1
|
Ilyin NP, Shevlyakov AD, Boyko GA, Moskalenko AM, Ikrin AN, Galstyan DS, Kolesnikova TO, Katolikova NV, Chekrygin SA, Lim LW, Yang L, De Abreu MS, Yenkoyan KB, Kalueff AV, Demin KA. Neurotranscriptomic and behavioral effects of ISRIB, and its therapeutic effects in the traumatic brain injury model in zebrafish. Brain Res 2025; 1848:149329. [PMID: 39537125 DOI: 10.1016/j.brainres.2024.149329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 10/05/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
Traumatic brain injury (TBI) is a global medical concern and has a lasting impact on brain activity with high risks of mortality. Current treatments are inadequate for repairing damaged brain cells or correcting cognitive and behavioral disabilities in TBI patients. Mounting evidence links TBI to the activation of the Integrated Stress Response (ISR) signaling in the brain. A novel small molecule, ISRIB, is an effective inhibitor of the ISR pathway, offering potential advantages for brain health. Here, we investigated how ISRIB affects brain transcriptome and behavior in zebrafish TBI model evoked by telencephalic brain injury. Overall, while TBI diminished memory and social behavior in zebrafish, administering ISRIB post-injury markedly reduced these behavioral deficits, and modulated brain gene expression, rescuing TBI-activated pathways related to inflammation and brain cell development. Collectively, this supports the role of brain ISR in TBI, and suggests potential utility of ISRIB for the treatment of TBI-related states.
Collapse
Affiliation(s)
- Nikita P Ilyin
- Almazov National Medical Research Centre, St. Petersburg, Russia; Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Anton D Shevlyakov
- Neurobiology Program, Sirius University of Science and Technology, Sochi, Russia
| | - Galina A Boyko
- Almazov National Medical Research Centre, St. Petersburg, Russia; Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | | | - Aleksey N Ikrin
- Neurobiology Program, Sirius University of Science and Technology, Sochi, Russia
| | - David S Galstyan
- Almazov National Medical Research Centre, St. Petersburg, Russia; Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | | | - Nataliia V Katolikova
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Sergei A Chekrygin
- Core Facility Center "Center Bio-Bank", Saint Petersburg University, St. Petersburg, Russia
| | - Lee Wei Lim
- Department of Biociences and Bioinformatics, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China; Suzhou Key Municipal Laboratory of Neurobiology and Cell Signaling, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - LongEn Yang
- Department of Biociences and Bioinformatics, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China; Suzhou Key Municipal Laboratory of Neurobiology and Cell Signaling, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Murilo S De Abreu
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil; Western Caspian University, Baku, Azerbaijan
| | - Konstantin B Yenkoyan
- Neuroscience Laboratory, Cobrain Center, M. Heratsi Yerevan State Medical University, Yerevan, Armenia; Biochemistry Department, M. Heratsi Yerevan State Medical University, Yerevan, Armenia
| | - Allan V Kalueff
- Almazov National Medical Research Centre, St. Petersburg, Russia; Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Department of Biociences and Bioinformatics, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China; Suzhou Key Municipal Laboratory of Neurobiology and Cell Signaling, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China.
| | - Konstantin A Demin
- Almazov National Medical Research Centre, St. Petersburg, Russia; Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia.
| |
Collapse
|
2
|
Yoshikawa MH, Brasil S, Solla DJF, Amorim RL, Godoy DA, Kolias A, Paiva WS. Long-Term Functional Recovery after Severe Traumatic Brain Injury with Type II Diffuse Injury. Neurotrauma Rep 2025; 6:13-19. [PMID: 40008052 PMCID: PMC11848058 DOI: 10.1089/neur.2024.0052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025] Open
Abstract
This study aims to describe the late clinical outcomes of patients with severe traumatic brain injury (sTBI) and the risk factors associated with it. Patients were enrolled between April 2012 and January 2015 and followed until January 2022. The inclusion criteria were age 16-65 years, Glasgow Coma Scale ≤8 on admission, diagnosis of blunt TBI with Marshall diffuse injury type II on initial computerized tomography (CT), and alive at discharge. Clinical, laboratory, and radiological data from admission were collected. Glasgow Outcome Scale Extended (GOSE), Functional Independence Measure, and Zarit Burden Interview (ZBI) were assessed in the follow-up. Sixty-five patients were included, with a median follow-up time of 8 years. Nineteen (29.2%) patients had good recovery (GOSE 7-8), and 10 (15.3%) had moderate-to-severe sequelae (GOSE 4-6). Thirty-six (55.4%) patients died after discharge, and most of them in the first 3 months after discharge (n = 26; 72.2%). Despite the early mortality rate being the highest, the 6-month score is explained in the text (CRASH-CT) score on admission was not associated with death in the follow-up (p = 0.25). In the multivariate statistical analysis, only prothrombin time was associated with GOSE (p = 0.01). Twelve (41.3%) patients were independent for basic activities of daily living, and the most common cause of dependence was memory impairment (n = 12; 41.3%). The median ZBI score reported by caregivers was 23.5 (range 5-48), indicating mild overload. In this study, patients with sTBI sustaining Marshall II lesions had a significant mortality rate after discharge, and we found coagulation impairment as a potential predictor of poor outcomes. Around 30% experienced functional dependence and inability to return to social and work activities. Current instruments used to predict outcomes of TBI patients had poor predictive performance in this low- and middle-income country population, suggesting the need for new models to properly guide clinical decision-making and counseling family members.
Collapse
Affiliation(s)
| | - Sérgio Brasil
- Division of Neurosurgery, Department of Neurology, University of Sao Paulo, Brazil
| | | | - Robson Luís Amorim
- Division of Neurosurgery, Department of Neurology, University of Sao Paulo, Brazil
| | | | - Angelos Kolias
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge and Addenbrooke’s Hospital, UK
| | | |
Collapse
|
3
|
Svirsky SE, Henchir J, Parry M, Holets E, Zhang T, Gittes GK, Carlson SW, Dixon CE. Viral-mediated increased hippocampal neurogranin modulate synapses at one month in a rat model of controlled cortical impact. Sci Rep 2024; 14:28998. [PMID: 39578516 PMCID: PMC11584851 DOI: 10.1038/s41598-024-77682-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 10/24/2024] [Indexed: 11/24/2024] Open
Abstract
Reductions of neurogranin (Ng), a calcium-sensitive calmodulin-binding protein, result in significant impairment across various hippocampal-dependent learning and memory tasks. Conversely, increasing levels of Ng facilitates synaptic plasticity, increases synaptogenesis and boosts cognitive abilities. Controlled cortical impact (CCI), an experimental traumatic brain injury (TBI) model, results in significantly reduced hippocampal Ng protein expression up to 4 weeks post-injury, supporting a strategy to increase Ng to improve function. In this study, hippocampal Ng expression was increased in adult, male Sham and CCI injured animals using intraparenchymal injection of adeno-associated virus (AAV) 30 min post-injury, thereby also affording the ability to differentiate endogenous and exogenous Ng. At 4 weeks, molecular, anatomical, and behavioral measures of synaptic plasticity were evaluated to determine the therapeutic potential of Ng modulation post-TBI. Increasing Ng had a TBI-dependent effect on hippocampal expression of synaptic proteins and dendritic spine morphology. Increasing Ng did not improve behavior across all outcomes in both Sham and CCI groups at the 4 week time-point. Overall, increasing Ng expression modulated protein expression and dendritic spine morphology, but exerted limited functional benefit after CCI. This study furthers our understanding of Ng, and mechanisms of cognitive dysfunction within the synapse sub-acutely after TBI.
Collapse
Affiliation(s)
- Sarah E Svirsky
- Center for Neuroscience, University of Pittsburgh, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
- Department of Neurological Surgery, University of Pittsburgh Medical Center, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - Jeremy Henchir
- Department of Neurological Surgery, University of Pittsburgh Medical Center, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - Madison Parry
- Department of Neurological Surgery, University of Pittsburgh Medical Center, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - Erik Holets
- Department of Neurological Surgery, University of Pittsburgh Medical Center, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - Ting Zhang
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - George K Gittes
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - Shaun W Carlson
- Department of Neurological Surgery, University of Pittsburgh Medical Center, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - C Edward Dixon
- Center for Neuroscience, University of Pittsburgh, 4401 Penn Ave, Pittsburgh, PA, 15224, USA.
- Department of Neurological Surgery, University of Pittsburgh Medical Center, 4401 Penn Ave, Pittsburgh, PA, 15224, USA.
- V.A. Pittsburgh Healthcare System, 4401 Penn Ave, Pittsburgh, PA, 15224, USA.
| |
Collapse
|
4
|
Franczak Ł, Podwalski P, Wysocki P, Dawidowski B, Jędrzejewski A, Jabłoński M, Samochowiec J. Impulsivity in ADHD and Borderline Personality Disorder: A Systematic Review of Gray and White Matter Variations. J Clin Med 2024; 13:6906. [PMID: 39598050 PMCID: PMC11594719 DOI: 10.3390/jcm13226906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 11/10/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
Introduction: Impulsivity is one of the overlapping symptoms common to borderline personality disorder (BPD) and attention deficit hyperactivity disorder (ADHD), but the neurobiological basis of these disorders remains uncertain. This systematic review aims to identify abnormalities in the gray and white matter associated with impulsivity in BPD and ADHD. Methods: We conducted a systematic search of the PubMed, Embase, and SCOPUS databases, adhering to PRISMA guidelines. Studies that investigated gray and white matter alterations in BPD or ADHD populations and their relationship with impulsivity were included. We reviewed information from 23 studies involving 992 participants, which included findings from structural MRI and DTI. Results: The review identified various nonhomogeneous changes associated with impulsivity in BPD and ADHD. BPD was mainly associated with abnormalities in the prefrontal cortex (PFC) and limbic areas, which correlated negatively with impulsivity. In contrast, impulsivity associated with ADHD was associated with structural changes in the caudate nucleus and frontal-striatal pathways. Despite the overlapping symptoms of impulsivity, the neurobiological mechanisms appeared to differ between the two disorders. Conclusions: These findings emphasize the distinct neurostructural correlates of impulsivity in BPD and ADHD. While both disorders show impulsivity as one of their main symptoms, the fundamental brain structures associated with this trait are different. BPD is primarily associated with abnormalities in the prefrontal cortex and limbic system, whereas the alterations seen in ADHD tend to focus on the caudate nucleus and frontostriatal pathways. Further research is needed to clarify these differences and their implications for treatment.
Collapse
Affiliation(s)
- Łukasz Franczak
- Department of Psychiatry, Pomeranian Medical University, Broniewskiego 26 Street, 71-460 Szczecin, Poland; (Ł.F.); (P.W.); (B.D.); (M.J.); (J.S.)
| | - Piotr Podwalski
- Department of Psychiatry, Pomeranian Medical University, Broniewskiego 26 Street, 71-460 Szczecin, Poland; (Ł.F.); (P.W.); (B.D.); (M.J.); (J.S.)
| | - Patryk Wysocki
- Department of Psychiatry, Pomeranian Medical University, Broniewskiego 26 Street, 71-460 Szczecin, Poland; (Ł.F.); (P.W.); (B.D.); (M.J.); (J.S.)
| | - Bartosz Dawidowski
- Department of Psychiatry, Pomeranian Medical University, Broniewskiego 26 Street, 71-460 Szczecin, Poland; (Ł.F.); (P.W.); (B.D.); (M.J.); (J.S.)
| | - Adam Jędrzejewski
- Independent Clinical Psychology Unit, Pomeranian Medical University, Broniewskiego 26 Street, 71-460 Szczecin, Poland;
| | - Marcin Jabłoński
- Department of Psychiatry, Pomeranian Medical University, Broniewskiego 26 Street, 71-460 Szczecin, Poland; (Ł.F.); (P.W.); (B.D.); (M.J.); (J.S.)
| | - Jerzy Samochowiec
- Department of Psychiatry, Pomeranian Medical University, Broniewskiego 26 Street, 71-460 Szczecin, Poland; (Ł.F.); (P.W.); (B.D.); (M.J.); (J.S.)
| |
Collapse
|
5
|
Zima L, Moore AN, Smolen P, Kobori N, Noble B, Robinson D, Hood KN, Homma R, Al Mamun A, Redell JB, Dash PK. The evolving pathophysiology of TBI and the advantages of temporally-guided combination therapies. Neurochem Int 2024; 180:105874. [PMID: 39366429 PMCID: PMC12011104 DOI: 10.1016/j.neuint.2024.105874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/26/2024] [Accepted: 10/01/2024] [Indexed: 10/06/2024]
Abstract
Several clinical and experimental studies have demonstrated that traumatic brain injury (TBI) activates cascades of biochemical, molecular, structural, and pathological changes in the brain. These changes combine to contribute to the various outcomes observed after TBI. Given the breadth and complexity of changes, combination treatments may be an effective approach for targeting multiple detrimental pathways to yield meaningful improvements. In order to identify targets for therapy development, the temporally evolving pathophysiology of TBI needs to be elucidated in detail at both the cellular and molecular levels, as it has been shown that the mechanisms contributing to cognitive dysfunction change over time. Thus, a combination of individual mechanism-based therapies is likely to be effective when maintained based on the time courses of the cellular and molecular changes being targeted. In this review, we will discuss the temporal changes of some of the key clinical pathologies of human TBI, the underlying cellular and molecular mechanisms, and the results from preclinical and clinical studies aimed at mitigating their consequences. As most of the pathological events that occur after TBI are likely to have subsided in the chronic stage of the disease, combination treatments aimed at attenuating chronic conditions such as cognitive dysfunction may not require the initiation of individual treatments at a specific time. We propose that a combination of acute, subacute, and chronic interventions may be necessary to maximally improve health-related quality of life (HRQoL) for persons who have sustained a TBI.
Collapse
Affiliation(s)
- Laura Zima
- Departments of Neurosurgery, The University of Texas McGovern Medical School, Houston, TX, USA
| | - Anthony N Moore
- Departments of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, USA
| | - Paul Smolen
- Departments of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, USA
| | - Nobuhide Kobori
- Departments of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, USA
| | - Brian Noble
- Departments of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, USA
| | - Dustin Robinson
- Departments of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, USA
| | - Kimberly N Hood
- Departments of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, USA
| | - Ryota Homma
- Departments of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, USA
| | - Amar Al Mamun
- Departments of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, USA
| | - John B Redell
- Departments of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, USA
| | - Pramod K Dash
- Departments of Neurosurgery, The University of Texas McGovern Medical School, Houston, TX, USA; Departments of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, USA.
| |
Collapse
|
6
|
Smith AM, Grayson BE. A strike to the head: Parallels between the pediatric and adult human and the rodent in traumatic brain injury. J Neurosci Res 2024; 102:e25364. [PMID: 38953607 DOI: 10.1002/jnr.25364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 06/05/2024] [Accepted: 06/16/2024] [Indexed: 07/04/2024]
Abstract
Traumatic brain injury (TBI) is a condition that occurs commonly in children from infancy through adolescence and is a global health concern. Pediatric TBI presents with a bimodal age distribution, with very young children (0-4 years) and adolescents (15-19 years) more commonly injured. Because children's brains are still developing, there is increased vulnerability to the effects of head trauma, which results in entirely different patterns of injury than in adults. Pediatric TBI has a profound and lasting impact on a child's development and quality of life, resulting in long-lasting consequences to physical, cognitive, and emotional development. Chronic issues like learning disabilities, behavioral problems, and emotional disturbances can develop. Early intervention and ongoing support are critical for minimizing these long-term deficits. Many animal models of TBI exist, and each varies significantly, displaying different characteristics of clinical TBI. The neurodevelopment differs in the rodent from the human in timing and effect, so TBI outcomes in the juvenile rodent can thus vary from the human child. The current review compares findings from preclinical TBI work in juvenile and adult rodents to clinical TBI research in pediatric and adult humans. We focus on the four brain regions most affected by TBI: the prefrontal cortex, corpus callosum, hippocampus, and hypothalamus. Each has its unique developmental projections and thus is impacted by TBI differently. This review aims to compare the healthy neurodevelopment of these four brain regions in humans to the developmental processes in rodents.
Collapse
Affiliation(s)
- Allie M Smith
- Department of Neurology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Bernadette E Grayson
- Department of Neurology, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Department of Population Health Science, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Department of Anesthesiology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
7
|
Regniez M, Dufort-Gervais J, Provost C, Mongrain V, Martinez M. Characterization of Sleep, Emotional, and Cognitive Functions in a New Rat Model of Concomitant Spinal Cord and Traumatic Brain Injuries. J Neurotrauma 2024; 41:1044-1059. [PMID: 37885242 DOI: 10.1089/neu.2023.0387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2023] Open
Abstract
Traumatic injuries to the spinal cord or the brain have serious medical consequences and lead to long-term disability. The epidemiology, medical complications, and prognosis of isolated spinal cord injury (SCI) and traumatic brain injury (TBI) have been well described. However, there are limited data on patients suffering from concurrent SCI and TBI, even if a large proportion of SCI patients have concomitant TBI. The complications associated with this "dual-diagnosis" such as cognitive or behavioral dysfunction are well known in the rehabilitation setting, but evidence-based and standardized approaches for diagnosis and treatment are lacking. Our goal was to develop and characterize a pre-clinical animal model of concurrent SCI and TBI to help identifying "dual-diagnosis" tools. Female rats received a unilateral contusive SCI at the thoracic level alone (SCI group) or combined with a TBI centered on the contralateral sensorimotor cortex (SCI-TBI group). We first validated that the SCI extent was comparable between SCI-TBI and SCI groups, and that hindlimb function was impaired. We characterized various neurological outcomes, including locomotion, sleep architecture, brain activity during sleep, depressive- and anxiety-like behaviors, and working memory. We report that SCI-TBI and SCI groups show similar impairments in global locomotor function. While wake/sleep amount and distribution and anxiety- and depression-like symptoms were not affected in SCI-TBI and SCI groups in comparison to the control group (laminectomy and craniotomy only), working memory was impaired only in SCI-TBI rats. This pre-clinical model of concomitant SCI and TBI, including more severe variations of it, shows a translational value for the identification of biomarkers to refine the "dual-diagnosis" of neurotrauma in humans.
Collapse
Affiliation(s)
- Morgane Regniez
- Department of Neuroscience, Université de Montreal, Montréal, Québec, Canada
- Recherche CIUSSS-NIM, Montréal, Québec, Canada
| | | | | | - Valérie Mongrain
- Department of Neuroscience, Université de Montreal, Montréal, Québec, Canada
- Recherche CIUSSS-NIM, Montréal, Québec, Canada
- Research Center of the CHUM, Montréal, Québec, Canada
| | - Marina Martinez
- Department of Neuroscience, Université de Montreal, Montréal, Québec, Canada
- Recherche CIUSSS-NIM, Montréal, Québec, Canada
- Groupe de recherche sur la Signalisation Neurale et la Circuiterie, Université de Montreal, Montréal, Québec, Canada
- Centre interdisciplinaire de recherche sur le cerveau et l'apprentissage, Université de Montreal, Montréal, Québec, Canada
| |
Collapse
|
8
|
Witkin JM, Shafique H, Cerne R, Smith JL, Marini AM, Lipsky RH, Delery E. Mechanistic and therapeutic relationships of traumatic brain injury and γ-amino-butyric acid (GABA). Pharmacol Ther 2024; 256:108609. [PMID: 38369062 DOI: 10.1016/j.pharmthera.2024.108609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 01/18/2024] [Accepted: 02/01/2024] [Indexed: 02/20/2024]
Abstract
Traumatic brain injury (TBI) is a highly prevalent medical condition for which no medications specific for the prophylaxis or treatment of the condition as a whole exist. The spectrum of symptoms includes coma, headache, seizures, cognitive impairment, depression, and anxiety. Although it has been known for years that the inhibitory neurotransmitter γ-amino-butyric acid (GABA) is involved in TBI, no novel therapeutics based upon this mechanism have been introduced into clinical practice. We review the neuroanatomical, neurophysiological, neurochemical, and neuropharmacological relationships of GABA neurotransmission to TBI with a view toward new potential GABA-based medicines. The long-standing idea that excitatory and inhibitory (GABA and others) balances are disrupted by TBI is supported by the experimental data but has failed to invent novel methods of restoring this balance. The slow progress in advancing new treatments is due to the complexity of the disorder that encompasses multiple dynamically interacting biological processes including hemodynamic and metabolic systems, neurodegeneration and neurogenesis, major disruptions in neural networks and axons, frank brain lesions, and a multitude of symptoms that have differential neuronal and neurohormonal regulatory mechanisms. Although the current and ongoing clinical studies include GABAergic drugs, no novel GABA compounds are being explored. It is suggested that filling the gap in understanding the roles played by specific GABAA receptor configurations within specific neuronal circuits could help define new therapeutic approaches. Further research into the temporal and spatial delivery of GABA modulators should also be useful. Along with GABA modulation, research into the sequencing of GABA and non-GABA treatments will be needed.
Collapse
Affiliation(s)
- Jeffrey M Witkin
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent Hospital, Indianapolis, IN, USA; Departments of Neuroscience and Trauma Research, Ascension St. Vincent Hospital, Indianapolis, IN, USA; RespireRx Pharmaceuticals Inc, Glen Rock, NJ, USA.
| | | | - Rok Cerne
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent Hospital, Indianapolis, IN, USA; RespireRx Pharmaceuticals Inc, Glen Rock, NJ, USA; Department of Anatomy and Cell Biology, Indiana University/Purdue University, Indianapolis, IN, USA
| | - Jodi L Smith
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent Hospital, Indianapolis, IN, USA
| | - Ann M Marini
- Department of Neurology, Program in Neuroscience, and Molecular and Cellular Biology Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Robert H Lipsky
- Department of Neurology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Elizabeth Delery
- College of Osteopathic Medicine, Marian University, Indianapolis, IN, USA.
| |
Collapse
|
9
|
Jia M, Guo X, Liu R, Sun L, Wang Q, Wu J. Overexpress miR-132 in the Brain Parenchyma by a Non-invasive Way Improves Tissue Repairment and Releases Memory Impairment After Traumatic Brain Injury. Cell Mol Neurobiol 2023; 44:5. [PMID: 38104297 PMCID: PMC11397820 DOI: 10.1007/s10571-023-01435-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 11/08/2023] [Indexed: 12/19/2023]
Abstract
Traumatic brain injury (TBI) is a serious public health problem worldwide, which could lead to an extremely high percentage of mortality and disability. Current treatment strategies mainly concentrate on neuronal protection and reconstruction, among them, exogenous neural stem cell (NSC) transplantation has long been regarded as the most effective curative treatment. However, due to secondary trauma, transplant rejection, and increased incidence of brain malignant tumor, a non-invasive therapy that enhanced endogenous neurogenesis was more suitable for TBI treatment. Our previous work has shown that miR-132 overexpression could improve neuronal differentiation of NSCs in vitro and in vivo. So, we engineered a new kind of AAV vector named AAV-PHP.eB which can transfect brain parenchyma through intravenous injection to overexpress miR-132 in brain after TBI. We found that miR-132 overexpression could reduce impact volume, promote neurogenesis in the dentate gyrus (DG), accelerate neuroblast migrating into the impact cortex, ameliorate microglia-mediated inflammatory reaction, and ultimately restore learning memory function. Our results revealed that AAV-PHP.eB-based miR-132 overexpression could improve endogenous tissue repairment and release clinical symptoms after traumatic brain injury. This work would provide a new therapeutic strategy for TBI treatment and other neurological disorders characterized by markable neuronal loss and memory impairment. miR-132 overexpression accelerates endogenous neurogenesis and releases TBI-induced tissue repairment and memory impairment. Controlled cortical impact onto the cortex would induce serious cortical injury and microglia accumulation in both cortex and hippocampus. Moreover, endogenous neuroblast could migrate around the injury core. miR-132 overexpression could accelerate neuroblast migration toward the injury core and decreased microglia accumulation in the ipsilateral cortex and hippocampus. miR-132 could be a suitable target on neuroprotective therapy after TBI.
Collapse
Affiliation(s)
- Meng Jia
- Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100070, China
| | - Xi Guo
- Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100070, China
| | - Ru Liu
- Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100070, China
| | - Lei Sun
- Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100070, China
| | - Qun Wang
- Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
| | - Jianping Wu
- Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China.
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100070, China.
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China.
| |
Collapse
|
10
|
Arnsten AFT, Ishizawa Y, Xie Z. Scientific rationale for the use of α2A-adrenoceptor agonists in treating neuroinflammatory cognitive disorders. Mol Psychiatry 2023; 28:4540-4552. [PMID: 37029295 PMCID: PMC10080530 DOI: 10.1038/s41380-023-02057-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 03/22/2023] [Accepted: 03/24/2023] [Indexed: 04/09/2023]
Abstract
Neuroinflammatory disorders preferentially impair the higher cognitive and executive functions of the prefrontal cortex (PFC). This includes such challenging disorders as delirium, perioperative neurocognitive disorder, and the sustained cognitive deficits from "long-COVID" or traumatic brain injury. There are no FDA-approved treatments for these symptoms; thus, understanding their etiology is important for generating therapeutic strategies. The current review describes the molecular rationale for why PFC circuits are especially vulnerable to inflammation, and how α2A-adrenoceptor (α2A-AR) actions throughout the nervous and immune systems can benefit the circuits in PFC needed for higher cognition. The layer III circuits in the dorsolateral PFC (dlPFC) that generate and sustain the mental representations needed for higher cognition have unusual neurotransmission and neuromodulation. They are wholly dependent on NMDAR neurotransmission, with little AMPAR contribution, and thus are especially vulnerable to kynurenic acid inflammatory signaling which blocks NMDAR. Layer III dlPFC spines also have unusual neuromodulation, with cAMP magnification of calcium signaling in spines, which opens nearby potassium channels to rapidly weaken connectivity and reduce neuronal firing. This process must be tightly regulated, e.g. by mGluR3 or α2A-AR on spines, to prevent loss of firing. However, the production of GCPII inflammatory signaling reduces mGluR3 actions and markedly diminishes dlPFC network firing. Both basic and clinical studies show that α2A-AR agonists such as guanfacine can restore dlPFC network firing and cognitive function, through direct actions in the dlPFC, but also by reducing the activity of stress-related circuits, e.g. in the locus coeruleus and amygdala, and by having anti-inflammatory actions in the immune system. This information is particularly timely, as guanfacine is currently the focus of large clinical trials for the treatment of delirium, and in open label studies for the treatment of cognitive deficits from long-COVID.
Collapse
Affiliation(s)
- Amy F T Arnsten
- Department Neuroscience, Yale University School of Medicine, New Haven, CT, 056510, USA.
| | - Yumiko Ishizawa
- Department Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Zhongcong Xie
- Department Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| |
Collapse
|
11
|
Fesharaki Zadeh A, Arnsten AFT, Wang M. Scientific Rationale for the Treatment of Cognitive Deficits from Long COVID. Neurol Int 2023; 15:725-742. [PMID: 37368329 PMCID: PMC10303664 DOI: 10.3390/neurolint15020045] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/26/2023] [Accepted: 05/11/2023] [Indexed: 06/28/2023] Open
Abstract
Sustained cognitive deficits are a common and debilitating feature of "long COVID", but currently there are no FDA-approved treatments. The cognitive functions of the dorsolateral prefrontal cortex (dlPFC) are the most consistently afflicted by long COVID, including deficits in working memory, motivation, and executive functioning. COVID-19 infection greatly increases kynurenic acid (KYNA) and glutamate carboxypeptidase II (GCPII) in brain, both of which can be particularly deleterious to PFC function. KYNA blocks both NMDA and nicotinic-alpha-7 receptors, the two receptors required for dlPFC neurotransmission, and GCPII reduces mGluR3 regulation of cAMP-calcium-potassium channel signaling, which weakens dlPFC network connectivity and reduces dlPFC neuronal firing. Two agents approved for other indications may be helpful in restoring dlPFC physiology: the antioxidant N-acetyl cysteine inhibits the production of KYNA, and the α2A-adrenoceptor agonist guanfacine regulates cAMP-calcium-potassium channel signaling in dlPFC and is also anti-inflammatory. Thus, these agents may be helpful in treating the cognitive symptoms of long COVID.
Collapse
Affiliation(s)
- Arman Fesharaki Zadeh
- Departments of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Departments of Psychiatry, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Amy F. T. Arnsten
- Departments of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA;
| | - Min Wang
- Departments of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA;
| |
Collapse
|
12
|
He RH, Fan JZ, Qian FF, He YH, Du XH, Lu HX. Repetitive transcranial magnetic stimulation promotes neurological functional recovery in rats with traumatic brain injury by upregulating synaptic plasticity-related proteins. Neural Regen Res 2023; 18:368-374. [PMID: 35900432 PMCID: PMC9396518 DOI: 10.4103/1673-5374.346548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Studies have shown that repetitive transcranial magnetic stimulation (rTMS) can enhance synaptic plasticity and improve neurological dysfunction. However, the mechanism through which rTMS can improve moderate traumatic brain injury remains poorly understood. In this study, we established rat models of moderate traumatic brain injury using Feeney’s weight-dropping method and treated them using rTMS. To help determine the mechanism of action, we measured levels of several important brain activity-related proteins and their mRNA. On the injured side of the brain, we found that rTMS increased the protein levels and mRNA expression of brain-derived neurotrophic factor, tropomyosin receptor kinase B, N-methyl-D-aspartic acid receptor 1, and phosphorylated cAMP response element binding protein, which are closely associated with the occurrence of long-term potentiation. rTMS also partially reversed the loss of synaptophysin after injury and promoted the remodeling of synaptic ultrastructure. These findings suggest that upregulation of synaptic plasticity-related protein expression is the mechanism through which rTMS promotes neurological function recovery after moderate traumatic brain injury.
Collapse
|
13
|
Adegoke MA, Teter O, Meaney DF. Flexibility of in vitro cortical circuits influences resilience from microtrauma. Front Cell Neurosci 2022; 16:991740. [PMID: 36589287 PMCID: PMC9803265 DOI: 10.3389/fncel.2022.991740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
Background Small clusters comprising hundreds to thousands of neurons are an important level of brain architecture that correlates single neuronal properties to fulfill brain function, but the specific mechanisms through which this scaling occurs are not well understood. In this study, we developed an in vitro experimental platform of small neuronal circuits (islands) to probe the importance of structural properties for their development, physiology, and response to microtrauma. Methods Primary cortical neurons were plated on a substrate patterned to promote attachment in clusters of hundreds of cells (islands), transduced with GCaMP6f, allowed to mature until 10-13 days in vitro (DIV), and monitored with Ca2+ as a non-invasive proxy for electrical activity. We adjusted two structural factors-island size and cellular density-to evaluate their role in guiding spontaneous activity and network formation in neuronal islands. Results We found cellular density, but not island size, regulates of circuit activity and network function in this system. Low cellular density islands can achieve many states of activity, while high cellular density biases islands towards a limited regime characterized by low rates of activity and high synchronization, a property we summarized as "flexibility." The injury severity required for an island to lose activity in 50% of its population was significantly higher in low-density, high flexibility islands. Conclusion Together, these studies demonstrate flexible living cortical circuits are more resilient to microtrauma, providing the first evidence that initial circuit state may be a key factor to consider when evaluating the consequences of trauma to the cortex.
Collapse
Affiliation(s)
- Modupe A. Adegoke
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, United States
| | - Olivia Teter
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, United States
| | - David F. Meaney
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, United States,Department of Neurosurgery, Penn Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States,*Correspondence: David F. Meaney,
| |
Collapse
|
14
|
Verga L, Schwartze M, Stapert S, Winkens I, Kotz SA. Dysfunctional Timing in Traumatic Brain Injury Patients: Co-occurrence of Cognitive, Motor, and Perceptual Deficits. Front Psychol 2021; 12:731898. [PMID: 34733208 PMCID: PMC8558219 DOI: 10.3389/fpsyg.2021.731898] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/27/2021] [Indexed: 11/21/2022] Open
Abstract
Timing is an essential part of human cognition and of everyday life activities, such as walking or holding a conversation. Previous studies showed that traumatic brain injury (TBI) often affects cognitive functions such as processing speed and time-sensitive abilities, causing long-term sequelae as well as daily impairments. However, the existing evidence on timing capacities in TBI is mostly limited to perception and the processing of isolated intervals. It is therefore open whether the observed deficits extend to motor timing and to continuous dynamic tasks that more closely match daily life activities. The current study set out to answer these questions by assessing audio motor timing abilities and their relationship with cognitive functioning in a group of TBI patients (n = 15) and healthy matched controls. We employed a comprehensive set of tasks aiming at testing timing abilities across perception and production and from single intervals to continuous auditory sequences. In line with previous research, we report functional impairments in TBI patients concerning cognitive processing speed and perceptual timing. Critically, these deficits extended to motor timing: The ability to adjust to tempo changes in an auditory pacing sequence was impaired in TBI patients, and this motor timing deficit covaried with measures of processing speed. These findings confirm previous evidence on perceptual and cognitive timing deficits resulting from TBI and provide first evidence for comparable deficits in motor behavior. This suggests basic co-occurring perceptual and motor timing impairments that may factor into a wide range of daily activities. Our results thus place TBI into the wider range of pathologies with well-documented timing deficits (such as Parkinson’s disease) and encourage the search for novel timing-based therapeutic interventions (e.g., employing dynamic and/or musical stimuli) with high transfer potential to everyday life activities.
Collapse
Affiliation(s)
- Laura Verga
- Research Group Comparative Bioacoustics, Max Planck Institute for Psycholinguistics, Nijmegen, Netherlands.,Department of Neuropsychology and Psychopharmacology, Maastricht University, Maastricht, Netherlands
| | - Michael Schwartze
- Department of Neuropsychology and Psychopharmacology, Maastricht University, Maastricht, Netherlands
| | - Sven Stapert
- Department of Neuropsychology and Psychopharmacology, Maastricht University, Maastricht, Netherlands.,Zuyderland Medical Centre, Department of Medical Psychology, Sittard, Netherlands
| | - Ieke Winkens
- Department of Neuropsychology and Psychopharmacology, Maastricht University, Maastricht, Netherlands.,Limburg Brain Injury Center, Maastricht University, Maastricht, Netherlands
| | - Sonja A Kotz
- Department of Neuropsychology and Psychopharmacology, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
15
|
Woo E, Sansing LH, Arnsten AFT, Datta D. Chronic Stress Weakens Connectivity in the Prefrontal Cortex: Architectural and Molecular Changes. CHRONIC STRESS 2021; 5:24705470211029254. [PMID: 34485797 PMCID: PMC8408896 DOI: 10.1177/24705470211029254] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 06/14/2021] [Indexed: 12/26/2022]
Abstract
Chronic exposure to uncontrollable stress causes loss of spines and dendrites in the prefrontal cortex (PFC), a recently evolved brain region that provides top-down regulation of thought, action, and emotion. PFC neurons generate top-down goals through recurrent excitatory connections on spines. This persistent firing is the foundation for higher cognition, including working memory, and abstract thought. However, exposure to acute uncontrollable stress drives high levels of catecholamine release in the PFC, which activates feedforward calcium-cAMP signaling pathways to open nearby potassium channels, rapidly weakening synaptic connectivity to reduce persistent firing. Chronic stress exposures can further exacerbate these signaling events leading to loss of spines and resulting in marked cognitive impairment. In this review, we discuss how stress signaling mechanisms can lead to spine loss, including changes to BDNF-mTORC1 signaling, calcium homeostasis, actin dynamics, and mitochondrial actions that engage glial removal of spines through inflammatory signaling. Stress signaling events may be amplified in PFC spines due to cAMP magnification of internal calcium release. As PFC dendritic spine loss is a feature of many cognitive disorders, understanding how stress affects the structure and function of the PFC will help to inform strategies for treatment and prevention.
Collapse
Affiliation(s)
- Elizabeth Woo
- Department of Neuroscience, Yale Medical School, New Haven, CT, USA.,Department of Neurology, Yale Medical School, New Haven, CT, USA
| | - Lauren H Sansing
- Department of Neurology, Yale Medical School, New Haven, CT, USA
| | - Amy F T Arnsten
- Department of Neuroscience, Yale Medical School, New Haven, CT, USA
| | - Dibyadeep Datta
- Department of Neuroscience, Yale Medical School, New Haven, CT, USA
| |
Collapse
|
16
|
Faillot M, Chaillet A, Palfi S, Senova S. Rodent models used in preclinical studies of deep brain stimulation to rescue memory deficits. Neurosci Biobehav Rev 2021; 130:410-432. [PMID: 34437937 DOI: 10.1016/j.neubiorev.2021.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 11/28/2022]
Abstract
Deep brain stimulation paradigms might be used to treat memory disorders in patients with stroke or traumatic brain injury. However, proof of concept studies in animal models are needed before clinical translation. We propose here a comprehensive review of rodent models for Traumatic Brain Injury and Stroke. We systematically review the histological, behavioral and electrophysiological features of each model and identify those that are the most relevant for translational research.
Collapse
Affiliation(s)
- Matthieu Faillot
- Neurosurgery department, Henri Mondor University Hospital, APHP, DMU CARE, Université Paris Est Créteil, Mondor Institute for Biomedical Research, INSERM U955, Team 15, Translational Neuropsychiatry, France
| | - Antoine Chaillet
- Laboratoire des Signaux et Systèmes (L2S-UMR8506) - CentraleSupélec, Université Paris Saclay, Institut Universitaire de France, France
| | - Stéphane Palfi
- Neurosurgery department, Henri Mondor University Hospital, APHP, DMU CARE, Université Paris Est Créteil, Mondor Institute for Biomedical Research, INSERM U955, Team 15, Translational Neuropsychiatry, France
| | - Suhan Senova
- Neurosurgery department, Henri Mondor University Hospital, APHP, DMU CARE, Université Paris Est Créteil, Mondor Institute for Biomedical Research, INSERM U955, Team 15, Translational Neuropsychiatry, France.
| |
Collapse
|
17
|
Sanchez CM, Titus DJ, Wilson NM, Freund JE, Atkins CM. Early Life Stress Exacerbates Outcome after Traumatic Brain Injury. J Neurotrauma 2021; 38:555-565. [PMID: 32862765 PMCID: PMC8020564 DOI: 10.1089/neu.2020.7267] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The neurocognitive impairments associated with mild traumatic brain injury (TBI) often resolve within 1-2 weeks; however, a subset of people exhibit persistent cognitive dysfunction for weeks to months after injury. The factors that contribute to these persistent deficits are unknown. One potential risk factor for worsened outcome after TBI is a history of stress experienced by a person early in life. Early life stress (ELS) includes maltreatment such as neglect, and interferes with the normal construction of cortical and hippocampal circuits. We hypothesized that a history of ELS contributes to persistent learning and memory dysfunction following a TBI. To explore this interaction, we modeled ELS by separating Sprague Dawley pups from their nursing mothers from post-natal days 2-14 for 3 h daily. At 2 months of age, male rats received sham surgery or mild to moderate parasagittal fluid-percussion brain injury. We found that the combination of ELS with TBI in adulthood impaired hippocampal-dependent learning, as assessed with contextual fear conditioning, the water maze task, and spatial working memory. Cortical atrophy was significantly exacerbated in TBI animals exposed to ELS compared with normal-reared TBI animals. Changes in corticosterone in response to restraint stress were prolonged in TBI animals that received ELS compared with TBI animals that were normally reared or sham animals that received ELS. Our findings indicate that ELS is a risk factor for worsened outcome after TBI, and results in persistent learning and memory deficits, worsened cortical pathology, and an exacerbation of the hormonal stress response.
Collapse
Affiliation(s)
- Chantal M. Sanchez
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - David J. Titus
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Nicole M. Wilson
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Julie E. Freund
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Coleen M. Atkins
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
18
|
N-docosahexaenoylethanolamine reduces neuroinflammation and cognitive impairment after mild traumatic brain injury in rats. Sci Rep 2021; 11:756. [PMID: 33436960 PMCID: PMC7804312 DOI: 10.1038/s41598-020-80818-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/28/2020] [Indexed: 12/14/2022] Open
Abstract
At present, there is a growing interest in the study of the neurotropic activity of polyunsaturated fatty acids ethanolamides (N-acylethanolamines). N-docosahexaenoylethanolamine (DHEA, synaptamide) is an endogenous metabolite and structural analogue of anandamide, a widely studied endocannabinoid derived from arachidonic acid. The results of this study demonstrate that DHEA, when administered subcutaneously (10 mg/kg/day, 7 days), promotes cognitive recovery in rats subjected to mild traumatic brain injury (mTBI). In the cerebral cortex of experimental animals, we analyzed the dynamics of Iba-1-positive microglia activity changes and the expression of pro-inflammatory markers (IL1β, IL6, CD86). We used immortalized mouse microglial cells (SIM-A9) to assess the effects of DHEA on LPS-induced cytokines/ROS/NO/nitrite, as well as on CD206 (anti-inflammatory microglia) and the antioxidant enzyme superoxide dismutase (SOD) production. In vivo and in vitro experiments showed that DHEA: (1) improves indicators of anxiety and long-term memory; (2) inhibits the pro-inflammatory microglial cells activity; (3) decrease the level of pro-inflammatory cytokines/ROS/NO/nitrites; (4) increase CD206 and SOD production. In general, the results of this study indicate that DHEA has a complex effect on the neuroinflammation processes, which indicates its high therapeutic potential.
Collapse
|
19
|
Honig MG, Dorian CC, Worthen JD, Micetich AC, Mulder IA, Sanchez KB, Pierce WF, Del Mar NA, Reiner A. Progressive long-term spatial memory loss following repeat concussive and subconcussive brain injury in mice, associated with dorsal hippocampal neuron loss, microglial phenotype shift, and vascular abnormalities. Eur J Neurosci 2020; 54:5844-5879. [PMID: 32090401 PMCID: PMC7483557 DOI: 10.1111/ejn.14711] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 02/18/2020] [Accepted: 02/19/2020] [Indexed: 12/14/2022]
Abstract
There is considerable concern about the long‐term deleterious effects of repeat head trauma on cognition, but little is known about underlying mechanisms and pathology. To examine this, we delivered four air blasts to the left side of the mouse cranium, a week apart, with an intensity that causes deficits when delivered singly and considered “concussive,” or an intensity that does not yield significant deficits when delivered singly and considered “subconcussive.” Neither repeat concussive nor subconcussive blast produced spatial memory deficits at 4 months, but both yielded deficits at 14 months, and dorsal hippocampal neuron loss. Hierarchical cluster analysis of dorsal hippocampal microglia across the three groups based on morphology and expression of MHCII, CX3CR1, CD68 and IBA1 revealed five distinct phenotypes. Types 1A and 1B microglia were more common in sham mice, linked to better neuron survival and memory, and appeared mildly activated. By contrast, 2B and 2C microglia were more common in repeat concussive and subconcussive mice, linked to poorer neuron survival and memory, and characterized by low expression levels and attenuated processes, suggesting they were de‐activated and dysfunctional. In addition, endothelial cells in repeat concussive mice exhibited reduced CD31 and eNOS expression, which was correlated with the prevalence of type 2B and 2C microglia. Our findings suggest that both repeat concussive and subconcussive head injury engender progressive pathogenic processes, possibly through sustained effects on microglia that over time lead to increased prevalence of dysfunctional microglia, adversely affecting neurons and blood vessels, and thereby driving neurodegeneration and memory decline.
Collapse
Affiliation(s)
- Marcia G Honig
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Conor C Dorian
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - John D Worthen
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Anthony C Micetich
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Isabelle A Mulder
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Katelyn B Sanchez
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - William F Pierce
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Nobel A Del Mar
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Anton Reiner
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA.,Department of Ophthalmology, The University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
20
|
Sun YY, Zhu L, Sun ZL, Feng DF. CRMP2 improves memory deficits by enhancing the maturation of neuronal dendritic spines after traumatic brain injury. Exp Neurol 2020; 328:113253. [PMID: 32084454 DOI: 10.1016/j.expneurol.2020.113253] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 02/12/2020] [Accepted: 02/17/2020] [Indexed: 11/28/2022]
Abstract
Our recent study investigated the role of collapsin response mediator protein-2 (CRMP2) on dendritic spine morphology and memory function after traumatic brain injury (TBI). First, we examined the density and morphology of dendritic spines in Thy1-GFP mice on the 1 st day (P1D) and 7th day (P7D) after controlled cortical impact injury (CCI). The dendritic spine density in the hippocampus was decreased on P1D, in which mainly mushroom-type and thin-type spines were lost. The density of dendritic spines was increased on P7D, most of which were of the thin type. Next, we explored the expression of CRMP2 on P1D and P7D. CRMP2 expression was decreased on P1D, but the levels of the CRMP2 breakdown product were increased. On P7D, the expression pattern was the opposite. Then, we constructed CRMP2 overexpression and knockdown plasmids and transfected them into cultured neurons in vitro. CRMP2 increased the dendritic spine density of cultured neurons and the proportion of mushroom-type spines, while CRMP2-shRNA reduced the dendritic spine density and the proportion of mushroom-type spines. To determine the role of CRMP2 in dendritic spines after TBI, we stereotactically injected the CRMP2 overexpression and knockdown viruses into the hippocampus and found that CRMP2 increased the dendritic spine density and the proportion of mushroom-type spines after TBI. Meanwhile, as suggested by the morphological changes, fear conditioning behavioral experiments confirmed that CRMP2 improved memory deficits after TBI.
Collapse
Affiliation(s)
- Yi-Yu Sun
- Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201999, China
| | - Liang Zhu
- Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201999, China
| | - Zhao-Liang Sun
- Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201999, China
| | - Dong-Fu Feng
- Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201999, China; Institute of Traumatic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 201999, China.
| |
Collapse
|
21
|
Effect of mild blast-induced TBI on dendritic architecture of the cortex and hippocampus in the mouse. Sci Rep 2020; 10:2206. [PMID: 32042033 PMCID: PMC7010659 DOI: 10.1038/s41598-020-59252-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 01/20/2020] [Indexed: 11/09/2022] Open
Abstract
Traumatic brain injury (TBI) has been designated as a signature injury of modern military conflicts. Blast trauma, in particular, has come to make up a significant portion of the TBIs which are sustained in warzones. Though most TBIs are mild, even mild TBI can induce long term effects, including cognitive and memory deficits. In our study, we utilized a mouse model of mild blast-related TBI (bTBI) to investigate TBI-induced changes within the cortex and hippocampus. We performed rapid Golgi staining on the layer IV and V pyramidal neurons of the parietal cortex and the CA1 basilar tree of the hippocampus and quantified dendritic branching and distribution. We found decreased dendritic branching within both the cortex and hippocampus in injured mice. Within parietal cortex, this decreased branching was most evident within the middle region, while outer and inner regions resembled that of control mice. This study provides important knowledge in the study of how the shockwave associated with a blast explosion impacts different brain regions.
Collapse
|
22
|
Avila JA, Kiprowska M, Jean-Louis T, Rockwell P, Figueiredo-Pereira ME, Serrano PA. PACAP27 mitigates an age-dependent hippocampal vulnerability to PGJ2-induced spatial learning deficits and neuroinflammation in mice. Brain Behav 2020; 10:e01465. [PMID: 31769222 PMCID: PMC6955932 DOI: 10.1002/brb3.1465] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 08/20/2019] [Accepted: 10/13/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Inflammation in the brain is mediated by the cyclooxygenase pathway, which leads to the production of prostaglandins. Prostaglandin (PG) D2, the most abundant PG in the brain, increases under pathological conditions and is spontaneously metabolized to PGJ2. PGJ2 is highly neurotoxic, with the potential to transition neuroinflammation into a chronic state and contribute to neurodegeneration as seen in many neurological diseases. Conversely, PACAP27 is a lipophilic peptide that raises intracellular cAMP and is an anti-inflammatory agent. The aim of our study was to investigate the therapeutic potential of PACAP27 to counter the behavioral and neurotoxic effects of PGJ2 observed in aged subjects. METHODS PGJ2 was injected bilaterally into the hippocampal CA1 region of 53-week-old and 12-week-old C57BL/6N male mice, once per week over 3 weeks (three total infusions) and included co-infusions of PACAP27 within respective treatment groups. Our behavioral assessments looked at spatial learning and memory performance on the 8-arm radial maze, followed by histological analyses of fixed hippocampal tissue using Fluoro-Jade C and fluorescent immunohistochemistry focused on IBA-1 microglia. RESULTS Aged mice treated with PGJ2 exhibited spatial learning and long-term memory deficits, as well as neurodegeneration in CA3 pyramidal neurons. Aged mice that received co-infusions of PACAP27 exhibited remediated learning and memory performance and decreased neurodegeneration in CA3 pyramidal neurons. Moreover, microglial activation in the CA3 region was also reduced in aged mice cotreated with PACAP27. CONCLUSIONS Our data show that PGJ2 can produce a retrograde spread of damage not observed in PGJ2-treated young mice, leading to age-dependent neurodegeneration of hippocampal neurons producing learning and memory deficits. PACAP27 can remediate the behavioral and neurodegenerative effects that PGJ2 produces in aged subjects. Targeting specific neurotoxic prostaglandins, such as PGJ2, offers great promise as a new therapeutic strategy downstream of cyclooxygenases, to combat the neuronal deficits induced by chronic inflammation.
Collapse
Affiliation(s)
- Jorge A Avila
- Department of Psychology, Hunter College, City University of New York, New York, NY, USA.,The Graduate Center of CUNY, New York, NY, USA
| | - Magdalena Kiprowska
- The Graduate Center of CUNY, New York, NY, USA.,Department of Biological Sciences, Hunter College, City University of New York, New York, NY, USA
| | - Teneka Jean-Louis
- The Graduate Center of CUNY, New York, NY, USA.,Department of Biological Sciences, Hunter College, City University of New York, New York, NY, USA
| | - Patricia Rockwell
- The Graduate Center of CUNY, New York, NY, USA.,Department of Biological Sciences, Hunter College, City University of New York, New York, NY, USA
| | - Maria E Figueiredo-Pereira
- The Graduate Center of CUNY, New York, NY, USA.,Department of Biological Sciences, Hunter College, City University of New York, New York, NY, USA
| | - Peter A Serrano
- Department of Psychology, Hunter College, City University of New York, New York, NY, USA.,The Graduate Center of CUNY, New York, NY, USA
| |
Collapse
|
23
|
Pioglitazone improves working memory performance when administered in chronic TBI. Neurobiol Dis 2019; 132:104611. [PMID: 31513844 DOI: 10.1016/j.nbd.2019.104611] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 09/02/2019] [Accepted: 09/06/2019] [Indexed: 01/26/2023] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of long-term disability in the United States. Even in comparatively mild injuries, cognitive and behavioral symptoms can persist for years, and there are currently no established strategies for mitigating symptoms in chronic injury. A key feature of TBI-induced damage in acute and chronic injury is disruption of metabolic pathways. As neurotransmission, and therefore cognition, are highly dependent on the supply of energy, we hypothesized that modulating metabolic activity could help restore behavioral performance even when treatment was initiated weeks after TBI. We treated rats with pioglitazone, a FDA-approved drug for diabetes, beginning 46 days after lateral fluid percussion injury and tested working memory performance in the radial arm maze (RAM) after 14 days of treatment. Pioglitazone treated TBI rats performed significantly better in the RAM test than untreated TBI rats, and similarly to control animals. While hexokinase activity in hippocampus was increased by pioglitazone treatment, there was no upregulation of either the neuronal glucose transporter or hexokinase enzyme expression. Expression of glial markers GFAP and Iba-1 were also not influenced by pioglitazone treatment. These studies suggest that targeting brain metabolism, in particular hippocampal metabolism, may be effective in alleviating cognitive symptoms in chronic TBI.
Collapse
|
24
|
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: 47] [Impact Index Per Article: 7.8] [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
|
25
|
Deng-Bryant Y, Leung LY, Madathil S, Flerlage J, Yang F, Yang W, Gilsdorf J, Shear D. Chronic Cognitive Deficits and Associated Histopathology Following Closed-Head Concussive Injury in Rats. Front Neurol 2019; 10:699. [PMID: 31312174 PMCID: PMC6614177 DOI: 10.3389/fneur.2019.00699] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 06/14/2019] [Indexed: 12/13/2022] Open
Abstract
Close-head concussive injury, as one of the most common forms of traumatic brain injury (TBI), has been shown to induce cognitive deficits that are long lasting. A concussive impact model was previously established in our lab that produces clinically relevant signs of concussion and induced acute pathological changes in rats. To evaluate the long-term effects of repeated concussions in this model, we utilized a comprehensive Morris water maze (MWM) paradigm for cognitive assessments at 1 and 6 months following repeated concussive impacts in rats. As such, adult Sprague-Dawley rats received either anesthesia (sham) or repeated concussive impacts (4 consecutive impacts at 1 h interval). At 1 month post-injury, results of the spatial learning task showed that the average latencies to locate the hidden "escape" platform were significantly longer in the injured rats over the last 2 days of the MWM testing compared to sham controls (p < 0.05). In the memory retention task, rats subjected to repeated concussive impacts also spent significantly less time in the platform zone searching for the missing platform during the probe trial (p < 0.05). On the working memory task, the injured rats showed a trend toward worse performance, but this failed to reach statistical significance compared to sham controls (p = 0.07). At 6 months post-injury, no differences were detected between the injured group and sham controls in either the spatial learning or probe trials. However, rats with repeated concussive impacts exhibited significantly worsened working memory performance compared to sham controls (p < 0.05). In addition, histopathological assessments for axonal neurodegeneration using silver stain showed that repeated concussive impacts induced significantly more axonal degeneration in the corpus callosum compared to sham controls (p < 0.05) at 1 month post-injury, whereas such difference was not observed at 6 months post-injury. Overall, the results show that repeated concussive impacts in our model produced significant cognitive deficits in both spatial learning abilities and in working memory abilities in a time-dependent fashion that may be indicative of progressive pathology and warrant further investigation.
Collapse
Affiliation(s)
- Ying Deng-Bryant
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Lai Yee Leung
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States.,Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Sindhu Madathil
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Jesse Flerlage
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Fangzhou Yang
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Weihong Yang
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Janice Gilsdorf
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Deborah Shear
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| |
Collapse
|
26
|
Gálvez-Rosas A, Avila-Luna A, Valdés-Flores M, Montes S, Bueno-Nava A. GABAergic imbalance is normalized by dopamine D 1 receptor activation in the striatum contralateral to the cortical injury in motor deficit-recovered rats. Psychopharmacology (Berl) 2019; 236:2211-2222. [PMID: 30859334 DOI: 10.1007/s00213-019-05215-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 03/01/2019] [Indexed: 02/07/2023]
Abstract
RATIONALE The sensorimotor cortex and the striatum are interconnected by the corticostriatal pathway, suggesting that cortical injury alters the striatal function, which may be modulated by dopamine. OBJECTIVES We studied whether the activation of dopamine D1 receptors (D1Rs) modulates the γ-aminobutyric acid (GABA) and glutamate levels in the striatum of recovered rats at 192 h after cortical injury. METHODS The D1R agonist SKF-38393 (0, 2, 3, or 4 mg/kg) was administered at 24, 48, 96, and 192 h post-injury, and then rats were decapitated to determine GABA and glutamate levels and the levels of D1R mRNA on both sides of the striatum. RESULTS GABAergic imbalance in the striatum contralateral to the injury site was normalized by the administration of the D1R agonist, but this treatment did not produce a significant effect on glutamate levels, suggesting that glutamate was metabolized into GABA. The administration of SKF-38393 (2 mg/kg) decreased the levels of D1R mRNA in the striatum contralateral to the injury, and this effect was blocked by the coadministration of the D1R antagonist SCH-23390 (2 mg/kg). In the striatum ipsilateral to the injury, the D1R agonist increased the D1R mRNA levels, an effect that was blocked by SCH-23390. CONCLUSION The reversal of the GABAergic imbalance in the striatum contralateral to the cortical injury can be modulated by extrastriatal D1R activation, and the D1R agonist-induced increases in the D1R mRNA levels in the striatum ipsilateral to the injury suggest that the striatum may be necessary to achieve functional recovery.
Collapse
Affiliation(s)
- Arturo Gálvez-Rosas
- Lab. Neurofisiología Química de la Discapacidad, División de Neurociencias, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, SSa, Calz. México-Xochimilco 289, Col. Arenal de Guadalupe, 14389, Mexico City, Mexico
| | - Alberto Avila-Luna
- Lab. Neurofisiología Química de la Discapacidad, División de Neurociencias, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, SSa, Calz. México-Xochimilco 289, Col. Arenal de Guadalupe, 14389, Mexico City, Mexico
| | - Margarita Valdés-Flores
- Departamento de Genética y Medicina Genómica, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, SSa, Calz. México-Xochimilco 289, Col. Arenal de Guadalupe, 14389, Mexico City, Mexico
| | - Sergio Montes
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, SSa, Insurgentes Sur 3877, 14269, Mexico City, Mexico
| | - Antonio Bueno-Nava
- Lab. Neurofisiología Química de la Discapacidad, División de Neurociencias, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, SSa, Calz. México-Xochimilco 289, Col. Arenal de Guadalupe, 14389, Mexico City, Mexico.
| |
Collapse
|
27
|
McGuire JL, Ngwenya LB, McCullumsmith RE. Neurotransmitter changes after traumatic brain injury: an update for new treatment strategies. Mol Psychiatry 2019; 24:995-1012. [PMID: 30214042 DOI: 10.1038/s41380-018-0239-6] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 08/15/2018] [Accepted: 08/20/2018] [Indexed: 12/12/2022]
Abstract
Traumatic brain injury (TBI) is a pervasive problem in the United States and worldwide, as the number of diagnosed individuals is increasing yearly and there are no efficacious therapeutic interventions. A large number of patients suffer with cognitive disabilities and psychiatric conditions after TBI, especially anxiety and depression. The constellation of post-injury cognitive and behavioral symptoms suggest permanent effects of injury on neurotransmission. Guided in part by preclinical studies, clinical trials have focused on high-yield pathophysiologic mechanisms, including protein aggregation, inflammation, metabolic disruption, cell generation, physiology, and alterations in neurotransmitter signaling. Despite successful treatment of experimental TBI in animal models, clinical studies based on these findings have failed to translate to humans. The current international effort to reshape TBI research is focusing on redefining the taxonomy and characterization of TBI. In addition, as the next round of clinical trials is pending, there is a pressing need to consider what the field has learned over the past two decades of research, and how we can best capitalize on this knowledge to inform the hypotheses for future innovations. Thus, it is critically important to extend our understanding of the pathophysiology of TBI, particularly to mechanisms that are associated with recovery versus development of chronic symptoms. In this review, we focus on the pathology of neurotransmission after TBI, reflecting on what has been learned from both the preclinical and clinical studies, and we discuss new directions and opportunities for future work.
Collapse
Affiliation(s)
- Jennifer L McGuire
- Department of Neurosurgery, University of Cincinnati, Cincinnati, OH, USA.
| | - Laura B Ngwenya
- Department of Neurosurgery, University of Cincinnati, Cincinnati, OH, USA.,Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH, USA.,Neurotrauma Center, University of Cincinnati Gardner Neuroscience Institute, Cincinnati, OH, 45219, USA
| | - Robert E McCullumsmith
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, USA.,Department of Psychiatry, Cincinnati Veterans Administration Medical Center, Cincinnati, OH, USA
| |
Collapse
|
28
|
Neville IS, Zaninotto AL, Hayashi CY, Rodrigues PA, Galhardoni R, Ciampi de Andrade D, Brunoni AR, Amorim RLO, Teixeira MJ, Paiva WS. Repetitive TMS does not improve cognition in patients with TBI: A randomized double-blind trial. Neurology 2019; 93:e190-e199. [PMID: 31175209 PMCID: PMC6656650 DOI: 10.1212/wnl.0000000000007748] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 03/01/2019] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To determine whether high-frequency repetitive transcranial magnetic stimulation (rTMS) improves cognition in patients with severe traumatic brain injury. METHODS A single-center, randomized, double-blind, placebo-controlled study of rTMS was conducted in patients aged 18-60 years with chronic (>12 months postinjury) diffuse axonal injury (DAI). Patients were randomized to either a sham or real group in a 1:1 ratio. A 10-session rTMS protocol was used with 10-Hz stimulation over the left dorsolateral prefrontal cortex (DLPFC). Neuropsychological assessments were performed at 3 time points: at baseline, after the 10th rTMS session, and 90 days after intervention. The primary outcome was change in executive function evaluated using the Trail Making Test Part B. RESULTS Thirty patients with chronic DAI met the study criteria. Between-group comparisons of performance on TMT Part B at baseline and after the 10th rTMS session did not differ between groups (p = 0.680 and p = 0.341, respectively). No significant differences were observed on other neuropsychological tests. No differences in adverse events between treatment groups were observed. CONCLUSIONS Cognitive function in individuals with chronic DAI is not improved by high-frequency rTMS over the left DLPFC, though it appears safe and well-tolerated in this population. CLINICALTRIALSGOV IDENTIFIER NCT02167971. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that for individuals with chronic DAI, high-frequency rTMS over the left DLPFC does not significantly improve cognition.
Collapse
Affiliation(s)
- Iuri Santana Neville
- From the Division of Neurosurgery/LIM-62 (I.S.N., A.L.Z., C.Y.H., P.A.R., R.G., D.C.d.A., R.L.O.A., M.J.T., W.S.P.) and Department of Psychiatry, Instituto de Psiquiatria (A.R.B.), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, and Service of Interdisciplinary Neuromodulation (I.S.N., C.Y.H., R.G., D.C.d.A., A.R.B., M.J.T., W.S.P.), Universidade de Sao Paulo, Brazil; Neuromodulation Center (A.L.Z.), Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA; School of Medicine (R.G.), Universidade da Cidade de Sao Paulo UNICID, Sao Paulo; and Department of Neurology (R.L.O.A.), Universidade Federal do Amazonas, Manaus, Brazil.
| | - Ana Luiza Zaninotto
- From the Division of Neurosurgery/LIM-62 (I.S.N., A.L.Z., C.Y.H., P.A.R., R.G., D.C.d.A., R.L.O.A., M.J.T., W.S.P.) and Department of Psychiatry, Instituto de Psiquiatria (A.R.B.), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, and Service of Interdisciplinary Neuromodulation (I.S.N., C.Y.H., R.G., D.C.d.A., A.R.B., M.J.T., W.S.P.), Universidade de Sao Paulo, Brazil; Neuromodulation Center (A.L.Z.), Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA; School of Medicine (R.G.), Universidade da Cidade de Sao Paulo UNICID, Sao Paulo; and Department of Neurology (R.L.O.A.), Universidade Federal do Amazonas, Manaus, Brazil
| | - Cintya Yukie Hayashi
- From the Division of Neurosurgery/LIM-62 (I.S.N., A.L.Z., C.Y.H., P.A.R., R.G., D.C.d.A., R.L.O.A., M.J.T., W.S.P.) and Department of Psychiatry, Instituto de Psiquiatria (A.R.B.), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, and Service of Interdisciplinary Neuromodulation (I.S.N., C.Y.H., R.G., D.C.d.A., A.R.B., M.J.T., W.S.P.), Universidade de Sao Paulo, Brazil; Neuromodulation Center (A.L.Z.), Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA; School of Medicine (R.G.), Universidade da Cidade de Sao Paulo UNICID, Sao Paulo; and Department of Neurology (R.L.O.A.), Universidade Federal do Amazonas, Manaus, Brazil
| | - Priscila Aparecida Rodrigues
- From the Division of Neurosurgery/LIM-62 (I.S.N., A.L.Z., C.Y.H., P.A.R., R.G., D.C.d.A., R.L.O.A., M.J.T., W.S.P.) and Department of Psychiatry, Instituto de Psiquiatria (A.R.B.), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, and Service of Interdisciplinary Neuromodulation (I.S.N., C.Y.H., R.G., D.C.d.A., A.R.B., M.J.T., W.S.P.), Universidade de Sao Paulo, Brazil; Neuromodulation Center (A.L.Z.), Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA; School of Medicine (R.G.), Universidade da Cidade de Sao Paulo UNICID, Sao Paulo; and Department of Neurology (R.L.O.A.), Universidade Federal do Amazonas, Manaus, Brazil
| | - Ricardo Galhardoni
- From the Division of Neurosurgery/LIM-62 (I.S.N., A.L.Z., C.Y.H., P.A.R., R.G., D.C.d.A., R.L.O.A., M.J.T., W.S.P.) and Department of Psychiatry, Instituto de Psiquiatria (A.R.B.), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, and Service of Interdisciplinary Neuromodulation (I.S.N., C.Y.H., R.G., D.C.d.A., A.R.B., M.J.T., W.S.P.), Universidade de Sao Paulo, Brazil; Neuromodulation Center (A.L.Z.), Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA; School of Medicine (R.G.), Universidade da Cidade de Sao Paulo UNICID, Sao Paulo; and Department of Neurology (R.L.O.A.), Universidade Federal do Amazonas, Manaus, Brazil
| | - Daniel Ciampi de Andrade
- From the Division of Neurosurgery/LIM-62 (I.S.N., A.L.Z., C.Y.H., P.A.R., R.G., D.C.d.A., R.L.O.A., M.J.T., W.S.P.) and Department of Psychiatry, Instituto de Psiquiatria (A.R.B.), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, and Service of Interdisciplinary Neuromodulation (I.S.N., C.Y.H., R.G., D.C.d.A., A.R.B., M.J.T., W.S.P.), Universidade de Sao Paulo, Brazil; Neuromodulation Center (A.L.Z.), Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA; School of Medicine (R.G.), Universidade da Cidade de Sao Paulo UNICID, Sao Paulo; and Department of Neurology (R.L.O.A.), Universidade Federal do Amazonas, Manaus, Brazil
| | - Andre Russowsky Brunoni
- From the Division of Neurosurgery/LIM-62 (I.S.N., A.L.Z., C.Y.H., P.A.R., R.G., D.C.d.A., R.L.O.A., M.J.T., W.S.P.) and Department of Psychiatry, Instituto de Psiquiatria (A.R.B.), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, and Service of Interdisciplinary Neuromodulation (I.S.N., C.Y.H., R.G., D.C.d.A., A.R.B., M.J.T., W.S.P.), Universidade de Sao Paulo, Brazil; Neuromodulation Center (A.L.Z.), Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA; School of Medicine (R.G.), Universidade da Cidade de Sao Paulo UNICID, Sao Paulo; and Department of Neurology (R.L.O.A.), Universidade Federal do Amazonas, Manaus, Brazil
| | - Robson L Oliveira Amorim
- From the Division of Neurosurgery/LIM-62 (I.S.N., A.L.Z., C.Y.H., P.A.R., R.G., D.C.d.A., R.L.O.A., M.J.T., W.S.P.) and Department of Psychiatry, Instituto de Psiquiatria (A.R.B.), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, and Service of Interdisciplinary Neuromodulation (I.S.N., C.Y.H., R.G., D.C.d.A., A.R.B., M.J.T., W.S.P.), Universidade de Sao Paulo, Brazil; Neuromodulation Center (A.L.Z.), Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA; School of Medicine (R.G.), Universidade da Cidade de Sao Paulo UNICID, Sao Paulo; and Department of Neurology (R.L.O.A.), Universidade Federal do Amazonas, Manaus, Brazil
| | - Manoel Jacobsen Teixeira
- From the Division of Neurosurgery/LIM-62 (I.S.N., A.L.Z., C.Y.H., P.A.R., R.G., D.C.d.A., R.L.O.A., M.J.T., W.S.P.) and Department of Psychiatry, Instituto de Psiquiatria (A.R.B.), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, and Service of Interdisciplinary Neuromodulation (I.S.N., C.Y.H., R.G., D.C.d.A., A.R.B., M.J.T., W.S.P.), Universidade de Sao Paulo, Brazil; Neuromodulation Center (A.L.Z.), Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA; School of Medicine (R.G.), Universidade da Cidade de Sao Paulo UNICID, Sao Paulo; and Department of Neurology (R.L.O.A.), Universidade Federal do Amazonas, Manaus, Brazil
| | - Wellingson Silva Paiva
- From the Division of Neurosurgery/LIM-62 (I.S.N., A.L.Z., C.Y.H., P.A.R., R.G., D.C.d.A., R.L.O.A., M.J.T., W.S.P.) and Department of Psychiatry, Instituto de Psiquiatria (A.R.B.), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, and Service of Interdisciplinary Neuromodulation (I.S.N., C.Y.H., R.G., D.C.d.A., A.R.B., M.J.T., W.S.P.), Universidade de Sao Paulo, Brazil; Neuromodulation Center (A.L.Z.), Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA; School of Medicine (R.G.), Universidade da Cidade de Sao Paulo UNICID, Sao Paulo; and Department of Neurology (R.L.O.A.), Universidade Federal do Amazonas, Manaus, Brazil
| |
Collapse
|
29
|
Cognitive and neuropsychiatric impairments vary as a function of injury severity at 12 months post-experimental diffuse traumatic brain injury: Implications for dementia development. Behav Brain Res 2019; 365:66-76. [PMID: 30826298 DOI: 10.1016/j.bbr.2019.02.045] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 02/20/2019] [Accepted: 02/27/2019] [Indexed: 12/14/2022]
Abstract
Traumatic brain injury (TBI) is a common risk factor for later neurodegeneration, which can manifest as dementia. Despite this, little is known about the time-course of development of functional deficits, particularly cognitive and neuropsychiatric impairments, and whether these differ depending on the nature of the initiating insult. Therefore, this study investigated long term functional impairment at 12 months post-injury following diffuse TBI of different severities. Male Sprague-Dawley rats (420-480 g; 10-12 weeks) were either given a sham surgery (n = 14) or subjected to Marmarou's impact acceleration model of diffuse TBI for a single mild TBI (n = 12), repetitive mild TBI (3 mild diffuse injuries at 5 day intervals) (n = 14) or moderate to severe TBI (n = 14). At 12 months after injury, they were tested on a functional battery encompassing motor, neuropsychiatric (anxiety and depressive-like) and cognitive function. Our results showed that moderate to severe TBI animals exhibited significant impairments in cognitive flexibility (p = 0.009) on the Barnes maze when compared to age-matched sham animals. Neither repetitive mild TBI nor single mild TBI animals showed significant functional impairments when compared to shams. Thus, this study provides the first insight into chronic functional impairments associated with different severities of diffuse TBI, with moderate to severe TBI being a higher risk factor for impaired cognitive function at 12 months post-injury. Taken together, this may have implications for risk of dementia development following different severities of injury.
Collapse
|
30
|
Fortress AM, Avcu P, Wagner AK, Dixon CE, Pang KCH. Experimental traumatic brain injury results in estrous cycle disruption, neurobehavioral deficits, and impaired GSK3β/β-catenin signaling in female rats. Exp Neurol 2019; 315:42-51. [PMID: 30710530 DOI: 10.1016/j.expneurol.2019.01.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 01/11/2019] [Accepted: 01/29/2019] [Indexed: 12/25/2022]
Abstract
An estimated 2.8 million traumatic brain injuries (TBI) occur within the United States each year. Approximately 40% of new TBI cases are female, however few studies have investigated the effects of TBI on female subjects. In addition to typical neurobehavioral sequelae observed after TBI, such as poor cognition, impaired behavior, and somatic symptoms, women with TBI report amenorrhea or irregular menstrual cycles suggestive of disruptions in the hypothalamic-pituitary-gonadal (HPG) axis. HPG dysfunction following TBI has been linked to poor functional outcome in men and women, but the mechanisms by which this may occur or relate to behavior has not been fully developed or ascertained. The present study determined if TBI resulted in HPG axis perturbations in young adult female Sprague Dawley rats, and whether TBI was associated with cognitive and sensorimotor deficits. Following lateral fluid percussion injury, injured females spent significantly more time in diestrus compared to sham females, consistent with a persistent low sex-steroid hormone state. Injured females displayed significantly reduced 17β-estradiol (E2) and luteinizing hormone levels. Concomitantly, injured females were impaired in spatial working memory compared to shams. Impaired GSK3β/β-catenin signaling related to synaptic changes was evident one-week post-injury in the hippocampus among injured females compared to sham females, and this impairment paralleled the deficits in spatial working memory. Sensorimotor function, as evidenced by suppression of the acoustic startle response, was chronically impaired even after normal estrous cycling resumed. These data demonstrate that TBI results in estrous cycle impairments, memory dysfunction, and perturbations in GSK3β/β-catenin signaling, suggesting a potential mechanism for HPG-mediated cognitive impairment following TBI.
Collapse
Affiliation(s)
- Ashley M Fortress
- NeuroBehavioral Research Laboratory, Department of Veterans Affairs, New Jersey Health Care System, East Orange, NJ, USA; VA Pittsburgh Healthcare System, Mailstop 151, University Drive C, Pittsburgh, PA 15240, USA.
| | - Pelin Avcu
- Graduate School of Biomedical Sciences, Rutgers Biomedical and Health Sciences, 65 Bergen Street, Newark, NJ 07103, USA
| | - Amy K Wagner
- Safar Center for Resuscitation Research, Center for Neuroscience, 3471 Fifth Avenue Suite 202, Kaufman BuildingUniversity of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA; Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA.
| | - C Edward Dixon
- VA Pittsburgh Healthcare System, Mailstop 151, University Drive C, Pittsburgh, PA 15240, USA; Safar Center for Resuscitation Research, Center for Neuroscience, 3471 Fifth Avenue Suite 202, Kaufman BuildingUniversity of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Kevin C H Pang
- NeuroBehavioral Research Laboratory, Department of Veterans Affairs, New Jersey Health Care System, East Orange, NJ, USA; Graduate School of Biomedical Sciences, Rutgers Biomedical and Health Sciences, 65 Bergen Street, Newark, NJ 07103, USA; Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers Biomedical and Health Science, Newark, NJ, USA.
| |
Collapse
|
31
|
Froudist-Walsh S, Browning PG, Young JJ, Murphy KL, Mars RB, Fleysher L, Croxson PL. Macro-connectomics and microstructure predict dynamic plasticity patterns in the non-human primate brain. eLife 2018; 7:34354. [PMID: 30462609 PMCID: PMC6249000 DOI: 10.7554/elife.34354] [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: 12/14/2017] [Accepted: 09/14/2018] [Indexed: 12/12/2022] Open
Abstract
The brain displays a remarkable ability to adapt following injury by altering its connections through neural plasticity. Many of the biological mechanisms that underlie plasticity are known, but there is little knowledge as to when, or where in the brain plasticity will occur following injury. This knowledge could guide plasticity-promoting interventions and create a more accurate roadmap of the recovery process following injury. We causally investigated the time-course of plasticity after hippocampal lesions using multi-modal MRI in monkeys. We show that post-injury plasticity is highly dynamic, but also largely predictable on the basis of the functional connectivity of the lesioned region, gradients of cell densities across the cortex and the pre-lesion network structure of the brain. The ability to predict which brain areas will plastically adapt their functional connectivity following injury may allow us to decipher why some brain lesions lead to permanent loss of cognitive function, while others do not. The brain has the ability to adapt after injury, a process known as plasticity. When one area sustains damage, for example following a car accident or stroke, other areas change their activity and structure to compensate. Understanding how this happens is critical to helping people recover from brain injuries. Certain factors may affect how well the brain can repair itself. These include how much the damaged area interacts with other areas, and which cell types different areas of the brain contain. Froudist-Walsh et al. set out to determine how these factors influence recovery from brain injury in monkeys, whose brains are similar to our own. The monkeys had damage to a structure called the hippocampus. This part of the brain has a key role in memory, which is often impaired in patients with brain injuries. The hippocampus cannot repair itself because the brain has only a limited capacity to grow new neurons. Instead, the brain attempts to compensate for disruption to the hippocampus via changes in other, undamaged areas. Using brain imaging, Froudist-Walsh et al. show that the types of changes that occur depend on how much time has passed since the injury. In the first three months, many areas of the brain change how much they coordinate their activity with other areas. Highly connected areas reduce their communication with other areas the most. In the long-term, the responses of brain areas depend more on which cell types they contain. Areas with more support cells known as “glia” – which supply nutrients and energy to neurons – are better able to adapt their connectivity up to a year after the injury. These findings may ultimately benefit people who have suffered brain injuries after accidents or stroke. They suggest that stimulating intact brain areas may be helpful in the months immediately after an injury. By contrast, long-term therapy may need to focus more on structural repair. Future studies must build on these results to discover the best ways to induce successful recovery from brain injury.
Collapse
Affiliation(s)
- Sean Froudist-Walsh
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, United States.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Philip Gf Browning
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, United States.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, United States.,Laboratory of Neuropsychology, National Institute of Mental Health, Bethesda, United States
| | - James J Young
- Department of Neurology, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Kathy L Murphy
- Comparative Biology Centre, Medical School, Newcastle University, United Kingdom
| | - Rogier B Mars
- Centre for Functional MRI of the Brain, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom.,Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Lazar Fleysher
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Paula L Croxson
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, United States.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, United States.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, United States
| |
Collapse
|
32
|
The Effect of Frontal Lobe Traumatic Brain Injury on Sentence Comprehension and Working Memory. Trauma Mon 2018. [DOI: 10.5812/traumamon.74353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
33
|
Marchesseault ER, Nguyen D, Spahr L, Beals C, Razak B, Rosene JM. Head impacts and cognitive performance in men's lacrosse. PHYSICIAN SPORTSMED 2018; 46:324-330. [PMID: 29733264 DOI: 10.1080/00913847.2018.1470888] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
OBJECTIVES The purpose of this investigation was to record head impacts and assess cognitive function throughout a NCAA Division III men's lacrosse season. METHODS Fifteen NCAA Div III men's lacrosse players (age = 21.1 ± 1.5 years; height = 179 ± 7.00cm; weight = 80.74 ± 8.00kg) wore Smart Impact Monitors (SIM) (Triax Technologies, Inc., Norwalk, CT) within headbands, in 28 practices and 9 home games of one season. The SIM devices communicated with the Triax Technologies SKYi, which confirmed activated SIMs and obtained data of linear acceleration, rotational acceleration, rotational velocity, direction, and location of each head impact. A minimum threshold of 15g of force was set for head impacts to register with the SIMs. The Comprehensive Trail Making Test and Stroop Color and Word Test were administered at preseason, midseason, and postseason to assess cognitive function performance. RESULTS There was no significant difference found between all measures of frequency and magnitude of head impacts between games and practices. There was also no significant difference for peak linear acceleration of head impacts between different positions and no significant difference between the magnitude of force and the location of impact on the head. There was a significant increase in CTMT performance from preseason to midseason, from midseason to postseason, and preseason to postseason. There was no significant difference in Stroop test performance throughout the season. CONCLUSION Subconcussive head impacts in men's lacrosse appear to occur at the same magnitude in practices and games, and do not appear to be position dependent, nor head location dependent. Men's lacrosse athletes' cognitive function as measured by the CTMT, can improve, while cognitive function as measured by the Stroop test remained unchanged.
Collapse
Affiliation(s)
| | - Daniel Nguyen
- a Exercise and Sport Performance , University of New England , Biddeford , ME , USA
| | - Lee Spahr
- a Exercise and Sport Performance , University of New England , Biddeford , ME , USA
| | - Caroline Beals
- b Health Wellness and Occupational Studies , University of New England , Biddeford , ME , USA
| | - Brian Razak
- a Exercise and Sport Performance , University of New England , Biddeford , ME , USA
| | - John M Rosene
- a Exercise and Sport Performance , University of New England , Biddeford , ME , USA
| |
Collapse
|
34
|
Arulsamy A, Teng J, Colton H, Corrigan F, Collins-Praino L. Evaluation of early chronic functional outcomes and their relationship to pre-frontal cortex and hippocampal pathology following moderate-severe traumatic brain injury. Behav Brain Res 2018; 348:127-138. [DOI: 10.1016/j.bbr.2018.04.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 03/20/2018] [Accepted: 04/06/2018] [Indexed: 01/02/2023]
|
35
|
Semple BD, Zamani A, Rayner G, Shultz SR, Jones NC. Affective, neurocognitive and psychosocial disorders associated with traumatic brain injury and post-traumatic epilepsy. Neurobiol Dis 2018; 123:27-41. [PMID: 30059725 DOI: 10.1016/j.nbd.2018.07.018] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 07/08/2018] [Accepted: 07/16/2018] [Indexed: 12/13/2022] Open
Abstract
Survivors of traumatic brain injury (TBI) often develop chronic neurological, neurocognitive, psychological, and psychosocial deficits that can have a profound impact on an individual's wellbeing and quality of life. TBI is also a common cause of acquired epilepsy, which is itself associated with significant behavioral morbidity. This review considers the clinical and preclinical evidence that post-traumatic epilepsy (PTE) acts as a 'second-hit' insult to worsen chronic behavioral outcomes for brain-injured patients, across the domains of emotional, cognitive, and psychosocial functioning. Surprisingly, few well-designed studies have specifically examined the relationship between seizures and behavioral outcomes after TBI. The complex mechanisms underlying these comorbidities remain incompletely understood, although many of the biological processes that precipitate seizure occurrence and epileptogenesis may also contribute to the development of chronic behavioral deficits. Further, the relationship between PTE and behavioral dysfunction is increasingly recognized to be a bidirectional one, whereby premorbid conditions are a risk factor for PTE. Clinical studies in this arena are often challenged by the confounding effects of anti-seizure medications, while preclinical studies have rarely examined an adequately extended time course to fully capture the time course of epilepsy development after a TBI. To drive the field forward towards improved treatment strategies, it is imperative that both seizures and neurobehavioral outcomes are assessed in parallel after TBI, both in patient populations and preclinical models.
Collapse
Affiliation(s)
- Bridgette D Semple
- Department of Neuroscience, Monash University, 99 Commercial Road, Melbourne, VIC, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Royal Parade, Parkville, VIC, Australia.
| | - Akram Zamani
- Department of Neuroscience, Monash University, 99 Commercial Road, Melbourne, VIC, Australia.
| | - Genevieve Rayner
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre (Austin Campus), Heidelberg, VIC, Australia; Melbourne School of Psychological Sciences, The University of Melbourne, Parkville, VIC, Australia; Comprehensive Epilepsy Program, Alfred Health, Australia.
| | - Sandy R Shultz
- Department of Neuroscience, Monash University, 99 Commercial Road, Melbourne, VIC, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Royal Parade, Parkville, VIC, Australia.
| | - Nigel C Jones
- Department of Neuroscience, Monash University, 99 Commercial Road, Melbourne, VIC, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Royal Parade, Parkville, VIC, Australia.
| |
Collapse
|
36
|
Thau-Zuchman O, Gomes RN, Dyall SC, Davies M, Priestley JV, Groenendijk M, De Wilde MC, Tremoleda JL, Michael-Titus AT. Brain Phospholipid Precursors Administered Post-Injury Reduce Tissue Damage and Improve Neurological Outcome in Experimental Traumatic Brain Injury. J Neurotrauma 2018; 36:25-42. [PMID: 29768974 PMCID: PMC6306688 DOI: 10.1089/neu.2017.5579] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Traumatic brain injury (TBI) leads to cellular loss, destabilization of membranes, disruption of synapses and altered brain connectivity, and increased risk of neurodegenerative disease. A significant and long-lasting decrease in phospholipids (PLs), essential membrane constituents, has recently been reported in plasma and brain tissue, in human and experimental TBI. We hypothesized that supporting PL synthesis post-injury could improve outcome post-TBI. We tested this hypothesis using a multi-nutrient combination designed to support the biosynthesis of PLs and available for clinical use. The multi-nutrient, Fortasyn® Connect (FC), contains polyunsaturated omega-3 fatty acids, choline, uridine, vitamins, cofactors required for PL biosynthesis, and has been shown to have significant beneficial effects in early Alzheimer's disease. Male C57BL/6 mice received a controlled cortical impact injury and then were fed a control diet or a diet enriched with FC for 70 days. FC led to a significantly improved sensorimotor outcome and cognition, reduced lesion size and oligodendrocyte loss, and it restored myelin. It reversed the loss of the synaptic protein synaptophysin and decreased levels of the axon growth inhibitor, Nogo-A, thus creating a permissive environment. It decreased microglia activation and the rise in ß-amyloid precursor protein and restored the depressed neurogenesis. The effects of this medical multi-nutrient suggest that support of PL biosynthesis post-TBI, a new treatment paradigm, has significant therapeutic potential in this neurological condition for which there is no satisfactory treatment. The multi-nutrient tested has been used in dementia patients and is safe and well tolerated, which would enable rapid clinical exploration in TBI.
Collapse
Affiliation(s)
- Orli Thau-Zuchman
- 1 Centre for Neuroscience and Trauma, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Rita N Gomes
- 1 Centre for Neuroscience and Trauma, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Simon C Dyall
- 3 Bournemouth University, Royal London House, Bournemouth, United Kingdom
| | - Meirion Davies
- 1 Centre for Neuroscience and Trauma, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - John V Priestley
- 1 Centre for Neuroscience and Trauma, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Martine Groenendijk
- 2 Nutricia Research-Nutricia Advanced Medical Nutrition, Utrecht, The Netherlands
| | - Martijn C De Wilde
- 2 Nutricia Research-Nutricia Advanced Medical Nutrition, Utrecht, The Netherlands
| | - Jordi L Tremoleda
- 1 Centre for Neuroscience and Trauma, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Adina T Michael-Titus
- 1 Centre for Neuroscience and Trauma, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
37
|
Grossberg S. Desirability, availability, credit assignment, category learning, and attention: Cognitive-emotional and working memory dynamics of orbitofrontal, ventrolateral, and dorsolateral prefrontal cortices. Brain Neurosci Adv 2018; 2:2398212818772179. [PMID: 32166139 PMCID: PMC7058233 DOI: 10.1177/2398212818772179] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Accepted: 03/16/2018] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND The prefrontal cortices play an essential role in cognitive-emotional and working memory processes through interactions with multiple brain regions. METHODS This article further develops a unified neural architecture that explains many recent and classical data about prefrontal function and makes testable predictions. RESULTS Prefrontal properties of desirability, availability, credit assignment, category learning, and feature-based attention are explained. These properties arise through interactions of orbitofrontal, ventrolateral prefrontal, and dorsolateral prefrontal cortices with the inferotemporal cortex, perirhinal cortex, parahippocampal cortices; ventral bank of the principal sulcus, ventral prearcuate gyrus, frontal eye fields, hippocampus, amygdala, basal ganglia, hypothalamus, and visual cortical areas V1, V2, V3A, V4, middle temporal cortex, medial superior temporal area, lateral intraparietal cortex, and posterior parietal cortex. Model explanations also include how the value of visual objects and events is computed, which objects and events cause desired consequences and which may be ignored as predictively irrelevant, and how to plan and act to realise these consequences, including how to selectively filter expected versus unexpected events, leading to movements towards, and conscious perception of, expected events. Modelled processes include reinforcement learning and incentive motivational learning; object and spatial working memory dynamics; and category learning, including the learning of object categories, value categories, object-value categories, and sequence categories, or list chunks. CONCLUSION This article hereby proposes a unified neural theory of prefrontal cortex and its functions.
Collapse
Affiliation(s)
- Stephen Grossberg
- Center for Adaptive Systems, Graduate Program in Cognitive and Neural Systems, Departments of Mathematics & Statistics, Psychological & Brain Sciences, Biomedical Engineering, Boston University, Boston, MA, USA
| |
Collapse
|
38
|
Kaplan GB, Leite-Morris KA, Wang L, Rumbika KK, Heinrichs SC, Zeng X, Wu L, Arena DT, Teng YD. Pathophysiological Bases of Comorbidity: Traumatic Brain Injury and Post-Traumatic Stress Disorder. J Neurotrauma 2017; 35:210-225. [PMID: 29017388 DOI: 10.1089/neu.2016.4953] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The high rates of traumatic brain injury (TBI) and post-traumatic stress disorder (PTSD) diagnoses encountered in recent years by the United States Veterans Affairs Healthcare System have increased public awareness and research investigation into these conditions. In this review, we analyze the neural mechanisms underlying the TBI/PTSD comorbidity. TBI and PTSD present with common neuropsychiatric symptoms including anxiety, irritability, insomnia, personality changes, and memory problems, and this overlap complicates diagnostic differentiation. Interestingly, both TBI and PTSD can be produced by overlapping pathophysiological changes that disrupt neural connections termed the "connectome." The neural disruptions shared by PTSD and TBI and the comorbid condition include asymmetrical white matter tract abnormalities and gray matter changes in the basolateral amygdala, hippocampus, and prefrontal cortex. These neural circuitry dysfunctions result in behavioral changes that include executive function and memory impairments, fear retention, fear extinction deficiencies, and other disturbances. Pathophysiological etiologies can be identified using experimental models of TBI, such as fluid percussion or blast injuries, and for PTSD, using models of fear conditioning, retention, and extinction. In both TBI and PTSD, there are discernible signs of neuroinflammation, excitotoxicity, and oxidative damage. These disturbances produce neuronal death and degeneration, axonal injury, and dendritic spine dysregulation and changes in neuronal morphology. In laboratory studies, various forms of pharmacological or psychological treatments are capable of reversing these detrimental processes and promoting axonal repair, dendritic remodeling, and neurocircuitry reorganization, resulting in behavioral and cognitive functional enhancements. Based on these mechanisms, novel neurorestorative therapeutics using anti-inflammatory, antioxidant, and anticonvulsant agents may promote better outcomes for comorbid TBI and PTSD.
Collapse
Affiliation(s)
- Gary B Kaplan
- 1 Mental Health Service , VA Boston Healthcare System, Brockton, Massachusetts.,2 Department of Psychiatry, Boston University School of Medicine , Boston, Massachusetts.,3 Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine , Boston, Massachusetts
| | - Kimberly A Leite-Morris
- 2 Department of Psychiatry, Boston University School of Medicine , Boston, Massachusetts.,3 Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine , Boston, Massachusetts.,4 Research Service, VA Boston Healthcare System , Jamaica Plain, Massachusetts
| | - Lei Wang
- 5 Division of Spinal Cord Injury Research, VA Boston Healthcare System , West Roxbury, Massachusetts.,6 Departments of Physical Medicine and Rehabilitation and Neurosurgery, Harvard Medical School , Boston, Massachusetts
| | - Kendra K Rumbika
- 7 Research Service, VA Boston Healthcare System , West Roxbury, Massachusetts
| | - Stephen C Heinrichs
- 7 Research Service, VA Boston Healthcare System , West Roxbury, Massachusetts
| | - Xiang Zeng
- 5 Division of Spinal Cord Injury Research, VA Boston Healthcare System , West Roxbury, Massachusetts.,6 Departments of Physical Medicine and Rehabilitation and Neurosurgery, Harvard Medical School , Boston, Massachusetts
| | - Liquan Wu
- 5 Division of Spinal Cord Injury Research, VA Boston Healthcare System , West Roxbury, Massachusetts.,6 Departments of Physical Medicine and Rehabilitation and Neurosurgery, Harvard Medical School , Boston, Massachusetts
| | - Danielle T Arena
- 7 Research Service, VA Boston Healthcare System , West Roxbury, Massachusetts
| | - Yang D Teng
- 5 Division of Spinal Cord Injury Research, VA Boston Healthcare System , West Roxbury, Massachusetts.,6 Departments of Physical Medicine and Rehabilitation and Neurosurgery, Harvard Medical School , Boston, Massachusetts
| |
Collapse
|
39
|
Froudist-Walsh S, López-Barroso D, José Torres-Prioris M, Croxson PL, Berthier ML. Plasticity in the Working Memory System: Life Span Changes and Response to Injury. Neuroscientist 2017; 24:261-276. [PMID: 28691573 DOI: 10.1177/1073858417717210] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Working memory acts as a key bridge between perception, long-term memory, and action. The brain regions, connections, and neurotransmitters that underlie working memory undergo dramatic plastic changes during the life span, and in response to injury. Early life reliance on deep gray matter structures fades during adolescence as increasing reliance on prefrontal and parietal cortex accompanies the development of executive aspects of working memory. The rise and fall of working memory capacity and executive functions parallels the development and loss of neurotransmitter function in frontal cortical areas. Of the affected neurotransmitters, dopamine and acetylcholine modulate excitatory-inhibitory circuits that underlie working memory, are important for plasticity in the system, and are affected following preterm birth and adult brain injury. Pharmacological interventions to promote recovery of working memory abilities have had limited success, but hold promise if used in combination with behavioral training and brain stimulation. The intense study of working memory in a range of species, ages and following injuries has led to better understanding of the intrinsic plasticity mechanisms in the working memory system. The challenge now is to guide these mechanisms to better improve or restore working memory function.
Collapse
Affiliation(s)
- Sean Froudist-Walsh
- 1 Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Diana López-Barroso
- 2 Cognitive Neurology and Aphasia Unit and Cathedra ARPA of Aphasia, Centro de Investigaciones Médico-Sanitarias (CIMES) and Instituto de Investigación Biomédica de Malaga, University of Malaga, Malaga, Spain.,3 Area of Psychobiology, Faculty of Psychology, University of Malaga, Malaga, Spain
| | - María José Torres-Prioris
- 2 Cognitive Neurology and Aphasia Unit and Cathedra ARPA of Aphasia, Centro de Investigaciones Médico-Sanitarias (CIMES) and Instituto de Investigación Biomédica de Malaga, University of Malaga, Malaga, Spain.,3 Area of Psychobiology, Faculty of Psychology, University of Malaga, Malaga, Spain
| | - Paula L Croxson
- 1 Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,4 Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marcelo L Berthier
- 2 Cognitive Neurology and Aphasia Unit and Cathedra ARPA of Aphasia, Centro de Investigaciones Médico-Sanitarias (CIMES) and Instituto de Investigación Biomédica de Malaga, University of Malaga, Malaga, Spain
| |
Collapse
|
40
|
Paterno R, Folweiler KA, Cohen AS. Pathophysiology and Treatment of Memory Dysfunction After Traumatic Brain Injury. Curr Neurol Neurosci Rep 2017; 17:52. [PMID: 28500417 PMCID: PMC5861722 DOI: 10.1007/s11910-017-0762-x] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Memory is fundamental to everyday life, and cognitive impairments resulting from traumatic brain injury (TBI) have devastating effects on TBI survivors. A contributing component to memory impairments caused by TBI is alteration in the neural circuits associated with memory function. In this review, we aim to bring together experimental findings that characterize behavioral memory deficits and the underlying pathophysiology of memory-involved circuits after TBI. While there is little doubt that TBI causes memory and cognitive dysfunction, it is difficult to conclude which memory phase, i.e., encoding, maintenance, or retrieval, is specifically altered by TBI. This is most likely due to variation in behavioral protocols and experimental models. Additionally, we review a selection of experimental treatments that hold translational potential to mitigate memory dysfunction following injury.
Collapse
Affiliation(s)
- Rosalia Paterno
- Center for Sleep and Circadian Neurobiology, Perelman School of Medicine, University of Pennsylvania, 3615 Civic Center Boulevard, Abramson Research Center, Rm. 816-h, Philadelphia, PA, 19104, USA.
| | - Kaitlin A Folweiler
- Department of Anesthesiology and Critical Care Medicine, Joseph Stokes, Jr. Research Institute, Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Abramson Research Center, Rm. 816-h, Philadelphia, PA, 19104, USA
| | - Akiva S Cohen
- Department of Anesthesiology and Critical Care Medicine, Joseph Stokes, Jr. Research Institute, Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Abramson Research Center, Rm. 816-h, Philadelphia, PA, 19104, USA
- Department of Anesthesiology and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, 3615 Civic Center Boulevard, Abramson Research Center, Rm. 816-h, Philadelphia, PA, 19104, USA
| |
Collapse
|
41
|
Wilke S, List J, Mekle R, Lindenberg R, Bukowski M, Ott S, Schubert F, Ittermann B, Flöel A. No Effect of Anodal Transcranial Direct Current Stimulation on Gamma-Aminobutyric Acid Levels in Patients with Recurrent Mild Traumatic Brain Injury. J Neurotrauma 2017; 34:281-290. [DOI: 10.1089/neu.2016.4399] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Skadi Wilke
- Department of Neurology, Charité–University Hospital, Berlin, Germany
| | - Jonathan List
- Department of Neurology, Charité–University Hospital, Berlin, Germany
| | - Ralf Mekle
- Physikalisch-Technische Bundesanstalt (PTB), Braunschweig und Berlin, Berlin, Germany
| | - Robert Lindenberg
- Department of Neurology, Charité–University Hospital, Berlin, Germany
| | - Martin Bukowski
- Department of Neurology, Charité–University Hospital, Berlin, Germany
| | - Stefanie Ott
- Department of Neurology, Charité–University Hospital, Berlin, Germany
| | - Florian Schubert
- Physikalisch-Technische Bundesanstalt (PTB), Braunschweig und Berlin, Berlin, Germany
| | - Bernd Ittermann
- Physikalisch-Technische Bundesanstalt (PTB), Braunschweig und Berlin, Berlin, Germany
| | - Agnes Flöel
- Department of Neurology, Charité–University Hospital, Berlin, Germany
- Center for Stroke Research Berlin, Charité–University Hospital, Berlin, Germany
- NeuroCure Cluster of Excellence, Charité–University Hospital, Berlin, Germany
| |
Collapse
|
42
|
Caruso JP, Susick LL, Charlton JL, Henson EL, Conti AC. Region-specific disruption of synapsin phosphorylation following ethanol administration in brain-injured mice. Brain Circ 2016; 2:183-188. [PMID: 30276296 PMCID: PMC6126228 DOI: 10.4103/2394-8108.195284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 07/13/2016] [Accepted: 07/18/2016] [Indexed: 11/11/2022] Open
Abstract
Introduction: Civilians and military personnel develop a range of physical and psychosocial impairments following traumatic brain injury (TBI), including alcohol abuse. As a consequence, increased rates of alcohol misuse magnify TBI-induced pathologies and impede rehabilitation efforts. Therefore, a developed understanding of the mechanisms that foster susceptibility of the injured brain to alcohol sensitivity and the response of the injured brain to alcohol is imperative for the treatment of TBI patients. Alcohol sensitivity has been demonstrated to be increased following experimental TBI and, in additional studies, regulated by presynaptic vesicle release mechanisms, including synapsin phosphorylation. Materials and Methods: Mice were exposed to controlled midline impact of the intact skull and assessed for cortical, hippocampal, and striatal expression of phosphorylated synapsin I and II in response to high-dose ethanol exposure administered 14 days following injury, a time point at which injured mice demonstrate increased sedation after ethanol exposure. Results and Discussion: Immunoblot quantitation revealed that TBI alone, compared to sham controls, significantly increased phosphorylated synapsin I and II protein expression in the striatum. In sham controls, ethanol administration significantly increased phosphorylated synapsin I and II protein expression compared to saline-treated sham controls; however, no significant increase in ethanol-induced phosphorylated synapsin I and II protein expression was observed in the striatum of injured mice compared to saline-treated TBI controls. A similar expression pattern was observed in the cortex although restricted to increases in phosphorylated synapsin II. Conclusion: These data show that increased phosphorylated synapsin expression in the injured striatum may reflect a compensatory neuroplastic response to TBI which is proposed to occur as a result of a compromised presynaptic response of the injured brain to high-dose ethanol. These results offer a mechanistic basis for the altered ethanol sensitivity observed following experimental TBI and contribute to our understanding of alcohol action in the injured brain.
Collapse
Affiliation(s)
- James P Caruso
- John D. Dingell VA Medical Center and Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Laura L Susick
- John D. Dingell VA Medical Center and Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Jennifer L Charlton
- John D. Dingell VA Medical Center and Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Emily L Henson
- John D. Dingell VA Medical Center and Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Alana C Conti
- John D. Dingell VA Medical Center and Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| |
Collapse
|
43
|
Sleep Modulation Alleviates Axonal Damage and Cognitive Decline after Rodent Traumatic Brain Injury. J Neurosci 2016; 36:3422-9. [PMID: 27013672 DOI: 10.1523/jneurosci.3274-15.2016] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 02/18/2016] [Indexed: 12/25/2022] Open
Abstract
UNLABELLED Traumatic brain injury (TBI) is a major cause of death and disability worldwide. It produces diffuse axonal injury (DAI), which contributes to cognitive impairment, but effective disease-modifying treatment strategies are missing. We have recently developed a rat model of closed skull TBI that reproduces human TBI consequences, including DAI and clinical sequelae such as memory impairment. Here, we investigated whether sleep modulation after trauma has an impact on DAI and memory outcome. We assessed cognition with the novel object recognition test and stained for amyloid precursor protein, a DAI marker. We found that both sleep induction and restriction acutely after TBI enhanced encephalographic slow-wave activity, markedly reduced diffuse axonal damage in the cortex and hippocampus, and improved memory impairment 2 weeks after trauma. These results suggest that enhancing slow-wave sleep acutely after trauma may have a beneficial disease-modifying effect in subjects with acute TBI. SIGNIFICANCE STATEMENT Traumatic brain injury (TBI) is a clinically important entity. Cognitive deficits belong to the most prevalent chronic posttraumatic symptoms, most likely due to diffuse axonal injury (DAI). A growing body of evidence suggests a role of sleep in the clearance of waste products in the brain, possibly including amyloid precursor protein (APP), a marker of DAI. In this study, we provide evidence that enhancement of slow-wave oscillatory activity in the delta-frequency range decreases the APP-immunoreactivity and preserves cognitive abilities after trauma, potentially offering novel, noninvasive treatment options for traumatic injury.
Collapse
|
44
|
Titus DJ, Wilson NM, Freund JE, Carballosa MM, Sikah KE, Furones C, Dietrich WD, Gurney ME, Atkins CM. Chronic Cognitive Dysfunction after Traumatic Brain Injury Is Improved with a Phosphodiesterase 4B Inhibitor. J Neurosci 2016; 36:7095-108. [PMID: 27383587 PMCID: PMC4938858 DOI: 10.1523/jneurosci.3212-15.2016] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 05/20/2016] [Accepted: 05/25/2016] [Indexed: 12/19/2022] Open
Abstract
UNLABELLED Learning and memory impairments are common in traumatic brain injury (TBI) survivors. However, there are no effective treatments to improve TBI-induced learning and memory impairments. TBI results in decreased cAMP signaling and reduced cAMP-response-element binding protein (CREB) activation, a critical pathway involved in learning and memory. TBI also acutely upregulates phosphodiesterase 4B2 (PDE4B2), which terminates cAMP signaling by hydrolyzing cAMP. We hypothesized that a subtype-selective PDE4B inhibitor could reverse the learning deficits induced by TBI. To test this hypothesis, adult male Sprague-Dawley rats received sham surgery or moderate parasagittal fluid-percussion brain injury. At 3 months postsurgery, animals were administered a selective PDE4B inhibitor or vehicle before cue and contextual fear conditioning, water maze training and a spatial working memory task. Treatment with the PDE4B inhibitor significantly reversed the TBI-induced deficits in cue and contextual fear conditioning and water maze retention. To further understand the underlying mechanisms of these memory impairments, we examined hippocampal long-term potentiation (LTP). TBI resulted in a significant reduction in basal synaptic transmission and impaired expression of LTP. Treatment with the PDE4B inhibitor significantly reduced the deficits in basal synaptic transmission and rescued LTP expression. The PDE4B inhibitor reduced tumor necrosis factor-α levels and increased phosphorylated CREB levels after TBI, suggesting that this drug inhibited molecular pathways in the brain known to be regulated by PDE4B. These results suggest that a subtype-selective PDE4B inhibitor is a potential therapeutic to reverse chronic learning and memory dysfunction and deficits in hippocampal synaptic plasticity following TBI. SIGNIFICANCE STATEMENT Currently, there are an estimated 3.2-5.3 million individuals living with disabilities from traumatic brain injury (TBI) in the United States, and 8 of 10 of these individuals report cognitive disabilities (Thurman et al., 1999; Lew et al., 2006; Zaloshnja et al., 2008). One of the molecular mechanisms associated with chronic cognitive disabilities is impaired cAMP signaling in the hippocampus. In this study, we report that a selective phosphodiesterase 4B (PDE4B) inhibitor reduces chronic cognitive deficits after TBI and rescues deficits in hippocampal long-term potentiation. These results suggest that PDE4B inhibition has the potential to improve learning and memory ability and overall functioning for people living with TBI.
Collapse
Affiliation(s)
- David J Titus
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida 33136, and
| | - Nicole M Wilson
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida 33136, and
| | - Julie E Freund
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida 33136, and
| | - Melissa M Carballosa
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida 33136, and
| | - Kevin E Sikah
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida 33136, and
| | - Concepcion Furones
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida 33136, and
| | - W Dalton Dietrich
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida 33136, and
| | - Mark E Gurney
- Tetra Discovery Partners, Grand Rapids, Michigan 49503
| | - Coleen M Atkins
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida 33136, and
| |
Collapse
|
45
|
Büchele F, Morawska MM, Schreglmann SR, Penner M, Muser M, Baumann CR, Noain D. Novel Rat Model of Weight Drop-Induced Closed Diffuse Traumatic Brain Injury Compatible with Electrophysiological Recordings of Vigilance States. J Neurotrauma 2016; 33:1171-80. [DOI: 10.1089/neu.2015.4001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Fabian Büchele
- Department of Neurology, University Hospital of Zurich, Zurich, Switzerland
| | - Marta M. Morawska
- Department of Neurology, University Hospital of Zurich, Zurich, Switzerland
- Neuroscience Centre Zurich ZNZ, University of Zurich, Zurich, Switzerland
| | | | - Marco Penner
- Department of Neurology, University Hospital of Zurich, Zurich, Switzerland
| | - Markus Muser
- Working Group on Accident Mechanics, Swiss Federal Institute of Technology, Zurich, Switzerland
| | | | - Daniela Noain
- Department of Neurology, University Hospital of Zurich, Zurich, Switzerland
| |
Collapse
|
46
|
Lee MS, Kim H. Development and application of cognitive-pragmatic language ability assessment protocol for traumatic brain injury. APPLIED NEUROPSYCHOLOGY-ADULT 2016; 23:436-48. [PMID: 27218873 DOI: 10.1080/23279095.2016.1178644] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The study aim was to introduce a newly-developed multifaceted cognitive-pragmatic language assessment protocol. This study was also designed to assess the reliability and validity of the assessment protocol in the discrimination between mild traumatic brain injury (mTBI) and normal control. Individuals in this study were 25 to 64 years old. Ten mTBIs and twenty-two control group were recruited for the preliminary study. Their mean ages were 45.20 and 41.23, respectively. For the main study, we recruited 39 mTBIs and 100 healthy individuals whose mean ages were 44.67 and 40.84, respectively. The newly-developed protocol was completed through a systematic review based on an item analysis. We administered the CAPTBI based on nine domains, 22 subcategories, and 57 items. All nine domains of the CAPTBI were found to be significant variables by which mTBI individuals can be distinguished from normal individuals (p < .001). We also presented the cut-off points by education level to maximize the validity of differentiating the two groups. This study is the first attempt to evaluate mTBI by means of the cognitive-linguistic protocol with multiple domains. The CAPTBI is an appropriate tool for differentiating the cognitive-pragmatic language abilities between mTBI and control group.
Collapse
Affiliation(s)
- Mi Sook Lee
- a Graduate School of Special Education (Speech Rehabilitation & Therapy) , Kongju National University , Gongju , Korea
| | - HyangHee Kim
- b Graduate Program in Speech-Language Pathology , Yonsei University , Seoul , Korea.,c Department & Research Institute of Rehabilitation Medicine , Yonsei University College of Medicine , Seoul , Korea
| |
Collapse
|
47
|
Fischer TD, Hylin MJ, Zhao J, Moore AN, Waxham MN, Dash PK. Altered Mitochondrial Dynamics and TBI Pathophysiology. Front Syst Neurosci 2016; 10:29. [PMID: 27065821 PMCID: PMC4811888 DOI: 10.3389/fnsys.2016.00029] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 03/15/2016] [Indexed: 11/13/2022] Open
Abstract
Mitochondrial function is intimately linked to cellular survival, growth, and death. Mitochondria not only generate ATP from oxidative phosphorylation, but also mediate intracellular calcium buffering, generation of reactive oxygen species (ROS), and apoptosis. Electron leakage from the electron transport chain, especially from damaged or depolarized mitochondria, can generate excess free radicals that damage cellular proteins, DNA, and lipids. Furthermore, mitochondrial damage releases pro-apoptotic factors to initiate cell death. Previous studies have reported that traumatic brain injury (TBI) reduces mitochondrial respiration, enhances production of ROS, and triggers apoptotic cell death, suggesting a prominent role of mitochondria in TBI pathophysiology. Mitochondria maintain cellular energy homeostasis and health via balanced processes of fusion and fission, continuously dividing and fusing to form an interconnected network throughout the cell. An imbalance of these processes, particularly an excess of fission, can be detrimental to mitochondrial function, causing decreased respiration, ROS production, and apoptosis. Mitochondrial fission is regulated by the cytosolic GTPase, dynamin-related protein 1 (Drp1), which translocates to the mitochondrial outer membrane (MOM) to initiate fission. Aberrant Drp1 activity has been linked to excessive mitochondrial fission and neurodegeneration. Measurement of Drp1 levels in purified hippocampal mitochondria showed an increase in TBI animals as compared to sham controls. Analysis of cryo-electron micrographs of these mitochondria also showed that TBI caused an initial increase in the length of hippocampal mitochondria at 24 h post-injury, followed by a significant decrease in length at 72 h. Post-TBI administration of Mitochondrial division inhibitor-1 (Mdivi-1), a pharmacological inhibitor of Drp1, prevented this decrease in mitochondria length. Mdivi-1 treatment also reduced the loss of newborn neurons in the hippocampus and improved novel object recognition (NOR) memory and context-specific fear memory. Taken together, our results show that TBI increases mitochondrial fission and that inhibition of fission improves hippocampal-dependent learning and memory, suggesting that strategies to reduce fission may have translational value after injury.
Collapse
Affiliation(s)
- Tara D Fischer
- Department of Neurobiology and Anatomy, McGovern Medical School, University of Texas Health Science Center at Houston Houston, TX, USA
| | - Michael J Hylin
- Department of Psychology, Southern Illinois University Carbondale, IL, USA
| | - Jing Zhao
- Department of Neurobiology and Anatomy, McGovern Medical School, University of Texas Health Science Center at Houston Houston, TX, USA
| | - Anthony N Moore
- Department of Neurobiology and Anatomy, McGovern Medical School, University of Texas Health Science Center at Houston Houston, TX, USA
| | - M Neal Waxham
- Department of Neurobiology and Anatomy, McGovern Medical School, University of Texas Health Science Center at Houston Houston, TX, USA
| | - Pramod K Dash
- Department of Neurobiology and Anatomy, McGovern Medical School, University of Texas Health Science Center at HoustonHouston, TX, USA; Vivian L. Smith Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at HoustonHouston, TX, USA
| |
Collapse
|
48
|
Guerriero RM, Giza CC, Rotenberg A. Glutamate and GABA imbalance following traumatic brain injury. Curr Neurol Neurosci Rep 2015; 15:27. [PMID: 25796572 DOI: 10.1007/s11910-015-0545-1] [Citation(s) in RCA: 328] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Traumatic brain injury (TBI) leads to multiple short- and long-term changes in neuronal circuits that ultimately conclude with an imbalance of cortical excitation and inhibition. Changes in neurotransmitter concentrations, receptor populations, and specific cell survival are important contributing factors. Many of these changes occur gradually, which may explain the vulnerability of the brain to multiple mild impacts, alterations in neuroplasticity, and delays in the presentation of posttraumatic epilepsy. In this review, we provide an overview of normal glutamate and GABA homeostasis and describe acute, subacute, and chronic changes that follow injury. We conclude by highlighting opportunities for therapeutic interventions in this paradigm.
Collapse
Affiliation(s)
- Réjean M Guerriero
- Division Epilepsy, Department of Neurology, Boston Children's Hospital and Harvard Medical School, 300 Longwood Ave, Boston, MA, 02115, USA,
| | | | | |
Collapse
|
49
|
Smith CJ, Xiong G, Elkind JA, Putnam B, Cohen AS. Brain Injury Impairs Working Memory and Prefrontal Circuit Function. Front Neurol 2015; 6:240. [PMID: 26617569 PMCID: PMC4643141 DOI: 10.3389/fneur.2015.00240] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 10/30/2015] [Indexed: 12/13/2022] Open
Abstract
More than 2.5 million Americans suffer a traumatic brain injury (TBI) each year. Even mild to moderate TBI causes long-lasting neurological effects. Despite its prevalence, no therapy currently exists to treat the underlying cause of cognitive impairment suffered by TBI patients. Following lateral fluid percussion injury (LFPI), the most widely used experimental model of TBI, we investigated alterations in working memory and excitatory/inhibitory synaptic balance in the prefrontal cortex. LFPI impaired working memory as assessed with a T-maze behavioral task. Field excitatory postsynaptic potentials recorded in the prefrontal cortex were reduced in slices derived from brain-injured mice. Spontaneous and miniature excitatory postsynaptic currents onto layer 2/3 neurons were more frequent in slices derived from LFPI mice, while inhibitory currents onto layer 2/3 neurons were smaller after LFPI. Additionally, an increase in action potential threshold and concomitant decrease in firing rate was observed in layer 2/3 neurons in slices from injured animals. Conversely, no differences in excitatory or inhibitory synaptic transmission onto layer 5 neurons were observed; however, layer 5 neurons demonstrated a decrease in input resistance and action potential duration after LFPI. These results demonstrate synaptic and intrinsic alterations in prefrontal circuitry that may underlie working memory impairment caused by TBI.
Collapse
Affiliation(s)
- Colin J. Smith
- Research Institute of Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Neuroscience Graduate Group, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Guoxiang Xiong
- Research Institute of Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jaclynn A. Elkind
- Research Institute of Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Brendan Putnam
- Research Institute of Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Akiva S. Cohen
- Research Institute of Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Anesthesiology and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
50
|
Gull S, Ingrisch I, Tausch S, Witte OW, Schmidt S. Consistent and reproducible staining of glia by a modified Golgi-Cox method. J Neurosci Methods 2015; 256:141-50. [PMID: 26365333 DOI: 10.1016/j.jneumeth.2015.08.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 08/24/2015] [Accepted: 08/26/2015] [Indexed: 10/23/2022]
Abstract
BACKGROUND Golgi-Cox staining is a powerful histochemical approach which has been used extensively to visualize the morphology of neurons and glia. However, its usage as a first-choice method is hindered by its uncertain nature, diminished consistency and lengthy staining duration. The FD Rapid GolgiStain™ Kit (FD Neurotechnologies, Inc., USA) has been developed by employing the Golgi-Cox approach. It is a simple, reliable and reproducible way of performing Golgi impregnation for the analysis of neuronal morphology. NEW METHOD We report here simple modifications to the manufacturer's protocol which enable reproducible and reliable staining of glial cells. RESULTS Exposure of brain tissue to 4% paraformaldehyde (PFA) during perfusion followed by postfixation with 8% glutaraldehyde in 4% PFA led to only glial cells being stained, whereas in the absence of postfixation both neurons and glia were stained with unclear morphology. Additionally, we found that impregnation at 26°C±1 was critical to attain uniform staining. COMPARISON WITH EXISTING METHOD Our modified Golgi-Cox approach is consistent and reproducible and affords uniform glial staining throughout the brain. CONCLUSION As this protocol stains only a small percentage of cells, it is suitable for the analysis of individual cells.
Collapse
Affiliation(s)
- S Gull
- Hans Berger Department of Neurology, Jena University Hospital, 07747 Jena, Germany
| | - I Ingrisch
- Hans Berger Department of Neurology, Jena University Hospital, 07747 Jena, Germany
| | - S Tausch
- Hans Berger Department of Neurology, Jena University Hospital, 07747 Jena, Germany
| | - O W Witte
- Hans Berger Department of Neurology, Jena University Hospital, 07747 Jena, Germany
| | - S Schmidt
- Hans Berger Department of Neurology, Jena University Hospital, 07747 Jena, Germany.
| |
Collapse
|