1
|
Green TRF, Carey SD, Mannino G, Craig JA, Rowe RK, Zielinski MR. Sleep, inflammation, and hemodynamics in rodent models of traumatic brain injury. Front Neurosci 2024; 18:1361014. [PMID: 38426017 PMCID: PMC10903352 DOI: 10.3389/fnins.2024.1361014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 01/29/2024] [Indexed: 03/02/2024] Open
Abstract
Traumatic brain injury (TBI) can induce dysregulation of sleep. Sleep disturbances include hypersomnia and hyposomnia, sleep fragmentation, difficulty falling asleep, and altered electroencephalograms. TBI results in inflammation and altered hemodynamics, such as changes in blood brain barrier permeability and cerebral blood flow. Both inflammation and altered hemodynamics, which are known sleep regulators, contribute to sleep impairments post-TBI. TBIs are heterogenous in cause and biomechanics, which leads to different molecular and symptomatic outcomes. Animal models of TBI have been developed to model the heterogeneity of TBIs observed in the clinic. This review discusses the intricate relationship between sleep, inflammation, and hemodynamics in pre-clinical rodent models of TBI.
Collapse
Affiliation(s)
- Tabitha R. F. Green
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - Sean D. Carey
- Veterans Affairs (VA) Boston Healthcare System, West Roxbury, MA, United States
- Department of Psychiatry, Harvard Medical School, West Roxbury, MA, United States
| | - Grant Mannino
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - John A. Craig
- Veterans Affairs (VA) Boston Healthcare System, West Roxbury, MA, United States
| | - Rachel K. Rowe
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - Mark R. Zielinski
- Veterans Affairs (VA) Boston Healthcare System, West Roxbury, MA, United States
- Department of Psychiatry, Harvard Medical School, West Roxbury, MA, United States
| |
Collapse
|
2
|
Wang D, Zhang HX, Yan GJ, Zhao HR, Dong XH, Tan YX, Li S, Lu MN, Mei R, Liu LN, Wang XY, Xiyang YB. Voluntary running wheel exercise induces cognitive improvement post traumatic brain injury in mouse model through redressing aberrant excitation regulated by voltage-gated sodium channels 1.1, 1.3, and 1.6. Exp Brain Res 2024; 242:205-224. [PMID: 37994916 PMCID: PMC10786980 DOI: 10.1007/s00221-023-06734-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 10/29/2023] [Indexed: 11/24/2023]
Abstract
Traumatic brain injury (TBI) leads to disturbed brain discharge rhythm, elevated excitability, anxiety-like behaviors, and decreased learning and memory capabilities. Cognitive dysfunctions severely affect the quality of life and prognosis of TBI patients, requiring effective rehabilitation treatment. Evidence indicates that moderate exercise after brain injury decreases TBI-induced cognitive decline. However, the underlying mechanism remains unelucidated. Our results demonstrate that TBI causes cognitive impairment behavior abnormalities and overexpression of Nav1.1, Nav1.3 and Nav1.6 proteins inside the hippocampus of mice models. Three weeks of voluntary running wheel (RW) exercise treatments before or/and post-injury effectively redressed the aberrant changes caused by TBI. Additionally, a 10% exercise-conditioned medium helped recover cell viability, neuronal sodium current and expressions of Nav1.1, Nav1.3 and Nav1.6 proteins across cultured neurons after injury. Therefore, the results validate the neuroprotection induced by voluntary RW exercise treatment before or/and post-TBI. The RW exercise-induced improvement in cognitive behaviors and neuronal excitability could be associated with correcting the Nav1.1, Nav1.3, and Nav1.6 expression levels. The current study proves that voluntary exercise is an effective treatment strategy against TBI. The study also highlights novel potential targets for rehabilitating TBI, including the Navs proteins.
Collapse
Affiliation(s)
- Dan Wang
- Institute of Neuroscience, Faculty of Basic Medical Science, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong, Kunming, Yunnan, 650500, People's Republic of China
| | - Hui-Xiang Zhang
- Institute of Neuroscience, Faculty of Basic Medical Science, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong, Kunming, Yunnan, 650500, People's Republic of China
| | - Guo-Ji Yan
- Institute of Neuroscience, Faculty of Basic Medical Science, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong, Kunming, Yunnan, 650500, People's Republic of China
| | - Hao-Ran Zhao
- Institute of Neuroscience, Faculty of Basic Medical Science, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong, Kunming, Yunnan, 650500, People's Republic of China
| | - Xiao-Han Dong
- Institute of Neuroscience, Faculty of Basic Medical Science, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong, Kunming, Yunnan, 650500, People's Republic of China
| | - Ya-Xin Tan
- Institute of Neuroscience, Faculty of Basic Medical Science, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong, Kunming, Yunnan, 650500, People's Republic of China
- Department of Pediatrics, The People's Liberation Army (PLA) Rocket Force Characteristic Medical Center, Beijing, 100088, China
| | - Shan Li
- Institute of Neuroscience, Faculty of Basic Medical Science, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong, Kunming, Yunnan, 650500, People's Republic of China
- Department of Anatomy, Changsha Medical University, Changsha, China
| | - Min-Nan Lu
- Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Rong Mei
- Department of Neurology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Li-Na Liu
- Institute of Neuroscience, Faculty of Basic Medical Science, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong, Kunming, Yunnan, 650500, People's Republic of China
| | - Xu-Yang Wang
- Department of Neurosurgery, Shanghai Sixth People' Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, People's Republic of China.
| | - Yan-Bin Xiyang
- Institute of Neuroscience, Faculty of Basic Medical Science, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong, Kunming, Yunnan, 650500, People's Republic of China.
| |
Collapse
|
3
|
Somach RT, Jean ID, Farrugia AM, Cohen AS. Mild Traumatic Brain Injury Affects Orexin/Hypocretin Physiology Differently in Male and Female Mice. J Neurotrauma 2023; 40:2146-2163. [PMID: 37476962 PMCID: PMC10701510 DOI: 10.1089/neu.2023.0125] [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: 07/22/2023] Open
Abstract
Traumatic brain injury (TBI) is known to affect the physiology of neural circuits in several brain regions, which can contribute to behavioral changes after injury. Disordered sleep is a behavior that is often seen after TBI, but there is little research into how injury affects the circuitry that contributes to disrupted sleep regulation. Orexin/hypocretin neurons (hereafter referred to as orexin neurons) located in the lateral hypothalamus normally stabilize wakefulness in healthy animals and have been suggested as a source of dysregulated sleep behavior. Despite this, few studies have examined how TBI affects orexin neuron circuitry. Further, almost no animal studies of orexin neurons after TBI have included female animals. Here, we address these gaps by studying changes to orexin physiology using ex vivo acute brain slices and whole-cell patch clamp recording. We hypothesized that orexin neurons would have reduced afferent excitatory activity after injury. Ultimately, this hypothesis was supported but there were additional physiological changes that occurred that we did not originally hypothesize. We studied physiological properties in orexin neurons approximately 1 week after mild traumatic brain injury (mTBI) in 6-8-week-old male and female mice. mTBI was performed with a lateral fluid percussion injury between 1.4 and 1.6 atmospheres. Mild TBI increased the size of action potential afterhyperpolarization in orexin neurons from female mice, but not male mice and reduced the action potential threshold in male mice, but not in female mice. Mild TBI reduced afferent excitatory activity and increased afferent inhibitory activity onto orexin neurons. Alterations in afferent excitatory activity occurred in different parameters in male and female animals. The increased afferent inhibitory activity after injury is more pronounced in recordings from female animals. Our results indicate that mTBI changes the physiology of orexin neuron circuitry and that these changes are not the same in male and female animals.
Collapse
Affiliation(s)
- Rebecca T. Somach
- Department of Anesthesiology and Critical Care Medicine, the Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Anesthesiology and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Neuroscience Graduate Group, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ian D. Jean
- Department of Anesthesiology and Critical Care Medicine, the Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Anthony M. Farrugia
- Department of Anesthesiology and Critical Care Medicine, the Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Anesthesiology and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Akiva S. Cohen
- Department of Anesthesiology and Critical Care Medicine, the Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Anesthesiology and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Neuroscience Graduate Group, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
4
|
Que M, Li Y, Wang X, Zhan G, Luo X, Zhou Z. Role of astrocytes in sleep deprivation: accomplices, resisters, or bystanders? Front Cell Neurosci 2023; 17:1188306. [PMID: 37435045 PMCID: PMC10330732 DOI: 10.3389/fncel.2023.1188306] [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: 03/17/2023] [Accepted: 06/05/2023] [Indexed: 07/13/2023] Open
Abstract
Sleep plays an essential role in all studied animals with a nervous system. However, sleep deprivation leads to various pathological changes and neurobehavioral problems. Astrocytes are the most abundant cells in the brain and are involved in various important functions, including neurotransmitter and ion homeostasis, synaptic and neuronal modulation, and blood-brain barrier maintenance; furthermore, they are associated with numerous neurodegenerative diseases, pain, and mood disorders. Moreover, astrocytes are increasingly being recognized as vital contributors to the regulation of sleep-wake cycles, both locally and in specific neural circuits. In this review, we begin by describing the role of astrocytes in regulating sleep and circadian rhythms, focusing on: (i) neuronal activity; (ii) metabolism; (iii) the glymphatic system; (iv) neuroinflammation; and (v) astrocyte-microglia cross-talk. Moreover, we review the role of astrocytes in sleep deprivation comorbidities and sleep deprivation-related brain disorders. Finally, we discuss potential interventions targeting astrocytes to prevent or treat sleep deprivation-related brain disorders. Pursuing these questions would pave the way for a deeper understanding of the cellular and neural mechanisms underlying sleep deprivation-comorbid brain disorders.
Collapse
Affiliation(s)
- Mengxin Que
- Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Department of Anesthesiology, Tongji Medical College, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yujuan Li
- Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Department of Anesthesiology, Tongji Medical College, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xuan Wang
- Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Department of Anesthesiology, Tongji Medical College, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Gaofeng Zhan
- Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Department of Anesthesiology, Tongji Medical College, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxiao Luo
- Department of Oncology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiqiang Zhou
- Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Department of Anesthesiology, Tongji Medical College, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
5
|
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
|
6
|
The Effect of Traumatic Brain Injury on Sleep Architecture and Circadian Rhythms in Mice—A Comparison of High-Frequency Head Impact and Controlled Cortical Injury. BIOLOGY 2022; 11:biology11071031. [PMID: 36101412 PMCID: PMC9312487 DOI: 10.3390/biology11071031] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/02/2022] [Accepted: 07/07/2022] [Indexed: 11/17/2022]
Abstract
Simple Summary Traumatic brain injury (TBI) is a significant risk factor for the development of sleep and circadian rhythm impairments. In order to understand if TBI models with different injury mechanism, severity and pathology have different sleep and circadian rhythm disruptions, we performed a detailed sleep and circadian analysis of the high-frequency head impact TBI model (a mouse model that mimics sports-related head impacts) and the controlled cortical impact TBI model (a mouse model that mimics severe brain trauma). We found that both TBI models disrupt the ability of brain cells to maintain circadian rhythms; however, both injury groups could still maintain circadian behavior patterns. Both the mild head impact model and the severe brain injury model had normal amount of sleep at 7 d after injury; however, the severe brain injury mice had disrupted brain wave patterns during sleep. We conclude that different types of TBI have different patterns of sleep disruptions. Abstract Traumatic brain injury (TBI) is a significant risk factor for the development of sleep and circadian rhythm impairments. In this study we compare the circadian rhythms and sleep patterns in the high-frequency head impact (HFHI) and controlled cortical impact (CCI) mouse models of TBI. These mouse models have different injury mechanisms key differences of pathology in brain regions controlling circadian rhythms and EEG wave generation. We found that both HFHI and CCI caused dysregulation in the diurnal expression of core circadian genes (Bmal1, Clock, Per1,2, Cry1,2) at 24 h post-TBI. CCI mice had reduced locomotor activity on running wheels in the first 7 d post-TBI; however, both CCI and HFHI mice were able to maintain circadian behavior cycles even in the absence of light cues. We used implantable EEG to measure sleep cycles and brain activity and found that there were no differences in the time spent awake, in NREM or REM sleep in either TBI model. However, in the sleep states, CCI mice have reduced delta power in NREM sleep and reduced theta power in REM sleep at 7 d post-TBI. Our data reveal that different types of brain trauma can result in distinct patterns of circadian and sleep disruptions and can be used to better understand the etiology of sleep disorders after TBI.
Collapse
|
7
|
Cho FS, Vainchtein ID, Voskobiynyk Y, Morningstar AR, Aparicio F, Higashikubo B, Ciesielska A, Broekaart DWM, Anink JJ, van Vliet EA, Yu X, Khakh BS, Aronica E, Molofsky AV, Paz JT. Enhancing GAT-3 in thalamic astrocytes promotes resilience to brain injury in rodents. Sci Transl Med 2022; 14:eabj4310. [PMID: 35857628 DOI: 10.1126/scitranslmed.abj4310] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Inflammatory processes induced by brain injury are important for recovery; however, when uncontrolled, inflammation can be deleterious, likely explaining why most anti-inflammatory treatments have failed to improve neurological outcomes after brain injury in clinical trials. In the thalamus, chronic activation of glial cells, a proxy of inflammation, has been suggested as an indicator of increased seizure risk and cognitive deficits that develop after cortical injury. Furthermore, lesions in the thalamus, more than other brain regions, have been reported in patients with viral infections associated with neurological deficits, such as SARS-CoV-2. However, the extent to which thalamic inflammation is a driver or by-product of neurological deficits remains unknown. Here, we found that thalamic inflammation in mice was sufficient to phenocopy the cellular and circuit hyperexcitability, enhanced seizure risk, and disruptions in cortical rhythms that develop after cortical injury. In our model, down-regulation of the GABA transporter GAT-3 in thalamic astrocytes mediated this neurological dysfunction. In addition, GAT-3 was decreased in regions of thalamic reactive astrocytes in mouse models of cortical injury. Enhancing GAT-3 in thalamic astrocytes prevented seizure risk, restored cortical states, and was protective against severe chemoconvulsant-induced seizures and mortality in a mouse model of traumatic brain injury, emphasizing the potential of therapeutically targeting this pathway. Together, our results identified a potential therapeutic target for reducing negative outcomes after brain injury.
Collapse
Affiliation(s)
- Frances S Cho
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA.,Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA.,Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ilia D Vainchtein
- Department of Psychiatry/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yuliya Voskobiynyk
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | | | - Francisco Aparicio
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA.,Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Bryan Higashikubo
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | | | - Diede W M Broekaart
- Amsterdam UMC location University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam 1105 AZ, Netherlands
| | - Jasper J Anink
- Amsterdam UMC location University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam 1105 AZ, Netherlands
| | - Erwin A van Vliet
- Amsterdam UMC location University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam 1105 AZ, Netherlands.,Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam 1098 XH, Netherlands
| | - Xinzhu Yu
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.,Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Baljit S Khakh
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.,Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Eleonora Aronica
- Amsterdam UMC location University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam 1105 AZ, Netherlands.,Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede 2103 SW, Netherlands
| | - Anna V Molofsky
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA.,Department of Psychiatry/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA.,Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Jeanne T Paz
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA.,Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA.,Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA.,Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
8
|
Andrade P, Lara-Valderrábano L, Manninen E, Ciszek R, Tapiala J, Ndode-Ekane XE, Pitkänen A. Seizure Susceptibility and Sleep Disturbance as Biomarkers of Epileptogenesis after Experimental TBI. Biomedicines 2022; 10:biomedicines10051138. [PMID: 35625875 PMCID: PMC9138230 DOI: 10.3390/biomedicines10051138] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 05/10/2022] [Accepted: 05/12/2022] [Indexed: 02/06/2023] Open
Abstract
Objectives: We investigated whether seizure susceptibility increases over weeks−months after experimental traumatic brain injury (TBI), and whether seizure susceptibility in rats predicts the development of post-traumatic epilepsy (PTE) or epileptiform activity. We further investigated whether rats develop chronic sleep disturbance after TBI, and whether sleep disturbance parameters—alone or in combination with pentylenetetrazol (PTZ) test parameters—could serve as novel biomarkers for the development of post-traumatic epileptogenesis. Methods: TBI was induced in adult male Sprague-Dawley rats with lateral fluid-percussion injury. Sham-operated experimental controls underwent craniectomy without exposure to an impact force. Seizure susceptibility was tested with a PTZ test (30 mg/kg, intraperitoneally) on day (D) 30, D60, D90, and D180 after TBI (n = 28) or sham operation (n = 16) under video electroencephalogram (vEEG). In the 7th post-injury month, rats underwent continuous vEEG monitoring to detect spontaneous seizures and assess sleep disturbances. At the end of the experiments, rats were perfused for brain histology. Results: In the TBI group, the percentage of rats with PTZ-induced seizures increased over time (adjusted p < 0.05 compared with D30). Combinations of three PTZ test parameters (latency to the first epileptiform discharge (ED), number of EDs, and number of PTZ-induced seizures) survived the leave-one-out validation for differentiating rats with or without epileptiform activity, indicating an area under the receiver operating curve (AUC) of 0.743 (95% CI 0.472−0.992, p = 0.05) with a misclassification rate of 36% on D90, and an AUC of 0.752 (95% CI 0.483−0.929, p < 0.05) with a misclassification rate of 32% on D180. Sleep analysis revealed that the number of transitions to N3 or rapid eye movement (REM) sleep, along with the total number of transitions, was increased in the TBI group during the lights-on period (all p < 0.05). The sleep fragmentation index during the lights-on period was greater in the TBI rats than in sham-operated rats (p < 0.05). A combination of sleep parameters showed promise as diagnostic biomarkers of prior TBI, with an AUC of 0.792 (95% CI 0.549−0.934, p < 0.01) and a misclassification rate of 28%. Rats with epilepsy or any epileptiform activity had more transitions from N3 to the awake stage (p < 0.05), and the number of N3−awake transitions differentiated rats with or without epileptiform activity, with an AUC of 0.857 (95% CI 0.651−1.063, p < 0.01). Combining sleep parameters with PTZ parameters did not improve the biomarker performance. Significance: This is the first attempt to monitor the evolution of seizure susceptibility over months in a well-described rat model of PTE. Our data suggest that assessment of seizure susceptibility and sleep disturbance can provide diagnostic biomarkers of prior TBI and prognostic biomarkers of post-traumatic epileptogenesis.
Collapse
|
9
|
van Alphen B, Stewart S, Iwanaszko M, Xu F, Li K, Rozenfeld S, Ramakrishnan A, Itoh TQ, Sisobhan S, Qin Z, Lear BC, Allada R. Glial immune-related pathways mediate effects of closed head traumatic brain injury on behavior and lethality in Drosophila. PLoS Biol 2022; 20:e3001456. [PMID: 35081110 PMCID: PMC8791498 DOI: 10.1371/journal.pbio.3001456] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 10/22/2021] [Indexed: 02/07/2023] Open
Abstract
In traumatic brain injury (TBI), the initial injury phase is followed by a secondary phase that contributes to neurodegeneration, yet the mechanisms leading to neuropathology in vivo remain to be elucidated. To address this question, we developed a Drosophila head-specific model for TBI termed Drosophila Closed Head Injury (dCHI), where well-controlled, nonpenetrating strikes are delivered to the head of unanesthetized flies. This assay recapitulates many TBI phenotypes, including increased mortality, impaired motor control, fragmented sleep, and increased neuronal cell death. TBI results in significant changes in the transcriptome, including up-regulation of genes encoding antimicrobial peptides (AMPs). To test the in vivo functional role of these changes, we examined TBI-dependent behavior and lethality in mutants of the master immune regulator NF-κB, important for AMP induction, and found that while sleep and motor function effects were reduced, lethality effects were enhanced. Similarly, loss of most AMP classes also renders flies susceptible to lethal TBI effects. These studies validate a new Drosophila TBI model and identify immune pathways as in vivo mediators of TBI effects.
Collapse
Affiliation(s)
- Bart van Alphen
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Samuel Stewart
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Marta Iwanaszko
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
- Department of Preventive Medicine—Biostatistics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Fangke Xu
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Keyin Li
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Sydney Rozenfeld
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Anujaianthi Ramakrishnan
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Taichi Q. Itoh
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Shiju Sisobhan
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Zuoheng Qin
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Bridget C. Lear
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Ravi Allada
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| |
Collapse
|
10
|
Mountney A, Blaze J, Wang Z, Umali M, Flerlage WJ, Dougherty J, Ge Y, Shear D, Haghighi F. Penetrating Ballistic Brain Injury Produces Acute Alterations in Sleep and Circadian-Related Genes in the Rodent Cortex: A Preliminary Study. Front Neurol 2021; 12:745330. [PMID: 34777213 PMCID: PMC8580116 DOI: 10.3389/fneur.2021.745330] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 09/22/2021] [Indexed: 11/23/2022] Open
Abstract
Traumatic brain injury (TBI) affects millions of Americans each year, with extremely high prevalence in the Veteran community, and sleep disturbance is one of the most commonly reported symptoms. Reduction in the quality and amount of sleep can negatively impact recovery and result in a wide range of behavioral and physiological symptoms, such as impaired cognition, mood and anxiety disorders, and cardiovascular effects. Thus, to improve long-term patient outcomes and develop novel treatments, it is essential to understand the molecular mechanisms involved in sleep disturbance following TBI. In this effort, we performed transcriptional profiling in an established rodent model of penetrating ballistic brain injury (PBBI) in conjunction with continuous sleep/wake EEG/EMG recording of the first 24 h after injury. Rats subjected to PBBI showed profound differences in sleep architecture. Injured animals spent significantly more time in slow wave sleep and less time in REM sleep compared to sham control animals. To identify PBBI-related transcriptional differences, we then performed transcriptome-wide gene expression profiling at 24 h post-injury, which identified a vast array of immune- related genes differentially expressed in the injured cortex as well as sleep-related genes. Further, transcriptional changes associated with total time spent in various sleep stages were identified. Such molecular changes may underlie the pathology and symptoms that emerge following TBI, including neurodegeneration, sleep disturbance, and mood disorders.
Collapse
Affiliation(s)
- Andrea Mountney
- Walter Reed Army Institute of Research (WRAIR), Silver Spring, MD, United States
| | - Jennifer Blaze
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Zhaoyu Wang
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Michelle Umali
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | | | - Jacqueline Dougherty
- Walter Reed Army Institute of Research (WRAIR), Silver Spring, MD, United States
| | - Yongchao Ge
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Deborah Shear
- Walter Reed Army Institute of Research (WRAIR), Silver Spring, MD, United States
| | - Fatemeh Haghighi
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Research and Development Service, James J. Peters Veterans Affairs Medical Center, Bronx, NY, United States
| |
Collapse
|
11
|
Necula D, Cho FS, He A, Paz JT. Secondary thalamic neuroinflammation after focal cortical stroke and traumatic injury mirrors corticothalamic functional connectivity. J Comp Neurol 2021; 530:998-1019. [PMID: 34633669 PMCID: PMC8957545 DOI: 10.1002/cne.25259] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 10/03/2021] [Accepted: 10/04/2021] [Indexed: 12/29/2022]
Abstract
While cortical injuries, such as traumatic brain injury (TBI) and neocortical stroke, acutely disrupt the neocortex, most of their consequent disabilities reflect secondary injuries that develop over time. Thalamic neuroinflammation has been proposed to be a biomarker of cortical injury and of the long-term cognitive and neurological deficits that follow. However, the extent to which thalamic neuroinflammation depends on the type of cortical injury or its location remains unknown. Using two mouse models of focal neocortical injury that do not directly damage subcortical structures-controlled cortical impact and photothrombotic ischemic stroke-we found that chronic neuroinflammation in the thalamic region mirrors the functional connections with the injured cortex, and that sensory corticothalamic regions may be more likely to sustain long-term damage than nonsensory circuits. Currently, heterogeneous clinical outcomes complicate treatment. Understanding how thalamic inflammation depends on the injury site can aid in predicting features of subsequent deficits and lead to more effective, customized therapies.
Collapse
Affiliation(s)
- Deanna Necula
- Gladstone Institute of Neurological Disease, San Francisco, California, USA.,Neuroscience Graduate Program, University of California, San Francisco, California, USA.,Department of Neurology and the Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, California, USA
| | - Frances S Cho
- Gladstone Institute of Neurological Disease, San Francisco, California, USA.,Neuroscience Graduate Program, University of California, San Francisco, California, USA.,Department of Neurology and the Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, California, USA
| | - Andrea He
- Gladstone Institute of Neurological Disease, San Francisco, California, USA
| | - Jeanne T Paz
- Gladstone Institute of Neurological Disease, San Francisco, California, USA.,Neuroscience Graduate Program, University of California, San Francisco, California, USA.,Department of Neurology and the Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
12
|
Leng Y, Byers AL, Barnes DE, Peltz CB, Li Y, Yaffe K. Traumatic Brain Injury and Incidence Risk of Sleep Disorders in Nearly 200,000 US Veterans. Neurology 2021; 96:e1792-e1799. [PMID: 33658328 PMCID: PMC8055309 DOI: 10.1212/wnl.0000000000011656] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 12/23/2020] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To test the hypothesis that veterans with traumatic brain injury (TBI) have an increased subsequent risk of sleep disorders, we studied the longitudinal association between TBI and incident sleep disorders in nearly 200,000 veterans. METHODS We performed a cohort study of all patients diagnosed with a TBI in the Veterans Health Administration system from October 1, 2001, to September 30, 2015, who were age-matched 1:1 to veterans without TBI. Veterans with prevalent sleep disorders at baseline were excluded. Development of sleep disorders was defined as any inpatient or outpatient diagnosis of sleep apnea, hypersomnia, insomnia, or sleep-related movement disorders based on ICD-9 codes after the first TBI diagnosis or the random selection date for those without TBI. We restricted the analysis to those with at least 1 year of follow-up. We used Cox proportional hazards models to examine the association between TBI and subsequent risk of sleep disorders. RESULTS The study included 98,709 veterans with TBI and 98,709 age-matched veterans without TBI (age 49 ± 20 years). After an average follow-up of 5 (1-14) years, 23,127 (19.6%) veterans developed sleep disorders. After adjustment for demographics, education, income, and medical and psychiatric conditions, those who had TBI compared to those without TBI were 41% more likely to develop any sleep disorders (hazard ratio 1.41 [95% confidence interval 1.37-1.44]), including sleep apnea (1.28 [1.24-1.32]), insomnia (1.50 [1.45-1.55]), hypersomnia (1.50 [1.39-1.61]), and sleep-related movement disorders (1.33 [1.16-1.52]). The association was stronger for mild TBIs, did not differ appreciably by presence of posttraumatic stress disorder, and remained after a 2-year time lag. CONCLUSION In 197,418 veterans without sleep disorders, those with diagnosed TBI had an increased risk of incident sleep disorders over 14 years. Improved prevention and long-term management strategies for sleep are needed for veterans with TBI.
Collapse
Affiliation(s)
- Yue Leng
- From the Department of Psychiatry (Y. Leng, A.L.B., D.E.B., K.Y.), Department of Epidemiology and Biostatistics (A.L.B., D.E.B., K.Y.), and Department of Neurology (K.Y.), University of California, San Francisco; and San Francisco Veterans Affairs Health Care System (A.L.B., D.E.B., C.B.P., Y. Li., K.Y.), CA.
| | - Amy L Byers
- From the Department of Psychiatry (Y. Leng, A.L.B., D.E.B., K.Y.), Department of Epidemiology and Biostatistics (A.L.B., D.E.B., K.Y.), and Department of Neurology (K.Y.), University of California, San Francisco; and San Francisco Veterans Affairs Health Care System (A.L.B., D.E.B., C.B.P., Y. Li., K.Y.), CA
| | - Deborah E Barnes
- From the Department of Psychiatry (Y. Leng, A.L.B., D.E.B., K.Y.), Department of Epidemiology and Biostatistics (A.L.B., D.E.B., K.Y.), and Department of Neurology (K.Y.), University of California, San Francisco; and San Francisco Veterans Affairs Health Care System (A.L.B., D.E.B., C.B.P., Y. Li., K.Y.), CA
| | - Carrie B Peltz
- From the Department of Psychiatry (Y. Leng, A.L.B., D.E.B., K.Y.), Department of Epidemiology and Biostatistics (A.L.B., D.E.B., K.Y.), and Department of Neurology (K.Y.), University of California, San Francisco; and San Francisco Veterans Affairs Health Care System (A.L.B., D.E.B., C.B.P., Y. Li., K.Y.), CA
| | - Yixia Li
- From the Department of Psychiatry (Y. Leng, A.L.B., D.E.B., K.Y.), Department of Epidemiology and Biostatistics (A.L.B., D.E.B., K.Y.), and Department of Neurology (K.Y.), University of California, San Francisco; and San Francisco Veterans Affairs Health Care System (A.L.B., D.E.B., C.B.P., Y. Li., K.Y.), CA
| | - Kristine Yaffe
- From the Department of Psychiatry (Y. Leng, A.L.B., D.E.B., K.Y.), Department of Epidemiology and Biostatistics (A.L.B., D.E.B., K.Y.), and Department of Neurology (K.Y.), University of California, San Francisco; and San Francisco Veterans Affairs Health Care System (A.L.B., D.E.B., C.B.P., Y. Li., K.Y.), CA.
| |
Collapse
|
13
|
Konduru SS, Wallace EP, Pfammatter JA, Rodrigues PV, Jones MV, Maganti RK. Sleep-wake characteristics in a mouse model of severe traumatic brain injury: Relation to posttraumatic epilepsy. Epilepsia Open 2021; 6:181-194. [PMID: 33681661 PMCID: PMC7918302 DOI: 10.1002/epi4.12462] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 12/01/2022] Open
Abstract
Study objectives Traumatic brain injury (TBI) results in sequelae that include posttraumatic epilepsy (PTE) and sleep-wake disturbances. Here, we sought to determine whether sleep characteristics could predict development of PTE in a model of severe TBI. Methods Following controlled cortical impact (CCI) or sham injury (craniotomy only), CD-1 mice were implanted with epidural electroencephalography (EEG) and nuchal electromyography (EMG) electrodes. Acute (1st week) and chronic (months 1, 2, or 3) 1-week-long video-EEG recordings were performed after the injury to examine epileptiform activity. High-amplitude interictal events were extracted from EEG using an automated method. After scoring sleep-wake patterns, sleep spindles and EEG delta power were derived from nonrapid eye movement (NREM) sleep epochs. Brain CTs (computerized tomography) were performed in sham and CCI cohorts to quantify the brain lesions. We then employed a no craniotomy (NC) control to perform 1-week-long EEG recordings at week 1 and month 1 after surgery. Results Posttraumatic seizures were seen in the CCI group only, whereas interictal epileptiform activity was seen in CCI or sham. Sleep-wake disruptions consisted of shorter wake or NREM bout lengths and shorter duration or lower power for spindles in CCI and sham. NREM EEG delta power increased in CCI and sham groups compared with NC though the CCI group with posttraumatic seizures had lower power at a chronic time point compared with those without. Follow-up brain CTs showed a small lesion in the sham injury group suggesting a milder form of TBI that may account for their interictal activity and sleep changes. Significance In our TBI model, tracking changes in NREM delta power distinguishes between CCI acutely and animals that will eventually develop PTE, but further work is necessary to identify sleep biomarkers of PTE. Employing NC controls together with sham controls should be considered in future TBI studies.
Collapse
Affiliation(s)
- Sai Sruthi Konduru
- Department of NeurologyUniversity of Wisconsin School of Medicine and Public HealthMadisonWIUSA
| | - Eli P. Wallace
- Department of NeurologyUniversity of Wisconsin School of Medicine and Public HealthMadisonWIUSA
- Department of NeuroscienceUniversity of Wisconsin School of Medicine and Public HealthMadisonWIUSA
- Cellular and Molecular Pathology Graduate ProgramUniversity of Wisconsin School of Medicine and Public HealthMadisonWIUSA
| | - Jesse A. Pfammatter
- Department of NeuroscienceUniversity of Wisconsin School of Medicine and Public HealthMadisonWIUSA
| | - Paulo V. Rodrigues
- Department of NeurologyUniversity of Wisconsin School of Medicine and Public HealthMadisonWIUSA
| | - Mathew V. Jones
- Department of NeuroscienceUniversity of Wisconsin School of Medicine and Public HealthMadisonWIUSA
| | - Rama K. Maganti
- Department of NeurologyUniversity of Wisconsin School of Medicine and Public HealthMadisonWIUSA
| |
Collapse
|
14
|
Krishna G, Bromberg C, Connell EC, Mian E, Hu C, Lifshitz J, Adelson PD, Thomas TC. Traumatic Brain Injury-Induced Sex-Dependent Changes in Late-Onset Sensory Hypersensitivity and Glutamate Neurotransmission. Front Neurol 2020; 11:749. [PMID: 32849211 PMCID: PMC7419702 DOI: 10.3389/fneur.2020.00749] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 06/17/2020] [Indexed: 01/15/2023] Open
Abstract
Women approximate one-third of the annual 2.8 million people in the United States who sustain traumatic brain injury (TBI). Several clinical reports support or refute that menstrual cycle-dependent fluctuations in sex hormones are associated with severity of persisting post-TBI symptoms. Previously, we reported late-onset sensory hypersensitivity to whisker stimulation that corresponded with changes in glutamate neurotransmission at 1-month following diffuse TBI in male rats. Here, we incorporated intact age-matched naturally cycling females into the experimental design while monitoring daily estrous cycle. We hypothesized that sex would not influence late-onset sensory hypersensitivity and associated in vivo amperometric extracellular recordings of glutamate neurotransmission within the behaviorally relevant thalamocortical circuit. At 28 days following midline fluid percussion injury (FPI) or sham surgery, young adult Sprague-Dawley rats were tested for hypersensitivity to whisker stimulation using the whisker nuisance task (WNT). As predicted, both male and female rats showed significantly increased sensory hypersensitivity to whisker stimulation after FPI, with females having an overall decrease in whisker nuisance scores (sex effect), but no injury and sex interaction. In males, FPI increased potassium chloride (KCl)-evoked glutamate overflow in primary somatosensory barrel cortex (S1BF) and ventral posteromedial nucleus of the thalamus (VPM), while in females the FPI effect was discernible only within the VPM. Similar to our previous report, we found the glutamate clearance parameters were not influenced by FPI, while a sex-specific effect was evident with female rats showing a lower uptake rate constant both in S1BF and VPM and longer clearance time (in S1BF) in comparison to male rats. Fluctuations in estrous cycle were evident among brain-injured females with longer diestrus (low circulating hormone) phase of the cycle over 28 days post-TBI. Together, these findings add to growing evidence indicating both similarities and differences between sexes in a chronic response to TBI. A better understanding of the influence of gonadal hormones on behavior, neurotransmission, secondary injury and repair processes after TBI is needed both clinically and translationally, with potential impact on acute treatment, rehabilitation, and symptom management.
Collapse
Affiliation(s)
- Gokul Krishna
- Department of Child Health, University of Arizona College of Medicine – Phoenix, Phoenix, AZ, United States
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States
| | - Caitlin Bromberg
- Department of Child Health, University of Arizona College of Medicine – Phoenix, Phoenix, AZ, United States
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States
| | - Emily Charlotte Connell
- Department of Child Health, University of Arizona College of Medicine – Phoenix, Phoenix, AZ, United States
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Erum Mian
- Department of Child Health, University of Arizona College of Medicine – Phoenix, Phoenix, AZ, United States
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States
| | - Chengcheng Hu
- Department of Epidemiology and Biostatistics, University of Arizona, Tucson, AZ, United States
| | - Jonathan Lifshitz
- Department of Child Health, University of Arizona College of Medicine – Phoenix, Phoenix, AZ, United States
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States
- Phoenix VA Health Care System, Phoenix, AZ, United States
| | - P. David Adelson
- Department of Child Health, University of Arizona College of Medicine – Phoenix, Phoenix, AZ, United States
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States
| | - Theresa Currier Thomas
- Department of Child Health, University of Arizona College of Medicine – Phoenix, Phoenix, AZ, United States
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States
- Phoenix VA Health Care System, Phoenix, AZ, United States
| |
Collapse
|
15
|
Uddin O, Studlack PE, Parihar S, Keledjian K, Cruz A, Farooq T, Shin N, Gerzanich V, Simard JM, Keller A. Chronic pain after blast-induced traumatic brain injury in awake rats. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2019; 6:100030. [PMID: 31223145 PMCID: PMC6565615 DOI: 10.1016/j.ynpai.2019.100030] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 02/14/2019] [Accepted: 04/01/2019] [Indexed: 12/14/2022]
Abstract
Explosive blast-induced traumatic brain injury (blast-TBI) in military personnel is a leading cause of injury and persistent neurological abnormalities, including chronic pain. We previously demonstrated that chronic pain after spinal cord injury results from central sensitization in the posterior thalamus (PO). The presence of persistent headaches and back pain in veterans with blast-TBI suggests a similar involvement of thalamic sensitization. Here, we tested the hypothesis that pain after blast-TBI is associated with abnormal increases in activity of neurons in PO thalamus. We developed a novel model with two unique features: (1) blast-TBI was performed in awake, un-anesthetized rats, to simulate the human experience and to eliminate confounds of anesthesia and surgery inherent in other models; (2) only the cranium, rather than the entire body, was exposed to a collimated blast wave, with the blast wave striking the posterior cranium in the region of the occipital crest and foramen magnum. Three weeks after blast-TBI, rats developed persistent, ongoing spontaneous pain. Contrary to our hypothesis, we found no significant differences in the activity of PO neurons, or of neurons in the spinal trigeminal nucleus. There were also no significant changes in gliosis in either of these structures. This novel model will allow future studies on the pathophysiology of chronic pain after blast-TBI.
Collapse
Affiliation(s)
- Olivia Uddin
- Department of Anatomy and Neurobiology and Program in Neuroscience, University of Maryland School of Medicine, 20 Penn St, HSF-II S251, Baltimore, MD, USA
| | - Paige E. Studlack
- Department of Anatomy and Neurobiology and Program in Neuroscience, University of Maryland School of Medicine, 20 Penn St, HSF-II S251, Baltimore, MD, USA
| | - Saitu Parihar
- Department of Anatomy and Neurobiology and Program in Neuroscience, University of Maryland School of Medicine, 20 Penn St, HSF-II S251, Baltimore, MD, USA
| | - Kaspar Keledjian
- Department of Neurosurgery, University of Maryland School of Medicine, 10 S Pine St, MSTF 634B, Baltimore, MD, USA
| | - Alexis Cruz
- Department of Anatomy and Neurobiology and Program in Neuroscience, University of Maryland School of Medicine, 20 Penn St, HSF-II S251, Baltimore, MD, USA
| | - Tayyiaba Farooq
- Department of Anatomy and Neurobiology and Program in Neuroscience, University of Maryland School of Medicine, 20 Penn St, HSF-II S251, Baltimore, MD, USA
| | - Naomi Shin
- Department of Anatomy and Neurobiology and Program in Neuroscience, University of Maryland School of Medicine, 20 Penn St, HSF-II S251, Baltimore, MD, USA
| | - Volodymyr Gerzanich
- Department of Neurosurgery, University of Maryland School of Medicine, 10 S Pine St, MSTF 634B, Baltimore, MD, USA
| | - J. Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, 10 S Pine St, MSTF 634B, Baltimore, MD, USA
- Department of Pathology, University of Maryland School of Medicine, 10 S Pine St, MSTF, Room 634B, Baltimore, MD, USA
- Department of Physiology, University of Maryland School of Medicine, 10 S Pine St, MSTF, Room 634B, Baltimore, MD, USA
| | - Asaf Keller
- Department of Anatomy and Neurobiology and Program in Neuroscience, University of Maryland School of Medicine, 20 Penn St, HSF-II S251, Baltimore, MD, USA
| |
Collapse
|
16
|
Thomasy HE, Opp MR. Hypocretin Mediates Sleep and Wake Disturbances in a Mouse Model of Traumatic Brain Injury. J Neurotrauma 2019; 36:802-814. [PMID: 30136622 PMCID: PMC6387567 DOI: 10.1089/neu.2018.5810] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Traumatic brain injury (TBI) is a major cause of disability worldwide. Post-TBI sleep and wake disturbances are extremely common and difficult for patients to manage. Sleep and wake disturbances contribute to poor functional and emotional outcomes from TBI, yet effective therapies remain elusive. A more comprehensive understanding of mechanisms underlying post-TBI sleep and wake disturbance will facilitate development of effective pharmacotherapies. Previous research in human patients and animal models indicates that altered hypocretinergic function may be a major contributor to sleep-wake disturbance after TBI. In this study, we further elucidate the role of hypocretin by determining the impact of TBI on sleep-wake behavior of hypocretin knockout (HCRT KO) mice. Adult male C57BL/6J and HCRT KO mice were implanted with electroencephalography recording electrodes, and pre-injury baseline recordings were obtained. Mice were then subjected to either moderate TBI or sham surgery. Additional recordings were obtained and sleep-wake behavior determined at 3, 7, 15, and 30 days after TBI or sham procedures. At baseline, HCRT KO mice had a significantly different sleep-wake phenotype than control C57BL/6J mice. Post-TBI sleep-wake behavior was altered in a genotype-dependent manner: sleep of HCRT KO mice was not altered by TBI, whereas C57BL/6J mice had more non-rapid eye movement sleep, less wakefulness, and more short wake bouts and fewer long wake bouts. Numbers of hypocretin-positive cells were reduced in C57BL/6J mice by TBI. Collectively, these data indicate that the hypocretinergic system is involved in the alterations in sleep-wake behavior that develop after TBI in this model, and suggest potential therapeutic interventions.
Collapse
Affiliation(s)
- Hannah E. Thomasy
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington
| | - Mark R. Opp
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington
- Graduate Program in Neurobiology and Behavior, University of Washington, Seattle, Washington
| |
Collapse
|
17
|
Zhou X, Oishi Y, Cherasse Y, Korkutata M, Fujii S, Lee CY, Lazarus M. Extracellular adenosine and slow-wave sleep are increased after ablation of nucleus accumbens core astrocytes and neurons in mice. Neurochem Int 2019; 124:256-263. [PMID: 30690114 DOI: 10.1016/j.neuint.2019.01.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 01/07/2019] [Accepted: 01/19/2019] [Indexed: 02/06/2023]
Abstract
Sleep and wakefulness are controlled by a wide range of neuronal populations in the mammalian brain. Activation of adenosine A2A receptor (A2AR)-expressing neurons in the nucleus accumbens (NAc) core promotes slow-wave sleep (SWS). The neuronal mechanism by which activation of NAc A2AR neurons induces SWS, however, is unknown. We hypothesized that the ability of NAc activation to induce sleep is mediated by the classic somnogen adenosine, which can be formed by various processes in all types of cells. Here, to investigate whether astrocytes are involved in the ability of the NAc to regulate SWS, we ablated glial fibrillary acidic protein (GFAP)-positive cells in the NAc core of mice by virus-mediated expression of diphtheria toxin (DT) receptors and intraperitoneal administration of DT. Analysis of electroencephalogram and electromyogram recordings of DT-treated wild-type mice revealed that SWS was remarkably increased at 1 week after DT treatment, whereas sleep-wake behavior was unchanged in DT-treated A2AR knockout mice. Cell ablation was associated with an increased number of GFAP-positive cells and activation of microglia in the NAc. In-vivo microdialysis revealed significantly increased levels of extracellular adenosine in the NAc at 1 week after DT treatment. Our findings suggest that elevated adenosine levels in the NAc core promote SWS by acting on A2ARs and provide the first evidence that adenosine is an endogenous candidate for activating NAc A2AR neurons that have the ability to induce SWS.
Collapse
Affiliation(s)
- Xuzhao Zhou
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan; Doctoral Program of Biomedical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Yo Oishi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Yoan Cherasse
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Mustafa Korkutata
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan; PhD Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, Tsukuba, Ibaraki, 305-0005, Japan
| | - Shinya Fujii
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan; Doctoral Program of Biomedical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Chia-Ying Lee
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan; PhD Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, Tsukuba, Ibaraki, 305-0005, Japan
| | - Michael Lazarus
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan.
| |
Collapse
|
18
|
Yu M, Wang M, Yang D, Wei X, Li W. Dynamics of blood brain barrier permeability and tissue microstructure following controlled cortical impact injury in rat: A dynamic contrast-enhanced magnetic resonance imaging and diffusion kurtosis imaging study. Magn Reson Imaging 2019; 62:1-9. [PMID: 30660704 DOI: 10.1016/j.mri.2019.01.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 01/15/2019] [Accepted: 01/16/2019] [Indexed: 01/09/2023]
Abstract
OBJECTIVE The blood-brain barrier (BBB) and cerebral tissue microstructure can be impaired following traumatic brain injury (TBI). However, the spatiotemporal changes of BBB leakage and tissue microstructure are not completely understood. In this study, we evaluated the spatiotemporal changes of BBB leakage and tissue microstructure using dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) and diffusion kurtosis imaging (DKI) in controlled cortical impact (CCI) rats. MATERIALS AND METHODS The DCE-MRI parameters volume transfer coefficient (Ktrans) and DKI parameters were longitudinally measured in bilateral cortex, hippocampus, thalamus and corpus callosum (CC) at baseline (D0), acute stage (D1, D3), and subacute stage (D7, D14 and D28) post-injury. Immunohistochemistry analysis was performed at D28 after MRI scanning. Repeated-measures ANOVA was used to assess the temporal changes of MRI parameters. RESULTS Ktrans abnormality was only localized to ipsilateral perilesional cortex with a significant temporal change (F = 144.2, p < 0.0001). Compared to baseline, increased mean kurtosis (MK) was observed in ipsilateral regions of cortex and hippocampus and CC for all the time points (p < 0.05 for all). Increased MK was also observed in ipsilateral thalamus (p = 0.005) at subacute stage but not at acute stage while no change was observed with MD and FA (p > 0.05 for both). In ipsilateral cortex, the overall Ktrans value of D0, D1, D3, D7, D14, and D28 post-injury were significantly correlated with MK value (r = 0.84, p < 0.0001). The CCI group showed higher staining of glial fibrillary acidic protein (GFAP) and ionized calcium binding adaptor molecule 1 (Iba-1) and lower staining of neuron-specific nuclear protein (NeuN) and myelin basic protein (MBP) in ipsilateral regions of cortex, hippocampus, thalamus and CC (p < 0.05 for all) as compared to control group. There were no significant differences in the contralateral regions by immunohistochemistry. CONCLUSION The BBB disruption reflected by Ktrans correlated well with MK value in ipsilateral cortex. In addition, MK could detect the delayed microstructural changes in thalamus. DCE-MRI and DKI could be used to assess the BBB breakdown and cerebral microstructural changes of TBI.
Collapse
Affiliation(s)
- Mengmeng Yu
- Department of Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Mingliang Wang
- Department of Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Dianxu Yang
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Xiaoer Wei
- Department of Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Wenbin Li
- Department of Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China; Imaging Center, Kashgar Prefecture Second People's Hospital, Kashgar 844000, Xinjiang, China.
| |
Collapse
|
19
|
Corne R, Leconte C, Ouradou M, Fassina V, Zhu Y, Déou E, Besson V, Plotkine M, Marchand-Leroux C, Mongeau R. Spontaneous resurgence of conditioned fear weeks after successful extinction in brain injured mice. Prog Neuropsychopharmacol Biol Psychiatry 2019; 88:276-286. [PMID: 30096331 DOI: 10.1016/j.pnpbp.2018.07.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 06/28/2018] [Accepted: 07/29/2018] [Indexed: 12/25/2022]
Abstract
Mild traumatic brain injury (TBI) is a major risk factor for post-traumatic stress disorder (PTSD), and both disorders share common symptoms and neurobiological defects. Relapse after successful treatment, known as long-term fear resurgence, is common in PTSD patients and a major therapeutic hurdle. We induced a mild focal TBI by controlled cortical impact (CCI) in male C57BL/6 J mice and used fear conditioning to assess PTSD-like behaviors and concomitant alterations in the fear circuitry. We found for the first time that mild TBI, and to a lesser extent sham (craniotomy), mice displayed a spontaneous resurgence of conditioned fear when tested for fear extinction memory recall, despite having effectively acquired and extinguished conditioned fear 6 weeks earlier in the same context. Other characteristic symptoms of PTSD are risk-taking behaviors and cognitive deficits. CCI mice displayed risk-taking behaviors, behavioral inflexibility and reductions in processing speed compared to naïve mice. In conjunction with these changes there were alterations in amygdala morphology 3 months post-trauma, and decreased myelin basic protein density at the primary lesion site and in distant secondary sites such as the hippocampus, thalamus, and amygdala, compared to sham mice. Furthermore, activity-dependent brain-derived neurotrophic factor (BDNF) transcripts were decreased in the prefrontal cortex, a key region for fear extinction consolidation, following fear extinction training in both TBI and, to a lesser extent, sham mice. This study shows for the first time that a mild brain injury can generate a spontaneous resurgence of conditioned fear associated with defective BDNF signalling in the prefrontal cortex, PTSD-like behaviors, and have enduring effects on the brain.
Collapse
Affiliation(s)
- R Corne
- EA4475 - Pharmacologie de la Circulation Cérébrale, Faculté de Pharmacie de Paris, Université Paris Descartes, Paris, France
| | - C Leconte
- EA4475 - Pharmacologie de la Circulation Cérébrale, Faculté de Pharmacie de Paris, Université Paris Descartes, Paris, France
| | - M Ouradou
- EA4475 - Pharmacologie de la Circulation Cérébrale, Faculté de Pharmacie de Paris, Université Paris Descartes, Paris, France
| | - V Fassina
- EA4475 - Pharmacologie de la Circulation Cérébrale, Faculté de Pharmacie de Paris, Université Paris Descartes, Paris, France
| | - Y Zhu
- EA4475 - Pharmacologie de la Circulation Cérébrale, Faculté de Pharmacie de Paris, Université Paris Descartes, Paris, France
| | - E Déou
- EA4475 - Pharmacologie de la Circulation Cérébrale, Faculté de Pharmacie de Paris, Université Paris Descartes, Paris, France
| | - V Besson
- EA4475 - Pharmacologie de la Circulation Cérébrale, Faculté de Pharmacie de Paris, Université Paris Descartes, Paris, France
| | - M Plotkine
- EA4475 - Pharmacologie de la Circulation Cérébrale, Faculté de Pharmacie de Paris, Université Paris Descartes, Paris, France
| | - C Marchand-Leroux
- EA4475 - Pharmacologie de la Circulation Cérébrale, Faculté de Pharmacie de Paris, Université Paris Descartes, Paris, France
| | - R Mongeau
- EA4475 - Pharmacologie de la Circulation Cérébrale, Faculté de Pharmacie de Paris, Université Paris Descartes, Paris, France.
| |
Collapse
|
20
|
Zhou Y, Greenwald BD. Update on Insomnia after Mild Traumatic Brain Injury. Brain Sci 2018; 8:brainsci8120223. [PMID: 30551607 PMCID: PMC6315624 DOI: 10.3390/brainsci8120223] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 12/08/2018] [Accepted: 12/11/2018] [Indexed: 12/16/2022] Open
Abstract
Sleep disturbance after traumatic brain injury (TBI) has received growing interest in recent years, garnering many publications. Insomnia is highly prevalent within the mild traumatic brain injury (mTBI) population and is a subtle, frequently persistent complaint that often goes undiagnosed. For individuals with mTBI, problems with sleep can compromise the recovery process and impede social reintegration. This article updates the evidence on etiology, epidemiology, prognosis, consequences, differential diagnosis, and treatment of insomnia in the context of mild TBI. This article aims to increase awareness about insomnia following mTBI in the hopes that it may improve diagnosis, evaluation, and treatment of sleeping disturbance in this population while revealing areas for future research.
Collapse
Affiliation(s)
- Yi Zhou
- Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA.
| | | |
Collapse
|
21
|
Chronic Upregulation of Cleaved-Caspase-3 Associated with Chronic Myelin Pathology and Microvascular Reorganization in the Thalamus after Traumatic Brain Injury in Rats. Int J Mol Sci 2018; 19:ijms19103151. [PMID: 30322151 PMCID: PMC6214127 DOI: 10.3390/ijms19103151] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 10/06/2018] [Accepted: 10/09/2018] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) is associated with long-term disabilities and devastating chronic neurological complications including problems with cognition, motor function, sensory processing, as well as behavioral deficits and mental health problems such as anxiety, depression, personality change and social unsuitability. Clinical data suggest that disruption of the thalamo-cortical system including anatomical and metabolic changes in the thalamus following TBI might be responsible for some chronic neurological deficits following brain trauma. Detailed mechanisms of these pathological processes are not completely understood. The goal of this study was to evaluate changes in the thalamus following TBI focusing on cleaved-caspase-3, a specific effector of caspase pathway activation and myelin and microvascular pathologies using immuno- and histochemistry at different time points from 24 h to 3 months after controlled cortical impact (CCI) in adult Sprague-Dawley rats. Significant increases in cleaved-caspase-3 immunoreactivity in the thalamus were observed starting one month and persisting for at least three months following experimental TBI. Further, the study demonstrated an association of cleaved-caspase-3 with the demyelination of neuronal processes and tissue degeneration in the gray matter in the thalamus, as reflected in alterations of myelinated fiber integrity (luxol fast blue) and decreases in myelin basic protein (MBP) immunoreactivity. The immunofluorescent counterstaining of cleaved-caspase-3 with endothelial barrier antigen (EBA), a marker of blood-brain barrier, revealed limited direct and indirect associations of cleaved caspase-3 with blood-brain barrier damage. These results demonstrate for the first time a significant chronic upregulation of cleaved-caspase-3 in selected thalamic regions associated with cortical regions directly affected by CCI injury. Further, our study is also the first to report that significant upregulation of cleaved-caspase-3 in selected ipsilateral thalamic regions is associated with microvascular reorganization reflected in the significant increases in the number of microvessels with blood-brain barrier alterations detected by EBA staining. These findings provide new insights into potential mechanisms of TBI cell death involving chronic activation of caspase-3 associated with disrupted cortico-thalamic and thalamo-cortical connectivity. Moreover, this study offers the initial evidence that this upregulation of activated caspase-3, delayed degeneration of myelinated nerve fibers and microvascular reorganization with impaired blood-brain barrier integrity in the thalamus might represent reciprocal pathological processes affecting neuronal networks and brain function at the chronic stages of TBI.
Collapse
|
22
|
Elliott MB, Ward SJ, Abood ME, Tuma RF, Jallo JI. Understanding the endocannabinoid system as a modulator of the trigeminal pain response to concussion. Concussion 2018; 2:CNC49. [PMID: 30202590 PMCID: PMC6122691 DOI: 10.2217/cnc-2017-0010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 08/29/2017] [Indexed: 12/29/2022] Open
Abstract
Post-traumatic headache is the most common symptom of postconcussion syndrome and becomes a chronic neurological disorder in a substantial proportion of patients. This review provides a brief overview of the epidemiology of postconcussion headache, research models used to study this disorder, as well as the proposed mechanisms. An objective of this review is to enhance the understanding of how the endogenous cannabinoid system is essential for maintaining the balance of the CNS and regulating inflammation after injury, and in turn making the endocannabinoid system a potential modulator of the trigeminal response to concussion. The review describes the role of endocannabinoid modulation of pain and the potential for use of phytocannabinoids to treat pain, migraine and concussion.
Collapse
Affiliation(s)
- Melanie B Elliott
- Department of Neurosurgery, Vickie & Jack Farber Institute for Neuroscience Thomas Jefferson University, PA 19107, USA.,Department of Neurosurgery, Vickie & Jack Farber Institute for Neuroscience Thomas Jefferson University, PA 19107, USA
| | - Sara J Ward
- Department of Pharmacology, Lewis Katz School of Medicine, Temple University, PA 19140, USA.,Department of Pharmacology, Lewis Katz School of Medicine, Temple University, PA 19140, USA
| | - Mary E Abood
- Department of Anatomy & Cell Biology, Lewis Katz School of Medicine, Temple University, PA 19140, USA.,Department of Anatomy & Cell Biology, Lewis Katz School of Medicine, Temple University, PA 19140, USA
| | - Ronald F Tuma
- Department of Physiology Lewis Katz School of Medicine, Temple University, PA 19140, USA.,Department of Physiology Lewis Katz School of Medicine, Temple University, PA 19140, USA
| | - Jack I Jallo
- Department of Neurosurgery, Vickie & Jack Farber Institute for Neuroscience Thomas Jefferson University, PA 19107, USA.,Department of Neurosurgery, Vickie & Jack Farber Institute for Neuroscience Thomas Jefferson University, PA 19107, USA
| |
Collapse
|
23
|
Falnikar A, Stratton J, Lin R, Andrews CE, Tyburski A, Trovillion VA, Gottschalk C, Ghosh B, Iacovitti L, Elliott MB, Lepore AC. Differential Response in Novel Stem Cell Niches of the Brain after Cervical Spinal Cord Injury and Traumatic Brain Injury. J Neurotrauma 2018; 35:2195-2207. [PMID: 29471717 DOI: 10.1089/neu.2017.5497] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Populations of neural stem cells (NSCs) reside in a number of defined niches in the adult central nervous system (CNS) where they continually give rise to mature cell types throughout life, including newly born neurons. In addition to the prototypical niches of the subventricular zone (SVZ) and subgranular zone (SGZ) of the hippocampal dentate gyrus, novel stem cell niches that are also neurogenic have recently been identified in multiple midline structures, including circumventricular organs (CVOs) of the brain. These resident NSCs serve as a homeostatic source of new neurons and glial cells under intact physiological conditions. Importantly, they may also have the potential for reparative processes in pathological states such as traumatic spinal cord injury (SCI) and traumatic brain injury (TBI). As the response in these novel CVO stem cell niches has been characterized after stroke but not following SCI or TBI, we quantitatively assessed cell proliferation and the neuronal and glial lineage fate of resident NSCs in three CVO nuclei-area postrema (AP), median eminence (ME), and subfornical organ (SFO) -in rat models of cervical contusion-type SCI and controlled cortical impact (CCI)-induced TBI. Using bromodeoxyuridine (BrdU) labeling of proliferating cells, we find that TBI significantly enhanced proliferation in AP, ME, and SFO, whereas cervical SCI had no effects at early or chronic time-points post-injury. In addition, SCI did not alter NSC differentiation profile into doublecortin-positive neuroblasts, GFAP-expressing astrocytes, or Olig2-labeled cells of the oligodendrocyte lineage within AP, ME, or SFO at both time-points. In contrast, CCI induced a pronounced increase in Sox2- and doublecortin-labeled cells in the AP and Iba1-labeled microglia in the SFO. Lastly, plasma derived from CCI animals significantly increased NSC expansion in an in vitro neurosphere assay, whereas plasma from SCI animals did not exert such an effect, suggesting that signaling factors present in blood may be relevant to stimulating CVO niches after CNS injury and may explain the differential in vivo effects of SCI and TBI on the novel stem cell niches.
Collapse
Affiliation(s)
- Aditi Falnikar
- 1 Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Jarred Stratton
- 2 Department of Neurological Surgery, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Ruihe Lin
- 1 Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Carrie E Andrews
- 1 Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Ashley Tyburski
- 2 Department of Neurological Surgery, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Victoria A Trovillion
- 1 Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Chelsea Gottschalk
- 1 Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Biswarup Ghosh
- 1 Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Lorraine Iacovitti
- 1 Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Melanie B Elliott
- 1 Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia, Pennsylvania.,2 Department of Neurological Surgery, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Angelo C Lepore
- 1 Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia, Pennsylvania
| |
Collapse
|
24
|
Noain D, Büchele F, Schreglmann SR, Valko PO, Gavrilov YV, Morawska MM, Imbach LL, Baumann CR. Increased Sleep Need and Reduction of Tuberomammillary Histamine Neurons after Rodent Traumatic Brain Injury. J Neurotrauma 2018; 35:85-93. [DOI: 10.1089/neu.2017.5067] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Daniela Noain
- Department of Neurology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Fabian Büchele
- Department of Neurology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Sebastian R. Schreglmann
- Department of Neurology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Philipp O. Valko
- Department of Neurology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Yuri V. Gavrilov
- Department of Neurology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Marta M. Morawska
- Department of Neurology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Lukas L. Imbach
- Department of Neurology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Christian R. Baumann
- Department of Neurology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
25
|
Effects of experimental traumatic brain injury and impaired glutamate transport on cortical spreading depression. Exp Neurol 2017; 295:155-161. [DOI: 10.1016/j.expneurol.2017.05.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 04/14/2017] [Accepted: 05/03/2017] [Indexed: 12/21/2022]
|
26
|
Kim Y, Fu AH, Tucker LB, Liu J, McCabe JT. Characterization of controlled cortical impact devices by high-speed image analysis. J Neurosci Res 2017; 96:501-511. [DOI: 10.1002/jnr.24099] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 04/27/2017] [Accepted: 05/16/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Yeonho Kim
- Department of Anatomy, Physiology & Genetics, F.E. Hébert School of Medicine; Uniformed Services University of the Health Sciences; Bethesda MD 20814 USA
- Pre-Clinical Studies Core, Center for Neuroscience and Regenerative Medicine, F.E. Hébert School of Medicine; Uniformed Services University of the Health Sciences; Bethesda MD 20814 USA
| | - Amanda H. Fu
- Department of Anatomy, Physiology & Genetics, F.E. Hébert School of Medicine; Uniformed Services University of the Health Sciences; Bethesda MD 20814 USA
- Pre-Clinical Studies Core, Center for Neuroscience and Regenerative Medicine, F.E. Hébert School of Medicine; Uniformed Services University of the Health Sciences; Bethesda MD 20814 USA
| | - Laura B. Tucker
- Department of Anatomy, Physiology & Genetics, F.E. Hébert School of Medicine; Uniformed Services University of the Health Sciences; Bethesda MD 20814 USA
- Pre-Clinical Studies Core, Center for Neuroscience and Regenerative Medicine, F.E. Hébert School of Medicine; Uniformed Services University of the Health Sciences; Bethesda MD 20814 USA
| | - Jiong Liu
- Department of Anatomy, Physiology & Genetics, F.E. Hébert School of Medicine; Uniformed Services University of the Health Sciences; Bethesda MD 20814 USA
| | - Joseph T. McCabe
- Department of Anatomy, Physiology & Genetics, F.E. Hébert School of Medicine; Uniformed Services University of the Health Sciences; Bethesda MD 20814 USA
- Pre-Clinical Studies Core, Center for Neuroscience and Regenerative Medicine, F.E. Hébert School of Medicine; Uniformed Services University of the Health Sciences; Bethesda MD 20814 USA
| |
Collapse
|
27
|
Sandsmark DK, Elliott JE, Lim MM. Sleep-Wake Disturbances After Traumatic Brain Injury: Synthesis of Human and Animal Studies. Sleep 2017; 40:3074241. [PMID: 28329120 PMCID: PMC6251652 DOI: 10.1093/sleep/zsx044] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2017] [Indexed: 12/23/2022] Open
Abstract
Sleep-wake disturbances following traumatic brain injury (TBI) are increasingly recognized as a serious consequence following injury and as a barrier to recovery. Injury-induced sleep-wake disturbances can persist for years, often impairing quality of life. Recently, there has been a nearly exponential increase in the number of primary research articles published on the pathophysiology and mechanisms underlying sleep-wake disturbances after TBI, both in animal models and in humans, including in the pediatric population. In this review, we summarize over 200 articles on the topic, most of which were identified objectively using reproducible online search terms in PubMed. Although these studies differ in terms of methodology and detailed outcomes; overall, recent research describes a common phenotype of excessive daytime sleepiness, nighttime sleep fragmentation, insomnia, and electroencephalography spectral changes after TBI. Given the heterogeneity of the human disease phenotype, rigorous translation of animal models to the human condition is critical to our understanding of the mechanisms and of the temporal course of sleep-wake disturbances after injury. Arguably, this is most effectively accomplished when animal and human studies are performed by the same or collaborating research programs. Given the number of symptoms associated with TBI that are intimately related to, or directly stem from sleep dysfunction, sleep-wake disorders represent an important area in which mechanistic-based therapies may substantially impact recovery after TBI.
Collapse
Affiliation(s)
| | - Jonathan E Elliott
- VA Portland Health Care System, Portland, OR
- Department of Neurology, Oregon Health & Science University, Portland, OR
| | - Miranda M Lim
- VA Portland Health Care System, Portland, OR
- Department of Neurology, Oregon Health & Science University, Portland, OR
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR; Department of Behavioral Neuroscience, Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR
| |
Collapse
|
28
|
Tyburski AL, Cheng L, Assari S, Darvish K, Elliott MB. Frequent mild head injury promotes trigeminal sensitivity concomitant with microglial proliferation, astrocytosis, and increased neuropeptide levels in the trigeminal pain system. J Headache Pain 2017; 18:16. [PMID: 28176234 PMCID: PMC5296267 DOI: 10.1186/s10194-017-0726-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 01/18/2017] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Frequent mild head injuries or concussion along with the presence of headache may contribute to the persistence of concussion symptoms. METHODS In this study, the acute effects of recovery between mild head injuries and the frequency of injuries on a headache behavior, trigeminal allodynia, was assessed using von Frey testing up to one week after injury, while histopathological changes in the trigeminal pain pathway were evaluated using western blot, ELISA and immunohistochemistry. RESULTS: A decreased recovery time combined with an increased mild closed head injury (CHI) frequency results in reduced trigeminal allodynia thresholds compared to controls. The repetitive CHI group with the highest injury frequency showed the greatest reduction in trigeminal thresholds along with greatest increased levels of calcitonin gene-related peptide (CGRP) in the trigeminal nucleus caudalis. Repetitive CHI resulted in astrogliosis in the central trigeminal system, increased GFAP protein levels in the sensory barrel cortex, and an increased number of microglia cells in the trigeminal nucleus caudalis. CONCLUSIONS Headache behavior in rats is dependent on the injury frequency and recovery interval between mild head injuries. A worsening of headache behavior after repetitive mild head injuries was concomitant with increases in CGRP levels, the presence of astrocytosis, and microglia proliferation in the central trigeminal pathway. Signaling between neurons and proliferating microglia in the trigeminal pain system may contribute to the initiation of acute headache after concussion or other traumatic brain injuries.
Collapse
Affiliation(s)
- Ashley L Tyburski
- Department of Neurosurgery, Thomas Jefferson University, 1020 Locust Street, Philadelphia, PA, 19107, USA
| | - Lan Cheng
- Department of Neurosurgery, Thomas Jefferson University, 1020 Locust Street, Philadelphia, PA, 19107, USA
| | | | | | - Melanie B Elliott
- Department of Neurosurgery, Thomas Jefferson University, 1020 Locust Street, Philadelphia, PA, 19107, USA.
| |
Collapse
|
29
|
Moye LS, Pradhan AA. From blast to bench: A translational mini-review of posttraumatic headache. J Neurosci Res 2017; 95:1347-1354. [PMID: 28151589 DOI: 10.1002/jnr.24001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 11/07/2016] [Accepted: 11/28/2016] [Indexed: 12/25/2022]
Abstract
Current events within the military and professional sports have resulted in an increased recognition of the long-term and debilitating consequences of traumatic brain injury. Mild traumatic brain injury accounts for the majority of head injuries, and posttraumatic headache is the most common adverse effect. It is estimated that between 30% to 90% of traumatic brain injuries result in posttraumatic headache, and for a significant number of people this headache disorder can continue for up to and over a year post injury. Often, the most severe and chronic posttraumatic headache has a migraine-like phenotype and is difficult to resolve. In this review we discuss the preclinical findings from animal models of posttraumatic headache. We also describe potential mechanisms by which traumatic brain injury leads to chronic posttraumatic headache, including neuroinflammatory mediators and migraine-associated neuropeptides. There are surprisingly few preclinical studies that have investigated overlapping mechanisms between posttraumatic headache and migraine, especially considering the prevalence and debilitating nature of posttraumatic headache. Given this context, posttraumatic headache is a field with many emerging opportunities for growth. The frequency of posttraumatic headache in the general and military population is rising, and further preclinical research is required to understand, ameliorate, and treat this disabling disorder. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Laura S Moye
- Department of Psychiatry, University of Illinois at Chicago
| | | |
Collapse
|
30
|
Muccigrosso MM, Ford J, Benner B, Moussa D, Burnsides C, Fenn AM, Popovich PG, Lifshitz J, Walker FR, Eiferman DS, Godbout JP. Cognitive deficits develop 1month after diffuse brain injury and are exaggerated by microglia-associated reactivity to peripheral immune challenge. Brain Behav Immun 2016; 54:95-109. [PMID: 26774527 PMCID: PMC4828283 DOI: 10.1016/j.bbi.2016.01.009] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 01/05/2016] [Accepted: 01/12/2016] [Indexed: 01/07/2023] Open
Abstract
UNLABELLED Traumatic brain injury (TBI) elicits immediate neuroinflammatory events that contribute to acute cognitive, motor, and affective disturbance. Despite resolution of these acute complications, significant neuropsychiatric and cognitive issues can develop and progress after TBI. We and others have provided novel evidence that these complications are potentiated by repeated injuries, immune challenges and stressors. A key component to this may be increased sensitization or priming of glia after TBI. Therefore, our objectives were to determine the degree to which cognitive deterioration occurred after diffuse TBI (moderate midline fluid percussion injury) and ascertain if glial reactivity induced by an acute immune challenge potentiated cognitive decline 30 days post injury (dpi). In post-recovery assessments, hippocampal-dependent learning and memory recall were normal 7 dpi, but anterograde learning was impaired by 30 dpi. Examination of mRNA and morphological profiles of glia 30 dpi indicated a low but persistent level of inflammation with elevated expression of GFAP and IL-1β in astrocytes and MHCII and IL-1β in microglia. Moreover, an acute immune challenge 30 dpi robustly interrupted memory consolidation specifically in TBI mice. These deficits were associated with exaggerated microglia-mediated inflammation with amplified (IL-1β, CCL2, TNFα) and prolonged (TNFα) cytokine/chemokine expression, and a marked reactive morphological profile of microglia in the CA3 of the hippocampus. Collectively, these data indicate that microglia remain sensitized 30 dpi after moderate TBI and a secondary inflammatory challenge elicits robust microglial reactivity that augments cognitive decline. STATEMENT OF SIGNIFICANCE Traumatic brain injury (TBI) is a major risk factor in development of neuropsychiatric problems long after injury, negatively affecting quality of life. Mounting evidence indicates that inflammatory processes worsen with time after a brain injury and are likely mediated by glia. Here, we show that primed microglia and astrocytes developed in mice 1 month following moderate diffuse TBI, coinciding with cognitive deficits that were not initially evident after injury. Additionally, TBI-induced glial priming may adversely affect the ability of glia to appropriately respond to immune challenges, which occur regularly across the lifespan. Indeed, we show that an acute immune challenge augmented microglial reactivity and cognitive deficits. This idea may provide new avenues of clinical assessments and treatments following TBI.
Collapse
Affiliation(s)
- Megan M. Muccigrosso
- Department of Neuroscience, The Ohio State University, 333 W. 10th Ave, Columbus, OH
| | - Joni Ford
- Department of Neuroscience, The Ohio State University, 333 W. 10th Ave, Columbus, OH
| | - Brooke Benner
- Department of Neuroscience, The Ohio State University, 333 W. 10th Ave, Columbus, OH
| | - Daniel Moussa
- Department of Neuroscience, The Ohio State University, 333 W. 10th Ave, Columbus, OH
| | - Christopher Burnsides
- Department of Neuroscience, The Ohio State University, 333 W. 10th Ave, Columbus, OH
| | - Ashley M. Fenn
- Department of Neuroscience, The Ohio State University, 333 W. 10th Ave, Columbus, OH
| | - Phillip G. Popovich
- Department of Neuroscience, The Ohio State University, 333 W. 10th Ave, Columbus, OH,Center for Brain and Spinal Cord Repair, The Ohio State University, 460 W. 12th Ave, Columbus, OH,Institute for Behavioral Medicine Research, The Ohio State University, 460 Medical Center Dr., Columbus, OH
| | - Jonathan Lifshitz
- Barrow Neurological Institute at Phoenix Children’s Hospital, Department of Child Health, University of Arizona, College of Medicine-Phoenix, Phoenix, AZ
| | - Fredrick Rohan Walker
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan 2308, South Wales, Australia
| | - Daniel S. Eiferman
- Department of Surgery, The Ohio State University, 395 W. 12th Avenue, Columbus, OH
| | - Jonathan P. Godbout
- Department of Neuroscience, The Ohio State University, 333 W. 10th Ave, Columbus, OH,Center for Brain and Spinal Cord Repair, The Ohio State University, 460 W. 12th Ave, Columbus, OH,Institute for Behavioral Medicine Research, The Ohio State University, 460 Medical Center Dr., Columbus, OH,To whom correspondence should be addressed: J.P. Godbout, 259 IBMR Bldg., 460 Medical Center Dr., The Ohio State University, Columbus, OH 43210, USA. Tel: (614) 293-3456 Fax: (614) 366-2097,
| |
Collapse
|
31
|
Hazra A, Corbett BF, You JC, Aschmies S, Zhao L, Li K, Lepore AC, Marsh ED, Chin J. Corticothalamic network dysfunction and behavioral deficits in a mouse model of Alzheimer's disease. Neurobiol Aging 2016; 44:96-107. [PMID: 27318137 DOI: 10.1016/j.neurobiolaging.2016.04.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 04/21/2016] [Accepted: 04/22/2016] [Indexed: 10/21/2022]
Abstract
Alzheimer's disease is associated with cognitive decline and seizures. Growing evidence indicates that seizures contribute to cognitive deficits early in disease, but how they develop and impact cognition are unclear. To investigate potential mechanisms, we studied a mouse model that overexpresses mutant human amyloid precursor protein with high levels of amyloid beta (Aβ). These mice develop generalized epileptiform activity, including nonconvulsive seizures, consistent with alterations in corticothalamic network activity. Amyloid precursor protein mice exhibited reduced activity marker expression in the reticular thalamic nucleus, a key inhibitory regulatory nucleus, and increased activity marker expression in downstream thalamic relay targets that project to cortex and limbic structures. Slice recordings revealed impaired cortical inputs to the reticular thalamic nucleus that may contribute to corticothalamic dysfunction. These results are consistent with our findings of impaired sleep maintenance in amyloid precursor protein mice. Finally, the severity of sleep impairments predicted the severity of deficits in Morris water maze, suggesting corticothalamic dysfunction may relate to hippocampal dysfunction, and may be a pathophysiological mechanism underlying multiple behavioral and cognitive alterations in Alzheimer's disease.
Collapse
Affiliation(s)
- Anupam Hazra
- Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107.,Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107
| | - Brian F Corbett
- Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107.,Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107
| | - Jason C You
- Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107.,Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107
| | - Suzan Aschmies
- Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107.,Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107
| | - Lijuan Zhao
- Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107.,Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107
| | - Ke Li
- Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107.,Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107
| | - Angelo C Lepore
- Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107.,Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107
| | - Eric D Marsh
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia and the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104
| | - Jeannie Chin
- Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107.,Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107.,Memory & Brain Research Center, Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
32
|
Thomasy HE, Febinger HY, Ringgold KM, Gemma C, Opp MR. Hypocretinergic and cholinergic contributions to sleep-wake disturbances in a mouse model of traumatic brain injury. Neurobiol Sleep Circadian Rhythms 2016; 2:71-84. [PMID: 31236496 PMCID: PMC6575582 DOI: 10.1016/j.nbscr.2016.03.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 03/25/2016] [Accepted: 03/28/2016] [Indexed: 12/24/2022] Open
Abstract
Disorders of sleep and wakefulness occur in the majority of individuals who have experienced traumatic brain injury (TBI), with increased sleep need and excessive daytime sleepiness often reported. Behavioral and pharmacological therapies have limited efficacy, in part, because the etiology of post-TBI sleep disturbances is not well understood. Severity of injuries resulting from head trauma in humans is highly variable, and as a consequence so are their sequelae. Here, we use a controlled laboratory model to investigate the effects of TBI on sleep-wake behavior and on candidate neurotransmitter systems as potential mediators. We focus on hypocretin and melanin-concentrating hormone (MCH), hypothalamic neuropeptides important for regulating sleep and wakefulness, and two potential downstream effectors of hypocretin actions, histamine and acetylcholine. Adult male C57BL/6 mice (n=6-10/group) were implanted with EEG recording electrodes and baseline recordings were obtained. After baseline recordings, controlled cortical impact was used to induce mild or moderate TBI. EEG recordings were obtained from the same animals at 7 and 15 days post-surgery. Separate groups of animals (n=6-8/group) were used to determine effects of TBI on the numbers of hypocretin and MCH-producing neurons in the hypothalamus, histaminergic neurons in the tuberomammillary nucleus, and cholinergic neurons in the basal forebrain. At 15 days post-TBI, wakefulness was decreased and NREM sleep was increased during the dark period in moderately injured animals. There were no differences between groups in REM sleep time, nor were there differences between groups in sleep during the light period. TBI effects on hypocretin and cholinergic neurons were such that more severe injury resulted in fewer cells. Numbers of MCH neurons and histaminergic neurons were not altered under the conditions of this study. Thus, we conclude that moderate TBI in mice reduces wakefulness and increases NREM sleep during the dark period, effects that may be mediated by hypocretin-producing neurons and/or downstream cholinergic effectors in the basal forebrain.
Collapse
Affiliation(s)
- Hannah E Thomasy
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, United States
| | - Heidi Y Febinger
- Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA, United States
| | - Kristyn M Ringgold
- Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA, United States
| | - Carmelina Gemma
- Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA, United States
| | - Mark R Opp
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, United States.,Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA, United States
| |
Collapse
|
33
|
Daiutolo BV, Tyburski A, Clark SW, Elliott MB. Trigeminal Pain Molecules, Allodynia, and Photosensitivity Are Pharmacologically and Genetically Modulated in a Model of Traumatic Brain Injury. J Neurotrauma 2015; 33:748-60. [PMID: 26472135 DOI: 10.1089/neu.2015.4087] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The pain-signaling molecules, nitric oxide synthase (NOS) and calcitonin gene-related peptide (CGRP), are implicated in the pathophysiology of post-traumatic headache (PTH) as they are for migraine. This study assessed the changes of inducible NOS (iNOS) and its cellular source in the trigeminal pain circuit, as well as the relationship between iNOS and CGRP after controlled cortical impact (CCI) injury in mice. The effects of a CGRP antagonist (MK8825) and sumatriptan on iNOS messenger RNA (mRNA) and protein were compared to vehicle at 2 weeks postinjury. Changes in CGRP levels in the trigeminal nucleus caudalis (TNC) in iNOS knockouts with CCI were compared to wild-type (WT) mice at 3 days and 2 weeks post injury. Trigeminal allodynia and photosensitivity were measured. MK8825 and sumatriptan increased allodynic thresholds in CCI groups compared to vehicle (p < 0.01), whereas iNOS knockouts were not different from WT. Photosensitivity was attenuated in MK8825 mice and iNOS knockouts compared to WT (p < 0.05). MK8825 and sumatriptan reduced levels of iNOS mRNA and iNOS immunoreactivity in the TNC and ganglia (p < 0.01). Differences in iNOS cellular localization were found between the trigeminal ganglia and TNC. Although the knockout of iNOS attenuated CGRP at 3 days (p < 0.05), it did not reduce CGRP at 2 weeks. CGRP immunoreactivity was found in the meningeal layers post-CCI, while negligible in controls. Findings support the importance of interactions between CGRP and iNOS in mediating allodynia, as well as the individual roles in photosensitivity. Mitigating prolonged increases in CGRP may be a promising intervention for treating acute PTH.
Collapse
Affiliation(s)
- Brittany V Daiutolo
- Department of Neurological Surgery, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Ashley Tyburski
- Department of Neurological Surgery, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Shannon W Clark
- Department of Neurological Surgery, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Melanie B Elliott
- Department of Neurological Surgery, Thomas Jefferson University , Philadelphia, Pennsylvania
| |
Collapse
|
34
|
Impact of traumatic brain injury on sleep structure, electrocorticographic activity and transcriptome in mice. Brain Behav Immun 2015; 47:118-30. [PMID: 25576803 DOI: 10.1016/j.bbi.2014.12.023] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 12/22/2014] [Accepted: 12/22/2014] [Indexed: 12/21/2022] Open
Abstract
Traumatic brain injury (TBI), including mild TBI (mTBI), is importantly associated with vigilance and sleep complaints. Because sleep is required for learning, plasticity and recovery, we here evaluated the bidirectional relationship between mTBI and sleep with two specific objectives: (1) Test that mTBI rapidly impairs sleep-wake architecture and the dynamics of the electrophysiological marker of sleep homeostasis (i.e., non-rapid eye movement sleep delta (1-4Hz) activity); (2) evaluate the impact of sleep loss following mTBI on the expression of plasticity markers that have been linked to sleep homeostasis and on genome-wide gene expression. A closed-head injury model was used to perform a 48h electrocorticographic (ECoG) recording in mice submitted to mTBI or Sham surgery. mTBI was found to immediately decrease the capacity to sustain long bouts of wakefulness as well as the amplitude of the time course of ECoG delta activity during wakefulness. Significant changes in ECoG spectral activity during wakefulness, non-rapid eye movement and rapid eye movement sleep were observed mainly on the second recorded day. A second experiment was performed to measure gene expression in the cerebral cortex and hippocampus after a mTBI followed either by two consecutive days of 6h sleep deprivation (SD) or of undisturbed behavior (quantitative PCR and next-generation sequencing). mTBI modified the expression of genes involved in immunity, inflammation and glial function (e.g., chemokines, glial markers) and SD changed that of genes linked to circadian rhythms, synaptic activity/neuronal plasticity, neuroprotection and cell death and survival. SD appeared to affect gene expression in the cerebral cortex more importantly after mTBI than Sham surgery including that of the astrocytic marker Gfap, which was proposed as a marker of clinical outcome after TBI. Interestingly, SD impacted the hippocampal expression of the plasticity elements Arc and EfnA3 only after mTBI. Overall, our findings reveal alterations in spectral signature across all vigilance states in the first days after mTBI, and show that sleep loss post-mTBI reprograms the transcriptome in a brain area-specific manner and in a way that could be deleterious to brain recovery.
Collapse
|
35
|
Harrison JL, Rowe RK, Ellis TW, Yee NS, O’Hara BF, Adelson PD, Lifshitz J. Resolvins AT-D1 and E1 differentially impact functional outcome, post-traumatic sleep, and microglial activation following diffuse brain injury in the mouse. Brain Behav Immun 2015; 47:131-40. [PMID: 25585137 PMCID: PMC4468045 DOI: 10.1016/j.bbi.2015.01.001] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 12/22/2014] [Accepted: 01/01/2015] [Indexed: 02/09/2023] Open
Abstract
Traumatic brain injury (TBI) is induced by mechanical forces which initiate a cascade of secondary injury processes, including inflammation. Therapies which resolve the inflammatory response may promote neural repair without exacerbating the primary injury. Specific derivatives of omega-3 fatty acids loosely grouped as specialized pro-resolving lipid mediators (SPMs) and termed resolvins promote the active resolution of inflammation. In the current study, we investigate the effect of two resolvin molecules, RvE1 and AT-RvD1, on post-traumatic sleep and functional outcome following diffuse TBI through modulation of the inflammatory response. Adult, male C57BL/6 mice were injured using a midline fluid percussion injury (mFPI) model (6-10min righting reflex time for brain-injured mice). Experimental groups included mFPI administered RvE1 (100ng daily), AT-RvD1 (100ng daily), or vehicle (sterile saline) and counterbalanced with uninjured sham mice. Resolvins or saline were administered daily for seven consecutive days beginning 3days prior to TBI to evaluate proof-of-principle to improve outcome. Immediately following diffuse TBI, post-traumatic sleep was recorded for 24h post-injury. For days 1-7 post-injury, motor outcome was assessed by rotarod. Cognitive function was measured at 6days post-injury using novel object recognition (NOR). At 7days post-injury, microglial activation was quantified using immunohistochemistry for Iba-1. In the diffuse brain-injured mouse, AT-RvD1 treatment, but not RvE1, mitigated motor and cognitive deficits. RvE1 treatment significantly increased post-traumatic sleep in brain-injured mice compared to all other groups. RvE1 treated mice displayed a higher proportion of ramified microglia and lower proportion of activated rod microglia in the cortex compared to saline or AT-RvD1 treated brain-injured mice. Thus, RvE1 treatment modulated post-traumatic sleep and the inflammatory response to TBI, albeit independently of improvement in motor and cognitive outcome as seen in AT-RvD1-treated mice. This suggests AT-RvD1 may impart functional benefit through mechanisms other than resolution of inflammation alone.
Collapse
Affiliation(s)
- Jordan L. Harrison
- BARROW Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ
,Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ
,Interdisciplinary Graduate Program in Neuroscience, Arizona State University, Tempe, AZ
| | - Rachel K. Rowe
- BARROW Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ
,Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ
,Phoenix Veteran Affairs Healthcare System, Phoenix, AZ
| | - Timothy W. Ellis
- BARROW Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ
,Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ
,College of Osteopathic Medicine, Midwestern University, Glendale, AZ
| | - Nicole S. Yee
- BARROW Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ
,Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ
| | - Bruce F. O’Hara
- Department of Biology, University of Kentucky College of Arts and Sciences, Lexington, KY
,Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY, USA
| | - P. David Adelson
- BARROW Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ
,Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ
,Interdisciplinary Graduate Program in Neuroscience, Arizona State University, Tempe, AZ
| | - Jonathan Lifshitz
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA; Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA; Interdisciplinary Graduate Program in Neuroscience, Arizona State University, Tempe, AZ, USA; Phoenix Veteran Affairs Healthcare System, Phoenix, AZ, USA.
| |
Collapse
|
36
|
Abstract
More than one-half million patients are hospitalized annually for traumatic brain injury (TBI). One-quarter demonstrate sleep-disordered breathing, up to 50% experience insomnia, and half have hypersomnia. Sleep disturbances after TBI may result from injury to sleep-regulating brain tissue, nonspecific neurohormonal responses to systemic injury, ICU environmental interference, and medication side effects. A diagnosis of sleep disturbances requires a high index of suspicion and appropriate testing. Treatment starts with a focus on making the ICU environment conducive to normal sleep. Treating sleep-disordered breathing likely has outcome benefits in TBI. The use of sleep promoting sedative-hypnotics and anxiolytics should be judicious.
Collapse
|
37
|
Smith D, Brooks D, Wohlgehagen E, Rau T, Poulsen D. Temporal and Spatial Changes in the Pattern of Iba1 and CD68 Staining in the Rat Brain Following Severe Traumatic Brain Injury. ACTA ACUST UNITED AC 2015. [DOI: 10.4236/mri.2015.42002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
38
|
Skopin MD, Kabadi SV, Viechweg SS, Mong JA, Faden AI. Chronic decrease in wakefulness and disruption of sleep-wake behavior after experimental traumatic brain injury. J Neurotrauma 2014; 32:289-96. [PMID: 25242371 DOI: 10.1089/neu.2014.3664] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Traumatic brain injury (TBI) can cause sleep-wake disturbances and excessive daytime sleepiness. The pathobiology of sleep disorders in TBI, however, is not well understood, and animal models have been underused in studying such changes and potential underlying mechanisms. We used the rat lateral fluid percussion (LFP) model to analyze sleep-wake patterns as a function of time after injury. Rapid-eye movement (REM) sleep, non-REM (NREM) sleep, and wake bouts during light and dark phases were measured with electroencephalography and electromyography at an early as well as chronic time points after LFP. Moderate TBI caused disturbances in the ability to maintain consolidated wake bouts during the active phase and chronic loss of wakefulness. Further, TBI resulted in cognitive impairments and depressive-like symptoms, and reduced the number of orexin-A-positive neurons in the lateral hypothalamus.
Collapse
Affiliation(s)
- Mark D Skopin
- Department of Anesthesiology, Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine , Baltimore, Maryland
| | | | | | | | | |
Collapse
|