| | | |
Collapse
|
128
|
Jha SK, Jha NK, Kumar D, Sharma R, Shrivastava A, Ambasta RK, Kumar P. Stress-Induced Synaptic Dysfunction and Neurotransmitter Release in Alzheimer's Disease: Can Neurotransmitters and Neuromodulators be Potential Therapeutic Targets? J Alzheimers Dis 2018; 57:1017-1039. [PMID: 27662312 DOI: 10.3233/jad-160623] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The communication between neurons at synaptic junctions is an intriguing process that monitors the transmission of various electro-chemical signals in the central nervous system. Albeit any aberration in the mechanisms associated with transmission of these signals leads to loss of synaptic contacts in both the neocortex and hippocampus thereby causing insidious cognitive decline and memory dysfunction. Compelling evidence suggests that soluble amyloid-β (Aβ) and hyperphosphorylated tau serve as toxins in the dysfunction of synaptic plasticity and aberrant neurotransmitter (NT) release at synapses consequently causing a cognitive decline in Alzheimer's disease (AD). Further, an imbalance between excitatory and inhibitory neurotransmission systems induced by impaired redox signaling and altered mitochondrial integrity is also amenable for such abnormalities. Defective NT release at the synaptic junction causes several detrimental effects associated with altered activity of synaptic proteins, transcription factors, Ca2+ homeostasis, and other molecules critical for neuronal plasticity. These detrimental effects further disrupt the normal homeostasis of neuronal cells and thereby causing synaptic loss. Moreover, the precise mechanistic role played by impaired NTs and neuromodulators (NMs) and altered redox signaling in synaptic dysfunction remains mysterious, and their possible interlink still needs to be investigated. Therefore, this review elucidates the intricate role played by both defective NTs/NMs and altered redox signaling in synaptopathy. Further, the involvement of numerous pharmacological approaches to compensate neurotransmission imbalance has also been discussed, which may be considered as a potential therapeutic approach in synaptopathy associated with AD.
Collapse
|
129
|
Wang CC, Wee HY, Hu CY, Chio CC, Kuo JR. The Effects of Memantine on Glutamic Receptor-Associated Nitrosative Stress in a Traumatic Brain Injury Rat Model. World Neurosurg 2018; 112:e719-e731. [PMID: 29382619 DOI: 10.1016/j.wneu.2018.01.140] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 01/17/2018] [Accepted: 01/18/2018] [Indexed: 02/01/2023]
Abstract
BACKGROUND The main aim of this study is to elucidate whether the neuroprotective effect of memantine, a noncompetitive N-methyl-d-aspartate receptor 2B (NR2B) antagonist, affects neuronal nitrosative stress, apoptosis, and NR2B expression and improves functional outcomes. METHODS Immediately after the onset of fluid percussion traumatic brain injury (TBI), anesthetized male Sprague-Dawley rats were divided into sham-operated, TBI + vehicle, and TBI + memantine groups. TBI rats were treated with a memantine intraperitoneal injection dose of 20 mg/kg intraperitoneally and then 1 mg/kg every 12 hours intraperitoneally for 6 doses. The motor function, proprioception, infarction volume, and neuronal apoptosis were then measured. Immunofluorescence was used to evaluate astrogliosis, microgliosis, nitrosative stress, and NR2A and NR2B expression in cortical cells. All the parameters were assessed 72 hours after TBI. RESULTS Compared with the sham-operated controls, the TBI-induced motor and proprioception deficits, and increased infraction volume after TBI were significantly attenuated by memantine therapy. The TBI-induced neuronal apoptosis, astrogliosis, and microgliosis, the numbers of neuronal NO synthase and 3-nitro-l-tyrosine expression in neurons, and inducible NO synthase expression in microglia and astrocyte cells in the ischemic cortex after TBI were significantly improved by memantine therapy. Simultaneously, without affecting the NR2A expression in neuronal cells, the NR2B expression significantly decreased after memantine therapy, as evaluated by an immunofluorescence stain. CONCLUSIONS Intraperitoneal injection of memantine in the acute stage may ameliorate TBI in rats by affecting NR2B expression and decreasing neuronal apoptosis and nitrosative stress in the injured cortex. These effects might represent 1 mechanism by which functional recovery occurred.
Collapse
Affiliation(s)
- Che-Chuan Wang
- Department of Neurosurgery, Chi-Mei Medical Center, Tainan, Taiwan; Center for General Education, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Hsiao-Yue Wee
- Department of Neurosurgery, Chi-Mei Medical Center, Tainan, Taiwan; Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Chiao-Ya Hu
- Department of Medical Research, Chi-Mei Medical Center, Tainan, Taiwan
| | - Chung-Ching Chio
- Department of Neurosurgery, Chi-Mei Medical Center, Tainan, Taiwan
| | - Jinn-Rung Kuo
- Department of Neurosurgery, Chi-Mei Medical Center, Tainan, Taiwan; Department of Medical Research, Chi-Mei Medical Center, Tainan, Taiwan; Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan.
| |
Collapse
|
130
|
Profound deficits in hippocampal synaptic plasticity after traumatic brain injury and seizure is ameliorated by prophylactic levetiracetam. Oncotarget 2018; 9:11515-11527. [PMID: 29545916 PMCID: PMC5837755 DOI: 10.18632/oncotarget.23923] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 10/29/2017] [Indexed: 11/25/2022] Open
Abstract
Aim To determine the precise effects of post-traumatic seizure activity on hippocampal processes, we induced seizures at various intervals after traumatic brain injury (TBI) and analyzed plasticity at CA1 Schaffer collateral synapses. Material and Methods Rats were initially separated into two groups; one exposed solely to fluid percussion injury (FPI) at 2 Psi and the other only receiving kainic acid (KA)-induced seizures without FPI. Electrophysiological (ePhys) studies including paired-pulse stimulation for short-term presynaptic plasticity and long-term potentiation (LTP) of CA1 Schaffer collateral synapses of the hippocampus for post-synaptic function survey were followed at post-event 1 hour, 3 and 7 days respectively. Additional rats were exposed to three seizures at weekly intervals starting 1 week or 2 weeks after TBI and compared with seizures without TBI, TBI without seizures, and uninjured animals. An additional group placed under the same control variables were treated with levetiracetam prior to seizure induction. The ePhys studies related to post-TBI induced seizures were also followed in these additional groups. Results Seizures affected the short- and long-term synaptic plasticity of the hippocampal CA3-CA1 pathway. FPI itself suppressed LTP and field excitatory post synaptic potentials (fEPSP) in the CA1 Schaffer collateral synapses; KA-induced seizures that followed FPI further suppressed synaptic plasticity. The impairments in both short-term presynaptic and long-term plasticity were worse in the rats in which early post-TBI seizures were induced than those in which later post-TBI seizures were induced. Finally, prophylactic infusion of levetiracetam for one week after FPI reduced the synaptic plasticity deficits in early post-TBI seizure animals. Conclusion Our data indicates that synaptic plasticity (i.e., both presynaptic and postsynaptic) suppression occurs in TBI followed by a seizure and that the interval between the TBI and seizure is an important factor in the severity of the resulting deficits. Furthermore, the infusion of prophylactic levetiracetam could partially reverse the suppression of synaptic plasticity.
Collapse
|
131
|
Leinonen V, Vanninen R, Rauramaa T. Raised intracranial pressure and brain edema. HANDBOOK OF CLINICAL NEUROLOGY 2018; 145:25-37. [PMID: 28987174 DOI: 10.1016/b978-0-12-802395-2.00004-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Acutely increased intracranial pressure (ICP) is a life-threatening neurosurgical emergency. Optimal management strategy is selected according to the causative process. Typical causes are intracranial bleeds like traumatic subdural, epidural, or intracerebral hematoma (ICH); spontaneous ICH, intraventricular hemorrhage, subarachnoid hemorrhage, and hydrocephalus. When occurring without significant brain injury and treated effectively before herniation, a full recovery can be expected. In intraparenchymal injuries a full recovery is unlikely since dead cells in the central nervous system leave an "empty hole," to be replaced by cerebrospinal fluid. The clinical recovery is based on the surviving cells that are able to make new synapses. Surgery may decrease ICP by removing significant mass effect. In all conditions, when notable injury of brain parenchyma occurs, brain edema may gradually increase ICP and further worsen the clinical condition. This is seen typically in large brain infarctions when the formation of brain edema may lead to increased ICP for hours and days. Brain edema is traditionally classified as vasogenic or cytotoxic but according to current knowledge is rather a continuum, starting with cytotoxic cell swelling followed by ionic edema and then vasogenic edema. Here we review the causes of increased ICP, including mechanisms of brain edema, with clinical examples.
Collapse
Affiliation(s)
- Ville Leinonen
- Department of Neurosurgery, Institute of Clinical Medicine, University of Eastern Finland and Department of Neurosurgery, NeuroCenter, Kuopio University Hospital, Kuopio, Finland.
| | - Ritva Vanninen
- Department of Radiology, Institute of Clinical Medicine, University of Eastern Finland and Department of Radiology, Kuopio University Hospital, Kuopio, Finland
| | - Tuomas Rauramaa
- Department of Pathology, Institute of Clinical Medicine, University of Eastern Finland and Department of Pathology, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
132
|
DeWalt GJ, Mahajan B, Foster AR, Thompson LDE, Marttini AA, Schmidt EV, Mansuri S, D'Souza D, Patel SB, Tenenbaum M, Brandao-Viruet KI, Thompson D, Duong B, Smith DH, Blute TA, Eldred WD. Region-specific alterations in astrocyte and microglia morphology following exposure to blasts in the mouse hippocampus. Neurosci Lett 2017; 664:160-166. [PMID: 29133177 DOI: 10.1016/j.neulet.2017.11.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 11/07/2017] [Accepted: 11/07/2017] [Indexed: 12/28/2022]
Abstract
Traumatic brain injury (TBI) is a serious public health concern, especially injuries from repetitive insults. The main objective of this study was to immunocytochemically examine morphological alterations in astrocytes and microglia in the hippocampus 48h following a single blast versus multiple blasts in adult C57BL/6 mice. The effects of ketamine and xylazine (KX), two common anesthetic agents used in TBI research, were also evaluated due to the confounding effect of anesthetics on injury outcome. Results showed a significant increase in hypertrophic microglia that was limited to the outer molecular layer of the dentate gyrus, but only in the absence of KX. Although the presence or absence of KX had no effect on astrocytes following a single blast, a significant decrease in astrocytic immunoreactivity was observed in the stratum lacunosum moleculare following multiple blasts in the absence of KX. The morphological changes in astrocytes and microglia reported in this study reveal region-specific differences in the absence of KX that could have significant implications for our interpretation of glial alterations in animal models of injury.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Sara Mansuri
- Boston University, Department of Biology, United States
| | | | - Shama B Patel
- Boston University, Department of Biology, United States
| | | | | | | | - Bryan Duong
- Boston University, Department of Biology, United States
| | | | - Todd A Blute
- Boston University, Department of Biology, United States
| | | |
Collapse
|
133
|
Boone DR, Leek JM, Falduto MT, Torres KEO, Sell SL, Parsley MA, Cowart JC, Uchida T, Micci MA, DeWitt DS, Prough DS, Hellmich HL. Effects of AAV-mediated knockdown of nNOS and GPx-1 gene expression in rat hippocampus after traumatic brain injury. PLoS One 2017; 12:e0185943. [PMID: 29016640 PMCID: PMC5634593 DOI: 10.1371/journal.pone.0185943] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 09/21/2017] [Indexed: 02/06/2023] Open
Abstract
Virally mediated RNA interference (RNAi) to knock down injury-induced genes could improve functional outcome after traumatic brain injury (TBI); however, little is known about the consequences of gene knockdown on downstream cell signaling pathways and how RNAi influences neurodegeneration and behavior. Here, we assessed the effects of adeno-associated virus (AAV) siRNA vectors that target two genes with opposing roles in TBI pathogenesis: the allegedly detrimental neuronal nitric oxide synthase (nNOS) and the potentially protective glutathione peroxidase 1 (GPx-1). In rat hippocampal progenitor cells, three siRNAs that target different regions of each gene (nNOS, GPx-1) effectively knocked down gene expression. However, in vivo, in our rat model of fluid percussion brain injury, the consequences of AAV-siRNA were variable. One nNOS siRNA vector significantly reduced the number of degenerating hippocampal neurons and showed a tendency to improve working memory. GPx-1 siRNA treatment did not alter TBI-induced neurodegeneration or working memory deficits. Nevertheless, microarray analysis of laser captured, virus-infected neurons showed that knockdown of nNOS or GPx-1 was specific and had broad effects on downstream genes. Since nNOS knockdown only modestly ameliorated TBI-induced working memory deficits, despite widespread genomic changes, manipulating expression levels of single genes may not be sufficient to alter functional outcome after TBI.
Collapse
Affiliation(s)
- Deborah R. Boone
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Jeanna M. Leek
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | | | | | - Stacy L. Sell
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Margaret A. Parsley
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Jeremy C. Cowart
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Tatsuo Uchida
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Maria-Adelaide Micci
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Douglas S. DeWitt
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Donald S. Prough
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Helen L. Hellmich
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
- * E-mail:
| |
Collapse
|
134
|
Adams SM, Conley YP, Wagner AK, Jha RM, Clark RSB, Poloyac SM, Kochanek PM, Empey PE. The pharmacogenomics of severe traumatic brain injury. Pharmacogenomics 2017; 18:1413-1425. [PMID: 28975867 PMCID: PMC5694019 DOI: 10.2217/pgs-2017-0073] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 07/06/2017] [Indexed: 01/08/2023] Open
Abstract
Pharmacotherapy for traumatic brain injury (TBI) is focused on resuscitation, prevention of secondary injury, rehabilitation and recovery. Pharmacogenomics may play a role in TBI for predicting therapies for sedation, analgesia, seizure prevention, intracranial pressure-directed therapy and neurobehavioral/psychiatric symptoms. Research into genetic predictors of outcomes and susceptibility to complications may also help clinicians to tailor therapeutics for high-risk individuals. Additionally, the expanding use of genomics in the drug development pipeline has provided insight to novel investigational and repurposed medications that may be useful in the treatment of TBI and its complications. Genomics in the context of treatment and prognostication for patients with TBI is a promising area for clinical progress of pharmacogenomics.
Collapse
Affiliation(s)
- Solomon M Adams
- Department of Pharmaceutical Sciences, Center for Clinical Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Clinical & Translational Science Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Yvette P Conley
- Health Promotion & Development, School of Nursing, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Amy K Wagner
- Department of Physical Medicine & Rehabilitation, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Ruchira M Jha
- Clinical & Translational Science Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15224, USA
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Neurological Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Robert SB Clark
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15224, USA
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Division of Pediatric Critical Care Medicine, Children’s Hospital of Pittsburgh of UPMC, Pittsburgh, PA 15224, USA
| | - Samuel M Poloyac
- Department of Pharmaceutical Sciences, Center for Clinical Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Clinical & Translational Science Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Patrick M Kochanek
- Clinical & Translational Science Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15224, USA
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Philip E Empey
- Clinical & Translational Science Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15224, USA
- Department of Pharmacy & Therapeutics, Center for Clinical Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
135
|
ApoE4-associated phospholipid dysregulation contributes to development of Tau hyper-phosphorylation after traumatic brain injury. Sci Rep 2017; 7:11372. [PMID: 28900205 PMCID: PMC5595858 DOI: 10.1038/s41598-017-11654-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 08/25/2017] [Indexed: 12/14/2022] Open
Abstract
The apolipoprotein E4 (ApoE4) genotype combines with traumatic brain injury (TBI) to increase the risk of developing Alzheimer's Disease (AD). However, the underlying mechanism(s) is not well-understood. We found that after exposure to repetitive blast-induced TBI, phosphoinositol biphosphate (PIP2) levels in hippocampal regions of young ApoE3 mice were elevated and associated with reduction in expression of a PIP2 degrading enzyme, synaptojanin 1 (synj1). In contrast, hippocampal PIP2 levels in ApoE4 mice did not increase after blast TBI. Following blast TBI, phospho-Tau (pTau) levels were unchanged in ApoE3 mice, whereas in ApoE4 mice, levels of pTau were significantly increased. To determine the causal relationship between changes in pTau and PIP2/synj1 levels after TBI, we tested if down-regulation of synj1 prevented blast-induced Tau hyper-phosphorylation. Knockdown of synj1 decreased pTau levels in vitro, and abolished blast-induced elevation of pTau in vivo. Blast TBI increased glycogen synthase kinase (GSK)-3β activities in ApoE4 mice, and synj1 knockdown inhibited GSK3β phosphorylation of Tau. Together, these data suggest that ApoE proteins regulate brain phospholipid homeostasis in response to TBI and that the ApoE4 isoform is dysfunctional in this process. Down-regulation of synj1 rescues blast-induced phospholipid dysregulation and prevents development of Tau hyper-phosphorylation in ApoE4 carriers.
Collapse
|
136
|
Ercole A, Magnoni S, Vegliante G, Pastorelli R, Surmacki J, Bohndiek SE, Zanier ER. Current and Emerging Technologies for Probing Molecular Signatures of Traumatic Brain Injury. Front Neurol 2017; 8:450. [PMID: 28912750 PMCID: PMC5582086 DOI: 10.3389/fneur.2017.00450] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 08/14/2017] [Indexed: 01/10/2023] Open
Abstract
Traumatic brain injury (TBI) is understood as an interplay between the initial injury, subsequent secondary injuries, and a complex host response all of which are highly heterogeneous. An understanding of the underlying biology suggests a number of windows where mechanistically inspired interventions could be targeted. Unfortunately, biologically plausible therapies have to-date failed to translate into clinical practice. While a number of stereotypical pathways are now understood to be involved, current clinical characterization is too crude for it to be possible to characterize the biological phenotype in a truly mechanistically meaningful way. In this review, we examine current and emerging technologies for fuller biochemical characterization by the simultaneous measurement of multiple, diverse biomarkers. We describe how clinically available techniques such as cerebral microdialysis can be leveraged to give mechanistic insights into TBI pathobiology and how multiplex proteomic and metabolomic techniques can give a more complete description of the underlying biology. We also describe spatially resolved label-free multiplex techniques capable of probing structural differences in chemical signatures. Finally, we touch on the bioinformatics challenges that result from the acquisition of such large amounts of chemical data in the search for a more mechanistically complete description of the TBI phenotype.
Collapse
Affiliation(s)
- Ari Ercole
- Division of Anaesthesia, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Sandra Magnoni
- Department of Anesthesiology and Intensive Care, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Gloria Vegliante
- Laboratory of Acute Brain Injury and Therapeutic Strategies, Department of Neuroscience, IRCCS – Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Roberta Pastorelli
- Unit of Gene and Protein Biomarkers, Laboratory of Mass Spectrometry, IRCCS – Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Jakub Surmacki
- Department of Physics, University of Cambridge, Cambridge, United Kingdom
| | - Sarah Elizabeth Bohndiek
- Department of Physics, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Elisa R. Zanier
- Laboratory of Acute Brain Injury and Therapeutic Strategies, Department of Neuroscience, IRCCS – Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| |
Collapse
|
137
|
Qiu X, Shi L, Zhuang H, Zhang H, Wang J, Wang L, Sun P, Yu L, Liu L. Cerebrovascular Protective Effect of Boldine Against Neural Apoptosis via Inhibition of Mitochondrial Bax Translocation and Cytochrome C Release. Med Sci Monit 2017; 23:4109-4116. [PMID: 28841638 PMCID: PMC5584841 DOI: 10.12659/msm.903040] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND In the present study, we explored the protective effect and mechanism of action of boldine (BOL) against neural apoptosis, which is a mediator of TBI. MATERIAL AND METHODS The effect of BOL on mitochondrial and cytosol proteins of extracted from cerebral cortical tissue of mice was evaluated. The grip test was used to assess the neurological deficit and brain water content of the subjects after administration of BOL to assess its effect on SOD, GSH, and MDA activity in brain ischemic tissues. To further confirm the effect of the BOL, the histopathological analysis and morphology of neurons were studied by Nissl staining. The effect of BOL against TBI-induced neural apoptosis by immuno-histochemistry and Western blotting assay were also studied. RESULTS BOL showed significant improvement against TBI in a dose-dependent manner. In the BOL-treated group, the apoptotic index was significantly reduced, but the level of caspase-3 was greatly diminished. Additionally, the level of the Bax in mitochondria (mit) and cytosol was elevated in the TBI-treated group as compared to the sham group. Further BOL at the test dose causes significant reduction in the level of mitochondrial MDA together with increase in SOD activity as compared to the TBI alone group. CONCLUSIONS BOL showed a cerebroprotective effect against TBI by attenuating the oxidative stress and the mitochondrial apoptotic pathway. It also inhibited mitochondrial Bax translocation and cytochrome c release.
Collapse
Affiliation(s)
- Xiaozhong Qiu
- Department of Neurosurgery, The Affiliated Hiser Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Ling Shi
- Department of Neurosurgery, The Affiliated Hiser Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Hanting Zhuang
- Department of Neurosurgery, The Affiliated Hiser Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Hongtao Zhang
- Department of Neurosurgery, The Affiliated Hiser Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Juan Wang
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Lijun Wang
- Department of Neurosurgery, The Affiliated Hiser Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Peng Sun
- Department of Neurosurgery, The Affiliated Hiser Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Lili Yu
- Department of Neurosurgery, The Affiliated Hiser Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Longxi Liu
- Department of Neurosurgery, The Affiliated Hiser Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| |
Collapse
|
138
|
Wirth P, Yu W, Kimball AL, Liao J, Berkner P, Glenn MJ. New method to induce mild traumatic brain injury in rodents produces differential outcomes in female and male Sprague Dawley rats. J Neurosci Methods 2017; 290:133-144. [PMID: 28780369 DOI: 10.1016/j.jneumeth.2017.07.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 07/27/2017] [Accepted: 07/28/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND Mild traumatic brain injuries (mTBI) are an increasing health concern due to persistent behavioral and neurological effects. To better understand these effects, researchers frequently rely on animal injury models. Existing models, however, may not adequately reproduce the mechanism of injury as it occurs in humans. NEW METHOD Our new model for inducing mTBI in rodents entails acceleration of the animal toward a stationary impact zone to produce rapid rotational movement of the head. The aim of the present experiment was to characterize the effects of this injury in female and male rats on behavior, cognition, and neural plasticity. RESULTS mTBI produced the most widespread effects in females: they were more active during recovery within minutes of mTBI and more active in the center of the open field 4days after mTBI. Spatial learning deficits in the water maze were mild but persistent and accompanied by reduced numbers of immature neurons in the hippocampus along with reductions in sera levels of the neurotrophin, BDNF. By contrast, male mTBI rats mainly exhibited mild spatial learning deficits, with no other observed effects. COMPARISON WITH EXISTING METHODS Our model induced effects on behavior and biology in rats that aligned with existing models. However, new patterns were observed, particularly when comparing females and males. CONCLUSIONS Taken together, these findings confirm the validity of this model and point to key differences between females and males in symptom severity and type. Additionally, our model adds a novel injury mechanism that complements existing rodent models.
Collapse
Affiliation(s)
- Peter Wirth
- Department of Biology, Colby College, Waterville, ME, 04901, USA; Maine Concussion Management Initiative, Colby College, Waterville, ME, 04901, USA.
| | - Waylin Yu
- Department of Psychology, Colby College, Waterville, ME, 04901, USA.
| | - Amanda L Kimball
- Department of Psychology, Colby College, Waterville, ME, 04901, USA.
| | - Jennifer Liao
- Department of Biology, Colby College, Waterville, ME, 04901, USA; Maine Concussion Management Initiative, Colby College, Waterville, ME, 04901, USA.
| | - Paul Berkner
- Maine Concussion Management Initiative, Colby College, Waterville, ME, 04901, USA.
| | - Melissa J Glenn
- Department of Psychology, Colby College, Waterville, ME, 04901, USA; Maine Concussion Management Initiative, Colby College, Waterville, ME, 04901, USA.
| |
Collapse
|
139
|
Du X, West MB, Cai Q, Cheng W, Ewert DL, Li W, Floyd RA, Kopke RD. Antioxidants reduce neurodegeneration and accumulation of pathologic Tau proteins in the auditory system after blast exposure. Free Radic Biol Med 2017; 108:627-643. [PMID: 28438658 DOI: 10.1016/j.freeradbiomed.2017.04.343] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 04/17/2017] [Accepted: 04/21/2017] [Indexed: 12/31/2022]
Abstract
Cochlear neurodegeneration commonly accompanies hair cell loss resulting from aging, ototoxicity, or exposures to intense noise or blast overpressures. However, the precise pathophysiological mechanisms that drive this degenerative response have not been fully elucidated. Our laboratory previously demonstrated that non-transgenic rats exposed to blast overpressures exhibited marked somatic accumulation of neurotoxic variants of the microtubule-associated protein, Tau, in the hippocampus. In the present study, we extended these analyses to examine neurodegeneration and pathologic Tau accumulation in the auditory system in response to blast exposure and evaluated the potential therapeutic efficacy of antioxidants on short-circuiting this pathological process. Blast injury induced ribbon synapse loss and retrograde neurodegeneration in the cochlea in untreated animals. An accompanying perikaryal accumulation of neurofilament light chain and pathologic Tau oligomers were observed in neurons from both the peripheral and central auditory system, spanning from the spiral ganglion to the auditory cortex. Due to its coincident accumulation pattern and well-documented neurotoxicity, our results suggest that the accumulation of pathologic Tau oligomers may actively contribute to blast-induced cochlear neurodegeneration. Therapeutic intervention with a combinatorial regimen of 2,4-disulfonyl α-phenyl tertiary butyl nitrone (HPN-07) and N-acetylcysteine (NAC) significantly reduced both pathologic Tau accumulation and indications of ongoing neurodegeneration in the cochlea and the auditory cortex. These results demonstrate that a combination of HPN-07 and NAC administrated shortly after a blast exposure can serve as a potential therapeutic strategy for preserving auditory function among military personnel or civilians with blast-induced traumatic brain injuries.
Collapse
Affiliation(s)
| | | | | | | | | | - Wei Li
- Hough Ear Institute, Oklahoma City, OK, USA
| | - Robert A Floyd
- Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Richard D Kopke
- Hough Ear Institute, Oklahoma City, OK, USA; Oklahoma Medical Research Foundation, Oklahoma City, OK, USA; Departments of Physiology and Otolaryngology, University of Oklahoma Health Sciences Center, Oklahoma City 73014, USA.
| |
Collapse
|
140
|
DeWalt GJ, Eldred WD. Visual system pathology in humans and animal models of blast injury. J Comp Neurol 2017; 525:2955-2967. [PMID: 28560719 DOI: 10.1002/cne.24252] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 05/12/2017] [Accepted: 05/14/2017] [Indexed: 12/20/2022]
Abstract
Injury from blast exposure is becoming a more prevalent cause of death and disability worldwide. The devastating neurological impairments that result from blasts are significant and lifelong. Progress in the development of effective therapies to treat injury has been slowed by its heterogeneous pathology and the dearth of information regarding the cellular mechanisms involved. Within the last decade, a number of studies have documented visual dysfunction following injury. This brief review examines damage to the visual system in both humans and animal models of blast injury. The in vivo use of the retina as a surrogate to evaluate brain injury following exposure to blast is also highlighted.
Collapse
Affiliation(s)
- Gloria J DeWalt
- Department of Biology, Boston University, Boston, Massachusetts
| | | |
Collapse
|
141
|
Erythropoietin ameliorates diabetes-associated cognitive dysfunction in vitro and in vivo. Sci Rep 2017; 7:2801. [PMID: 28584284 PMCID: PMC5459814 DOI: 10.1038/s41598-017-03137-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 04/20/2017] [Indexed: 01/15/2023] Open
Abstract
Several studies indicate that erythropoietin (EPO) has remarkable neuroprotective effects in various central nervous system disorders, while little is known about the effects of EPO in diabetes-associated cognitive dysfunction. Therefore, the present study aimed to investigate whether EPO ameliorates diabetes-associated cognitive dysfunction in vivo and in vitro. We investigated the protective effects of EPO on high-glucose (HG)-induced PC12 cell death and oxidative stress. The effects of EPO (300 U/kg administered three times a week for 4 weeks) on diabetes-associated cognitive decline were investigated in diabetic rats. EPO significantly increased cell viability, increased the activity of superoxide dismutase, decreased the production of malondialdehyde and reactive oxygen species, and decreased the apoptosis rate. Additionally, LY294002, a phosphatidylinositol 3-kinase (PI3K) inhibitor, abolished the protective effects of EPO in HG-treated PC12 cells. In diabetic rats, EPO prevented deficits in spatial learning and memory in the Morris water maze test. The results of real-time PCR and Western blotting showed that EPO upregulated EPO receptor, PI3K, and phosphorylated Akt2 relative to unphosphorylated Akt2 (p-Akt2/Akt2) and downregulated glycogen synthase kinase-3β (GSK-3β). These studies demonstrate that EPO is an effective neuroprotective agent in the context of diabetes-associated cognitive dysfunction and show that this effect involves the PI3K/Akt/GSK-3β pathway.
Collapse
|
142
|
Bragin DE, Kameneva MV, Bragina OA, Thomson S, Statom GL, Lara DA, Yang Y, Nemoto EM. Rheological effects of drag-reducing polymers improve cerebral blood flow and oxygenation after traumatic brain injury in rats. J Cereb Blood Flow Metab 2017; 37:762-775. [PMID: 28155574 PMCID: PMC5363490 DOI: 10.1177/0271678x16684153] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Cerebral ischemia has been clearly demonstrated after traumatic brain injury (TBI); however, neuroprotective therapies have not focused on improvement of the cerebral microcirculation. Blood soluble drag-reducing polymers (DRP), prepared from high molecular weight polyethylene oxide, target impaired microvascular perfusion by altering the rheological properties of blood and, until our recent reports, has not been applied to the brain. We hypothesized that DRP improve cerebral microcirculation and oxygenation after TBI. DRP were studied in healthy and traumatized rat brains and compared to saline controls. Using in-vivo two-photon laser scanning microscopy over the parietal cortex, we showed that after TBI, nanomolar concentrations of intravascular DRP significantly enhanced microvascular perfusion and tissue oxygenation in peri-contusional areas, preserved blood-brain barrier integrity and protected neurons. The mechanisms of DRP effects were attributable to reduction of the near-vessel wall cell-free layer which increased near-wall blood flow velocity, microcirculatory volume flow, and number of erythrocytes entering capillaries, thereby reducing capillary stasis and tissue hypoxia as reflected by a reduction in NADH. Our results indicate that early reduction in CBF after TBI is mainly due to ischemia; however, metabolic depression of contused tissue could be also involved.
Collapse
Affiliation(s)
- Denis E Bragin
- 1 Department of Neurosurgery, School of Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Marina V Kameneva
- 2 McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,3 Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA.,4 Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Olga A Bragina
- 1 Department of Neurosurgery, School of Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Susan Thomson
- 1 Department of Neurosurgery, School of Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Gloria L Statom
- 1 Department of Neurosurgery, School of Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Devon A Lara
- 1 Department of Neurosurgery, School of Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Yirong Yang
- 5 College of Pharmacy, University of New Mexico, Albuquerque, NM, USA
| | - Edwin M Nemoto
- 1 Department of Neurosurgery, School of Medicine, University of New Mexico, Albuquerque, NM, USA
| |
Collapse
|
143
|
Tamargo IA, Bader M, Li Y, Yu SJ, Wang Y, Talbot K, DiMarchi RD, Pick CG, Greig NH. Novel GLP-1R/GIPR co-agonist "twincretin" is neuroprotective in cell and rodent models of mild traumatic brain injury. Exp Neurol 2017; 288:176-186. [PMID: 27845037 PMCID: PMC5878017 DOI: 10.1016/j.expneurol.2016.11.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 11/08/2016] [Accepted: 11/10/2016] [Indexed: 12/12/2022]
Abstract
Several single incretin receptor agonists that are approved for the treatment of type 2 diabetes mellitus (T2DM) have been shown to be neuroprotective in cell and animal models of neurodegeneration. Recently, a synthetic dual incretin receptor agonist, nicknamed "twincretin," was shown to improve upon the metabolic benefits of single receptor agonists in mouse and monkey models of T2DM. In the current study, the neuroprotective effects of twincretin are probed in cell and mouse models of mild traumatic brain injury (mTBI), a prevalent cause of neurodegeneration in toddlers, teenagers and the elderly. Twincretin is herein shown to have activity at two different receptors, dose-dependently increase levels of intermediates in the neurotrophic CREB pathway and enhance viability of human neuroblastoma cells exposed to toxic concentrations of glutamate and hydrogen peroxide, insults mimicking the inflammatory conditions in the brain post-mTBI. Additionally, twincretin is shown to improve upon the neurotrophic effects of single incretin receptor agonists in these same cells. Finally, a clinically translatable dose of twincretin, when administered post-mTBI, is shown to fully restore the visual and spatial memory deficits induced by mTBI, as evaluated in a mouse model of weight drop close head injury. These results establish twincretin as a novel neuroprotective agent and suggest that it may improve upon the effects of the single incretin receptor agonists via dual agonism.
Collapse
MESH Headings
- Animals
- Body Temperature/drug effects
- Brain Injuries, Traumatic/complications
- Brain Injuries, Traumatic/drug therapy
- CREB-Binding Protein/metabolism
- Cell Line, Tumor
- Cells, Cultured
- Disease Models, Animal
- Embryo, Mammalian
- Glucagon-Like Peptide 1/metabolism
- Glucagon-Like Peptide-1 Receptor/agonists
- Glucagon-Like Peptide-1 Receptor/metabolism
- Humans
- Incretins/therapeutic use
- Male
- Maze Learning/drug effects
- Memory Disorders/etiology
- Memory Disorders/prevention & control
- Mice
- Mice, Inbred ICR
- Neuroblastoma/pathology
- Neuroprotective Agents/therapeutic use
- Rats
- Rats, Sprague-Dawley
- Reactive Oxygen Species/metabolism
- Receptors, Gastrointestinal Hormone/agonists
- Receptors, Gastrointestinal Hormone/metabolism
- Recognition, Psychology/drug effects
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Ian A Tamargo
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| | - Miaad Bader
- Department of Anatomy and Anthropology, Sackler School 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 21224, USA
| | - Seong-Jin Yu
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan
| | - Yun Wang
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan
| | | | | | - 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
| | - Nigel H Greig
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| |
Collapse
|
144
|
Serum Proteome Alterations in Patients with Cognitive Impairment after Traumatic Brain Injury Revealed by iTRAQ-Based Quantitative Proteomics. BIOMED RESEARCH INTERNATIONAL 2017; 2017:8572509. [PMID: 28251161 PMCID: PMC5303854 DOI: 10.1155/2017/8572509] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 12/01/2016] [Accepted: 12/13/2016] [Indexed: 12/26/2022]
Abstract
Background. Cognitive impairment is the leading cause of traumatic brain injury- (TBI-) related disability; however, the underlying pathogenesis of this dysfunction is not completely understood. Methods. Using an isobaric tagging for relative and absolute quantitation- (iTRAQ-) based quantitative proteomic approach, serum samples from healthy control subjects, TBI patients with cognitive impairment, and TBI patients without cognitive impairment were analysed to identify differentially expressed proteins (DEPs) related to post-TBI cognitive impairment. In addition, DEPs were further analysed using bioinformatic platforms and validated using enzyme-linked immunosorbent assays (ELISA). Results. A total of 56 DEPs were identified that were specifically related to TBI-induced cognitive impairment. Bioinformatic analysis revealed that a wide variety of cellular and metabolic processes and some signaling pathways were involved in the pathophysiology of cognitive deficits following TBI. Five randomly selected DEPs were validated using ELISA in an additional 105 cases, and the results also supported the experimental findings. Conclusions. Despite limitations, our findings will facilitate further studies of the pathological mechanisms underlying TBI-induced cognitive impairment and provide new methods for the research and development of neuroprotective agents. However, further investigation on a large cohort is warranted.
Collapse
|
145
|
Abou-El-Hassan H, Sukhon F, Assaf EJ, Bahmad H, Abou-Abbass H, Jourdi H, Kobeissy FH. Degradomics in Neurotrauma: Profiling Traumatic Brain Injury. Methods Mol Biol 2017; 1598:65-99. [PMID: 28508358 DOI: 10.1007/978-1-4939-6952-4_4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Degradomics has recently emerged as a subdiscipline in the omics era with a focus on characterizing signature breakdown products implicated in various disease processes. Driven by promising experimental findings in cancer, neuroscience, and metabolomic disorders, degradomics has significantly promoted the notion of disease-specific "degradome." A degradome arises from the activation of several proteases that target specific substrates and generate signature protein fragments. Several proteases such as calpains, caspases, cathepsins, and matrix metalloproteinases (MMPs) are involved in the pathogenesis of numerous diseases that disturb the physiologic balance between protein synthesis and protein degradation. While regulated proteolytic activities are needed for development, growth, and regeneration, uncontrolled proteolysis initiated under pathological conditions ultimately culminates into apoptotic and necrotic processes. In this chapter, we aim to review the protease-substrate repertoires in neural injury concentrating on traumatic brain injury. A striking diversity of protease substrates, essential for neuronal and brain structural and functional integrity, namely, encryptic biomarker neoproteins, have been characterized in brain injury. These include cytoskeletal proteins, transcription factors, cell cycle regulatory proteins, synaptic proteins, and cell junction proteins. As these substrates are subject to proteolytic fragmentation, they are ceaselessly exposed to activated proteases. Characterization of these molecules allows for a surge of "possible" therapeutic approaches of intervention at various levels of the proteolytic cascade.
Collapse
Affiliation(s)
- Hadi Abou-El-Hassan
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon.
| | - Fares Sukhon
- Faculty of Medicine, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Edwyn Jeremy Assaf
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Hisham Bahmad
- Faculty of Medical, Neuroscience Research Center, Beirut Arab University, Beirut, Lebanon
- Faculty of Medicine, Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut, Lebanon
| | - Hussein Abou-Abbass
- Faculty of Medical Sciences, Neuroscience Research Center, Lebanese University, Beirut, Lebanon
- Faculty of Medicine, Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Hussam Jourdi
- Faculty of Science¸ Department of Biology, University of Balamand, Souk-el-Gharb Campus, Aley, Lebanon
| | - Firas H Kobeissy
- Faculty of Medicine, Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon.
- Department of Psychiatry, Center for Neuroproteomics and Biomarkers Research, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
146
|
Tripodis Y, Alosco ML, Zirogiannis N, Gavett BE, Chaisson C, Martin B, McClean MD, Mez J, Kowall N, Stern RA. The Effect of Traumatic Brain Injury History with Loss of Consciousness on Rate of Cognitive Decline Among Older Adults with Normal Cognition and Alzheimer's Disease Dementia. J Alzheimers Dis 2017; 59:251-263. [PMID: 28655133 PMCID: PMC5614490 DOI: 10.3233/jad-160585] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Traumatic brain injury (TBI) is thought to be a risk factor for dementia, including dementia due to Alzheimer's disease (AD). However, the influence of TBI history on the neuropsychological course of AD is unknown and, more broadly, the effect of TBI history on age-related cognitive change is poorly understood. We examined the relationship between history of TBI with loss of consciousness (LOC) history and cognitive change in participants with normal cognition and probable AD, stratified by APOEɛ4 allele status. The sample included 706 participants (432 with normal cognition; 274 probable AD) from the National Alzheimer's Coordinating Center (NACC) dataset that completed the Uniform Data Set evaluation between 2005 and 2014. Normal and probable AD participants with a history of TBI were matched to an equal number of demographically and clinically similar participants without a TBI history. In this dataset, TBI with LOC was defined as brain trauma with brief or extended unconsciousness. For the normal and probable AD cohorts, there was an average of 3.2±1.9 and 1.8±1.1 years of follow-up, respectively. 30.8% of the normal cohort were APOEɛ4 carriers, whereas 70.8% of probable AD participants were carriers. Mixed effects regressions showed TBI with LOC history did not affect rates of cognitive change in APOEɛ4 carriers and non-carriers. Findings from this study suggest that TBI with LOC may not alter the course of cognitive function in older adults with and without probable AD. Future studies that better characterize TBI (e.g., severity, number of TBIs, history of subconconcussive exposure) are needed to clarify the association between TBI and long-term neurocognitive outcomes.
Collapse
Affiliation(s)
| | - Michael L. Alosco
- Alzheimer’s Disease Center, Boston University School of Medicine, Boston, MA, USA
| | - Nikolaos Zirogiannis
- Indiana University School of Public and Environmental Affairs, Bloomington, IN, USA
| | - Brandon E. Gavett
- Department of Psychology, University of Colorado Colorado Springs, Colorado Springs, CO, USA
| | | | - Brett Martin
- Boston University School of Public Health, Boston, MA, USA
| | | | - Jesse Mez
- Alzheimer’s Disease Center, Boston University School of Medicine, Boston, MA, USA
| | - Neil Kowall
- Alzheimer’s Disease Center, Boston University School of Medicine, Boston, MA, USA
- Department of Psychology, University of Colorado Colorado Springs, Colorado Springs, CO, USA
| | | |
Collapse
|
147
|
Zhou J, Burns MP, Huynh L, Villapol S, Taub DD, Saavedra JM, Blackman MR. Temporal Changes in Cortical and Hippocampal Expression of Genes Important for Brain Glucose Metabolism Following Controlled Cortical Impact Injury in Mice. Front Endocrinol (Lausanne) 2017; 8:231. [PMID: 28955302 PMCID: PMC5601958 DOI: 10.3389/fendo.2017.00231] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 08/24/2017] [Indexed: 02/06/2023] Open
Abstract
Traumatic brain injury (TBI) causes transient increases and subsequent decreases in brain glucose utilization. The underlying molecular pathways are orchestrated processes and poorly understood. In the current study, we determined temporal changes in cortical and hippocampal expression of genes important for brain glucose/lactate metabolism and the effect of a known neuroprotective drug telmisartan on the expression of these genes after experimental TBI. Adult male C57BL/6J mice (n = 6/group) underwent sham or unilateral controlled cortical impact (CCI) injury. Their ipsilateral and contralateral cortex and hippocampus were collected 6 h, 1, 3, 7, 14, 21, and 28 days after injury. Expressions of several genes important for brain glucose utilization were determined by qRT-PCR. In results, (1) mRNA levels of three key enzymes in glucose metabolism [hexo kinase (HK) 1, pyruvate kinase, and pyruvate dehydrogenase (PDH)] were all increased 6 h after injury in the contralateral cortex, followed by decreases at subsequent times in the ipsilateral cortex and hippocampus; (2) capillary glucose transporter Glut-1 mRNA increased, while neuronal glucose transporter Glut-3 mRNA decreased, at various times in the ipsilateral cortex and hippocampus; (3) astrocyte lactate transporter MCT-1 mRNA increased, whereas neuronal lactate transporter MCT-2 mRNA decreased in the ipsilateral cortex and hippocampus; (4) HK2 (an isoform of hexokinase) expression increased at all time points in the ipsilateral cortex and hippocampus. GPR81 (lactate receptor) mRNA increased at various time points in the ipsilateral cortex and hippocampus. These temporal alterations in gene expression corresponded closely to the patterns of impaired brain glucose utilization reported in both TBI patients and experimental TBI rodents. The observed changes in hippocampal gene expression were delayed and prolonged, when compared with those in the cortex. The patterns of alterations were specific to different brain regions and exhibited different recovery periods following TBI. Oral administration of telmisartan (1 mg/kg, for 7 days, n = 10 per group) ameliorated cortical or hippocampal mRNA for Glut-1/3, MCT-1/2 and PDH in CCI mice. These data provide molecular evidence for dynamic alteration of multiple critical factors in brain glucose metabolism post-TBI and can inform further research for treating brain metabolic disorders post-TBI.
Collapse
Affiliation(s)
- June Zhou
- Research Service, Washington DC VA Medical Center, Washington, DC, United States
- Department of Biochemistry and Molecular Medicine, George Washington University School of Medicine, Washington, DC, United States
- *Correspondence: June Zhou,
| | - Mark P. Burns
- Department of Neuroscience, Georgetown University School of Medicine, Washington, DC, United States
| | - Linda Huynh
- Research Service, Washington DC VA Medical Center, Washington, DC, United States
| | - Sonia Villapol
- Department of Neuroscience, Georgetown University School of Medicine, Washington, DC, United States
| | - Daniel D. Taub
- Translational Medicine Section, Washington DC VA Medical Center, Washington, DC, United States
- Department of Biochemistry and Molecular and Cell Biology, Georgetown University School of Medicine, Washington, DC, United States
| | - Juan M. Saavedra
- Department of Pharmacology and Physiology, Georgetown University School of Medicine, Washington, DC, United States
| | - Marc R. Blackman
- Research Service, Washington DC VA Medical Center, Washington, DC, United States
- Department of Biochemistry and Molecular Medicine, George Washington University School of Medicine, Washington, DC, United States
- Department of Medicine George Washington University School of Medicine, Washington, DC, United States
- Department of Medicine, Georgetown University School of Medicine, Washington, DC, United States
| |
Collapse
|
148
|
Pu H, Jiang X, Wei Z, Hong D, Hassan S, Zhang W, Liu J, Meng H, Shi Y, Chen L, Chen J. Repetitive and Prolonged Omega-3 Fatty Acid Treatment After Traumatic Brain Injury Enhances Long-Term Tissue Restoration and Cognitive Recovery. Cell Transplant 2016; 26:555-569. [PMID: 27938482 DOI: 10.3727/096368916x693842] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Traumatic brain injury (TBI) is one of the most disabling clinical conditions that could lead to neurocognitive disorders in survivors. Our group and others previously reported that prophylactic enrichment of dietary omega-3 polyunsaturated fatty acids (n-3 PUFAs) markedly ameliorate cognitive deficits after TBI. However, it remains unclear whether a clinically relevant therapeutic regimen with n-3 PUFAs administered after TBI would still offer significant improvement of long-term cognitive recovery. In the present study, we employed the decline of spatial cognitive function as a main outcome after TBI to investigate the therapeutic efficacy of post-TBI n-3 PUFA treatment and the underlying mechanisms. Mice were subjected to sham operation or controlled cortical impact, followed by random assignment to receive the following four treatments: (1) vehicle control; (2) daily intraperitoneal injections of n-3 PUFAs for 2 weeks, beginning 2 h after TBI; (3) fish oil dietary supplementation throughout the study, beginning 1 day after TBI; or (4) combination of treatments (2) and (3). Spatial cognitive deficits and chronic brain tissue loss, as well as endogenous brain repair processes such as neurogenesis, angiogenesis, and oligodendrogenesis, were evaluated up to 35 days after TBI. The results revealed prominent spatial cognitive deficits and massive tissue loss caused by TBI. Among all mice receiving post-TBI n-3 PUFA treatments, the combined treatment of fish oil dietary supplement and n-3 PUFA injections demonstrated a reproducible beneficial effect in attenuating cognitive deficits although without reducing gross tissue loss. Mechanistically, the combined treatment promoted post-TBI restorative processes in the brain, including generation of immature neurons, microvessels, and oligodendrocytes, each of which was significantly correlated with the improved cognitive recovery. These results indicated that repetitive and prolonged n-3 PUFA treatments after TBI are capable of enhancing brain remodeling and could be developed as a potential therapy to treat TBI victims in the clinic.
Collapse
|
149
|
Chandran R, Sharma A, Bhomia M, Balakathiresan NS, Knollmann-Ritschel BE, Maheshwari RK. Differential expression of microRNAs in the brains of mice subjected to increasing grade of mild traumatic brain injury. Brain Inj 2016; 31:106-119. [PMID: 27819514 DOI: 10.1080/02699052.2016.1213420] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To investigate the effect of heterogeneity in mTBI on miRNA expression in mouse brain and to identify molecular pathways targeted by the modulated miRNAs. METHODS A weight drop device was used to induce four increasing grades of mTBI. MiRNA expression was evaluated using TaqMan rodent miRNA arrays. Bioinformatics analysis was done using the DIANA miRPath tool and Ingenuity Pathway Analysis software. Histology of brain sections was evaluated using H&E staining. RESULTS No histologic lesions were observed in the brains of injured mice; however, significant modulation in miRNA expression profile was observed. Global miRNA profiling indicated a trend of decrease in the number of modulated miRNAs from 24 hours to day 7 post-injury, except for the most severe grade of mTBI. Canonical pathways like calcium signalling, synaptic pathways and axon guidance pathway were the major targets of the modulated miRNAs. Network correlation analyses indicated an interaction between the modulated miRNAs and putative protein biomarkers of TBI. CONCLUSIONS The data demonstrated that varying intensities of mTBI induced a differential miRNA expression profile in the brain post-injury. Pathways such as calcium and synaptic signalling were major targets of modulated miRNAs and may play a role in the pathophysiology of mTBI.
Collapse
Affiliation(s)
- Raghavendar Chandran
- a Department of Pathology , Uniformed Services University of the Health Sciences , Bethesda , MD , USA.,b Biological Sciences Group , Birla Institute of Technology and Science , Pilani , Rajasthan , India
| | - Anuj Sharma
- a Department of Pathology , Uniformed Services University of the Health Sciences , Bethesda , MD , USA
| | - Manish Bhomia
- a Department of Pathology , Uniformed Services University of the Health Sciences , Bethesda , MD , USA
| | - Nagaraja S Balakathiresan
- a Department of Pathology , Uniformed Services University of the Health Sciences , Bethesda , MD , USA
| | | | - Radha K Maheshwari
- a Department of Pathology , Uniformed Services University of the Health Sciences , Bethesda , MD , USA
| |
Collapse
|
150
|
Veeramuthu V, Narayanan V, Ramli N, Hernowo A, Waran V, Bondi MW, Delano-Wood L, Ganesan D. Neuropsychological Outcomes in Patients with Complicated Versus Uncomplicated Mild Traumatic Brain Injury: 6-Month Follow-Up. World Neurosurg 2016; 97:416-423. [PMID: 27751922 DOI: 10.1016/j.wneu.2016.10.041] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Revised: 10/04/2016] [Accepted: 10/06/2016] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To compare the extent of persistent neuropsychological impairment in patients with complicated mild traumatic brain injury (mTBI) and those with uncomplicated mTBI. METHODS Sixty-one patients with mTBI (Glasgow Coma Scale score 13-15) were recruited prospectively, categorized according to baseline computed tomography findings, and subjected to neuropsychological assessment at initial admission (n = 61) as well as at a 6-month follow-up (n = 30). The paired t test, Cohen's d effect size calculation, and repeated-measures analysis of variance were used to establish the differences between the 2 groups in terms of neuropsychological performance. RESULTS A trend toward poorer neuropsychological performance among the patients with complicated mTBI was observed during admission; however, performance in this group improved over time. In contrast, the uncomplicated mTBI group showed slower recovery, especially in tasks of memory, visuospatial processing, and executive functions, at follow-up. CONCLUSIONS Our findings suggest that despite the broad umbrella designation of mTBI, the current classification schemes of injury severity for mild neurotrauma should be revisited. They also raise questions about the clinical relevance of both traumatic focal lesions and the absence of visible traumatic lesions on brain imaging studies in patients with milder forms of head trauma.
Collapse
Affiliation(s)
- Vigneswaran Veeramuthu
- Division of Neurosurgery, Department of Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Vairavan Narayanan
- Division of Neurosurgery, Department of Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.
| | - Norlisah Ramli
- Research Imaging Centre, University of Malaya, Kuala Lumpur, Malaysia
| | - Aditya Hernowo
- Division of Neurosurgery, Department of Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Vicknes Waran
- Division of Neurosurgery, Department of Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Mark W Bondi
- VA San Diego Healthcare System, San Diego, California, USA; Department of Psychiatry, University of California San Diego, La Jolla, California, USA
| | - Lisa Delano-Wood
- VA San Diego Healthcare System, San Diego, California, USA; Department of Psychiatry, University of California San Diego, La Jolla, California, USA
| | - Dharmendra Ganesan
- Division of Neurosurgery, Department of Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|