1
|
Jacquens A, Needham EJ, Zanier ER, Degos V, Gressens P, Menon D. Neuro-Inflammation Modulation and Post-Traumatic Brain Injury Lesions: From Bench to Bed-Side. Int J Mol Sci 2022; 23:11193. [PMID: 36232495 PMCID: PMC9570205 DOI: 10.3390/ijms231911193] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/14/2022] [Accepted: 09/15/2022] [Indexed: 11/16/2022] Open
Abstract
Head trauma is the most common cause of disability in young adults. Known as a silent epidemic, it can cause a mosaic of symptoms, whether neurological (sensory-motor deficits), psychiatric (depressive and anxiety symptoms), or somatic (vertigo, tinnitus, phosphenes). Furthermore, cranial trauma (CT) in children presents several particularities in terms of epidemiology, mechanism, and physiopathology-notably linked to the attack of an immature organ. As in adults, head trauma in children can have lifelong repercussions and can cause social and family isolation, difficulties at school, and, later, socio-professional adversity. Improving management of the pre-hospital and rehabilitation course of these patients reduces secondary morbidity and mortality, but often not without long-term disability. One hypothesized contributor to this process is chronic neuroinflammation, which could accompany primary lesions and facilitate their development into tertiary lesions. Neuroinflammation is a complex process involving different actors such as glial cells (astrocytes, microglia, oligodendrocytes), the permeability of the blood-brain barrier, excitotoxicity, production of oxygen derivatives, cytokine release, tissue damage, and neuronal death. Several studies have investigated the effect of various treatments on the neuroinflammatory response in traumatic brain injury in vitro and in animal and human models. The aim of this review is to examine the various anti-inflammatory therapies that have been implemented.
Collapse
Affiliation(s)
- Alice Jacquens
- Unité de Neuroanesthésie-Réanimation, Hôpital de la Pitié Salpêtrière 43-87, Boulevard de l’Hôpital, F-75013 Paris, France
- Inserm, Maladies Neurodéveloppementales et Neurovasculaires, Université Paris Cité, F-75019 Paris, France
| | - Edward J. Needham
- Division of Anaesthesia, Addenbrooke’s Hospital, University of Cambridge, Box 93, Hills Road, Cambridge CB2 2QQ, UK
| | - Elisa R. Zanier
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| | - Vincent Degos
- Unité de Neuroanesthésie-Réanimation, Hôpital de la Pitié Salpêtrière 43-87, Boulevard de l’Hôpital, F-75013 Paris, France
- Inserm, Maladies Neurodéveloppementales et Neurovasculaires, Université Paris Cité, F-75019 Paris, France
| | - Pierre Gressens
- Inserm, Maladies Neurodéveloppementales et Neurovasculaires, Université Paris Cité, F-75019 Paris, France
| | - David Menon
- Division of Anaesthesia, Addenbrooke’s Hospital, University of Cambridge, Box 93, Hills Road, Cambridge CB2 2QQ, UK
| |
Collapse
|
2
|
Golub VM, Reddy DS. Post-Traumatic Epilepsy and Comorbidities: Advanced Models, Molecular Mechanisms, Biomarkers, and Novel Therapeutic Interventions. Pharmacol Rev 2022; 74:387-438. [PMID: 35302046 PMCID: PMC8973512 DOI: 10.1124/pharmrev.121.000375] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Post-traumatic epilepsy (PTE) is one of the most devastating long-term, network consequences of traumatic brain injury (TBI). There is currently no approved treatment that can prevent onset of spontaneous seizures associated with brain injury, and many cases of PTE are refractory to antiseizure medications. Post-traumatic epileptogenesis is an enduring process by which a normal brain exhibits hypersynchronous excitability after a head injury incident. Understanding the neural networks and molecular pathologies involved in epileptogenesis are key to preventing its development or modifying disease progression. In this article, we describe a critical appraisal of the current state of PTE research with an emphasis on experimental models, molecular mechanisms of post-traumatic epileptogenesis, potential biomarkers, and the burden of PTE-associated comorbidities. The goal of epilepsy research is to identify new therapeutic strategies that can prevent PTE development or interrupt the epileptogenic process and relieve associated neuropsychiatric comorbidities. Therefore, we also describe current preclinical and clinical data on the treatment of PTE sequelae. Differences in injury patterns, latency period, and biomarkers are outlined in the context of animal model validation, pathophysiology, seizure frequency, and behavior. Improving TBI recovery and preventing seizure onset are complex and challenging tasks; however, much progress has been made within this decade demonstrating disease modifying, anti-inflammatory, and neuroprotective strategies, suggesting this goal is pragmatic. Our understanding of PTE is continuously evolving, and improved preclinical models allow for accelerated testing of critically needed novel therapeutic interventions in military and civilian persons at high risk for PTE and its devastating comorbidities.
Collapse
Affiliation(s)
- Victoria M Golub
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas
| | - Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas
| |
Collapse
|
3
|
McDonald BZ, Gee CC, Kievit FM. The Nanotheranostic Researcher’s Guide for Use of Animal Models of Traumatic Brain Injury. JOURNAL OF NANOTHERANOSTICS 2021; 2:224-268. [PMID: 35655793 PMCID: PMC9159501 DOI: 10.3390/jnt2040014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Traumatic brain injury (TBI) is currently the leading cause of injury-related morbidity and mortality worldwide, with an estimated global cost of USD 400 billion annually. Both clinical and preclinical behavioral outcomes associated with TBI are heterogeneous in nature and influenced by the mechanism and frequency of injury. Previous literature has investigated this relationship through the development of animal models and behavioral tasks. However, recent advancements in these methods may provide insight into the translation of therapeutics into a clinical setting. In this review, we characterize various animal models and behavioral tasks to provide guidelines for evaluating the therapeutic efficacy of treatment options in TBI. We provide a brief review into the systems utilized in TBI classification and provide comparisons to the animal models that have been developed. In addition, we discuss the role of behavioral tasks in evaluating outcomes associated with TBI. Our goal is to provide those in the nanotheranostic field a guide for selecting an adequate TBI animal model and behavioral task for assessment of outcomes to increase research in this field.
Collapse
|
4
|
Shultz SR, McDonald SJ, Corrigan F, Semple BD, Salberg S, Zamani A, Jones NC, Mychasiuk R. Clinical Relevance of Behavior Testing in Animal Models of Traumatic Brain Injury. J Neurotrauma 2020; 37:2381-2400. [DOI: 10.1089/neu.2018.6149] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Sandy R. Shultz
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Stuart J. McDonald
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Physiology, Anatomy, and Microbiology, La Trobe University, Melbourne, Victoria, Australia
| | - Frances Corrigan
- Department of Anatomy, University of South Australia, Adelaide, South Australia, Australia
| | - Bridgette D. Semple
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Sabrina Salberg
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Akram Zamani
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Nigel C. Jones
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
5
|
Abstract
Rodents are the most widely used experimental animals in stroke research due to their similar vascular anatomy, high reproductive rates, and availability of transgenic models. However, the difficulties in assessing higher brain functions, such as cognition and memory, in rodents decrease the translational potential of these studies. In this review, we summarize commonly used motor/sensorimotor and cognition tests in rodent models of stroke. Specifically, we first briefly introduce the objective and procedure of each behavioral test. Next, we summarize the application of each test in both ischemic stroke and hemorrhagic stroke. Last, the advantages and disadvantages of these tests in assessing stroke outcome are discussed. This review summarizes commonly used behavioral tests in stroke studies and compares their applications in different stroke types.
Collapse
Affiliation(s)
- Jingsong Ruan
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, 240 W Green Street, Athens, GA, USA
| | - Yao Yao
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, 240 W Green Street, Athens, GA, USA
| |
Collapse
|
6
|
Executive (dys)function after traumatic brain injury: special considerations for behavioral pharmacology. Behav Pharmacol 2019; 29:617-637. [PMID: 30215621 PMCID: PMC6155367 DOI: 10.1097/fbp.0000000000000430] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Executive function is an umbrella term that includes cognitive processes such as decision-making, impulse control, attention, behavioral flexibility, and working memory. Each of these processes depends largely upon monoaminergic (dopaminergic, serotonergic, and noradrenergic) neurotransmission in the frontal cortex, striatum, and hippocampus, among other brain areas. Traumatic brain injury (TBI) induces disruptions in monoaminergic signaling along several steps in the neurotransmission process - synthesis, distribution, and breakdown - and in turn, produces long-lasting deficits in several executive function domains. Understanding how TBI alters monoamingeric neurotransmission and executive function will advance basic knowledge of the underlying principles that govern executive function and potentially further treatment of cognitive deficits following such injury. In this review, we examine the influence of TBI on the following measures of executive function - impulsivity, behavioral flexibility, and working memory. We also describe monoaminergic-systems changes following TBI. Given that TBI patients experience alterations in monoaminergic signaling following injury, they may represent a unique population with regard to pharmacotherapy. We conclude this review by discussing some considerations for pharmacotherapy in the field of TBI.
Collapse
|
7
|
Magnesium Is a Key Player in Neuronal Maturation and Neuropathology. Int J Mol Sci 2019; 20:ijms20143439. [PMID: 31336935 PMCID: PMC6678825 DOI: 10.3390/ijms20143439] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 07/06/2019] [Accepted: 07/09/2019] [Indexed: 01/05/2023] Open
Abstract
Magnesium (Mg) is the second most abundant cation in mammalian cells, and it is essential for numerous cellular processes including enzymatic reactions, ion channel functions, metabolic cycles, cellular signaling, and DNA/RNA stabilities. Because of the versatile and universal nature of Mg2+, the homeostasis of intracellular Mg2+ is physiologically linked to growth, proliferation, differentiation, energy metabolism, and death of cells. On the cellular and tissue levels, maintaining Mg2+ within optimal levels according to the biological context, such as cell types, developmental stages, extracellular environments, and pathophysiological conditions, is crucial for development, normal functions, and diseases. Hence, Mg2+ is pathologically involved in cancers, diabetes, and neurodegenerative diseases, such as Parkinson's disease, Alzheimer's disease, and demyelination. In the research field regarding the roles and mechanisms of Mg2+ regulation, numerous controversies caused by its versatility and complexity still exist. As Mg2+, at least, plays critical roles in neuronal development, healthy normal functions, and diseases, appropriate Mg2+ supplementation exhibits neurotrophic effects in a majority of cases. Hence, the control of Mg2+ homeostasis can be a candidate for therapeutic targets in neuronal diseases. In this review, recent results regarding the roles of intracellular Mg2+ and its regulatory system in determining the cell phenotype, fate, and diseases in the nervous system are summarized, and an overview of the comprehensive roles of Mg2+ is provided.
Collapse
|
8
|
Semple BD, Zamani A, Rayner G, Shultz SR, Jones NC. Affective, neurocognitive and psychosocial disorders associated with traumatic brain injury and post-traumatic epilepsy. Neurobiol Dis 2018; 123:27-41. [PMID: 30059725 DOI: 10.1016/j.nbd.2018.07.018] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 07/08/2018] [Accepted: 07/16/2018] [Indexed: 12/13/2022] Open
Abstract
Survivors of traumatic brain injury (TBI) often develop chronic neurological, neurocognitive, psychological, and psychosocial deficits that can have a profound impact on an individual's wellbeing and quality of life. TBI is also a common cause of acquired epilepsy, which is itself associated with significant behavioral morbidity. This review considers the clinical and preclinical evidence that post-traumatic epilepsy (PTE) acts as a 'second-hit' insult to worsen chronic behavioral outcomes for brain-injured patients, across the domains of emotional, cognitive, and psychosocial functioning. Surprisingly, few well-designed studies have specifically examined the relationship between seizures and behavioral outcomes after TBI. The complex mechanisms underlying these comorbidities remain incompletely understood, although many of the biological processes that precipitate seizure occurrence and epileptogenesis may also contribute to the development of chronic behavioral deficits. Further, the relationship between PTE and behavioral dysfunction is increasingly recognized to be a bidirectional one, whereby premorbid conditions are a risk factor for PTE. Clinical studies in this arena are often challenged by the confounding effects of anti-seizure medications, while preclinical studies have rarely examined an adequately extended time course to fully capture the time course of epilepsy development after a TBI. To drive the field forward towards improved treatment strategies, it is imperative that both seizures and neurobehavioral outcomes are assessed in parallel after TBI, both in patient populations and preclinical models.
Collapse
Affiliation(s)
- Bridgette D Semple
- Department of Neuroscience, Monash University, 99 Commercial Road, Melbourne, VIC, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Royal Parade, Parkville, VIC, Australia.
| | - Akram Zamani
- Department of Neuroscience, Monash University, 99 Commercial Road, Melbourne, VIC, Australia.
| | - Genevieve Rayner
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre (Austin Campus), Heidelberg, VIC, Australia; Melbourne School of Psychological Sciences, The University of Melbourne, Parkville, VIC, Australia; Comprehensive Epilepsy Program, Alfred Health, Australia.
| | - Sandy R Shultz
- Department of Neuroscience, Monash University, 99 Commercial Road, Melbourne, VIC, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Royal Parade, Parkville, VIC, Australia.
| | - Nigel C Jones
- Department of Neuroscience, Monash University, 99 Commercial Road, Melbourne, VIC, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Royal Parade, Parkville, VIC, Australia.
| |
Collapse
|
9
|
Wang P, Wang ZY. Metal ions influx is a double edged sword for the pathogenesis of Alzheimer's disease. Ageing Res Rev 2017; 35:265-290. [PMID: 27829171 DOI: 10.1016/j.arr.2016.10.003] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Revised: 09/08/2016] [Accepted: 10/17/2016] [Indexed: 12/17/2022]
Abstract
Alzheimer's disease (AD) is a common form of dementia in aged people, which is defined by two pathological characteristics: β-amyloid protein (Aβ) deposition and tau hyperphosphorylation. Although the mechanisms of AD development are still being debated, a series of evidence supports the idea that metals, such as copper, iron, zinc, magnesium and aluminium, are involved in the pathogenesis of the disease. In particular, the processes of Aβ deposition in senile plaques (SP) and the inclusion of phosphorylated tau in neurofibrillary tangles (NFTs) are markedly influenced by alterations in the homeostasis of the aforementioned metal ions. Moreover, the mechanisms of oxidative stress, synaptic plasticity, neurotoxicity, autophagy and apoptosis mediate the effects of metal ions-induced the aggregation state of Aβ and phosphorylated tau on AD development. More importantly, imbalance of these mechanisms finally caused cognitive decline in different experiment models. Collectively, reconstructing the signaling network that regulates AD progression by metal ions may provide novel insights for developing chelators specific for metal ions to combat AD.
Collapse
Affiliation(s)
- Pu Wang
- College of Life and Health Sciences, Northeastern University, No. 3-11, Wenhua Road, Shenyang, 110819, PR China.
| | - Zhan-You Wang
- College of Life and Health Sciences, Northeastern University, No. 3-11, Wenhua Road, Shenyang, 110819, PR China.
| |
Collapse
|
10
|
Vonder Haar C, Peterson TC, Martens KM, Hoane MR. Vitamins and nutrients as primary treatments in experimental brain injury: Clinical implications for nutraceutical therapies. Brain Res 2016; 1640:114-129. [PMID: 26723564 PMCID: PMC4870112 DOI: 10.1016/j.brainres.2015.12.030] [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: 10/15/2015] [Revised: 12/14/2015] [Accepted: 12/15/2015] [Indexed: 02/07/2023]
Abstract
With the numerous failures of pharmaceuticals to treat traumatic brain injury in humans, more researchers have become interested in combination therapies. This is largely due to the multimodal nature of damage from injury, which causes excitotoxicity, oxidative stress, edema, neuroinflammation and cell death. Polydrug treatments have the potential to target multiple aspects of the secondary injury cascade, while many previous therapies focused on one particular aspect. Of specific note are vitamins, minerals and nutrients that can be utilized to supplement other therapies. Many of these have low toxicity, are already FDA approved and have minimal interactions with other drugs, making them attractive targets for therapeutics. Over the past 20 years, interest in supplementation and supraphysiologic dosing of nutrients for brain injury has increased and indeed many vitamins and nutrients now have a considerable body of the literature backing their use. Here, we review several of the prominent therapies in the category of nutraceutical treatment for brain injury in experimental models, including vitamins (B2, B3, B6, B9, C, D, E), herbs and traditional medicines (ginseng, Gingko biloba), flavonoids, and other nutrients (magnesium, zinc, carnitine, omega-3 fatty acids). While there is still much work to be done, several of these have strong potential for clinical therapies, particularly with regard to polydrug regimens. This article is part of a Special Issue entitled SI:Brain injury and recovery.
Collapse
|
11
|
Abstract
BACKGROUND The purpose of this systematic review was to evaluate the effect of magnesium sulfate in the treatment of acute traumatic brain injury. MATERIALS AND METHODS A systematic search of ClinicalTrials.gov, the Cochrane Library database, EMBASE, MEDLINE, Web of Science, and the World Health Organization trial registry, plus manual searches of gray literature, was undertaken in April 2013. Two reviewers independently extracted the data with a predefined data extraction form. RevMan 5 software was used to synthesize data and calculate the risk ratio for mortality with the 95% confidence interval. For the Glasgow Outcome Scale and posttreatment Glasgow Coma Scale data, the weighted mean difference was calculated with the 95% confidence interval. RESULTS A total of 8 randomized controlled trials with a total of 786 patients were included. Meta-analysis showed that there was no significant difference between the groups for mortality. The Glasgow Outcome Scale of the treatment group was higher than that of the control group, although the significance was borderline. The Glasgow Coma Scale score change posttreatment was significantly higher than that of the control. CONCLUSIONS The present meta-analysis of existing randomized controlled trials does not identify a significant beneficial effect in the mortality of traumatic brain injury patients; however, it suggests that magnesium sulfate shows a tendency to improve the Glasgow Outcome Scale and Glasgow Coma Scale scores, which is a promising result for traumatic brain injury therapy. Further effort is necessary to explore which subgroup of traumatic brain injury patients could benefit from magnesium sulfate.
Collapse
|
12
|
Balança B, Bapteste L, Lieutaud T, Ressnikoff D, Guy R, Bezin L, Marinesco S. Neuronal loss as evidenced by automated quantification of neuronal density following moderate and severe traumatic brain injury in rats. J Neurosci Res 2015; 94:39-49. [PMID: 26451689 DOI: 10.1002/jnr.23676] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 08/10/2015] [Accepted: 09/14/2015] [Indexed: 11/11/2022]
Abstract
Traumatic brain injury causes widespread neurological lesions that can be reproduced in animals with the lateral fluid percussion (LFP) model. The characterization of the pattern of neuronal death generated in this model remains unclear, involving both cortical and subcortical brain regions. Here, 7 days after moderate (3 atmospheres absolute [ATA]) or severe (3.8 ATA) LFP, we estimated neuronal loss by using immunohistochemistry together with a computer-assisted automated method for quantifying neuronal density in brain sections. Neuronal counts were performed ipsilateral to the impact, in the parietal cortex ventral to the site of percussion, in the temporal cortex, in the dorsal thalamus, and in the hippocampus. These results were compared with the counts observed at similar areas in sham animals. We found that neuronal density was severely decreased in the temporal cortex (-60%), in the dorsal thalamus (-63%), and in area CA3 of the hippocampus (-36%) of injured animals compared with controls but was not significantly modified in the cortices located immediately ventral to the impact. Total cellular density increased in brain structures displaying neuronal death, suggesting the presence of gliosis. The increase in the severity of LFP did not change the pattern of neuronal injury. This automated method simplified the study of neuronal loss following traumatic brain injury and allowed the identification of a pattern of neuronal loss that spreads from the dorsal thalamus to the temporal cortex, with the most severe lesions being in brain structures remote from the site of impact.
Collapse
Affiliation(s)
- Baptiste Balança
- INSERM U1028, CNRS UMR5292, Lyon Neuroscience Research Center, Team WAKING, Lyon, France.,Department of Anesthesiology and Intensive Care, P. Wertheimer Neurological Hospital, Hospices Civils de Lyon, Lyon, France.,University Claude Bernard Lyon 1, Lyon, France
| | - Lionel Bapteste
- Department of Anesthesiology and Intensive Care, P. Wertheimer Neurological Hospital, Hospices Civils de Lyon, Lyon, France.,University Claude Bernard Lyon 1, Lyon, France
| | - Thomas Lieutaud
- University Claude Bernard Lyon 1, Lyon, France.,INSERM U1028, CNRS UMR5292, Lyon Neuroscience Research Center, Team TIGER, Lyon, France
| | - Denis Ressnikoff
- University Claude Bernard Lyon 1, Lyon, France.,Centre d'Imagerie Quantitative Lyon Est, Lyon, France
| | - Rainui Guy
- INSERM U1028, CNRS UMR5292, Lyon Neuroscience Research Center, Team WAKING, Lyon, France.,University Claude Bernard Lyon 1, Lyon, France
| | - Laurent Bezin
- University Claude Bernard Lyon 1, Lyon, France.,INSERM U1028, CNRS UMR5292, Lyon Neuroscience Research Center, Team TIGER, Lyon, France
| | - Stéphane Marinesco
- INSERM U1028, CNRS UMR5292, Lyon Neuroscience Research Center, Team WAKING, Lyon, France.,University Claude Bernard Lyon 1, Lyon, France.,INSERM U1028, CNRS UMR5292, Lyon Neuroscience Research Center, AniRA-Neurochem Technological Platform, Lyon, France
| |
Collapse
|
13
|
Chang CZ, Wu SC, Kwan AL, Lin CL. Magnesium Lithospermate B Implicates 3'-5'-Cyclic Adenosine Monophosphate/Protein Kinase A Pathway and N-Methyl-d-Aspartate Receptors in an Experimental Traumatic Brain Injury. World Neurosurg 2015; 84:954-63. [PMID: 26093361 DOI: 10.1016/j.wneu.2015.05.075] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 05/12/2015] [Accepted: 05/14/2015] [Indexed: 10/23/2022]
Abstract
OBJECTIVE Decreased 3'-5'-cyclic adenosine monophosphate (cAMP), protein kinase A (PKA), and increased N-methyl-d-aspartate (NMDA) related apoptosis were observed in traumatic brain injury (TBI). It is of interest to examine the effect of magnesium lithospermate B (MLB) on cAMP/PKA pathway and NMDAR in TBI. METHODS A rodent weight-drop TBI model was used. Administration of MLB was initiated 1 week before (precondition) and 24 hours later (reversal). Cortical homogenates were harvested to measure cAMP (enzyme-linked immunosorbent assay), soluble guanylyl cyclases, PKA and NMDA receptor-2β (Western blot). In addition, cAMP kinase antagonist and H-89 dihydrochloride hydrate were used to test MLB's effect on the cytoplasm cAMP/PKA pathway after TBI. RESULTS Morphologically, vacuolated neuron and activated microglia were observed in the TBI groups but absent in the MLB preconditioning and healthy controls. Induced cAMP, soluble guanylyl cyclase α1, and PKA were observed in the MLB groups, when compared with the TBI group (P < 0.01) Administration of H-89 dihydrochloride hydrate reversed the effect of MLB on cortical PKA and NMDA-2β expression after TBI. CONCLUSIONS This study showed that MLB exerted an antioxidant effect on the enhancement of cytoplasm cAMP and PKA. This compound also decreased NMDA-2β levels, which may correspond to its neuroprotective effects. This finding lends credence to the presumption that MLB modulates the NMDA-2β neurotoxicity through a cAMP-dependent mechanism in the pathogenesis of TBI.
Collapse
Affiliation(s)
- Chih-Zen Chang
- Department of Surgery, Faculty of Medicine, School of Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Department of Surgery, Kaohsiung Municipal Ta Tung Hospital, Kaohsiung, Taiwan.
| | - Shu-Chuan Wu
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Aij-Lie Kwan
- Department of Surgery, Faculty of Medicine, School of Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Chih-Lung Lin
- Department of Surgery, Faculty of Medicine, School of Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| |
Collapse
|
14
|
Barbagallo M, Belvedere M, Sprini D, Dominguez LJ. Magnesium and Alzheimer’s Disease. DIET AND NUTRITION IN DEMENTIA AND COGNITIVE DECLINE 2015:585-592. [DOI: 10.1016/b978-0-12-407824-6.00054-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
15
|
Wilmott LA, Thompson LT. Sex- and dose-dependent effects of post-trial calcium channel blockade by magnesium chloride on memory for inhibitory avoidance conditioning. Behav Brain Res 2013; 257:49-53. [PMID: 24095881 DOI: 10.1016/j.bbr.2013.09.047] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 09/27/2013] [Accepted: 09/29/2013] [Indexed: 11/28/2022]
Abstract
Calcium influx through voltage-dependent Ca(2+) channels is critical for many neuronal processes required for learning and memory. Persistent increases in cytosolic intracellular Ca(2+) concentrations in aging neurons are associated with learning impairments, while small transient subcellular changes in intracellular calcium concentrations play critical roles in neural plasticity in young neurons. In the present study, young male and female Fisher 344 × Brown Norway (FBN) hybrid rats were administered different doses of magnesium chloride (0.0, 100.0, or 200.0mg/kg, i.p.) following a single inhibitory avoidance training trial. Extracellular magnesium ions can non-specifically block voltage-gated calcium channels, and/or reduce the calcium conductance gated via glutamate and serine's activation of neuronal NMDA receptors. In our study, magnesium chloride dose-dependently enhanced memory compared to controls (significantly increased latency to enter a dark compartment previously paired with an aversive stimulus) when tested 48 h later as compared to controls. A leftward shift in the dose response curve for memory enhancement by magnesium chloride was observed for male compared to female rats. These findings provide further insights into calcium-dependent modulation of aversive memory, and should be considered when assessing the design of effective treatment options for both male and female patients with dementia or other memory problems.
Collapse
Affiliation(s)
- Lynda A Wilmott
- The University of Texas at Dallas, Cognition & Neuroscience Program, School of Behavioral & Brain Sciences, 800 West Campbell Road, GR4.1, Richardson, TX 75080, USA
| | | |
Collapse
|
16
|
Tarazi FI, Riva MA. The preclinical profile of lurasidone: clinical relevance for the treatment of schizophrenia. Expert Opin Drug Discov 2013; 8:1297-307. [DOI: 10.1517/17460441.2013.815163] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
17
|
Cerio FGD, Lara-Celador I, Alvarez A, Hilario E. Neuroprotective therapies after perinatal hypoxic-ischemic brain injury. Brain Sci 2013; 3:191-214. [PMID: 24961314 PMCID: PMC4061821 DOI: 10.3390/brainsci3010191] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 02/13/2013] [Accepted: 02/22/2013] [Indexed: 12/29/2022] Open
Abstract
Hypoxic-ischemic (HI) brain injury is one of the main causes of disabilities in term-born infants. It is the result of a deprivation of oxygen and glucose in the neural tissue. As one of the most important causes of brain damage in the newborn period, the neonatal HI event is a devastating condition that can lead to long-term neurological deficits or even death. The pattern of this injury occurs in two phases, the first one is a primary energy failure related to the HI event and the second phase is an energy failure that takes place some hours later. Injuries that occur in response to these events are often manifested as severe cognitive and motor disturbances over time. Due to difficulties regarding the early diagnosis and treatment of HI injury, there is an increasing need to find effective therapies as new opportunities for the reduction of brain damage and its long term effects. Some of these therapies are focused on prevention of the production of reactive oxygen species, anti-inflammatory effects, anti-apoptotic interventions and in a later stage, the stimulation of neurotrophic properties in the neonatal brain which could be targeted to promote neuronal and oligodendrocyte regeneration.
Collapse
Affiliation(s)
- Felipe Goñi de Cerio
- Biotechnology Area, GAIKER Technology Centre, Parque Tecnológico de Zamudio Ed 202, 48170 Zamudio, Vizcaya, Spain.
| | - Idoia Lara-Celador
- Department of Cell Biology and Histology, School of Medicine and Dentistry, University of the Basque Country, 48949 Leioa, Bizkaia, Spain.
| | - Antonia Alvarez
- Department of Cell Biology and Histology, School of Medicine and Dentistry, University of the Basque Country, 48949 Leioa, Bizkaia, Spain.
| | - Enrique Hilario
- Department of Cell Biology and Histology, School of Medicine and Dentistry, University of the Basque Country, 48949 Leioa, Bizkaia, Spain.
| |
Collapse
|
18
|
Siddiq I, Park E, Liu E, Spratt SK, Surosky R, Lee G, Ando D, Giedlin M, Hare GMT, Fehlings MG, Baker AJ. Treatment of traumatic brain injury using zinc-finger protein gene therapy targeting VEGF-A. J Neurotrauma 2012; 29:2647-59. [PMID: 23016562 DOI: 10.1089/neu.2012.2444] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) plays a role in angiogenesis and has been shown to be neuroprotective following central nervous system trauma. In the present study we evaluated the pro-angiogenic and neuroprotective effects of an engineered zinc-finger protein transcription factor transactivator targeting the vascular endothelial growth factor A (VEGF-ZFP). We used two virus delivery systems, adeno-virus and adeno-associated virus, to examine the effects of early and delayed VEGF-A upregulation after brain trauma, respectively. Male Sprague-Dawley rats were subject to a unilateral fluid percussion injury (FPI) of moderate severity (2.2-2.5 atm) followed by intracerebral microinjection of either adenovirus vector (Adv) or an adeno-associated vector (AAV) carrying the VEGF-ZFP construct. Adv-VEGF-ZFP-treated animals had significantly fewer TUNEL positive cells in the injured penumbra of the cortex (p<0.001) and hippocampus (p=0.001) relative to untreated rats at 72 h post-injury. Adv-VEGF-ZFP treatment significantly improved fEPSP values (p=0.007) in the CA1 region relative to injury alone. Treatment with AAV2-VEGF-ZFP resulted in improved post-injury microvascular diameter and improved functional recovery on the balance beam and rotarod task at 30 days post-injury. Collectively, the results provide supportive evidence for the concept of acute and delayed treatment following TBI using VEGF-ZFP to induce angiogenesis, reduce cell death, and enhance functional recovery.
Collapse
Affiliation(s)
- Ishita Siddiq
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Pang TYC, Hannan AJ. Enhancement of cognitive function in models of brain disease through environmental enrichment and physical activity. Neuropharmacology 2012; 64:515-28. [PMID: 22766390 DOI: 10.1016/j.neuropharm.2012.06.029] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Revised: 06/06/2012] [Accepted: 06/15/2012] [Indexed: 12/21/2022]
Abstract
This review will provide an overview of the non-drug based approaches that have been demonstrated to enhance cognitive function of the compromised brain, primarily focussed on the two most widely adopted paradigms of environmental enrichment and enhanced physical exercise. Environmental enrichment involves the generation of novelty and complexity in animal housing conditions which facilitates enhanced sensory and cognitive stimulation as well as physical activity. In a wide variety of animal models of brain disorders, environmental enrichment and exercise have been found to have beneficial effects, including cognitive enhancement, delayed disease onset, enhanced cellular plasticity and associated molecular processes. Potential cellular and molecular mechanisms will also be discussed, which have relevance for the future development of 'enviromimetics', drugs which could mimic or enhance the beneficial effects of environmental stimulation. This article is part of a Special Issue entitled 'Cognitive Enhancers'.
Collapse
Affiliation(s)
- Terence Y C Pang
- Florey Neuroscience Institutes, Melbourne Brain Centre, University of Melbourne, Parkville, VIC 3010, Australia.
| | | |
Collapse
|
20
|
Marklund N, Hillered L. Animal modelling of traumatic brain injury in preclinical drug development: where do we go from here? Br J Pharmacol 2011; 164:1207-29. [PMID: 21175576 PMCID: PMC3229758 DOI: 10.1111/j.1476-5381.2010.01163.x] [Citation(s) in RCA: 178] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Revised: 12/02/2010] [Accepted: 12/06/2010] [Indexed: 11/26/2022] Open
Abstract
Traumatic brain injury (TBI) is the leading cause of death and disability in young adults. Survivors of TBI frequently suffer from long-term personality changes and deficits in cognitive and motor performance, urgently calling for novel pharmacological treatment options. To date, all clinical trials evaluating neuroprotective compounds have failed in demonstrating clinical efficacy in cohorts of severely injured TBI patients. The purpose of the present review is to describe the utility of animal models of TBI for preclinical evaluation of pharmacological compounds. No single animal model can adequately mimic all aspects of human TBI owing to the heterogeneity of clinical TBI. To successfully develop compounds for clinical TBI, a thorough evaluation in several TBI models and injury severities is crucial. Additionally, brain pharmacokinetics and the time window must be carefully evaluated. Although the search for a single-compound, 'silver bullet' therapy is ongoing, a combination of drugs targeting various aspects of neuroprotection, neuroinflammation and regeneration may be needed. In summary, finding drugs and prove clinical efficacy in TBI is a major challenge ahead for the research community and the drug industry. For a successful translation of basic science knowledge to the clinic to occur we believe that a further refinement of animal models and functional outcome methods is important. In the clinical setting, improved patient classification, more homogenous patient cohorts in clinical trials, standardized treatment strategies, improved central nervous system drug delivery systems and monitoring of target drug levels and drug effects is warranted.
Collapse
Affiliation(s)
- Niklas Marklund
- Department of Neuroscience, Neurosurgery, Uppsala University, Uppsala University Hospital, Uppsala, Sweden.
| | | |
Collapse
|
21
|
Park E, Gottlieb JJ, Cheung B, Shek PN, Baker AJ. A Model of Low-Level Primary Blast Brain Trauma Results in Cytoskeletal Proteolysis and Chronic Functional Impairment in the Absence of Lung Barotrauma. J Neurotrauma 2011; 28:343-57. [DOI: 10.1089/neu.2009.1050] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Affiliation(s)
- Eugene Park
- Cara Phelan Centre for Trauma Research, Keenan Research Centre in the Li Ka Shing Knowledge Institute at St. Michael's Hospital, Toronto, Ontario, Canada
| | - James J. Gottlieb
- University of Toronto Institute for Aerospace Studies, Toronto, Ontario, Canada
| | - Bob Cheung
- Defence Research and Development Canada (DRDC) Toronto, Ontario, Canada
| | - Pang N. Shek
- Defence Research and Development Canada (DRDC) Toronto, Ontario, Canada
| | - Andrew J. Baker
- Cara Phelan Centre for Trauma Research, Keenan Research Centre in the Li Ka Shing Knowledge Institute at St. Michael's Hospital, Toronto, Ontario, Canada
- Institute of Medical Science, Departments of Anesthesia and Surgery, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
22
|
Alkam T, Nitta A, Furukawa-Hibi Y, Niwa M, Mizoguchi H, Yamada K, Nabeshima T. Oral supplementation with Leu-Ile, a hydrophobic dipeptide, prevents the impairment of memory induced by amyloid beta in mice via restraining the hyperphosphorylation of extracellular signal-regulated kinase. Behav Brain Res 2010; 210:184-90. [DOI: 10.1016/j.bbr.2010.02.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2009] [Revised: 02/06/2010] [Accepted: 02/12/2010] [Indexed: 11/25/2022]
|
23
|
Nagamoto-Combs K, Morecraft RJ, Darling WG, Combs CK. Long-term gliosis and molecular changes in the cervical spinal cord of the rhesus monkey after traumatic brain injury. J Neurotrauma 2010; 27:565-85. [PMID: 20030560 DOI: 10.1089/neu.2009.0966] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Recovery of fine motor skills after traumatic brain injury (TBI) is variable, with some patients showing progressive improvements over time while others show poor recovery. We therefore studied possible cellular mechanisms accompanying the recovery process in a non-human primate model system, in which the lateral frontal motor cortex areas controlling the preferred upper limb were unilaterally lesioned, and the animals eventually regained fine hand motor function. Immunohistochemical staining of the cervical spinal cord, the site of compensatory sprouting and degeneration of corticospinal axons, showed profound increases in immunoreactivities for major histocompatibility complex class II molecule (MHC-II) and extracellular signal-regulated kinases (ERK1/2) up to 12 months post lesion, particularly within the lateral corticospinal tract (LCST). Double immunostaining demonstrated that phosphorylated ERK1/2 colocalized within the MCH-II + microglia, suggesting a trophic role of long-term microglia activation after TBI at the site of compensatory sprouting. Active sprouting was observed in the LCST as well as in the spinal gray matter of the lesioned animals, as illustrated by increases in growth associated protein 43. Upregulation of Nogo receptor and glutamate transporter expression was also observed in this region after TBI, suggesting possible mechanisms for controlling aberrant sprouting and/or synaptic formation en route and interstitial glutamate concentration changes at the site of axon degeneration, respectively. Taken together, these changes in the non-human primate spinal cord support a long-term trophic/tropic role for reactive microglia, in particular, during functional and structural recovery after TBI.
Collapse
Affiliation(s)
- Kumi Nagamoto-Combs
- Department of Anatomy and Cell Biology, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58202, USA
| | | | | | | |
Collapse
|
24
|
Magnesium sulfate reduces inflammation-associated brain injury in fetal mice. Am J Obstet Gynecol 2010; 202:292.e1-9. [PMID: 20207246 DOI: 10.1016/j.ajog.2010.01.022] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2009] [Revised: 12/24/2009] [Accepted: 01/14/2010] [Indexed: 01/28/2023]
Abstract
OBJECTIVE The purpose of this study was to investigate whether magnesium sulfate (MgSO(4)) prevents fetal brain injury in inflammation-associated preterm birth (PTB). STUDY DESIGN In a mouse model of PTB, mice exposed to lipopolysaccharide (LPS) or normal saline (NS) by intrauterine injection were randomized to intraperitoneal treatment with MgSO(4) or NS [corrected]. From the 4 treatment groups (NS + NS; LPS + NS; LPS + MgSO(4); and NS + MgSO(4)), fetal brains were collected for quantitative polymerase chain reaction studies and primary neuronal cultures. Messenger RNA expression of cytokines, cell death, and markers of neuronal and glial differentiation were assessed. Immunocytochemistry and confocal microscopy were performed. RESULTS There was no difference between the LPS + NS and LPS + MgSO(4) groups in the expression of proinflammatory cytokines, cell death markers, and markers of prooligodendrocyte and astrocyte development (P > .05 for all). Neuronal cultures from the LPS + NS group demonstrated morphologic changes; this neuronal injury was prevented by MgSO(4) (P < .001). CONCLUSION Amelioration of neuronal injury in inflammation-associated PTB may be a key mechanism by which MgSO(4) prevents cerebral palsy.
Collapse
|
25
|
Bales JW, Wagner AK, Kline AE, Dixon CE. Persistent cognitive dysfunction after traumatic brain injury: A dopamine hypothesis. Neurosci Biobehav Rev 2009; 33:981-1003. [PMID: 19580914 DOI: 10.1016/j.neubiorev.2009.03.011] [Citation(s) in RCA: 187] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2009] [Revised: 03/10/2009] [Accepted: 03/23/2009] [Indexed: 12/12/2022]
Abstract
Traumatic brain injury (TBI) represents a significant cause of death and disability in industrialized countries. Of particular importance to patients the chronic effect that TBI has on cognitive function. Therapeutic strategies have been difficult to evaluate because of the complexity of injuries and variety of patient presentations within a TBI population. However, pharmacotherapies targeting dopamine (DA) have consistently shown benefits in attention, behavioral outcome, executive function, and memory. Still it remains unclear what aspect of TBI pathology is targeted by DA therapies and what time-course of treatment is most beneficial for patient outcomes. Fortunately, ongoing research in animal models has begun to elucidate the pathophysiology of DA alterations after TBI. The purpose of this review is to discuss clinical and experimental research examining DAergic therapies after TBI, which will in turn elucidate the importance of DA for cognitive function/dysfunction after TBI as well as highlight the areas that require further study.
Collapse
Affiliation(s)
- James W Bales
- Brain Trauma Research Center, University of Pittsburgh, PA 15260, USA
| | | | | | | |
Collapse
|
26
|
Hoskison MM, Moore AN, Hu B, Orsi S, Kobori N, Dash PK. Persistent working memory dysfunction following traumatic brain injury: evidence for a time-dependent mechanism. Neuroscience 2009; 159:483-91. [PMID: 19167462 DOI: 10.1016/j.neuroscience.2008.12.050] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2008] [Accepted: 12/28/2008] [Indexed: 11/16/2022]
Abstract
The prefrontal cortex is highly vulnerable to traumatic brain injury (TBI) resulting in the dysfunction of many high-level cognitive and executive functions such as planning, information processing speed, language, memory, attention, and perception. All of these processes require some degree of working memory. Interestingly, in many cases, post-injury working memory deficits can arise in the absence of overt damage to the prefrontal cortex. Recently, excess GABA-mediated inhibition of prefrontal neuronal activity has been identified as a contributor to working memory dysfunction within the first month following cortical impact injury of rats. However, it has not been examined if these working memory deficits persist, and if so, whether they remain amenable to treatment by GABA antagonism. Our findings show that working memory dysfunction, assessed using both the delay match-to-place and delayed alternation T-maze tasks, following lateral cortical impact injury persists for at least 16 weeks post-injury. These deficits were found to be no longer the direct result of excess GABA-mediated inhibition of medial prefrontal cortex neuronal activity. Golgi staining of prelimbic pyramidal neurons revealed that TBI causes a significant shortening of layers V/VI basal dendrite arbors by 4 months post-injury, as well as an increase in the density of both basal and apical spines in these neurons. These changes were not observed in animals 14 days post-injury, a time point at which administration of GABA receptor antagonists improves working memory function. Taken together, the present findings, along with previously published reports, suggest that temporal considerations must be taken into account when designing mechanism-based therapies to improve working memory function in TBI patients.
Collapse
Affiliation(s)
- M M Hoskison
- Department of Physical Medicine and Rehabilitation, Baylor College of Medicine, The University of Texas Medical School, Houston, TX 77225, USA
| | | | | | | | | | | |
Collapse
|
27
|
Shi J, Miles DK, Orr BA, Massa SM, Kernie SG. Injury-induced neurogenesis in Bax-deficient mice: evidence for regulation by voltage-gated potassium channels. Eur J Neurosci 2007; 25:3499-512. [PMID: 17610570 DOI: 10.1111/j.1460-9568.2007.05624.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Adult neural stem and progenitor cells may help remodel the brain in response to injury. The pro-apoptotic molecule Bax has recently been identified as a key player in adult neural stem cell survival. In Bax-deficient mice that have undergone traumatic brain injury, we find increased numbers of neural progenitor cells in the dentate gyrus and improved remodeling of the hippocampus. Exogenous potassium chloride mimics spreading depression (SD)-like events in vitro, and Bax-deficient neural stem cells proliferate in response to these events more robustly than wild-type neural stem cells. Selective potassium channel blockers interrupt SD-mediated stimulation of stem cells. In addition, the potassium channel Kv4.1 is expressed within neural stem and progenitor cells in the dentate gyrus and is increased in Bax-deficiency. These data suggest that the neuroprotection observed after injury in Bax-deficiency may be due to increased neurogenesis via activation of the Kv4 family of potassium channels.
Collapse
Affiliation(s)
- Jian Shi
- Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, TX 75390-9133, USA
| | | | | | | | | |
Collapse
|
28
|
|