1
|
Beitchman JA, Chung JS, Jones JC, Hynan LS, Didehbani N, Cullum CM, Miller SM, Stokes M. Endophenotype presentation of athletes with concussion contingent on sex and time since injury. Brain Inj 2025:1-13. [PMID: 39787018 DOI: 10.1080/02699052.2025.2449934] [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: 09/24/2024] [Revised: 12/18/2024] [Accepted: 12/31/2024] [Indexed: 01/12/2025]
Abstract
INTRODUCTION Athletes with concussions experience heterogeneous symptoms and clinical trajectories. Subclassification provides diagnostic clarity that may improve prognostication and individualized treatments. METHODS We hypothesized that endophenotypes of adolescent athletes with concussions differ based on sex and time since injury. Post-concussive testing was performed for athletes (n = 1385) in the North Texas Concussion Registry (ConTex) at four timepoints: acute [0-3 days post-injury (DPI)], subacute-early (4-7DPI), subacute-late (8-28DPI), and persistent (29+DPI). Six endophenotypes (cognitive, headache, ocular-motor, vestibular, affective, sleep) were constructed by allocating post-concussion testing data elements described by the Concussion Subtype Workgroup. RESULTS Endophenotypes were defined using correlations between data elements and compared based on sex or time since injury. Correlograms revealed endophenotypes differed based on sex and time since injury. The affective endophenotype was dependent on the interaction between sex and time since injury and was more prevalent at the subacute-late and persistent timepoints. The sleep endophenotype became more prevalent at the persistent timepoint. Affective and sleep endophenotypes were interrelated with cognitive, vestibular, and headache endophenotypes at the persistent timepoint suggesting that dysregulated mood and sleep influence lingering symptoms. CONCLUSIONS Adolescent symptom-based concussion endophenotypes differ based on sex and time since injury. Clinical consideration may improve identification of separate trajectories following sport-related concussion and provide targeted care.
Collapse
Affiliation(s)
- Joshua A Beitchman
- Pediatric Neurology Residency Program, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jane S Chung
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Sports Medicine, Texas Scottish Rite Hospital for Children, Dallas, Texas, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jacob C Jones
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Sports Medicine, Texas Scottish Rite Hospital for Children, Dallas, Texas, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Linda S Hynan
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Nyaz Didehbani
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Physical Medicine and Rehabilitation, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - C Munro Cullum
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Shane M Miller
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Sports Medicine, Texas Scottish Rite Hospital for Children, Dallas, Texas, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Mathew Stokes
- Department of Sports Medicine, Texas Scottish Rite Hospital for Children, Dallas, Texas, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
2
|
Sabetta Z, Krishna G, Curry-Koski T, Lopez M, Adelson PD, Thomas TC. Sex-dependent temporal changes in astrocyte-vessel interactions following diffuse traumatic brain injury in rats. Front Physiol 2024; 15:1469073. [PMID: 39387100 PMCID: PMC11461938 DOI: 10.3389/fphys.2024.1469073] [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: 07/23/2024] [Accepted: 09/11/2024] [Indexed: 10/12/2024] Open
Abstract
Traumatic brain injury (TBI) is associated with diffuse axonal injury (DAI), a primary pathology linked to progressive neurodegeneration and neuroinflammation, including chronic astrogliosis, which influences long-term post-TBI recovery and morbidity. Sex-based differences in blood-brain barrier (BBB) permeability increases the risk of accelerated brain aging and early-onset neurodegeneration. However, few studies have evaluated chronic time course of astrocytic responses around cerebrovascular in the context of aging after TBI and sex dependence. We observed increased glial fibrillary acidic protein (GFAP)-labeled accessory processes branching near and connecting with GFAP-ensheathed cortical vessels, suggesting a critical nuance in astrocyte-vessel interactions after TBI. To quantify this observation, male and female Sprague Dawley rats (∼3 months old, n = 5-6/group) underwent either sham surgery or midline fluid percussion injury. Using immunohistochemical analysis, we quantified GFAP-labeled astrocyte primary and accessory processes that contacted GFAP-ensheathed vessels in the somatosensory barrel cortex at 7, 56, and 168 days post-injury (DPI). TBI significantly increased GFAP-positive primary processes at 7 DPI (P < 0.01) in both sexes. At 56 DPI, these vessel-process interactions remained significantly increased exclusively in males (P < 0.05). At 168 DPI, both sexes showed a significant reduction in vessel-process interactions compared to 7 DPI (P < 0.05); however, a modest but significant injury effect reemerged in females (P < 0.05). A similar sex-dependent pattern in the number of accessory processes provides novel evidence of long-term temporal changes in astrocyte-vessel interactions. TBI-induced changes in astrocyte-vessel interactions may indicate chronic BBB vulnerability and processes responsible for early onset vascular and neurodegenerative pathology.
Collapse
Affiliation(s)
- Zackary Sabetta
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
- A.T. Still University Kirksville College of Osteopathic Medicine, Kirksville, MO, United States
| | - Gokul Krishna
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
- Translational Neurotrauma and Neurochemistry, Barrow Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ, United States
| | - Tala Curry-Koski
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
- Translational Neurotrauma and Neurochemistry, Barrow Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ, United States
| | - Mackenzie Lopez
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
| | - P. David Adelson
- West Virginia University School of Medicine, Rockefeller Neuroscience Institute, Morgantown, WV, United States
| | - Theresa Currier Thomas
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
- Translational Neurotrauma and Neurochemistry, Barrow Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ, United States
- Phoenix VA Healthcare System, Phoenix, AZ, United States
| |
Collapse
|
3
|
Balleste AF, Sangadi A, Titus DJ, Johnstone T, Hogenkamp D, Gee KW, Atkins CM. Enhancing cognitive recovery in chronic traumatic brain injury through simultaneous allosteric modulation of α7 nicotinic acetylcholine and α5 GABA A receptors. Exp Neurol 2024; 379:114879. [PMID: 38942266 PMCID: PMC11283977 DOI: 10.1016/j.expneurol.2024.114879] [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: 03/11/2024] [Revised: 05/20/2024] [Accepted: 06/25/2024] [Indexed: 06/30/2024]
Abstract
Traumatic brain injury (TBI) leads to changes in the neural circuitry of the hippocampus that result in chronic learning and memory deficits. However, effective therapeutic strategies to ameliorate these chronic learning and memory impairments after TBI are limited. Two pharmacological targets for enhancing cognition are nicotinic acetylcholine receptors (nAChRs) and GABAA receptors (GABAARs), both of which regulate hippocampal network activity to form declarative memories. A promising compound, 522-054, both allosterically enhances α7 nAChRs and inhibits α5 subunit-containing GABAARs. Administration of 522-054 enhances long-term potentiation (LTP) and cognitive functioning in non-injured animals. In this study, we assessed the effects of 522-054 on hippocampal synaptic plasticity and learning and memory deficits in the chronic post-TBI recovery period. Adult male Sprague Dawley rats received moderate parasagittal fluid-percussion brain injury or sham surgery. At 12 wk after injury, we assessed basal synaptic transmission and LTP at the Schaffer collateral-CA1 synapse of the hippocampus. Bath application of 522-054 to hippocampal slices reduced deficits in basal synaptic transmission and recovered TBI-induced impairments in LTP. Moreover, treatment of animals with 522-054 at 12 wk post-TBI improved cue and contextual fear memory and water maze acquisition and retention without a measurable effect on cortical or hippocampal atrophy. These results suggest that dual allosteric modulation of α7 nAChR and α5 GABAAR signaling may be a potential therapy for treating cognitive deficits during chronic recovery from TBI.
Collapse
Affiliation(s)
- Alyssa F Balleste
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Akhila Sangadi
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - David J Titus
- Department of Psychiatry & Behavioral Sciences, University of Minnesota, Minneapolis, MN, USA
| | | | - Derk Hogenkamp
- Department of Pharmaceutical Sciences, University of California Irvine, Irvine, USA
| | - Kelvin W Gee
- Department of Pharmaceutical Sciences, University of California Irvine, Irvine, USA
| | - Coleen M Atkins
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
4
|
Fadaie F, Caldairou B, Gill RS, Foit NA, Hall JA, Bernhardt BC, Bernasconi N, Bernasconi A. Region-specific MRI predictors of surgical outcome in temporal lobe epilepsy. Neuroimage Clin 2024; 43:103658. [PMID: 39178601 PMCID: PMC11388716 DOI: 10.1016/j.nicl.2024.103658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/18/2024] [Accepted: 08/19/2024] [Indexed: 08/26/2024]
Abstract
OBJECTIVE In drug-resistant temporal lobe epilepsy (TLE), it is not well-established in how far surgery should target morphological anomalies to achieve seizure freedom. Here, we assessed interactions between structural brain compromise and surgery to identify region-specific predictors of seizure outcome. METHODS We obtained pre- and post-operative 3D T1-weighted MRI in 55 TLE patients who underwent selective amygdalo-hippocampectomy (SAH) or anterior temporal lobectomy (ATL) and 40 age and sex-matched healthy subjects. We measured surface-based morphological alterations of the mesiotemporal lobe structures (hippocampus, amygdala, entorhinal and piriform cortices), the neocortex and the thalamus on both pre- and post-operative MRI. Using precise co-registration, in each patient we mapped the surgical cavity onto the MRI acquired before surgery, thereby quantifying the amount of pathological tissue resected; these features, together with the preoperative morphometric data, served as input to a supervised classification algorithm for postsurgical outcome prediction. RESULTS On pre-operative MRI, patients who became seizure-free (TLE-SF) presented with severe ipsilateral amygdalar and hippocampal atrophy, while not seizure-free patients (TLE-NSF) displayed amygdalar hypertrophy. Stratifying patients based on the surgical approach, post-operative MRI showed similar patterns of mesiotemporal and thalamic changes, but divergent neocortical thinning affecting the parieto-temporo-occipital regions following ATL and the frontal lobes after SAH. Irrespective of the surgical approach, hippocampal atrophy on pre-operative MRI and its extent of resection were the most predictive features of seizure-freedom in 89% of patients (selected 100% across validations). SIGNIFICANCE Our study indicates a critical role of the extent of resection of MRI-derived hippocampal morphological anomalies on seizure outcome. Precise pre-operative quantification of the mesiotemporal lobe provides non-invasive prognostics for individualized surgery.
Collapse
Affiliation(s)
- Fatemeh Fadaie
- Neuroimaging of Epilepsy Laboratory, McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
| | - Benoit Caldairou
- Neuroimaging of Epilepsy Laboratory, McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
| | - Ravnoor S Gill
- Neuroimaging of Epilepsy Laboratory, McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
| | - Niels A Foit
- Neuroimaging of Epilepsy Laboratory, McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada; Freiburg Medical Center, Department of Neurosurgery, University of Freiburg, Freiburg, Germany
| | - Jeffery A Hall
- Department of Neurology and Neurosurgery, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
| | - Boris C Bernhardt
- Multimodal Imaging and Connectome Analysis Lab, McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
| | - Neda Bernasconi
- Neuroimaging of Epilepsy Laboratory, McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
| | - Andrea Bernasconi
- Neuroimaging of Epilepsy Laboratory, McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada.
| |
Collapse
|
5
|
Liu C, Guo Z, Pang J, Zhang Y, Yang Z, Cao J, Zhang T. Administration of Atosiban, an oxytocin receptor antagonist, ameliorates autistic-like behaviors in a female rat model of valproic acid-induced autism. Behav Brain Res 2024; 469:115052. [PMID: 38782096 DOI: 10.1016/j.bbr.2024.115052] [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: 12/24/2023] [Revised: 05/08/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024]
Abstract
Autism spectrum disorder (ASD) is a pervasive developmental disorder with gender differences. Oxytocin (OXT) is currently an important candidate drug for autism, but the lack of data on female autism is a big issue. It has been reported that the effect of OXT is likely to be different between male and female ASD patients. In the study, we specifically explored the role of the OXT signaling pathway in a VPA-induced female rat's model of autism. The data showed that there was an increase of either oxytocin or its receptor expressions in both the hippocampus and the prefrontal cortex of VPA-induced female offspring. To determine if the excess of OXT signaling contributed to autism symptoms in female rats, exogenous oxytocin and oxytocin receptor antagonists Atosiban were used in the experiment. It was found that exogenous oxytocin triggered autism-like behaviors in wild-type female rats by intranasal administration. More interestingly, several autism-like deficits including social interaction, anxiety, and repeat stereotypical sexual behavior in the VPA female offspring were significantly attenuated by oxytocin receptor antagonists Atosiban. Moreover, Atosiban also effectively improved the synaptic plasticity impairment induced by VPA in female offspring. Our results suggest that oxytocin receptor antagonists significantly improve autistic-like behaviors in a female rat model of valproic acid-induced autism.
Collapse
Affiliation(s)
- Chunhua Liu
- School of Medicine, Nankai University, Tianjin 300071, PR China
| | - Zhengyang Guo
- College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin 300071, PR China
| | - Jiyi Pang
- College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin 300071, PR China
| | - Yuying Zhang
- College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin 300071, PR China
| | - Zhuo Yang
- School of Medicine, Nankai University, Tianjin 300071, PR China
| | - Jianting Cao
- Graduate School of Engineering, Saitama Institute of Technology, Fukaya 369-0217, Japan
| | - Tao Zhang
- College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin 300071, PR China.
| |
Collapse
|
6
|
Wang S, Yu X, Cheng L, Ren W, Wen G, Wu X, Lou H, Ren X, Lu L, Hermenean A, Yao J, Li B, Lu Y, Wu X. Dexmedetomidine improves the circulatory dysfunction of the glymphatic system induced by sevoflurane through the PI3K/AKT/ΔFosB/AQP4 pathway in young mice. Cell Death Dis 2024; 15:448. [PMID: 38918408 PMCID: PMC11199640 DOI: 10.1038/s41419-024-06845-w] [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: 01/18/2024] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 06/27/2024]
Abstract
Multiple sevoflurane exposures may damage the developing brain. The neuroprotective function of dexmedetomidine has been widely confirmed in animal experiments and human studies. However, the effect of dexmedetomidine on the glymphatic system has not been clearly studied. We hypothesized that dexmedetomidine could alleviate sevoflurane-induced circulatory dysfunction of the glymphatic system in young mice. Six-day-old C57BL/6 mice were exposed to 3% sevoflurane for 2 h daily, continuously for 3 days. Intraperitoneal injection of either normal saline or dexmedetomidine was administered before every anaesthesia. Meanwhile the circulatory function of glymphatic system was detected by tracer injection at P8 and P32. On P30-P32, behavior tests including open field test, novel object recognition test, and Y-maze test were conducted. Primary astrocyte cultures were established and treated with the PI3K activator 740Y-P, dexmedetomidine, and small interfering RNA (siRNA) to silence ΔFosB. We propose for the first time that multiple exposure to sevoflurane induces circulatory dysfunction of the glymphatic system in young mice. Dexmedetomidine improves the circulatory capacity of the glymphatic system in young mice following repeated exposure to sevoflurane through the PI3K/AKT/ΔFosB/AQP4 signaling pathway, and enhances their long-term learning and working memory abilities.
Collapse
Affiliation(s)
- Shuying Wang
- School of Forensic Medicine, China Medical University, Shenyang, China
- Department of Anaesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Xiaojin Yu
- School of Forensic Medicine, China Medical University, Shenyang, China
- Department of Anaesthesiology, Affiliated Shengjing Hospital of China Medical University, Shenyang, China
| | - Lili Cheng
- School of Forensic Medicine, China Medical University, Shenyang, China
- Department of Anaesthesiology, Affiliated Shengjing Hospital of China Medical University, Shenyang, China
| | - Weishu Ren
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Gehua Wen
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Xue Wu
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Haoyang Lou
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Xinghua Ren
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Lei Lu
- Department of pediatrics Neonatology, University of Chicago, Chicago, IL, 60615, USA
| | - Anca Hermenean
- Faculty of Medicine, Vasile Goldis Western University of Arad, Arad, Romania
| | - Jun Yao
- School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China
- China Medical University Center of Forensic Investigation, Shenyang, China
| | - Baoman Li
- School of Forensic Medicine, China Medical University, Shenyang, China.
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China.
- China Medical University Center of Forensic Investigation, Shenyang, China.
| | - Yan Lu
- Key Laboratory of Health Ministry in Congenital Malformation, Affiliated Shengjing Hospital of China Medical University, Shenyang, China.
| | - Xu Wu
- School of Forensic Medicine, China Medical University, Shenyang, China.
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China.
- China Medical University Center of Forensic Investigation, Shenyang, China.
| |
Collapse
|
7
|
Ortiz JB, Tellez S, Rampal G, Mannino GS, Couillard N, Mendez M, Green TRF, Murphy SM, Rowe RK. Diffuse traumatic brain injury substantially alters plasma growth hormone in the juvenile rat. J Endocrinol 2024; 260:e230157. [PMID: 37855319 PMCID: PMC10692649 DOI: 10.1530/joe-23-0157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 10/18/2023] [Indexed: 10/20/2023]
Abstract
Traumatic brain injury (TBI) can damage the hypothalamus and cause improper activation of the growth hormone (GH) axis, leading to growth hormone deficiency (GHD). GHD is one of the most prevalent endocrinopathies following TBI in adults; however, the extent to which GHD affects juveniles remains understudied. We used postnatal day 17 rats (n = 83), which model the late infantile/toddler period, and assessed body weights, GH levels, and number of hypothalamic somatostatin neurons at acute (1, 7 days post injury (DPI)) and chronic (18, 25, 43 DPI) time points. We hypothesized that diffuse TBI would alter circulating GH levels because of damage to the hypothalamus, specifically somatostatin neurons. Data were analyzed with generalized linear and mixed effects models with fixed effects interactions between the injury and time. Despite similar growth rates over time with age, TBI rats weighed less than shams at 18 DPI (postnatal day 35; P = 0.03, standardized effect size [d] = 1.24), which is around the onset of puberty. Compared to shams, GH levels were lower in the TBI group during the acute period (P = 0.196; d = 12.3) but higher in the TBI group during the chronic period (P = 0.10; d = 52.1). Although not statistically significant, TBI-induced differences in GH had large standardized effect sizes, indicating biological significance. The mean number of hypothalamic somatostatin neurons (an inhibitor of GH) positively predicted GH levels in the hypothalamus but did not predict GH levels in the somatosensory cortex. Understanding TBI-induced alterations in the GH axis may identify therapeutic targets to improve the quality of life of pediatric survivors of TBI.
Collapse
Affiliation(s)
- J Bryce Ortiz
- Barrow Neurological Institute at Phoenix Children’s Hospital, Phoenix, Arizona, USA
- Department of Child Health, University of Arizona College of Medicine, Phoenix, Arizona, USA
| | - Sebastian Tellez
- Arizona State University, School of Life Sciences, Tempe, Arizona, USA
| | - Giri Rampal
- Department of Child Health, University of Arizona College of Medicine, Phoenix, Arizona, USA
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Grant S Mannino
- Department of Integrative Physiology, University of Colorado, Boulder, Colorado, USA
| | - Nicole Couillard
- Department of Integrative Physiology, University of Colorado, Boulder, Colorado, USA
| | - Matias Mendez
- Department of Integrative Physiology, University of Colorado, Boulder, Colorado, USA
| | - Tabitha R F Green
- Department of Integrative Physiology, University of Colorado, Boulder, Colorado, USA
| | - Sean M Murphy
- Department of Integrative Physiology, University of Colorado, Boulder, Colorado, USA
| | - Rachel K Rowe
- Department of Integrative Physiology, University of Colorado, Boulder, Colorado, USA
| |
Collapse
|
8
|
Peay DN, Acuna A, Reynolds CM, Willis C, Takalkar R, Bryce Ortiz J, Conrad CD. Chronic stress leads to persistent and contrasting stellate neuron dendritic hypertrophy in the amygdala of male and female rats, an effect not found in the hippocampus. Neurosci Lett 2023; 812:137403. [PMID: 37473795 DOI: 10.1016/j.neulet.2023.137403] [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: 04/07/2023] [Revised: 07/02/2023] [Accepted: 07/17/2023] [Indexed: 07/22/2023]
Abstract
In males, chronic stress enhances dendritic complexity in the amygdala, a region important in emotion regulation. An amygdalar subregion, the basolateral amygdala (BLA), is influenced by the hippocampus and prefrontal cortex to coordinate emotional learning and memory. This study quantified changes in dendritic complexity of BLA stellate neurons ten days after an unpredictable chronic stressor ended in both male and female rats. In addition, dendritic complexity of hippocampal neurons in male rats was assessed at a similar timepoint. Following Golgi processing, stressed male and female rats showed enhanced BLA dendritic complexity; increased arborization occurred near the soma in males and distally in females. As the brain was sampled ten days after chronic stress ended, BLA dendritic hypertrophy persisted in both sexes after the stressor had ended. For the hippocampus, CA3 dendritic complexity was similar for control and stressed males when assessed eight days after stress ended, suggesting that any stress-induced changes had resolved. These results show persistent enhancement of BLA dendritic arborization in both sexes following chronic stress, reveal sex differences in how BLA hypertrophy manifests, and suggest a putative neurobiological substrate by which chronic stress may create a vulnerable phenotype for emotional dysfunction.
Collapse
Affiliation(s)
- Dylan N Peay
- Department of Psychology, Arizona State University, Tempe, AZ, 85287-1104, United States
| | - Amanda Acuna
- Department of Psychology, Arizona State University, Tempe, AZ, 85287-1104, United States
| | - Cindy M Reynolds
- Department of Psychology, Arizona State University, Tempe, AZ, 85287-1104, United States
| | - Chris Willis
- Department of Psychology, Arizona State University, Tempe, AZ, 85287-1104, United States
| | - Rujuta Takalkar
- Department of Psychology, Arizona State University, Tempe, AZ, 85287-1104, United States
| | - J Bryce Ortiz
- Department of Psychology, Arizona State University, Tempe, AZ, 85287-1104, United States
| | - Cheryl D Conrad
- Department of Psychology, Arizona State University, Tempe, AZ, 85287-1104, United States.
| |
Collapse
|
9
|
Babb JA, Zuberer A, Heinrichs S, Rumbika KK, Alfiler L, Lakis GA, Leite-Morris KA, Kaplan GB. Disturbances in fear extinction learning after mild traumatic brain injury in mice are accompanied by alterations in dendritic plasticity in the medial prefrontal cortex and basolateral nucleus of the amygdala. Brain Res Bull 2023; 198:15-26. [PMID: 37031792 DOI: 10.1016/j.brainresbull.2023.04.001] [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: 01/10/2023] [Revised: 03/25/2023] [Accepted: 04/06/2023] [Indexed: 04/11/2023]
Abstract
Mild traumatic brain injury (mTBI) and post-traumatic stress disorder (PTSD) have emerged as the signature injuries of the U.S. veterans who served in Iraq and Afghanistan, and frequently co-occur in both military and civilian and populations. To better understand how fear learning and underlying neural systems might be altered after mTBI we examined the acquisition of cued fear conditioning and its extinction along with brain morphology and dendritic plasticity in a mouse model of mTBI. To induce mTBI in adult male C57BL/6J mice, a lateral fluid percussive injury (LFP 1.7) was produced using a fluid pulse of 1.7 atmosphere force to the right parietal lobe. Behavior in LFP 1.7 mice was compared to behavior in mice from two separate control groups: mice subjected to craniotomy without LFP injury (Sham) and mice that did not undergo surgery (Unoperated). Following behavioral testing, neural endpoints (dendritic structural plasticity and neuronal volume) were assessed in the basolateral nucleus of the amygdala (BLA), which plays a critical sensory role in fear learning, and medial prefrontal cortex (mPFC), responsible for executive functions and inhibition of fear behaviors. No gross motor abnormalities or increased anxiety-like behaviors were observed in LFP or Sham mice after surgery compared to Unoperated mice. We found that all mice acquired fear behavior, assessed as conditioned freezing to auditory cue in a single session of 6 trials, and acquisition was similar across treatment groups. Using a linear mixed effects analysis, we showed that fear behavior decreased overall over 6 days of extinction training with no effect of treatment group across extinction days. However, a significant interaction was demonstrated between the treatment groups during within-session freezing behavior (5 trials per day) during extinction training. Specifically, freezing behavior increased across within-session extinction trials in LFP 1.7 mice, whereas freezing behavior in control groups did not change on extinction test days, reflecting a dissociation between within-trial and between-trial fear extinction. Additionally, LFP mice demonstrated bilateral increases in dendritic spine density in the BLA and decreases in dendritic complexity in the PFC. The translational implications are that individuals with TBI undergoing fear extinction therapy may demonstrate within-session aberrant learning that could be targeted for more effective treatment interventions.
Collapse
Affiliation(s)
- Jessica A Babb
- Research Service, VA Boston Healthcare System, West Roxbury, MA, 02132 USA; Mental Health Service, VA Boston Healthcare System, West Roxbury, MA, 02132 USA; Department of Psychiatry, Harvard Medical School, Boston, MA, 02115 USA.
| | - Agnieszka Zuberer
- Department of Psychiatry and Psychotherapy, University of Tübingen, 72076 Tübingen, Germany; Department of Psychiatry and Psychotherapy, Jena University Hospital, 07743 Jena, Germany.
| | - Stephen Heinrichs
- Research Service, VA Boston Healthcare System, West Roxbury, MA, 02132 USA.
| | - Kendra K Rumbika
- Research Service, VA Boston Healthcare System, West Roxbury, MA, 02132 USA.
| | - Lauren Alfiler
- Research Service, VA Boston Healthcare System, West Roxbury, MA, 02132 USA.
| | - Gabrielle A Lakis
- Research Service, VA Boston Healthcare System, West Roxbury, MA, 02132 USA; Boston University Chobanian & Avedisian School of Medicine, Boston, MA, 02218 USA.
| | - Kimberly A Leite-Morris
- Research Service, VA Boston Healthcare System, West Roxbury, MA, 02132 USA; Department of Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, 02118 USA.
| | - Gary B Kaplan
- Research Service, VA Boston Healthcare System, West Roxbury, MA, 02132 USA; Mental Health Service, VA Boston Healthcare System, West Roxbury, MA, 02132 USA; Department of Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, 02118 USA; Department of Pharmacology & Experimental Therapeutics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, 02118 USA.
| |
Collapse
|
10
|
Sabetta Z, Krishna G, Curry T, Adelson PD, Thomas TC. Aging with TBI vs. Aging: 6-month temporal profiles for neuropathology and astrocyte activation converge in behaviorally relevant thalamocortical circuitry of male and female rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.06.527058. [PMID: 36798182 PMCID: PMC9934568 DOI: 10.1101/2023.02.06.527058] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Traumatic brain injury (TBI) manifests late-onset and persisting clinical symptoms with implications for sex differences and increased risk for the development of age-related neurodegenerative diseases. Few studies have evaluated chronic temporal profiles of neuronal and glial pathology that include sex as a biological variable. After experimental diffuse TBI, late-onset and persisting somatosensory hypersensitivity to whisker stimulation develops at one-month post-injury and persists to at least two months post-injury in male rats, providing an in vivo model to evaluate the temporal profile of pathology responsible for morbidity. Whisker somatosensation is dependent on signaling through the thalamocortical relays of the whisker barrel circuit made up of glutamatergic projections between the ventral posteromedial nucleus of the thalamus (VPM) and primary somatosensory barrel cortex (S1BF) with inhibitory (GABA) innervation from the thalamic reticular nucleus (TRN) to the VPM. To evaluate the temporal profiles of pathology, male and female Sprague Dawley rats ( n = 5-6/group) were subjected to sham surgery or midline fluid percussion injury (FPI). At 7-, 56-, and 168-days post-injury (DPI), brains were processed for amino-cupric silver stain and glial fibrillary acidic protein (GFAP) immunoreactivity, where pixel density of staining was quantified to determine the temporal profile of neuropathology and astrocyte activation in the VPM, S1BF, and TRN. FPI induced significant neuropathology in all brain regions at 7 DPI. At 168 DPI, neuropathology remained significantly elevated in the VPM and TRN, but returned to sham levels in the S1BF. GFAP immunoreactivity was increased as a function of FPI and DPI, with an FPI × DPI interaction in all regions and an FPI × Sex interaction in the S1BF. The interactions were driven by increased GFAP immunoreactivity in shams over time in the VPM and TRN. In the S1BF, GFAP immunoreactivity increased at 7 DPI and declined to age-matched sham levels by 168 DPI, while GFAP immunoreactivity in shams significantly increased between 7 and 168 days. The FPI × Sex interaction was driven by an overall greater level of GFAP immunoreactivity in FPI males compared to FPI females. Increased GFAP immunoreactivity was associated with an increased number of GFAP-positive soma, predominantly at 7 DPI. Overall, these findings indicate that FPI, time post-injury, sex, region, and aging with injury differentially contribute to chronic changes in neuronal pathology and astrocyte activation after diffuse brain injury. Thus, our results highlight distinct patterns of pathological alterations associated with the development and persistence of morbidity that supports chronic neuropathology, especially within the thalamus. Further, data indicate a convergence between TBI-induced and age-related pathology where further investigation may reveal a role for divergent astrocytic phenotypes associated with increased risk for neurodegenerative diseases.
Collapse
|
11
|
Bickart KC, Olsen A, Dennis EL, Babikian T, Hoffman AN, Snyder A, Sheridan CA, Fischer JT, Giza CC, Choe MC, Asarnow RF. Frontoamygdala hyperconnectivity predicts affective dysregulation in adolescent moderate-severe TBI. FRONTIERS IN REHABILITATION SCIENCES 2023; 3:1064215. [PMID: 36684686 PMCID: PMC9845889 DOI: 10.3389/fresc.2022.1064215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 11/29/2022] [Indexed: 01/06/2023]
Abstract
In survivors of moderate to severe traumatic brain injury (msTBI), affective disruptions often remain underdetected and undertreated, in part due to poor understanding of the underlying neural mechanisms. We hypothesized that limbic circuits are integral to affective dysregulation in msTBI. To test this, we studied 19 adolescents with msTBI 17 months post-injury (TBI: M age 15.6, 5 females) as well as 44 matched healthy controls (HC: M age 16.4, 21 females). We leveraged two previously identified, large-scale resting-state (rsfMRI) networks of the amygdala to determine whether connectivity strength correlated with affective problems in the adolescents with msTBI. We found that distinct amygdala networks differentially predicted externalizing and internalizing behavioral problems in patients with msTBI. Specifically, patients with the highest medial amygdala connectivity were rated by parents as having greater externalizing behavioral problems measured on the BRIEF and CBCL, but not cognitive problems. The most correlated voxels in that network localize to the rostral anterior cingulate (rACC) and posterior cingulate (PCC) cortices, predicting 48% of the variance in externalizing problems. Alternatively, patients with the highest ventrolateral amygdala connectivity were rated by parents as having greater internalizing behavioral problems measured on the CBCL, but not cognitive problems. The most correlated voxels in that network localize to the ventromedial prefrontal cortex (vmPFC), predicting 57% of the variance in internalizing problems. Both findings were independent of potential confounds including ratings of TBI severity, time since injury, lesion burden based on acute imaging, demographic variables, and other non-amygdalar rsfMRI metrics (e.g., rACC to PCC connectivity), as well as macro- and microstructural measures of limbic circuitry (e.g., amygdala volume and uncinate fasciculus fractional anisotropy). Supporting the clinical significance of these findings, patients with msTBI had significantly greater externalizing problem ratings than healthy control participants and all the brain-behavior findings were specific to the msTBI group in that no similar correlations were found in the healthy control participants. Taken together, frontoamygdala pathways may underlie chronic dysregulation of behavior and mood in patients with msTBI. Future work will focus on neuromodulation techniques to directly affect frontoamygdala pathways with the aim to mitigate such dysregulation problems.
Collapse
Affiliation(s)
- Kevin C. Bickart
- BrainSPORT, Department of Neurosurgery, UCLA, Los Angeles, CA, United States,Department of Neurology, UCLA, Los Angeles, CA, United States,Correspondence: Kevin C. Bickart
| | - Alexander Olsen
- Department of Psychiatry and Biobehavioral Sciences, UCLA, Los Angeles, CA, United States,Department of Psychology, Norwegian University of Science and Technology, Trondheim, Norway,Department of Physical Medicine and Rehabilitation, St. Olavs Hospital, University Hospital, Trondheim, Norway
| | - Emily L. Dennis
- TBI and Concussion Center, Department of Neurology, University of Utah, Salt Lake City, UT, United States
| | - Talin Babikian
- BrainSPORT, Department of Neurosurgery, UCLA, Los Angeles, CA, United States,Department of Psychiatry and Biobehavioral Sciences, UCLA, Los Angeles, CA, United States
| | - Ann N. Hoffman
- BrainSPORT, Department of Neurosurgery, UCLA, Los Angeles, CA, United States
| | - Aliyah Snyder
- BrainSPORT, Department of Neurosurgery, UCLA, Los Angeles, CA, United States,Department of Psychiatry and Biobehavioral Sciences, UCLA, Los Angeles, CA, United States
| | - Christopher A. Sheridan
- Wake Forest School of Medicine, Radiology Informatics and Image Processing Laboratory, Winston-Salem, NC, United States,Wake Forest School of Medicine, Department of Radiology, Section of Neuroradiology, Winston-Salem, NC, United States
| | - Jesse T. Fischer
- BrainSPORT, Department of Neurosurgery, UCLA, Los Angeles, CA, United States,Department of Psychiatry and Biobehavioral Sciences, UCLA, Los Angeles, CA, United States
| | - Christopher C. Giza
- BrainSPORT, Department of Neurosurgery, UCLA, Los Angeles, CA, United States,UCLA Mattel Children's Hospital, Department of Pediatrics, Division of Neurology, Los Angeles, CA, United States
| | - Meeryo C. Choe
- BrainSPORT, Department of Neurosurgery, UCLA, Los Angeles, CA, United States,UCLA Mattel Children's Hospital, Department of Pediatrics, Division of Neurology, Los Angeles, CA, United States
| | - Robert F. Asarnow
- BrainSPORT, Department of Neurosurgery, UCLA, Los Angeles, CA, United States,Department of Psychiatry and Biobehavioral Sciences, UCLA, Los Angeles, CA, United States
| |
Collapse
|
12
|
Hoffman AN, Watson S, Chavda N, Lam J, Hovda DA, Giza CC, Fanselow MS. Increased Fear Generalization and Amygdala AMPA Receptor Proteins in Chronic Traumatic Brain Injury. J Neurotrauma 2022; 39:1561-1574. [PMID: 35722903 PMCID: PMC9689770 DOI: 10.1089/neu.2022.0119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Cognitive impairments and emotional lability are common long-term consequences of traumatic brain injury (TBI). How TBI affects interactions between sensory, cognitive, and emotional systems may reveal mechanisms that underlie chronic mental health comorbidities. Previously, we reported changes in auditory-emotional network activity and enhanced fear learning early after TBI. In the current study, we asked whether TBI has long-term effects on fear learning and responses to novel stimuli. Four weeks following lateral fluid percussion injury (FPI) or sham surgery, adult male rats were fear conditioned to either white noise-shock or tone-shock pairing, or shock-only control and subsequently were tested for freezing to context and to the trained or novel auditory cues in a new context. FPI groups showed greater freezing to their trained auditory cue, indicating long-term TBI enhanced fear. Interestingly, FPI-Noise Shock animals displayed robust fear to the novel, untrained tone compared with Sham-Noise Shock across both experiments. Shock Only groups did not differ in freezing to either auditory stimulus. These findings suggest that TBI precipitates maladaptive associative fear generalization rather than non-associative sensitization. Basolateral amygdala (BLA) α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAr) subunits GluA1 and GluA2 levels were analyzed and the FPI-Noise Shock group had increased GluA1 (but not GluA2) levels that correlated with the level of tone fear generalization. This study illustrates a unique chronic TBI phenotype with both a cognitive impairment and increased fear and possibly altered synaptic transmission in the amygdala long after TBI, where stimulus generalization may underlie maladaptive fear and hyperarousal.
Collapse
Affiliation(s)
- Ann N. Hoffman
- Department of Neurosurgery, Brain Injury Research Center, University of California, Los Angeles, Los Angeles, California, USA
- Department of Psychology, University of California, Los Angeles, Los Angeles, California, USA
- Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, California, USA
- Staglin Center for Brain and Behavioral Health, University of California, Los Angeles, Los Angeles, California, USA
| | - Sonya Watson
- Department of Neurosurgery, Brain Injury Research Center, University of California, Los Angeles, Los Angeles, California, USA
- Department of Psychology, University of California, Los Angeles, Los Angeles, California, USA
| | - Nishtha Chavda
- Department of Psychology, University of California, Los Angeles, Los Angeles, California, USA
| | - Jamie Lam
- Department of Psychology, University of California, Los Angeles, Los Angeles, California, USA
| | - David A. Hovda
- Department of Neurosurgery, Brain Injury Research Center, University of California, Los Angeles, Los Angeles, California, USA
- Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, California, USA
- Department of Medical and Molecular Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
| | - Christopher C. Giza
- Department of Neurosurgery, Brain Injury Research Center, University of California, Los Angeles, Los Angeles, California, USA
- Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, California, USA
- Mattel Children's Hospital, University of California, Los Angeles, Los Angeles, California, USA
| | - Michael S. Fanselow
- Department of Psychology, University of California, Los Angeles, Los Angeles, California, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, California, USA
- Staglin Center for Brain and Behavioral Health, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
13
|
Zhuang Y, Dong J, Ge Q, Zhang B, Yang M, Lu S, Li H, Niu F, Xu X, Liu B. Contralateral synaptic changes following severe unilateral brain injury. Brain Res Bull 2022; 188:21-29. [PMID: 35868500 DOI: 10.1016/j.brainresbull.2022.07.010] [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: 05/10/2022] [Revised: 07/15/2022] [Accepted: 07/17/2022] [Indexed: 11/02/2022]
Abstract
The brain is highly integrated and thus unilateral injury can impact the contralateral hemisphere. However, further research is needed to clarify the changes in the response of the contralateral homotopic area to ipsilateral injury. We hypothesized that severe unilateral brain injury would be accompanied by contralateral synaptic changes that are related to functional recovery. To test this, we divided rats into sham and experimental groups. In the experimental group, we performed right motor cortex resection. These rats were further divided into three subgroups according to post-injury time: 7 days, 14 days, and 30 days post-injury. Rats in each group were evaluated using a beam walking test to quantify the recovery of motor function, and all rats received an injection of adeno-associated virus-containing green fluorescent protein (GFP). Finally, we conducted morphological and histological analyses to identify synaptic changes. Over time, the behavior of the rats that underwent right motor cortex resection recovered. Furthermore, in contrast to the sham group, the experimental groups exhibited an increase in the spine density and expression of synaptic proteins in layer V of the contralateral motor cortex, which was consistent with the GFP-labeled neurons. Moreover, more immature spines were observed 7 days post-injury. Notably, spine morphology matured from 7 to 30 days, and the increase in Synapsin-1 intensity in layer V peaked 14 days after the resection, whereas PSD-95 intensity continued to increase until day 30. Our findings suggested that following motor function recovery from unilateral brain injury, spine morphology and synaptic proteins change dynamically in the contralateral hemisphere.
Collapse
Affiliation(s)
- Yuan Zhuang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jinqian Dong
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Qianqian Ge
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Bin Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Mengshi Yang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shenghua Lu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Hao Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Fei Niu
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Xiaojian Xu
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
| | - Baiyun Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China; Center for Nerve Injury and Repair, Beijing Institute of Brain Disorders, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing, China.
| |
Collapse
|
14
|
Braga MFM, Juranek J, Eiden LE, Li Z, Figueiredo TH, de Araujo Furtado M, Marini AM. GABAergic circuits of the basolateral amygdala and generation of anxiety after traumatic brain injury. Amino Acids 2022; 54:1229-1249. [PMID: 35798984 DOI: 10.1007/s00726-022-03184-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/20/2022] [Indexed: 11/25/2022]
Abstract
Traumatic brain injury (TBI) has reached epidemic proportions around the world and is a major public health concern in the United States. Approximately 2.8 million individuals sustain a traumatic brain injury and are treated in an Emergency Department yearly in the U.S., and about 50,000 of them die. Persistent symptoms develop in 10-15% of the cases including neuropsychiatric disorders. Anxiety is the second most common neuropsychiatric disorder that develops in those with persistent neuropsychiatric symptoms after TBI. Abnormalities or atrophy in the temporal lobe has been shown in the overwhelming number of TBI cases. The basolateral amygdala (BLA), a temporal lobe structure that consolidates, stores and generates fear and anxiety-based behavioral outputs, is a critical brain region in the anxiety circuitry. In this review, we sought to capture studies that characterized the relationship between human post-traumatic anxiety and structural/functional alterations in the amygdala. We compared the human findings with results obtained with a reproducible mild TBI animal model that demonstrated a direct relationship between the alterations in the BLA and an anxiety-like phenotype. From this analysis, both preliminary insights, and gaps in knowledge, have emerged which may open new directions for the development of rational and more efficacious treatments.
Collapse
Affiliation(s)
- Maria F M Braga
- Department of Anatomy, Physiology and Genetics and Program in Neuroscience, Uniformed Services University of the Health Science School of Medicine, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Jenifer Juranek
- Department of Pediatric Surgery, McGovern Medical School at the University of Texas Health Science Center at Houston (UTHealth), Houston, TX, 77030, USA
| | - Lee E Eiden
- Section On Molecular Neuroscience, National Institute of Mental Health, Intramural Research Program, Bethesda, MD, 20814, USA
| | - Zheng Li
- Section On Synapse Development and Plasticity, National Institute of Mental Health, Intramural Research Program, Bethesda, MD, 20814, USA
| | - Taiza H Figueiredo
- Department of Anatomy, Physiology and Genetics and Program in Neuroscience, Uniformed Services University of the Health Science School of Medicine, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Marcio de Araujo Furtado
- Department of Anatomy, Physiology and Genetics and Program in Neuroscience, Uniformed Services University of the Health Science School of Medicine, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Ann M Marini
- Department of Neurology and Program in Neuroscience, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| |
Collapse
|
15
|
Weil ZM, White B, Whitehead B, Karelina K. The role of the stress system in recovery after traumatic brain injury: A tribute to Bruce S. McEwen. Neurobiol Stress 2022; 19:100467. [PMID: 35720260 PMCID: PMC9201063 DOI: 10.1016/j.ynstr.2022.100467] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 05/25/2022] [Accepted: 06/02/2022] [Indexed: 11/21/2022] Open
Abstract
Traumatic brain injury (TBI) represents a major public health concern. Although the majority of individuals that suffer mild-moderate TBI recover relatively quickly, a substantial subset of individuals experiences prolonged and debilitating symptoms. An exacerbated response to physiological and psychological stressors after TBI may mediate poor functional recovery. Individuals with TBI can suffer from poor stress tolerance, impairments in the ability to evaluate stressors, and poor initiation (and cessation) of neuroendocrine stress responses, all of which can exacerbate TBI-mediated dysfunction. Here, we pay tribute to the pioneering neuroendocrinologist Dr. Bruce McEwen by discussing the ways in which his work on stress physiology and allostatic loading impacts the TBI patient population both before and after their injuries. Specifically, we will discuss the modulatory role of hypothalamic-pituitary-adrenal axis responses immediately after TBI and later in recovery. We will also consider the impact of stressors and stress responses in promoting post-concussive syndrome and post-traumatic stress disorders, two common sequelae of TBI. Finally, we will explore the role of early life stressors, prior to brain injuries, as modulators of injury outcomes.
Collapse
Affiliation(s)
- Zachary M. Weil
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, 108 Biomedical Rd, Morgantown, WV, 26506, USA
| | - Brishti White
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, 108 Biomedical Rd, Morgantown, WV, 26506, USA
| | - Bailey Whitehead
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, 108 Biomedical Rd, Morgantown, WV, 26506, USA
| | - Kate Karelina
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, 108 Biomedical Rd, Morgantown, WV, 26506, USA
| |
Collapse
|
16
|
Giordano KR, Law LM, Henderson J, Rowe RK, Lifshitz J. Time Course of Remote Neuropathology Following Diffuse Traumatic Brain Injury in the Male Rat. Exp Neurobiol 2022; 31:105-115. [PMID: 35673999 PMCID: PMC9194637 DOI: 10.5607/en21027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 12/15/2021] [Accepted: 04/12/2022] [Indexed: 11/19/2022] Open
Abstract
Traumatic brain injury (TBI) can affect different regions throughout the brain. Regions near the site of impact are the most vulnerable to injury. However, damage to distal regions occurs. We investigated progressive neuropathology in the dorsal hippocampus (near the impact) and cerebellum (distal to the impact) after diffuse TBI. Adult male rats were subjected to midline fluid percussion injury or sham injury. Brain tissue was stained by the amino cupric silver stain. Neuropathology was quantified in sub-regions of the dorsal hippocampus at 1, 7, and 28 days post-injury (DPI) and coronal cerebellar sections at 1, 2, and 7 DPI. The highest observed neuropathology in the dentate gyrus occurred at 7 DPI which attenuated by 28 DPI, whereas the highest observed neuropathology was at 1 DPI in the CA3 region. There was no significant neuropathology in the CA1 region at any time point. Neuropathology was increased at 7 DPI in the cerebellum compared to shams and stripes of pathology were observed in the molecular layer perpendicular to the cerebellar cortical surface. Together these data show that diffuse TBI can result in neuropathology across the brain. By describing the time course of pathology in response to TBI, it is possible to build the temporal profile of disease progression.
Collapse
Affiliation(s)
- Katherine R Giordano
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ 85013, USA.,Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ 85004, USA.,Phoenix Veterans Affairs Health Care System, Phoenix, AZ 85012, USA
| | - L Matthew Law
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ 85013, USA.,Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ 85004, USA.,Phoenix Veterans Affairs Health Care System, Phoenix, AZ 85012, USA
| | - Jordan Henderson
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ 85013, USA.,Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ 85004, USA
| | - Rachel K Rowe
- Department of Integrative Physiology, University of Colorado, Boulder, CO 80309, USA
| | - Jonathan Lifshitz
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ 85013, USA.,Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ 85004, USA.,Phoenix Veterans Affairs Health Care System, Phoenix, AZ 85012, USA
| |
Collapse
|
17
|
Jacotte-Simancas A, Middleton JW, Stielper ZF, Edwards S, Molina PE, Gilpin NW. Brain Injury Effects on Neuronal Activation and Synaptic Transmission in the Basolateral Amygdala of Adult Male and Female Wistar Rats. J Neurotrauma 2022; 39:544-559. [PMID: 35081744 PMCID: PMC8978566 DOI: 10.1089/neu.2021.0270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Traumatic brain injury (TBI) is defined as brain damage produced by an external mechanical force that leads to behavioral, cognitive, and psychiatric sequelae. The basolateral amygdala (BLA) is involved in emotional regulation, and its function and morphology are altered following TBI. Little is known about potential sex-specific effects of TBI on BLA neuronal function, but it is critical for the field to identify potential sex differences in TBI effects on brain and behavior. Here, we hypothesized that TBI would produce sex-specific acute (1 h) effects on BLA neuronal activation, excitability, and synaptic transmission in adult male and female rats. Forty-nine Wistar rats (n = 23 males and 26 females) were randomized to TBI (using lateral fluid percussion) or Sham groups in two separate studies. Study 1 used in situ hybridization (i.e., RNAscope) to measure BLA expression of c-fos (a marker of cell activation), vGlut, and vGat (markers of glutamatergic and GABAergic neurons, respectively) messenger RNA (mRNA). Study 2 used slice electrophysiology to measure intrinsic excitability and excitatory/inhibitory synaptic transmission in putative pyramidal neurons in the BLA. Physiological measures of injury severity were collected from all animals. Our results show that females exhibit increased apnea duration and reduced respiratory rate post-TBI relative to males. In male and female rats, TBI increased c-fos expression in BLA glutamatergic cells but not in BLA GABAergic cells, and TBI increased firing rate in BLA pyramidal neurons. Further, TBI increased spontaneous excitatory and inhibitory postsynaptic current (sEPSC and sIPSC) amplitude in BLA neurons of females relative to all other groups. TBI increased sEPSC frequency in BLA neurons of females relative to males but did not alter sIPSC frequency. In summary, lateral fluid percussion produced different physiological responses in male and female rats, as well as sex-specific alterations in BLA neuronal activation, excitability, and synaptic transmission 1 h after injury.
Collapse
Affiliation(s)
- Alejandra Jacotte-Simancas
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
- Alcohol and Drug of Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Jason W. Middleton
- Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Zachary F. Stielper
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Scott Edwards
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
- Alcohol and Drug of Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Patricia E. Molina
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
- Alcohol and Drug of Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Nicholas W. Gilpin
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
- Alcohol and Drug of Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
- Southeast Louisiana VA Healthcare System, New Orleans, Louisiana, USA
| |
Collapse
|
18
|
Li B, Zhang D, Verkhratsky A. Astrocytes in Post-traumatic Stress Disorder. Neurosci Bull 2022; 38:953-965. [PMID: 35349095 PMCID: PMC8960712 DOI: 10.1007/s12264-022-00845-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 01/07/2022] [Indexed: 01/15/2023] Open
Abstract
Although posttraumatic stress disorder (PTSD) is on the rise, traumatic events and their consequences are often hidden or minimized by patients for reasons linked to PTSD itself. Traumatic experiences can be broadly classified into mental stress (MS) and traumatic brain injury (TBI), but the cellular mechanisms of MS- or TBI-induced PTSD remain unknown. Recent evidence has shown that the morphological remodeling of astrocytes accompanies and arguably contributes to fearful memories and stress-related disorders. In this review, we summarize the roles of astrocytes in the pathogenesis of MS-PTSD and TBI-PTSD. Astrocytes synthesize and secrete neurotrophic, pro- and anti-inflammatory factors and regulate the microenvironment of the nervous tissue through metabolic pathways, ionostatic control, and homeostatic clearance of neurotransmitters. Stress or trauma-associated impairment of these vital astrocytic functions contribute to the pathophysiological evolution of PTSD and may present therapeutic targets.
Collapse
Affiliation(s)
- Baoman Li
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, 110122, China
| | - Dianjun Zhang
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, 110122, China
| | - Alexei Verkhratsky
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, 110122, China.
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK.
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, 01102, Vilnius, Lithuania.
| |
Collapse
|
19
|
Shi Y, Wu X, Zhou J, Cui W, Wang J, Hu Q, Zhang S, Han L, Zhou M, Luo J, Wang Q, Liu H, Feng D, Ge S, Qu Y. Single-Nucleus RNA Sequencing Reveals that Decorin Expression in the Amygdala Regulates Perineuronal Nets Expression and Fear Conditioning Response after Traumatic Brain Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104112. [PMID: 35038242 PMCID: PMC8895134 DOI: 10.1002/advs.202104112] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/17/2021] [Indexed: 06/14/2023]
Abstract
Traumatic brain injury (TBI) is a risk factor for posttraumatic stress disorder (PTSD). Augmented fear is a defining characteristic of PTSD, and the amygdala is considered the main brain region to process fear. The mechanism by which the amygdala is involved in fear conditioning after TBI is still unclear. Using single-nucleus RNA sequencing (snRNA-seq), transcriptional changes in cells in the amygdala after TBI are investigated. In total, 72 328 nuclei are obtained from the sham and TBI groups. 7 cell types, and analysis of differentially expressed genes (DEGs) reveals widespread transcriptional changes in each cell type after TBI are identified. In in vivo experiments, it is demonstrated that Decorin (Dcn) expression in the excitatory neurons of the amygdala significantly increased after TBI, and Dcn knockout in the amygdala mitigates TBI-associated fear conditioning. Of note, this effect is caused by a Dcn-mediated decrease in the expression of perineuronal nets (PNNs), which affect the glutamate-γ-aminobutyric acid balance in the amygdala. Finally, the results suggest that Dcn functions by interacting with collagen VI α3 (Col6a3). Consequently, the findings reveal transcriptional changes in different cell types of the amygdala after TBI and provide direct evidence that Dcn relieves fear conditioning by regulating PNNs.
Collapse
Affiliation(s)
- Yingwu Shi
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Xun Wu
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Jinpeng Zhou
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Wenxing Cui
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Jin Wang
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Qing Hu
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Shenghao Zhang
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Liying Han
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Meixuan Zhou
- School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200240China
| | - Jianing Luo
- Department of NeurosurgeryWest Theater General HospitalChengduSichuan610083China
| | - Qiang Wang
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Haixiao Liu
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Dayun Feng
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Shunnan Ge
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Yan Qu
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| |
Collapse
|
20
|
Chronic Administration of 7,8-DHF Lessens the Depression-like Behavior of Juvenile Mild Traumatic Brain Injury Treated Rats at Their Adult Age. Pharmaceutics 2021; 13:pharmaceutics13122169. [PMID: 34959450 PMCID: PMC8704538 DOI: 10.3390/pharmaceutics13122169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/11/2021] [Accepted: 12/10/2021] [Indexed: 01/12/2023] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of mortality and morbidity among the global youth and commonly results in long-lasting sequelae, including paralysis, epilepsy, and a host of mental disorders such as major depressive disorder. Previous studies were mainly focused on severe TBI as it occurs in adults. This study explored the long-term adverse effect of mild TBI in juvenile animals (mTBI-J). Male Sprague Dawley rats received mTBI-J or sham treatment at six weeks old, then underwent behavioral, biochemical, and histological experiments three weeks later (at nine weeks old). TTC staining, H&E staining, and brain edema measurement were applied to evaluate the mTBI-J induced cerebral damage. The forced swimming test (FST) and sucrose preference test (SPT) were applied for measuring depression-like behavior. The locomotor activity test (LAT) was performed to examine mTBI-J treatment effects on motor function. After the behavioral experiments, the dorsal hippocampus (dHip) and ventral hippocampus (vHip) were dissected out for western blotting to examine the expression of brain-derived neurotrophic factor (BDNF) and tropomyosin receptor kinase B (TrkB). Finally, a TrkB agonist 7,8-DHF was injected intraperitoneally to evaluate its therapeutic effect on the mTBI-J induced behavioral abnormalities at the early adult age. Results showed that a mild brain edema occurred, but no significant neural damage was found in the mTBI-J treated animals. In addition, a significant increase of depression-like behaviors was observed in the mTBI-J treated animals; the FST revealed an increase in immobility, and a decrease in sucrose consumption was found in the mTBI-J treated animals. There were no differences observed in the total distance traveled of the LAT and the fall latency of the rotarod test. The hippocampal BDNF expression, but not the TrkB, were significantly reduced in mTBI-J, and the mTBI-J treatment-induced depression-like behavior was lessened after four weeks of 7,8-DHF administration. Collectively, these results indicate that even a mild juvenile TBI treatment that did not produce motor deficits or significant histological damage could have a long-term adverse effect that could be sustained to adulthood, which raises the depression-like behavior in the adult age. In addition, chronic administration of 7,8-DHF lessens the mTBI-J treatment-induced depression-like behaviors in adult rats. We suggest the potential usage of 7,8-DHF as a therapeutic agent for preventing the long-term adverse effect of mTBI-J.
Collapse
|
21
|
Buhlman LM, Krishna G, Jones TB, Thomas TC. Drosophila as a model to explore secondary injury cascades after traumatic brain injury. Biomed Pharmacother 2021; 142:112079. [PMID: 34463269 PMCID: PMC8458259 DOI: 10.1016/j.biopha.2021.112079] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/11/2021] [Accepted: 08/17/2021] [Indexed: 12/14/2022] Open
Abstract
Drosophilae are emerging as a valuable model to study traumatic brain injury (TBI)-induced secondary injury cascades that drive persisting neuroinflammation and neurodegenerative pathology that imposes significant risk for long-term neurological deficits. As in mammals, TBI in Drosophila triggers axonal injury, metabolic crisis, oxidative stress, and a robust innate immune response. Subsequent neurodegeneration stresses quality control systems and perpetuates an environment for neuroprotection, regeneration, and delayed cell death via highly conserved cell signaling pathways. Fly injury models continue to be developed and validated for both whole-body and head-specific injury to isolate, evaluate, and modulate these parallel pathways. In conjunction with powerful genetic tools, the ability for longitudinal evaluation, and associated neurological deficits that can be tested with established behavioral tasks, Drosophilae are an attractive model to explore secondary injury cascades and therapeutic intervention after TBI. Here, we review similarities and differences between mammalian and fly pathophysiology and highlight strategies for their use in translational neurotrauma research.
Collapse
Affiliation(s)
- Lori M Buhlman
- Biomedical Sciences Program, Midwestern University, Glendale, AZ, USA.
| | - Gokul Krishna
- Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA; Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA
| | - T Bucky Jones
- Department of Anatomy, Midwestern University, Glendale, AZ, USA
| | - Theresa Currier Thomas
- Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA; Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA; Phoenix VA Health Care System, Phoenix, AZ, USA.
| |
Collapse
|
22
|
Hoffman AN, Watson S, Fanselow MS, Hovda DA, Giza C. Region-Dependent Modulation of Neural Plasticity in Limbic Structures Early after Traumatic Brain Injury. Neurotrauma Rep 2021; 2:200-213. [PMID: 33937912 PMCID: PMC8086520 DOI: 10.1089/neur.2020.0045] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Traumatic brain injury (TBI)-induced disruptions in synaptic function within brain regions and across networks in the limbic system may underlie a vulnerability for maladaptive plasticity and contribute to behavioral comorbidities. In this study we measured how synaptic proteins respond to lateral fluid percussion injury (FPI) brain regions known to regulate emotion and memory, including the basolateral amygdala (BLA), dorsal and ventral hippocampus (DH, VH), and medial prefrontal cortex (PFC). We investigated proteins involved in regulating plasticity, including synaptic glutamatergic a-amino-3-hydroxy5-methyl-4-isoxazolepropionic acid (AMPA; GluA1, GluA2) and N-methyl-D-aspartate (NMDA; NR1, NR2A, NR2B) receptor subunits as well as inhibitory gamma-aminobutyric acid (GABA) synthetic enzymes (GAD67, GAD65) via western blot. Adult male rats received a mild-moderate lateral FPI or sham surgery and ipsi- and contralateral BLA, DH, VH, and PFC were collected 6 h, 24 h, 48 h, and 7 days post-injury. In the ipsilateral BLA, there was a significant decrease in NR1 and GluA2 24 h after injury, whereas NR2A and NR2B were increased in the contralateral BLA at 48 h compared with sham. GAD67 was increased ipsilaterally at 24 h, but decreased contralaterally at 48 h in the BLA. In the DH, both NMDA (NR2A, NR2B) and GABA-synthetic (GAD65, GAD67) proteins were increased acutely at 6 h compared with sham. GAD67 was also robustly increased in the ipsilateral VH at 6 h. In the contralateral VH, NR2A significantly increased between 6 h and 24 h after FPI, whereas GAD65 was decreased across the same time-points in the contralateral VH. In the medial PFC at 24 h we saw bilateral increases in GAD67 and a contralateral decrease in GluA1. Later, there was a significant decrease in GAD67 in contralateral PFC from 48 h to 7 days post-injury. Collectively, these data suggest that lateral FPI causes a dynamic homeostatic response across limbic networks, leading to an imbalance of the proteins involved in plasticity in neural systems underlying cognitive and emotional regulation.
Collapse
Affiliation(s)
- Ann N Hoffman
- Department of Neurosurgery, Brain Injury Research Center, University of California, Los Angeles, Los Angeles, California, USA.,Department of Psychology, University of California, Los Angeles, Los Angeles, California, USA.,Staglin Center for Brain and Behavioral Health, University of California, Los Angeles, Los Angeles, California, USA
| | - Sonya Watson
- Department of Neurosurgery, Brain Injury Research Center, University of California, Los Angeles, Los Angeles, California, USA.,Department of Psychology, University of California, Los Angeles, Los Angeles, California, USA
| | - Michael S Fanselow
- Department of Psychology, University of California, Los Angeles, Los Angeles, California, USA.,Staglin Center for Brain and Behavioral Health, University of California, Los Angeles, Los Angeles, California, USA.,Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, California, USA
| | - David A Hovda
- Department of Neurosurgery, Brain Injury Research Center, University of California, Los Angeles, Los Angeles, California, USA.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, California, USA.,Department of Medical and Molecular Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
| | - Christopher Giza
- Department of Neurosurgery, Brain Injury Research Center, University of California, Los Angeles, Los Angeles, California, USA.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, California, USA.,Mattel Children's Hospital, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
23
|
McCorkle TA, Barson JR, Raghupathi R. A Role for the Amygdala in Impairments of Affective Behaviors Following Mild Traumatic Brain Injury. Front Behav Neurosci 2021; 15:601275. [PMID: 33746719 PMCID: PMC7969709 DOI: 10.3389/fnbeh.2021.601275] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 01/29/2021] [Indexed: 11/30/2022] Open
Abstract
Mild traumatic brain injury (TBI) results in chronic affective disorders such as depression, anxiety, and fear that persist up to years following injury and significantly impair the quality of life for patients. Although a great deal of research has contributed to defining symptoms of mild TBI, there are no adequate drug therapies for brain-injured individuals. Preclinical studies have modeled these deficits in affective behaviors post-injury to understand the underlying mechanisms with a view to developing appropriate treatment strategies. These studies have also unveiled sex differences that contribute to the varying phenotypes associated with each behavior. Although clinical and preclinical studies have viewed these behavioral deficits as separate entities with unique neurobiological mechanisms, mechanistic similarities suggest that a novel approach is needed to advance research on drug therapy. This review will discuss the circuitry involved in the expression of deficits in affective behaviors following mild TBI in humans and animals and provide evidence that the manifestation of impairment in these behaviors stems from an amygdala-dependent emotional processing deficit. It will highlight mechanistic similarities between these different types of affective behaviors that can potentially advance mild TBI drug therapy by investigating treatments for the deficits in affective behaviors as one entity, requiring the same treatment.
Collapse
Affiliation(s)
- Taylor A. McCorkle
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences and Professional Studies, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Jessica R. Barson
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences and Professional Studies, Drexel University College of Medicine, Philadelphia, PA, United States
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Ramesh Raghupathi
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences and Professional Studies, Drexel University College of Medicine, Philadelphia, PA, United States
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
24
|
Beitchman JA, Lifshitz J, Harris NG, Thomas TC, Lafrenaye AD, Hånell A, Dixon CE, Povlishock JT, Rowe RK. Spatial Distribution of Neuropathology and Neuroinflammation Elucidate the Biomechanics of Fluid Percussion Injury. Neurotrauma Rep 2021; 2:59-75. [PMID: 34223546 PMCID: PMC8240834 DOI: 10.1089/neur.2020.0046] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Diffuse brain injury is better described as multi-focal, where pathology can be found adjacent to seemingly uninjured neural tissue. In experimental diffuse brain injury, pathology and pathophysiology have been reported far more lateral than predicted by the impact site. We hypothesized that local thickening of the rodent skull at the temporal ridges serves to focus the intracranial mechanical forces experienced during brain injury and generate predictable pathology. We demonstrated local thickening of the skull at the temporal ridges using contour analysis on magnetic resonance imaging. After diffuse brain injury induced by midline fluid percussion injury (mFPI), pathological foci along the anterior-posterior length of cortex under the temporal ridges were evident acutely (1, 2, and 7 days) and chronically (28 days) post-injury by deposition of argyophilic reaction product. Area CA3 of the hippocampus and lateral nuclei of the thalamus showed pathological change, suggesting that mechanical forces to or from the temporal ridges shear subcortical regions. A proposed model of mFPI biomechanics suggests that injury force vectors reflect off the skull base and radiate toward the temporal ridge, thereby injuring ventral thalamus, dorsolateral hippocampus, and sensorimotor cortex. Surgically thinning the temporal ridge before injury reduced injury-induced inflammation in the sensorimotor cortex. These data build evidence for temporal ridges of the rodent skull to contribute to the observed pathology, whether by focusing extracranial forces to enter the cranium or intracranial forces to escape the cranium. Pre-clinical investigations can take advantage of the predicted pathology to explore injury mechanisms and treatment efficacy.
Collapse
Affiliation(s)
- Joshua A Beitchman
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, Arizona, USA.,Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, USA.,Midwestern University, Glendale, Arizona, USA
| | - Jonathan Lifshitz
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, Arizona, USA.,Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, USA.,Arizona State University, Tempe, Arizona, USA.,Phoenix VA Health Care System, Phoenix, Arizona, USA
| | - Neil G Harris
- UCLA Brain Injury Research Center, Department of Neurosurgery, and Intellectual Development and Disabilities Research Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, USA
| | - Theresa Currier Thomas
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, Arizona, USA.,Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, USA.,Arizona State University, Tempe, Arizona, USA.,Phoenix VA Health Care System, Phoenix, Arizona, USA
| | | | - Anders Hånell
- Virginia Commonwealth University, Richmond, Virginia, USA.,Uppsala University Hospital, Uppsala, Sweden
| | | | | | - Rachel K Rowe
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, Arizona, USA.,Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, USA.,Phoenix VA Health Care System, Phoenix, Arizona, USA
| |
Collapse
|
25
|
Rowe RK, Ortiz JB, Thomas TC. Mild and Moderate Traumatic Brain Injury and Repeated Stress Affect Corticosterone in the Rat. Neurotrauma Rep 2020; 1:113-124. [PMID: 34223536 PMCID: PMC8240883 DOI: 10.1089/neur.2020.0019] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Traumatic brain injury (TBI) survivors suffer from a range of morbidities, including post-traumatic endocrinopathies that can cause physical and mental changes in patients, greatly compromising quality of life. This study tested the hypothesis that mild and moderate diffuse TBI leads to chronic deficiencies in corticosterone (CORT) regulation following repeated exposure to restraint stress over time. Young adult male rats (n = 9–11/group) were subjected to mild or moderate TBI induced by midline fluid percussion injury (mFPI) or control sham surgery. At 6 and 24 h post-injury, both mild and moderate TBI resulted in elevated resting plasma CORT levels compared with uninjured shams. Independent of TBI severity, all rats had lower resting plasma CORT levels at 7, 14, 28, and 54 days post-injury compared with pre-surgery baseline CORT. Circulating levels of CORT were also evaluated under restraint stress and in response to dexamethasone (DEX), a synthetic glucocorticoid. Independent of TBI severity, restraint stress elevated CORT at 30, 60, and 90 min post-stressor initiation at all post-injury time-points. A blunted CORT response to restraint stress was observed with lower CORT levels after restraint at 28 and 54 days compared with 7 days post-injury (DPI), indicative of habituation to the stressor. A high dose of DEX lowered CORT levels at 90 min post-restraint stress initiation compared with low-dose DEX, independent of TBI severity. These results support TBI-induced CORT dysregulation at acute time-points, but additional studies that investigate the onset and progression of endocrinopathies, controlling for habituation to repeated restraint stress, are needed to inform the diagnosis and treatment of such morbidities in TBI survivors.
Collapse
Affiliation(s)
- Rachel K Rowe
- Barrow Neurological Institute, Phoenix Children's Hospital, Phoenix, Arizona, USA.,Department of Child Health, University of Arizona College of Medicine Phoenix, Phoenix, Arizona, USA.,Phoenix Veteran Affairs Health Care System, Phoenix, Arizona, USA
| | - J Bryce Ortiz
- Barrow Neurological Institute, Phoenix Children's Hospital, Phoenix, Arizona, USA.,Department of Child Health, University of Arizona College of Medicine Phoenix, Phoenix, Arizona, USA
| | - Theresa Currier Thomas
- Barrow Neurological Institute, Phoenix Children's Hospital, Phoenix, Arizona, USA.,Department of Child Health, University of Arizona College of Medicine Phoenix, Phoenix, Arizona, USA.,Phoenix Veteran Affairs Health Care System, Phoenix, Arizona, USA
| |
Collapse
|
26
|
Bromberg CE, Condon AM, Ridgway SW, Krishna G, Garcia-Filion PC, Adelson PD, Rowe RK, Thomas TC. Sex-Dependent Pathology in the HPA Axis at a Sub-acute Period After Experimental Traumatic Brain Injury. Front Neurol 2020; 11:946. [PMID: 33101162 PMCID: PMC7554641 DOI: 10.3389/fneur.2020.00946] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 07/21/2020] [Indexed: 12/18/2022] Open
Abstract
Over 2.8 million traumatic brain injuries (TBIs) are reported in the United States annually, of which, over 75% are mild TBIs with diffuse axonal injury (DAI) as the primary pathology. TBI instigates a stress response that stimulates the hypothalamic-pituitary-adrenal (HPA) axis concurrently with DAI in brain regions responsible for feedback regulation. While the incidence of affective symptoms is high in both men and women, presentation is more prevalent and severe in women. Few studies have longitudinally evaluated the etiology underlying late-onset affective symptoms after mild TBI and even fewer have included females in the experimental design. In the experimental TBI model employed in this study, evidence of chronic HPA dysregulation has been reported at 2 months post-injury in male rats, with peak neuropathology in other regions of the brain at 7 days post-injury (DPI). We predicted that mechanisms leading to dysregulation of the HPA axis in male and female rats would be most evident at 7 DPI, the sub-acute time point. Young adult age-matched male and naturally cycling female Sprague Dawley rats were subjected to midline fluid percussion injury (mFPI) or sham surgery. Corticotropin releasing hormone, gliosis, and glucocorticoid receptor (GR) levels were evaluated in the hypothalamus and hippocampus, along with baseline plasma adrenocorticotropic hormone (ACTH) and adrenal gland weights. Microglial response in the paraventricular nucleus of the hypothalamus indicated mild neuroinflammation in males compared to sex-matched shams, but not females. Evidence of microglia activation in the dentate gyrus of the hippocampus was robust in both sexes compared with uninjured shams and there was evidence of a significant interaction between sex and injury regarding microglial cell count. GFAP intensity and astrocyte numbers increased as a function of injury, indicative of astrocytosis. GR protein levels were elevated 30% in the hippocampus of females in comparison to sex-matched shams. These data indicate sex-differences in sub-acute pathophysiology following DAI that precede late-onset HPA axis dysregulation. Further understanding of the etiology leading up to late-onset HPA axis dysregulation following DAI could identify targets to stabilize feedback, attenuate symptoms, and improve efficacy of rehabilitation and overall recovery.
Collapse
Affiliation(s)
- Caitlin E Bromberg
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States.,Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
| | - Andrew M Condon
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States.,Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Samantha W Ridgway
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States.,Department of Biology, School of Life Sciences, Arizona State University, Tempe, AZ, United States
| | - Gokul Krishna
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States.,Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
| | - Pamela C Garcia-Filion
- Department of Biomedical Informatics, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
| | - P David Adelson
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States.,Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States.,Department of Neurosurgery, Mayo Clinic School of Medicine, Phoenix, AZ, United States.,School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, United States
| | - Rachel K Rowe
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States.,Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States.,Phoenix VA Health Care System, Phoenix, AZ, United States
| | - Theresa Currier Thomas
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States.,Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States.,Phoenix VA Health Care System, Phoenix, AZ, United States
| |
Collapse
|
27
|
Stielper ZF, Fucich EA, Middleton JW, Hillard CJ, Edwards S, Molina PE, Gilpin NW. Traumatic Brain Injury and Alcohol Drinking Alter Basolateral Amygdala Endocannabinoids in Female Rats. J Neurotrauma 2020; 38:422-434. [PMID: 32838651 DOI: 10.1089/neu.2020.7175] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Traumatic brain injury (TBI) affects approximately 3 million Americans yearly and increases vulnerability to developing psychiatric comorbidities. Alcohol use disorder (AUD) is the most prevalent psychiatric diagnosis preceding injury and TBI may increase subsequent alcohol use. The basolateral amygdala (BLA) is a limbic structure commonly affected by TBI that is implicated in anxiety and AUD. Endocannabinoids (eCBs) regulate synaptic activity in the BLA, and BLA eCB modulation alters anxiety-like behavior and stress reactivity. Previous work from our laboratories showed that systemic eCB degradation inhibition ameliorates TBI-induced increases in anxiety-like behavior and motivation to respond for alcohol in male rats. Here, we used a lateral fluid percussion model to test moderate TBI effects on anxiety-like behavior, alcohol drinking, and eCB levels and cell signaling in BLA, as well as the effect of alcohol drinking on anxiety-like behavior and the BLA eCB system, in female rats. Our results show that TBI does not promote escalation of operant alcohol self-administration or increase anxiety-like behavior in female rats. In the BLA, TBI and alcohol drinking alter tissue amounts of 2-arachidonoylglycerol (2-AG) and N-arachidonoylethanolamine (anandamide; AEA) 1 h post-injury, and 2-AG levels remain low 11 days post-injury. Eleven days after injury, BLA pyramidal neurons were hyperexcitable, but measures of synaptic transmission and eCB signaling were unchanged. These data show that TBI impacts BLA 2-AG tissue levels, that this effect is modified by alcohol drinking, and also that TBI increases BLA cell excitability.
Collapse
Affiliation(s)
- Zachary F Stielper
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA.,Alcohol and Drug Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Elizabeth A Fucich
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA.,Alcohol and Drug Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA.,Neuroscience Program, Tulane University, New Orleans, Louisiana, USA
| | - Jason W Middleton
- Alcohol and Drug Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA.,Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Cecilia J Hillard
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Scott Edwards
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA.,Alcohol and Drug Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Patricia E Molina
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA.,Alcohol and Drug Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Nicholas W Gilpin
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA.,Alcohol and Drug Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA.,Southeast Louisiana VA Healthcare System, New Orleans, Louisiana, USA
| |
Collapse
|
28
|
Ratliff WA, Delic V, Pick CG, Citron BA. Dendritic arbor complexity and spine density changes after repetitive mild traumatic brain injury and neuroprotective treatments. Brain Res 2020; 1746:147019. [PMID: 32681835 DOI: 10.1016/j.brainres.2020.147019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 06/25/2020] [Accepted: 07/10/2020] [Indexed: 12/17/2022]
Abstract
Traumatic brain injury has been described as the signature affliction of recent military conflicts and repetitive TBIs, particularly associated with military and athletic activities, typically result in more severe clinical effects. The majority of TBIs are mild, but they can result in long term cognitive deficits for which there is no effective treatment. One of the most significant deficits observed in TBI patients is memory loss, which suggests that TBI can induce pathological changes within the hippocampus. tert-butylhydroquinone (tBHQ) and pioglitazone activate the Nrf2 and PPAR-γ transcription factors, respectively, and both have been shown to be neuroprotective in model systems. We examined the morphological changes within the hippocampus following repetitive mild TBI and simultaneous treatment with both factors. We utilized a closed head injury mouse model with five injuries over 5 weeks. Our results showed marked morphological changes among the dendrites and dendritic spines of the neurons of the dentate gyrus of the hippocampus. We observed decreases in overall dendritic length, as well as in the quantity and density of dendritic spines. Our treatment partially ameliorated these effects, suggesting that the Nrf2 and PPAR-γ transcription factors may be important targets for future drug development in the treatment of TBI in humans.
Collapse
Affiliation(s)
- Whitney A Ratliff
- Laboratory of Molecular Biology, Bay Pines VA Healthcare System, Research and Development 151, Bldg. 22 Rm. 123, 10000 Bay Pines Blvd, Bay Pines, FL 33744, United States
| | - Vedad Delic
- Laboratory of Molecular Biology, VA New Jersey Health Care System, Research & Development (15), Bldg. 16, Rm 16-176 385 Tremont Ave, East Orange, NJ 07018, United States
| | - Chaim G Pick
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel; Dr. Miriam and Sheldon G. Adelson Chair and Center for the Biology of Addictive Diseases, Tel Aviv University, Tel Aviv, Israel
| | - Bruce A Citron
- Laboratory of Molecular Biology, VA New Jersey Health Care System, Research & Development (15), Bldg. 16, Rm 16-176 385 Tremont Ave, East Orange, NJ 07018, United States; Laboratory of Molecular Biology, Bay Pines VA Healthcare System, Research and Development 151, Bldg. 22 Rm. 123, 10000 Bay Pines Blvd, Bay Pines, FL 33744, United States; Department of Pharmacology, Physiology & Neuroscience, Rutgers-New Jersey Medical School, Newark, NJ 07103, United States.
| |
Collapse
|
29
|
Balasubramanian N, Srivastava A, Pawar N, Sagarkar S, Sakharkar AJ. Repeated mild traumatic brain injury induces persistent variations in mitochondrial DNA copy number in mesocorticolimbic neurocircuitry of the rat. Neurosci Res 2020; 155:34-42. [DOI: 10.1016/j.neures.2019.06.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 06/26/2019] [Accepted: 06/27/2019] [Indexed: 12/18/2022]
|
30
|
Beitchman JA, Griffiths DR, Hur Y, Ogle SB, Bromberg CE, Morrison HW, Lifshitz J, Adelson PD, Thomas TC. Experimental Traumatic Brain Injury Induces Chronic Glutamatergic Dysfunction in Amygdala Circuitry Known to Regulate Anxiety-Like Behavior. Front Neurosci 2020; 13:1434. [PMID: 32038140 PMCID: PMC6985437 DOI: 10.3389/fnins.2019.01434] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 12/18/2019] [Indexed: 01/01/2023] Open
Abstract
Up to 50% of traumatic brain injury (TBI) survivors demonstrate persisting and late-onset anxiety disorders indicative of limbic system dysregulation, yet the pathophysiology underlying the symptoms is unclear. We hypothesize that the development of TBI-induced anxiety-like behavior in an experimental model of TBI is mediated by changes in glutamate neurotransmission within the amygdala. Adult, male Sprague-Dawley rats underwent midline fluid percussion injury or sham surgery. Anxiety-like behavior was assessed at 7 and 28 days post-injury (DPI) followed by assessment of real-time glutamate neurotransmission in the basolateral amygdala (BLA) and central nucleus of the amygdala (CeA) using glutamate-selective microelectrode arrays. The expression of anxiety-like behavior at 28 DPI coincided with decreased evoked glutamate release and slower glutamate clearance in the CeA, not BLA. Numerous factors contribute to the changes in glutamate neurotransmission over time. In two additional animal cohorts, protein levels of glutamatergic transporters (Glt-1 and GLAST) and presynaptic modulators of glutamate release (mGluR2, TrkB, BDNF, and glucocorticoid receptors) were quantified using automated capillary western techniques at 28 DPI. Astrocytosis and microglial activation have been shown to drive maladaptive glutamate signaling and were histologically assessed over 28 DPI. Alterations in glutamate neurotransmission could not be explained by changes in protein levels for glutamate transporters, mGluR2 receptors, astrocytosis, and microglial activation. Presynaptic modulators, BDNF and TrkB, were significantly decreased at 28 DPI in the amygdala. Dysfunction in presynaptic regulation of glutamate neurotransmission may contribute to anxiety-related behavior and serve as a therapeutic target to improve circuit function.
Collapse
Affiliation(s)
- Joshua A Beitchman
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States.,Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States.,College of Graduate Studies, Midwestern University, Glendale, AZ, United States
| | - Daniel R Griffiths
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States.,Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
| | - Yerin Hur
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States.,Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
| | - Sarah B Ogle
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States.,Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States.,Banner University Medical Center, Phoenix, AZ, United States
| | - Caitlin E Bromberg
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States.,Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
| | - Helena W Morrison
- College of Nursing, University of Arizona, Tucson, AZ, United States
| | - Jonathan Lifshitz
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States.,Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States.,Phoenix VA Health Care System, Phoenix, AZ, United States
| | - P David Adelson
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States.,Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
| | - Theresa Currier Thomas
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States.,Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States.,Phoenix VA Health Care System, Phoenix, AZ, United States
| |
Collapse
|
31
|
Stress reactivity after traumatic brain injury: implications for comorbid post-traumatic stress disorder. Behav Pharmacol 2020; 30:115-121. [PMID: 30640181 DOI: 10.1097/fbp.0000000000000461] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Most people have or will experience traumatic stress at some time over the lifespan, but only a subset of traumatized individuals develop post-traumatic stress disorder (PTSD). Clinical research supports high rates of traumatic brain injury (TBI)-PTSD comorbidity and demonstrates TBI as a significant predictor of the development of PTSD. Biological factors impacted following brain injury that may contribute to increased PTSD risk are unknown. Heightened stress reactivity and dysregulated hypothalamic-pituitary-adrenal (HPA) axis function are common to both TBI and PTSD, and affect amygdalar structure and function, which is implicated in PTSD. In this review, we summarize a growing body of literature that shows HPA axis dysregulation, as well as enhanced fear and amygdalar function after TBI. We present the hypothesis that altered stress reactivity as a result of brain injury impacts the amygdala and defense systems to be vulnerable to increased fear and PTSD development from traumatic stress. Identifying biological mechanisms that underlie this vulnerability, such as dysregulated HPA axis function, may lead to better targeted treatments and preventive measures to support psychological health after TBI.
Collapse
|
32
|
Fesharaki-Zadeh A, Miyauchi JT, St. Laurent-Arriot K, Tsirka SE, Bergold PJ. Increased Behavioral Deficits and Inflammation in a Mouse Model of Co-Morbid Traumatic Brain Injury and Post-Traumatic Stress Disorder. ASN Neuro 2020; 12:1759091420979567. [PMID: 33342261 PMCID: PMC7755938 DOI: 10.1177/1759091420979567] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/03/2020] [Accepted: 11/08/2020] [Indexed: 01/21/2023] Open
Abstract
Comorbid post-traumatic stress disorder with traumatic brain injury (TBI) produce more severe affective and cognitive deficits than PTSD or TBI alone. Both PTSD and TBI produce long-lasting neuroinflammation, which may be a key underlying mechanism of the deficits observed in co-morbid TBI/PTSD. We developed a model of co-morbid TBI/PTSD by combining the closed head (CHI) model of TBI with the chronic variable stress (CVS) model of PTSD and examined multiple behavioral and neuroinflammatory outcomes. Male C57/Bl6 mice received sham treatment, CHI, CVS, CHI then CVS (CHI → CVS) or CVS then CHI (CVS → CHI). The CVS → CHI group had deficits in Barnes maze or active place avoidance not seen in the other groups. The CVS → CHI, CVS and CHI → CVS groups displayed increased basal anxiety level, based on performance on elevated plus maze. The CVS → CHI had impaired performance on Barnes Maze, and Active Place Avoidance. These performance deficits were strongly correlated with increased hippocampal Iba-1 level an indication of activated MP/MG. These data suggest that greater cognitive deficits in the CVS → CHI group were due to increased inflammation. The increased deficits and neuroinflammation in the CVS → CHI group suggest that the order by which a subject experiences TBI and PTSD is a major determinant of the outcome of brain injury in co-morbid TBI/PTSD.
Collapse
Affiliation(s)
- Arman Fesharaki-Zadeh
- Department of Psychiatry, State University of New York, Downstate Medical Center, Brooklyn, New York
| | - Jeremy T. Miyauchi
- Department of Physiology, State University of New York, Downstate Medical Center, Brooklyn, New York
| | - Karrah St. Laurent-Arriot
- Department of Pharmacology, State University of New York, Downstate Medical Center, Brooklyn, New York
| | - Stella E. Tsirka
- Department of Physiology, State University of New York, Downstate Medical Center, Brooklyn, New York
| | - Peter J. Bergold
- Department of Pharmacology, State University of New York, Downstate Medical Center, Brooklyn, New York
- Department of Neurology, State University of New York, Downstate Medical Center, Brooklyn, New York
- Department of Pharmacological Sciences, Stony Brook Medicine, Stony Brook University, Stony Brook, New York
| |
Collapse
|
33
|
Hoffman AN, Lam J, Hovda DA, Giza CC, Fanselow MS. Sensory sensitivity as a link between concussive traumatic brain injury and PTSD. Sci Rep 2019; 9:13841. [PMID: 31554865 PMCID: PMC6761112 DOI: 10.1038/s41598-019-50312-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 09/10/2019] [Indexed: 12/12/2022] Open
Abstract
Traumatic brain injury (TBI) is one of the most common injuries to military personnel, a population often exposed to stressful stimuli and emotional trauma. Changes in sensory processing after TBI might contribute to TBI-post traumatic stress disorder (PTSD) comorbidity. Combining an animal model of TBI with an animal model of emotional trauma, we reveal an interaction between auditory sensitivity after TBI and fear conditioning where 75 dB white noise alone evokes a phonophobia-like phenotype and when paired with footshocks, fear is robustly enhanced. TBI reduced neuronal activity in the hippocampus but increased activity in the ipsilateral lateral amygdala (LA) when exposed to white noise. The white noise effect in LA was driven by increased activity in neurons projecting from ipsilateral auditory thalamus (medial geniculate nucleus). These data suggest that altered sensory processing within subcortical sensory-emotional circuitry after TBI results in neutral stimuli adopting aversive properties with a corresponding impact on facilitating trauma memories and may contribute to TBI-PTSD comorbidity.
Collapse
Affiliation(s)
- Ann N Hoffman
- UCLA, Neurosurgery; Brain Injury Research Center, Los Angeles, USA.
- UCLA, Psychology, Los Angeles, USA.
- UCLA Steve Tisch BrainSPORT Program, Los Angeles, USA.
- Staglin Center for Brain and Behavioral Health, Life Sciences, UCLA, Los Angeles, USA.
| | | | - David A Hovda
- UCLA, Neurosurgery; Brain Injury Research Center, Los Angeles, USA
- UCLA Steve Tisch BrainSPORT Program, Los Angeles, USA
- UCLA, Medical and Molecular Pharmacology, Los Angeles, USA
| | - Christopher C Giza
- UCLA, Neurosurgery; Brain Injury Research Center, Los Angeles, USA
- UCLA Steve Tisch BrainSPORT Program, Los Angeles, USA
- UCLA Mattel Children's Hospital, Los Angeles, USA
| | - Michael S Fanselow
- UCLA, Psychology, Los Angeles, USA
- UCLA, Psychiatry and Biobehavioral Sciences, Los Angeles, USA
- Staglin Center for Brain and Behavioral Health, Life Sciences, UCLA, Los Angeles, USA
| |
Collapse
|
34
|
Early TLR4 inhibition reduces hippocampal injury at puberty in a rat model of neonatal hypoxic-ischemic brain damage via regulation of neuroimmunity and synaptic plasticity. Exp Neurol 2019; 321:113039. [PMID: 31442443 DOI: 10.1016/j.expneurol.2019.113039] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 08/08/2019] [Accepted: 08/19/2019] [Indexed: 12/30/2022]
Abstract
Neonatal hypoxic-ischemic brain damage (HIBD) survivors present with long-term neurological disorders affecting their quality of life, and there remains a lack of effective treatment. Toll-like receptor 4 (TLR4) is widely distributed in nerve cells and its inhibition has a neuroprotective effect against brain injury. The present study aimed to evaluate the long-term neuroprotective effects of early inhibition of TLR4 during HIBD. Seven-day-old rat pups were subjected to left carotid artery ligation followed by 2 h of hypoxia (8.0% O2). A single dose of TAK-242 (0.5 mg/kg), a TLR4-specific antagonist, was intraperitoneally injected half an hour prior to hypoxic ischemia (HI). The long-term effects of TAK-242 inhibition on the induced hippocampal injury were investigated by assessing behaviour at P28, and then using a variety of methods to exploring the mechanism, including immunofluorescence, Golgi silver staining, Western blotting and real-time polymerase chain reaction (RT-PCR). TAK-242 treatment significantly reduced the expression levels of TLR4 and its downstream signalling molecules in the ipsilateral lesion of the hippocampus 24 h after HIBD. The Morris water maze (MWM) test demonstrated that TAK-242 treatment reduced the loss of HI-induced learning and memory functions. Immunofluorescence experiments showed that TAK-242 administration attenuated HI-induced loss of neurons, prevented the activation of microglia and astrocytes, and increased the expression of the glutamate receptor subtype, N-methyl d-aspartate 2A (NR2A) in the ipsilateral hippocampus region. Golgi silver staining revealed that TAK-242 prevented an HI-induced decline in spine density in the ipsilateral hippocampus. Western blot and RT-PCR results indicated that the expression of NR2A protein and mRNA in the ipsilateral hippocampi of adolescent rats decreased after neonatal HIBD; early TAK-242 administration may reverse these effects. In conclusion, our findings indicate that early inhibition of TLR4 signalling may improve the long-term prognosis of neonatal HIBD. The mechanisms contributing to this improvement involve reductions in neuronal loss, a decrease in glial cell activation, and an improvement in synaptic plasticity.
Collapse
|
35
|
Ratliff WA, Mervis RF, Citron BA, Schwartz B, Rubovitch V, Schreiber S, Pick CG. Mild blast-related TBI in a mouse model alters amygdalar neurostructure and circuitry. Exp Neurol 2019; 315:9-14. [PMID: 30711646 DOI: 10.1016/j.expneurol.2019.01.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/16/2019] [Accepted: 01/30/2019] [Indexed: 02/07/2023]
Abstract
Traumatic brain injury (TBI) continues to be a signature injury of our modern conflicts. Due in part to increased use of improvised explosive devices (IEDs), we have seen blast trauma make up a significant portion of TBIs sustained by deployed troops and civilians. In addition to the physical injury, TBI is also a common comorbidity with post-traumatic stress disorder (PTSD). Previous research suggests that PTSD is often associated with increased signaling within the amygdala, leading to feelings of fear and hyperarousal. In our study, we utilized a mouse model of mild blast-related TBI (bTBI) to investigate how TBI induces changes within the amygdala, which may provide favorable conditions for the development of PTSD. To do this, we performed Golgi staining on the stellate neurons of the basolateral amygdala and quantified dendritic amount, distribution, and complexity. We found increases in dendritic branching and in the density of dendritic spines in injured mice. Increases in spine density appears to be primarily due to increases in memory associated mushroom type dendritic spines. These changes observed in our bTBI model that are consistent with chronic stress models, suggesting an important connection between the physical changes induced by TBI and the neurological symptoms of PTSD.
Collapse
Affiliation(s)
- Whitney A Ratliff
- Bay Pines VA Healthcare System, Research and Development, 151, Bldg. 22 Rm. 123, 10000 Bay Pines Blvd, Bay Pines, FL 33744, USA; Department of Molecular Medicine, USF College of Medicine, 12901 Bruce B. Downs Blvd, MDC 7, Tampa, FL 33612, USA.
| | - Ronald F Mervis
- NeuroStructural Analytics, Inc., Columbus, OH, USA; Center for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 2 Tampa General Circle, Tampa, FL 33606, USA
| | - Bruce A Citron
- Bay Pines VA Healthcare System, Research and Development, 151, Bldg. 22 Rm. 123, 10000 Bay Pines Blvd, Bay Pines, FL 33744, USA; Department of Molecular Medicine, USF College of Medicine, 12901 Bruce B. Downs Blvd, MDC 7, Tampa, FL 33612, USA; VA New Jersey Health Care System, Research & Development, Mailstop 15, Bldg. 16, Rm. 16-176, 385 Tremont Ave, East Orange, NJ 07018, USA; Department of Pharmacology, Physiology & Neuroscience, Rutgers- New Jersey Medical School, 185 South Orange Ave., Newark, NJ 07101, USA
| | - Brian Schwartz
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Vardit Rubovitch
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Shaul Schreiber
- Department of Psychiatry, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; Department of Psychiatry, Tel Aviv University Sackler Faculty of Medicine, Tel Aviv 64239, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Chaim G Pick
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel; Dr. Miriam and Sheldon G. Adelson Chair, Center for the Biology of Addictive Diseases, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
36
|
Wellman CL, Moench KM. Preclinical studies of stress, extinction, and prefrontal cortex: intriguing leads and pressing questions. Psychopharmacology (Berl) 2019; 236:59-72. [PMID: 30225660 PMCID: PMC6374178 DOI: 10.1007/s00213-018-5023-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 09/03/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND Stress is associated with cognitive and emotional dysfunction, and increases risk for a variety of psychological disorders, including depression and posttraumatic stress disorder. Prefrontal cortex is critical for executive function and emotion regulation, is a target for stress hormones, and is implicated in many stress-influenced psychological disorders. Extinction of conditioned fear provides an excellent model system for examining how stress-induced changes in corticolimbic structure and function are related to stress-induced changes in neural function and behavior, as the neural circuitry underlying this behavior is well characterized. OBJECTIVES This review examines how acute and chronic stress influences extinction and describes how stress alters the structure and function of the medial prefrontal cortex, a potential neural substrate for these effects. In addition, we identify important unanswered questions about how stress-induced change in prefrontal cortex may mediate extinction deficits and avenues for future research. KEY FINDINGS A substantial body of work demonstrates deficits in extinction after either acute or chronic stress. A separate and substantial literature demonstrates stress-induced neuronal remodeling in medial prefrontal cortex, along with several key neurohormonal contributors to this remodeling, and there is substantial overlap in prefrontal mechanisms underlying extinction and the mechanisms implicated in stress-induced dysfunction of-and neuronal remodeling in-medial prefrontal cortex. However, data directly examining the contribution of changes in prefrontal structure and function to stress-induced extinction deficits is currently lacking. CONCLUSIONS Understanding how stress influences extinction and its neural substrates as well as individual differences in this effect will elucidate potential avenues for novel interventions for stress-sensitive disorders characterized by deficits in extinction.
Collapse
Affiliation(s)
- Cara L. Wellman
- Department of Psychological & Brain Sciences, Indiana University,Department of Psychological, Center for the Integrative Study of Animal Behavior, Indiana University,Department of Psychological, Program in Neuroscience, Indiana University
| | - Kelly M. Moench
- Department of Psychological & Brain Sciences, Indiana University,Department of Psychological, Center for the Integrative Study of Animal Behavior, Indiana University,Department of Psychological, Program in Neuroscience, Indiana University
| |
Collapse
|
37
|
Sharp BM. Basolateral amygdala, nicotinic cholinergic receptors, and nicotine: Pharmacological effects and addiction in animal models and humans. Eur J Neurosci 2018; 50:2247-2254. [PMID: 29802666 DOI: 10.1111/ejn.13970] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 05/10/2018] [Accepted: 05/14/2018] [Indexed: 01/08/2023]
Abstract
The amygdala is involved in processing incoming information about rewarding stimuli and emotions that denote danger such as anxiety and fear. Bi-directional neural connections between basolateral amygdala (BLA) and brain regions such as nucleus accumbens, prefrontal cortex, hippocampus, and hindbrain regions regulate motivation, cognition, and responses to stress. Altered local regulation of BLA excitability is pivotal to the behavioral disturbances characteristic of posttraumatic stress disorder, and relapse to drug use induced by stress. Herein, we review the physiological regulation of BLA by cholinergic inputs, emphasizing the role of BLA nicotinic receptors. We review BLA-dependent effects of nicotine on cognition, motivated behaviors, and emotional states, including memory, taking and seeking drugs, and anxiety and fear in humans and animal models. The alterations in BLA activity observed in animal studies inform human behavioral and brain imaging research by enabling a more exact understanding of altered BLA function. Converging evidence indicates that cholinergic signaling from basal forebrain projections to local nicotinic receptors is an important physiological regulator of BLA and that nicotine alters BLA function. In essence, BLA is necessary for behavioral responses to stimuli that evoke anxiety and fear; reinstatement of cue-induced drug seeking; responding to second-order cues conditioned to abused drugs; reacquisition of amplified nicotine self-administration due to chronic stress during abstinence; and to promote responding for natural reward.
Collapse
Affiliation(s)
- Burt M Sharp
- Department of Genetics, Genomics and Informatics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
38
|
Thomas TC, Stockhausen EM, Law LM, Khodadad A, Lifshitz J. Rehabilitation modality and onset differentially influence whisker sensory hypersensitivity after diffuse traumatic brain injury in the rat. Restor Neurol Neurosci 2018; 35:611-629. [PMID: 29036852 DOI: 10.3233/rnn-170753] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND As rehabilitation strategies advance as therapeutic interventions, the modality and onset of rehabilitation after traumatic brain injury (TBI) are critical to optimize treatment. Our laboratory has detected and characterized a late-onset, long-lasting sensory hypersensitivity to whisker stimulation in diffuse brain-injured rats; a deficit that is comparable to visual or auditory sensory hypersensitivity in humans with an acquired brain injury. OBJECTIVE We hypothesize that the modality and onset of rehabilitation therapies will differentially influence sensory hypersensitivity in response to the Whisker Nuisance Task (WNT) as well as WNT-induced corticosterone (CORT) stress response in diffuse brain-injured rats and shams. METHODS After midline fluid percussion brain injury (FPI) or sham surgery, rats were assigned to one of four rehabilitative interventions: (1) whisker sensory deprivation during week one or (2) week two or (3) whisker stimulation during week one or (4) week two. At 28 days following FPI and sham procedures, sensory hypersensitivity was assessed using the WNT. Plasma CORT was evaluated immediately following the WNT (aggravated levels) and prior to the pre-determined endpoint 24 hours later (non-aggravated levels). RESULTS Deprivation therapy during week two elicited significantly greater sensory hypersensitivity to the WNT compared to week one (p < 0.05), and aggravated CORT levels in FPI rats were significantly lower than sham levels. Stimulation therapy during week one resulted in low levels of sensory hypersensitivity to the WNT, similar to deprivation therapy and naïve controls, however, non-aggravated CORT levels in FPI rats were significantly higher than sham. CONCLUSION These data indicate that modality and onset of sensory rehabilitation can differentially influence FPI and sham rats, having a lasting impact on behavioral and stress responses to the WNT, emphasizing the necessity for continued evaluation of modality and onset of rehabilitation after TBI.
Collapse
Affiliation(s)
- Theresa Currier Thomas
- Barrow Neurological Institute at Phoenix Children's Hospital - Phoenix, AZ, USA.,Department of Child Health, University of Arizona College of Medicine - Phoenix, AZ, USA.,Phoenix VA Healthcare System - Phoenix, AZ, USA.,Spinal Cord and Brain Injury Research Center, University of Kentucky Chandler Medical Center - Lexington, KY, USA
| | - Ellen Magee Stockhausen
- Core Medical Group, Manchester, NH, USA.,Spinal Cord and Brain Injury Research Center, University of Kentucky Chandler Medical Center - Lexington, KY, USA
| | - L Matthew Law
- Barrow Neurological Institute at Phoenix Children's Hospital - Phoenix, AZ, USA.,Department of Child Health, University of Arizona College of Medicine - Phoenix, AZ, USA
| | - Aida Khodadad
- Barrow Neurological Institute at Phoenix Children's Hospital - Phoenix, AZ, USA.,Department of Child Health, University of Arizona College of Medicine - Phoenix, AZ, USA
| | - Jonathan Lifshitz
- Barrow Neurological Institute at Phoenix Children's Hospital - Phoenix, AZ, USA.,Department of Child Health, University of Arizona College of Medicine - Phoenix, AZ, USA.,Phoenix VA Healthcare System - Phoenix, AZ, USA.,Neuroscience Program, Arizona State University - Tempe, AZ, USA.,Spinal Cord and Brain Injury Research Center, University of Kentucky Chandler Medical Center - Lexington, KY, USA
| |
Collapse
|
39
|
Ortiz JB, Conrad CD. The impact from the aftermath of chronic stress on hippocampal structure and function: Is there a recovery? Front Neuroendocrinol 2018; 49:114-123. [PMID: 29428548 DOI: 10.1016/j.yfrne.2018.02.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 02/06/2018] [Accepted: 02/07/2018] [Indexed: 12/18/2022]
Abstract
Chronic stress results in functional and structural changes to the brain and especially the hippocampus. Decades of research have provided insights into the mechanisms by which chronic stress impairs hippocampal-mediated cognition and the corresponding reduction of hippocampal CA3 apical dendritic complexity. Yet, when chronic stress ends and time passes, which we refer to as a "post-stress rest period," hippocampal-mediated spatial memory deficits begin to improve and CA3 apical dendritic arbors increase in complexity. The processes by which the hippocampus improves from a chronically stressed state are not simply the reversal of the mechanisms that produced spatial memory deficits and CA3 apical dendritic retraction. This review will discuss our current understanding of how a chronically stressed hippocampus improves after a post-stress rest period. Untangling the mechanisms that allow for this post-stress plasticity is a critical next step in understanding how to promote resilience in the face of stressors.
Collapse
Affiliation(s)
- J Bryce Ortiz
- Department of Psychology, Arizona State University, Box 1104, Tempe, AZ 85287-1104, United States.
| | - Cheryl D Conrad
- Department of Psychology, Arizona State University, Box 1104, Tempe, AZ 85287-1104, United States
| |
Collapse
|
40
|
de la Tremblaye PB, O'Neil DA, LaPorte MJ, Cheng JP, Beitchman JA, Thomas TC, Bondi CO, Kline AE. Elucidating opportunities and pitfalls in the treatment of experimental traumatic brain injury to optimize and facilitate clinical translation. Neurosci Biobehav Rev 2018; 85:160-175. [PMID: 28576511 PMCID: PMC5709241 DOI: 10.1016/j.neubiorev.2017.05.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 05/12/2017] [Indexed: 12/19/2022]
Abstract
The aim of this review is to discuss the research presented in a symposium entitled "Current progress in characterizing therapeutic strategies and challenges in experimental CNS injury" which was presented at the 2016 International Behavioral Neuroscience Society annual meeting. Herein we discuss diffuse and focal traumatic brain injury (TBI) and ensuing chronic behavioral deficits as well as potential rehabilitative approaches. We also discuss the effects of stress on executive function after TBI as well as the response of the endocrine system and regulatory feedback mechanisms. The role of the endocannabinoids after CNS injury is also discussed. Finally, we conclude with a discussion of antipsychotic and antiepileptic drugs, which are provided to control TBI-induced agitation and seizures, respectively. The review consists predominantly of published data.
Collapse
Affiliation(s)
- Patricia B de la Tremblaye
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States
| | - Darik A O'Neil
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States
| | - Megan J LaPorte
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jeffrey P Cheng
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States
| | - Joshua A Beitchman
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States; Department of Child Health, University of Arizona College of Medicine, Phoenix, AZ, United States; Midwestern University, Glendale, AZ, United States
| | - Theresa Currier Thomas
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States; Department of Child Health, University of Arizona College of Medicine, Phoenix, AZ, United States; Phoenix VA Healthcare System, Phoenix, AZ, United States
| | - Corina O Bondi
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States; Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Anthony E Kline
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States; Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, United States; Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States; Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
41
|
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: 78] [Impact Index Per Article: 9.8] [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
|
42
|
Saykally JN, Ratliff WA, Keeley KL, Pick CG, Mervis RF, Citron BA. Repetitive Mild Closed Head Injury Alters Protein Expression and Dendritic Complexity in a Mouse Model. J Neurotrauma 2017; 35:139-148. [PMID: 28701108 DOI: 10.1089/neu.2017.5070] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Worldwide head injuries are a growing problem. In the United States alone, 1.7 million people suffer a head injury each year. While most of these injuries are mild, head injury sufferers still sustain symptoms that can have major medical and economical impacts. Moreover, repetitive mild head injuries, like those observed in active military personnel and athletes, have demonstrated a more severe and long-term set of consequences. In an effort to better understand the delayed pathological changes following multiple mild head injuries, we used a mouse model of mild closed head injury (with no motor deficits observed by rotarod testing) and measured dendritic complexity at 30 days after injury and potentially related factors up to 60 days post-injury. We found an increase in TDP-43 protein at 60 days post-injury in the hippocampus and a decrease in autophagy factors three days post-injury. Alterations in dendritic complexity were neuronal subtype and location specific. Measurements of neurotropic factors suggest that an increase in complexity in the cortex may be a consequence of neuronal loss of the less connected neurons.
Collapse
Affiliation(s)
- Jessica N Saykally
- 1 Laboratory of Molecular Biology, Research and Development 151, Bay Pines VA Healthcare System , Bay Pines, Florida.,2 Department of Molecular Medicine, University of South Florida College of Medicine , Tampa, Florida
| | - Whitney A Ratliff
- 1 Laboratory of Molecular Biology, Research and Development 151, Bay Pines VA Healthcare System , Bay Pines, Florida.,2 Department of Molecular Medicine, University of South Florida College of Medicine , Tampa, Florida
| | - Kristen L Keeley
- 1 Laboratory of Molecular Biology, Research and Development 151, Bay Pines VA Healthcare System , Bay Pines, Florida.,2 Department of Molecular Medicine, University of South Florida College of Medicine , Tampa, Florida
| | - Chaim G Pick
- 3 Department of Anatomy and Anthropology, Sackler School of Medicine, Tel Aviv University , Tel Aviv, Israel
| | - Ronald F Mervis
- 4 NeuroStructural Research Laboratories, Inc. , Tampa, Florida.,5 Center for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine , Tampa, Florida
| | - Bruce A Citron
- 1 Laboratory of Molecular Biology, Research and Development 151, Bay Pines VA Healthcare System , Bay Pines, Florida.,2 Department of Molecular Medicine, University of South Florida College of Medicine , Tampa, Florida
| |
Collapse
|
43
|
Does time heal all wounds? Experimental diffuse traumatic brain injury results in persisting histopathology in the thalamus. Behav Brain Res 2016; 340:137-146. [PMID: 28042008 DOI: 10.1016/j.bbr.2016.12.038] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 11/30/2016] [Accepted: 12/28/2016] [Indexed: 11/21/2022]
Abstract
BACKGROUND Thalamic dysfunction has been implicated in overall chronic neurological dysfunction after traumatic brain injury (TBI), however little is known about the underlying histopathology. In experimental diffuse TBI (dTBI), we hypothesize that persisting histopathological changes in the ventral posteromedial (VPM) nucleus of the thalamus is indicative of progressive circuit reorganization. Since circuit reorganization in the VPM impacts the whisker sensory system, the histopathology could explain the development of hypersensitivity to whisker stimulation by 28days post-injury; similar to light and sound hypersensitivity in human TBI survivors. METHODS Adult, male Sprague-Dawley rats underwent craniotomy and midline fluid percussion injury (FPI) (moderate severity; 1.8-2.0atm) or sham surgery. At 1d, 7d, and 28days post-FPI (d FPI) separate experiments confirmed the cytoarchitecture (Giemsa stain) and evaluated neuropathology (silver stain), activated astrocytes (GFAP), neuron morphology (Golgi stain) and microglial morphology (Iba-1) in the VPM. RESULTS Cytoarchitecture was unchanged throughout the time course, similar to previously published data; however, neuropathology and astrocyte activation were significantly increased at 7d and 28d and activated microglia were present at all time points. Neuron morphology was dynamic over the time course with decreased dendritic complexity (fewer branch points; decreased length of processes) at 7d FPI and return to sham values by 28d FPI. CONCLUSIONS These data indicate that dTBI results in persisting thalamic histopathology out to a chronic time point. While these changes can be indicative of either adaptive (recovery) or maladaptive (neurological dysfunction) circuit reorganization, they also provide a potential mechanism by which maladaptive circuit reorganization could contribute to the development of chronic neurological dysfunction. Understanding the processes that mediate circuit reorganization is critical to the development of future therapies for TBI patients.
Collapse
|
44
|
Rowe RK, Rumney BM, May HG, Permana P, Adelson PD, Harman SM, Lifshitz J, Thomas TC. Diffuse traumatic brain injury affects chronic corticosterone function in the rat. Endocr Connect 2016; 5:152-66. [PMID: 27317610 PMCID: PMC5002959 DOI: 10.1530/ec-16-0031] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 06/17/2016] [Indexed: 01/02/2023]
Abstract
As many as 20-55% of patients with a history of traumatic brain injury (TBI) experience chronic endocrine dysfunction, leading to impaired quality of life, impaired rehabilitation efforts and lowered life expectancy. Endocrine dysfunction after TBI is thought to result from acceleration-deceleration forces to the brain within the skull, creating enduring hypothalamic and pituitary neuropathology, and subsequent hypothalamic-pituitary endocrine (HPE) dysfunction. These experiments were designed to test the hypothesis that a single diffuse TBI results in chronic dysfunction of corticosterone (CORT), a glucocorticoid released in response to stress and testosterone. We used a rodent model of diffuse TBI induced by midline fluid percussion injury (mFPI). At 2months postinjury compared with uninjured control animals, circulating levels of CORT were evaluated at rest, under restraint stress and in response to dexamethasone, a synthetic glucocorticoid commonly used to test HPE axis regulation. Testosterone was evaluated at rest. Further, we assessed changes in injury-induced neuron morphology (Golgi stain), neuropathology (silver stain) and activated astrocytes (GFAP) in the paraventricular nucleus (PVN) of the hypothalamus. Resting plasma CORT levels were decreased at 2months postinjury and there was a blunted CORT increase in response to restraint induced stress. No changes in testosterone were measured. These changes in CORT were observed concomitantly with altered complexity of neuron processes in the PVN over time, devoid of neuropathology or astrocytosis. Results provide evidence that a single moderate diffuse TBI leads to changes in CORT function, which can contribute to the persistence of symptoms related to endocrine dysfunction. Future experiments aim to evaluate additional HP-related hormones and endocrine circuit pathology following diffuse TBI.
Collapse
Affiliation(s)
- Rachel K Rowe
- Phoenix Veterans Affairs Health Care SystemPhoenix, Arizona, USA BARROW Neurological Institute at Phoenix Children's HospitalPhoenix, Arizona, USA Department of Child HealthUniversity of Arizona College of Medicine - Phoenix, Phoenix, Arizona, USA
| | - Benjamin M Rumney
- BARROW Neurological Institute at Phoenix Children's HospitalPhoenix, Arizona, USA Department of Child HealthUniversity of Arizona College of Medicine - Phoenix, Phoenix, Arizona, USA Department of Biology and BiochemistryUniversity of Bath, UK
| | - Hazel G May
- BARROW Neurological Institute at Phoenix Children's HospitalPhoenix, Arizona, USA Department of Child HealthUniversity of Arizona College of Medicine - Phoenix, Phoenix, Arizona, USA Department of Biology and BiochemistryUniversity of Bath, UK
| | - Paska Permana
- Phoenix Veterans Affairs Health Care SystemPhoenix, Arizona, USA
| | - P David Adelson
- BARROW Neurological Institute at Phoenix Children's HospitalPhoenix, Arizona, USA Department of Child HealthUniversity of Arizona College of Medicine - Phoenix, Phoenix, Arizona, USA School of Biological and Health Systems EngineeringArizona State University, Tempe, Arizona, USA
| | | | - Jonathan Lifshitz
- Phoenix Veterans Affairs Health Care SystemPhoenix, Arizona, USA BARROW Neurological Institute at Phoenix Children's HospitalPhoenix, Arizona, USA Department of Child HealthUniversity of Arizona College of Medicine - Phoenix, Phoenix, Arizona, USA
| | - Theresa C Thomas
- Phoenix Veterans Affairs Health Care SystemPhoenix, Arizona, USA BARROW Neurological Institute at Phoenix Children's HospitalPhoenix, Arizona, USA Department of Child HealthUniversity of Arizona College of Medicine - Phoenix, Phoenix, Arizona, USA
| |
Collapse
|