1
|
Wang X, Qian J, Li Y, Meng Y, Cheng R, Ren N, Fei Y. Protective effects of forsythoside A against severe acute pancreatitis- induced brain injury in mice. Biomed Pharmacother 2024; 178:117301. [PMID: 39137650 DOI: 10.1016/j.biopha.2024.117301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 08/06/2024] [Accepted: 08/09/2024] [Indexed: 08/15/2024] Open
Abstract
OBJECTIVES This study aimed to evaluate the therapeutic effects of forsythoside A (FA) on brain injury induced by severe acute pancreatitis (SAP) using a murine model. METHODS Mice were induced with 3.5 % sodium taurocholate to model SAP-induced brain injury (SAP-IBI) and were randomly assigned to four distinct treatment regimens: the SAP-IBI model group (SAP-IBI), low-dose FA treatment group (FA L+SI), middle-dose FA treatment group (FA M+SI), and high-dose FA treatment group (FA H+SI). A sham-operation group (SO) served as a negative control. Serum levels of interleukin-1β (IL-1β) and IL-18 were quantified via ELISA, and serum amylase levels were assessed using optical turbidimetry. mRNA expression levels of AIM2, ASC, Caspase-1, and GAPDH in hippocampal brain tissue were measured by quantitative reverse transcription polymerase chain reaction (qRT-PCR). Protein levels of NLRP3, GSDMD, IL-1β, and IL-18 in hippocampal brain tissue were evaluated using Western blotting. Neurological function in surviving mice was assessed through modified neurological severity scores (mNSS). Transmission electron microscopy (TEM) provided ultrastructural analysis of the hippocampus. Additionally, water content and pathological changes in hippocampal brain tissue were examined 24 hours post-operation, along with other relevant indicators. RESULTS At 24 hours post-operation, the FA H+SI group exhibited significantly reduced levels of serum amylase, IL-1β, and IL-18, along with decreased expression of AIM2, ASC, and Caspase-1 mRNA. Furthermore, NLRP3 protein levels, water content, pancreas and hippocampal brain pathological scores, and mNSS were significantly lower compared to the SAP-IBI group (P<0.01). CONCLUSIONS FA demonstrates protective effects against SAP-IBI in mice, suggesting potential therapeutic benefits.
Collapse
Affiliation(s)
- Xiaohong Wang
- Department of Gastroenterology, Yizheng Hospital of Nanjing Drum Tower Hospital Group, Yizheng, Jiangsu 211900, China.
| | - Jing Qian
- Department of General Surgery, Yizheng Hospital of Nanjing Drum Tower Hospital Group, Yizheng, Jiangsu 211900, China
| | - Yanjie Li
- Department of Neurology, Yizheng Hospital of Nanjing Drum Tower Hospital Group, Yizheng, Jiangsu 211900, China
| | - Yun Meng
- Department of Gastroenterology, Yizheng Hospital of Nanjing Drum Tower Hospital Group, Yizheng, Jiangsu 211900, China
| | - Ruizhi Cheng
- Department of Gastroenterology, Yizheng Hospital of Nanjing Drum Tower Hospital Group, Yizheng, Jiangsu 211900, China
| | - Nana Ren
- Department of Gastroenterology, Yizheng Hospital of Nanjing Drum Tower Hospital Group, Yizheng, Jiangsu 211900, China
| | - Ying Fei
- Department of Gastroenterology, Yizheng Hospital of Nanjing Drum Tower Hospital Group, Yizheng, Jiangsu 211900, China
| |
Collapse
|
2
|
Rodriguez S, Sharma S, Tiarks G, Peterson Z, Jackson K, Thedens D, Wong A, Keffala-Gerhard D, Mahajan VB, Ferguson PJ, Newell EA, Glykys J, Nickl-Jockschat T, Bassuk AG. Neuroprotective effects of naltrexone in a mouse model of post-traumatic seizures. Sci Rep 2024; 14:13507. [PMID: 38867062 PMCID: PMC11169394 DOI: 10.1038/s41598-024-63942-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 06/03/2024] [Indexed: 06/14/2024] Open
Abstract
Traumatic Brain Injury (TBI) induces neuroinflammatory response that can initiate epileptogenesis, which develops into epilepsy. Recently, we identified anti-convulsive effects of naltrexone, a mu-opioid receptor (MOR) antagonist, used to treat drug addiction. While blocking opioid receptors can reduce inflammation, it is unclear if post-TBI seizures can be prevented by blocking MORs. Here, we tested if naltrexone prevents neuroinflammation and/or seizures post-TBI. TBI was induced by a modified Marmarou Weight-Drop (WD) method on 4-week-old C57BL/6J male mice. Mice were placed in two groups: non-telemetry assessing the acute effects or in telemetry monitoring for interictal events and spontaneous seizures both following TBI and naltrexone. Molecular, histological and neuroimaging techniques were used to evaluate neuroinflammation, neurodegeneration and fiber track integrity at 8 days and 3 months post-TBI. Peripheral immune responses were assessed through serum chemokine/cytokine measurements. Our results show an increase in MOR expression, nitro-oxidative stress, mRNA expression of inflammatory cytokines, microgliosis, neurodegeneration, and white matter damage in the neocortex of TBI mice. Video-EEG revealed increased interictal events in TBI mice, with 71% mice developing post-traumatic seizures (PTS). Naltrexone treatment ameliorated neuroinflammation, neurodegeneration, reduced interictal events and prevented seizures in all TBI mice, which makes naltrexone a promising candidate against PTS, TBI-associated neuroinflammation and epileptogenesis in a WD model of TBI.
Collapse
Affiliation(s)
- Saul Rodriguez
- Stead Family Department of Pediatrics , Carver College of Medicine, University of Iowa, 25 South Grand Ave, 2040 MedLabs, Iowa City, IA, 52242, USA
| | - Shaunik Sharma
- Stead Family Department of Pediatrics , Carver College of Medicine, University of Iowa, 25 South Grand Ave, 2040 MedLabs, Iowa City, IA, 52242, USA
| | - Grant Tiarks
- Stead Family Department of Pediatrics , Carver College of Medicine, University of Iowa, 25 South Grand Ave, 2040 MedLabs, Iowa City, IA, 52242, USA
| | - Zeru Peterson
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Kyle Jackson
- Stead Family Department of Pediatrics , Carver College of Medicine, University of Iowa, 25 South Grand Ave, 2040 MedLabs, Iowa City, IA, 52242, USA
| | - Daniel Thedens
- Department of Radiology, University of Iowa, Iowa City, IA, USA
| | - Angela Wong
- Stead Family Department of Pediatrics , Carver College of Medicine, University of Iowa, 25 South Grand Ave, 2040 MedLabs, Iowa City, IA, 52242, USA
| | - David Keffala-Gerhard
- Stead Family Department of Pediatrics , Carver College of Medicine, University of Iowa, 25 South Grand Ave, 2040 MedLabs, Iowa City, IA, 52242, USA
| | - Vinit B Mahajan
- Department of Ophthalmology, Stanford University, Palo Alto, CA, USA
| | - Polly J Ferguson
- Stead Family Department of Pediatrics , Carver College of Medicine, University of Iowa, 25 South Grand Ave, 2040 MedLabs, Iowa City, IA, 52242, USA
| | - Elizabeth A Newell
- Stead Family Department of Pediatrics , Carver College of Medicine, University of Iowa, 25 South Grand Ave, 2040 MedLabs, Iowa City, IA, 52242, USA
| | - Joseph Glykys
- Stead Family Department of Pediatrics , Carver College of Medicine, University of Iowa, 25 South Grand Ave, 2040 MedLabs, Iowa City, IA, 52242, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
- Department of Neurology, University of Iowa, Iowa City, IA, USA
| | - Thomas Nickl-Jockschat
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
- Department of Psychiatry and Psychotherapy, Otto-von-Guericke University, Magdeburg, Germany German Center for Mental Health (DZPG), partner site Halle-Jena-Magdeburg, Germany Center for Intervention and Research on adaptive and maladaptive brain Circuits underlying mental health (C-I-R-C), Halle-Jena-Magdeburg, Germany
| | - Alexander G Bassuk
- Stead Family Department of Pediatrics , Carver College of Medicine, University of Iowa, 25 South Grand Ave, 2040 MedLabs, Iowa City, IA, 52242, USA.
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA.
- Department of Neurology, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
3
|
Zhang Y, Zhao Y, Wang Y, Li J, Huang Y, Lyu F, Wang Y, Wei P, Yuan Y, Fu Y, Gao Y. Microglial histone deacetylase 2 is dispensable for functional and histological outcomes in a mouse model of traumatic brain injury. J Cereb Blood Flow Metab 2024; 44:817-835. [PMID: 38069842 PMCID: PMC11197137 DOI: 10.1177/0271678x231197173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/08/2023] [Accepted: 08/08/2023] [Indexed: 04/26/2024]
Abstract
The Class-I histone deacetylases (HDACs) mediate microglial inflammation and neurological dysfunction after traumatic brain injury (TBI). However, whether the individual Class-I HDACs play an indispensable role in TBI pathogenesis remains elusive. HDAC2 has been shown to upregulate pro-inflammatory genes in myeloid cells under brain injuries such as intracerebral hemorrhage, thereby worsening outcomes. Thus, we hypothesized that HDAC2 drives microglia toward a pro-inflammatory neurotoxic phenotype in a murine model of controlled cortical impact (CCI). Our results revealed that HDAC2 expression was highly induced in CD16/CD32+ pro-inflammatory microglia 3 and 7d after TBI. Surprisingly, microglia-targeted HDAC2 knockout (HDAC2 miKO) mice failed to demonstrate a beneficial phenotype after CCI/TBI compared to their wild-type (WT) littermates. HDAC2 miKO mice exhibited comparable levels of grey and white matter injury, efferocytosis, and sensorimotor and cognitive deficits after CCI/TBI as WT mice. RNA sequencing of isolated microglia 3d after CCI/TBI indicated the elevation of a panel of pro-inflammatory cytokines/chemokines in HDAC2 miKO mice over WT mice, and flow cytometry showed further elevated brain infiltration of neutrophils and B cells in HDAC2 miKO mice. Together, this study does not support a detrimental role for HDAC2 in microglial responses after TBI and calls for investigation into alternative mechanisms.
Collapse
Affiliation(s)
- Yue Zhang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yongfang Zhao
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yana Wang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Jiaying Li
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yichen Huang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Fan Lyu
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yangfan Wang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Pengju Wei
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yiwen Yuan
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yi Fu
- Department of Neurology & Institute of Neurology, Rui Jin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yanqin Gao
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
4
|
Fesharaki-Zadeh A, Datta D. An overview of preclinical models of traumatic brain injury (TBI): relevance to pathophysiological mechanisms. Front Cell Neurosci 2024; 18:1371213. [PMID: 38682091 PMCID: PMC11045909 DOI: 10.3389/fncel.2024.1371213] [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: 01/16/2024] [Accepted: 03/20/2024] [Indexed: 05/01/2024] Open
Abstract
Background Traumatic brain injury (TBI) is a major cause of morbidity and mortality, affecting millions annually worldwide. Although the majority of TBI patients return to premorbid baseline, a subset of patient can develop persistent and often debilitating neurocognitive and behavioral changes. The etiology of TBI within the clinical setting is inherently heterogenous, ranging from sport related injuries, fall related injuries and motor vehicle accidents in the civilian setting, to blast injuries in the military setting. Objective Animal models of TBI, offer the distinct advantage of controlling for injury modality, duration and severity. Furthermore, preclinical models of TBI have provided the necessary temporal opportunity to study the chronic neuropathological sequelae of TBI, including neurodegenerative sequelae such as tauopathy and neuroinflammation within the finite experimental timeline. Despite the high prevalence of TBI, there are currently no disease modifying regimen for TBI, and the current clinical treatments remain largely symptom based. The preclinical models have provided the necessary biological substrate to examine the disease modifying effect of various pharmacological agents and have imperative translational value. Methods The current review will include a comprehensive survey of well-established preclinical models, including classic preclinical models including weight drop, blast injury, fluid percussion injury, controlled cortical impact injury, as well as more novel injury models including closed-head impact model of engineered rotational acceleration (CHIMERA) models and closed-head projectile concussive impact model (PCI). In addition to rodent preclinical models, the review will include an overview of other species including large animal models and Drosophila. Results There are major neuropathological perturbations post TBI captured in various preclinical models, which include neuroinflammation, calcium dysregulation, tauopathy, mitochondrial dysfunction and oxidative stress, axonopathy, as well as glymphatic system disruption. Conclusion The preclinical models of TBI continue to offer valuable translational insight, as well as essential neurobiological basis to examine specific disease modifying therapeutic regimen.
Collapse
Affiliation(s)
- Arman Fesharaki-Zadeh
- Department of Neurology and Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Dibyadeep Datta
- Division of Aging and Geriatric Psychiatry, Alzheimer’s Disease Research Unit, Department of Psychiatry, New Haven, CT, United States
| |
Collapse
|
5
|
Wang J, Lu Y, Carr C, Dhandapani KM, Brann DW. Senolytic therapy is neuroprotective and improves functional outcome long-term after traumatic brain injury in mice. Front Neurosci 2023; 17:1227705. [PMID: 37575310 PMCID: PMC10416099 DOI: 10.3389/fnins.2023.1227705] [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: 05/23/2023] [Accepted: 07/10/2023] [Indexed: 08/15/2023] Open
Abstract
Introduction Chronic neuroinflammation can exist for months to years following traumatic brain injury (TBI), although the underlying mechanisms remain poorly understood. Methods In the current study, we used a controlled cortical impact mouse model of TBI to examine whether proinflammatory senescent cells are present in the brain long-term (months) after TBI and whether ablation of these cells via administration of senolytic drugs can improve long-term functional outcome after TBI. The results revealed that astrocytes and microglia in the cerebral cortex, hippocampus, corpus callosum and lateral posterior thalamus colocalized the senescent cell markers, p16Ink4a or p21Cip1/Waf1 at 5 weeks post injury (5wpi) and 4 months post injury (4mpi) in a controlled cortical impact (CCI) model. Intermittent administration of the senolytic drugs, dasatinib and quercetin (D + Q) beginning 1-month after TBI for 13 weeks significantly ablated p16Ink4a-positive- and p21Cip1/Waf1-positive-cells in the brain of TBI animals, and significantly reduced expression of the major senescence-associated secretory phenotype (SASP) pro-inflammatory factors, interleukin-1β and interleukin-6. Senolytic treatment also significantly attenuated neurodegeneration and enhanced neuron number at 18 weeks after TBI in the ipsilateral cortex, hippocampus, and lateral posterior thalamus. Behavioral testing at 18 weeks after TBI further revealed that senolytic therapy significantly rescued defects in spatial reference memory and recognition memory, as well as depression-like behavior in TBI mice. Discussion Taken as a whole, these findings indicate there is robust and widespread induction of senescent cells in the brain long-term after TBI, and that senolytic drug treatment begun 1-month after TBI can efficiently ablate the senescent cells, reduce expression of proinflammatory SASP factors, reduce neurodegeneration, and rescue defects in reference memory, recognition memory, and depressive behavior.
Collapse
Affiliation(s)
| | | | | | | | - Darrell W. Brann
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
6
|
Yang ZL, Liang ZY, Lin YK, Lin FB, Rao J, Xu XJ, Wang CH, Chen CM. Efficacy of extracellular vesicles of different cell origins in traumatic brain injury: A systematic review and network meta-analysis. Front Neurosci 2023; 17:1147194. [PMID: 37065922 PMCID: PMC10090410 DOI: 10.3389/fnins.2023.1147194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
BackgroundThere was still no effective treatment for traumatic brain injury (TBI). Recently, many preclinical studies had shown promising efficacy of extracellular vesicles (EVs) from various cell sources. Our aim was to compare which cell-derived EVs were most effective in treating TBI through a network meta-analysis.MethodsWe searched four databases and screened various cell-derived EVs for use in preclinical studies of TBI treatment. A systematic review and network meta-analysis were conducted for two outcome indicators, modified Neurological Severity Score (mNSS) and Morris Water Maze (MWM), and they were ranked by the surface under the cumulative ranking curves (SUCRA). Bias risk assessment was performed with SYRCLE. R software (version 4.1.3, Boston, MA, USA) was used for data analysis.ResultsA total of 20 studies were included in this study, involving 383 animals. Astrocyte-derived extracellular vesicles (AEVs) ranked first in response to mNSS at day 1 (SUCRA: 0.26%), day 3 (SUCRA: 16.32%), and day 7 (SUCRA: 9.64%) post-TBI. Extracellular vesicles derived from mesenchymal stem cells (MSCEVs) were most effective in mNSS assessment on day 14 (SUCRA: 21.94%) and day 28 (SUCRA: 6.26%), as well as MWM’s escape latency (SUCRA: 6.16%) and time spent in the target quadrant (SUCRA: 86.52%). The result of mNSS analysis on day 21 showed that neural stem cell-derived extracellular vesicles (NSCEVs) had the best curative effect (SUCRA: 6.76%).ConclusionAEVs may be the best choice to improve early mNSS recovery after TBI. The efficacy of MSCEVs may be the best in the late mNSS and MWM after TBI.Systematic review registrationhttps://www.crd.york.ac.uk/prospero/, identifier CRD42023377350.
Collapse
|
7
|
Smith SM, Garcia EL, Davidson CG, Thompson JJ, Lovett SD, Ferekides N, Federico Q, Bumanglag AV, Hernandez AR, Abisambra JF, Burke SN. Paired associates learning is disrupted after unilateral parietal lobe controlled cortical impact in rats: A trial-by-trial behavioral analysis. Behav Brain Res 2023; 437:114106. [PMID: 36089100 PMCID: PMC9927580 DOI: 10.1016/j.bbr.2022.114106] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 02/01/2023]
Abstract
Approximately 60-70 million people suffer from traumatic brain injury (TBI) each year. Animal models continue to be paramount in understanding mechanisms of cellular dysfunction and testing new treatments for TBI. Enhancing the translational potential of novel interventions therefore necessitates testing pre-clinical intervention strategies with clinically relevant cognitive assays. This study used a unilateral parietal lobe controlled cortical impact (CCI) model of TBI and tested rats on a touchscreen-based Paired Associates Learning (PAL) task, which is part of the Cambridge Neuropsychological Test Automated Battery. In humans, the PAL task has been used to assess cognitive deficits in the ability to form stimulus-location associations in a multitude of disease states, including TBI. Although the use of PAL in animal models could be important for understanding the clinical severity of cognitive impairment post-injury and throughout intervention, to date, the extent to which a rat model of TBI produces deficits in PAL task performance has not yet been reported. This study details the behavioral consequences of the CCI injury model with a Trial-by-Trial analysis of PAL performance that enables behavioral strategy use to be inferred. Following behavior, the extent of the injury was quantified with histology and staining for the presence of glial fibrillary acid protein and ionized calcium-binding adapter molecule 1. Rats that received unilateral CCI were impaired on the PAL task and showed more aberrant response-driven behavior. The magnitude of PAL impairment was also correlated with Iba1 staining in the thalamus. These observations suggest that PAL could be useful for pre-clinical assessments of novel interventions for treating TBI.
Collapse
Affiliation(s)
- Samantha M Smith
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, United States; Graduate Program in Biomedical Sciences, Neuroscience Concentration, University of Florida, United States
| | - Elena L Garcia
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, United States
| | - Caroline G Davidson
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, United States
| | - John J Thompson
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, United States
| | - Sarah D Lovett
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, United States
| | - Nedi Ferekides
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, United States
| | - Quinten Federico
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, United States
| | - Argyle V Bumanglag
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, United States
| | - Abbi R Hernandez
- Department of Medicine, Division of Gerontology, Geriatrics, and Palliative Care, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jose F Abisambra
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, United States; Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine, Gainesville, FL, United States
| | - Sara N Burke
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, United States; Institute on Aging, University of Florida, Gainesville, FL, United States.
| |
Collapse
|
8
|
Cheng J, Lin L, Yu J, Zhu X, Ma H, Zhao Y. N6-methyladenosine RNA is modified in the rat hippocampus following traumatic brain injury with hypothermia treatment. Front Neurosci 2023; 17:1069640. [PMID: 36875640 PMCID: PMC9975158 DOI: 10.3389/fnins.2023.1069640] [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: 10/14/2022] [Accepted: 01/16/2023] [Indexed: 02/17/2023] Open
Abstract
Recent studies have suggested a role for N6-methyladenosine (m6A) modification in neurological diseases. Hypothermia, a commonly used treatment for traumatic brain injury, plays a neuroprotective role by altering m6A modifications. In this study, methylated RNA immunoprecipitation sequencing (MeRIP-Seq) was applied to conduct a genome-wide analysis of RNA m6A methylation in the rat hippocampus of Sham and traumatic brain injury (TBI) groups. In addition, we identified the expression of mRNA in the rat hippocampus after TBI with hypothermia treatment. Compared with the Sham group, the sequencing results of the TBI group showed that 951 different m6A peaks and 1226 differentially expressed mRNAs were found. We performed cross-linking analysis of the data of the two groups. The result showed that 92 hyper-methylated genes were upregulated, 13 hyper-methylated genes were downregulated, 25 hypo-methylated genes were upregulated, and 10 hypo-methylated genes were downregulated. Moreover, a total of 758 differential peaks were identified between TBI and hypothermia treatment groups. Among these differential peaks, 173 peaks were altered by TBI and reversed by hypothermia treatment, including Plat, Pdcd5, Rnd3, Sirt1, Plaur, Runx1, Ccr1, Marveld1, Lmnb2, and Chd7. We found that hypothermia treatment transformed some aspects of the TBI-induced m6A methylation landscape of the rat hippocampus.
Collapse
Affiliation(s)
- Jin Cheng
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Clinical Research Center for Emergency and Resuscitation, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lian Lin
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, China.,Department of Emergency, Gansu Provincial People's Hospital, Lanzhou, China
| | - Jiangtao Yu
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xiaolu Zhu
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Haoli Ma
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yan Zhao
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Clinical Research Center for Emergency and Resuscitation, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
9
|
Hung SY, Chung HY, Luo ST, Chu YT, Chen YH, MacDonald IJ, Chien SY, Kotha P, Yang LY, Hwang LL, Dun NJ, Chuang DM, Chen YH. Electroacupuncture improves TBI dysfunction by targeting HDAC overexpression and BDNF-associated Akt/GSK-3β signaling. Front Cell Neurosci 2022; 16:880267. [PMID: 36016833 PMCID: PMC9396337 DOI: 10.3389/fncel.2022.880267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 06/27/2022] [Indexed: 11/18/2022] Open
Abstract
Background Acupuncture or electroacupuncture (EA) appears to be a potential treatment in acute clinical traumatic brain injury (TBI); however, it remains uncertain whether acupuncture affects post-TBI histone deacetylase (HDAC) expression or impacts other biochemical/neurobiological events. Materials and methods We used behavioral testing, Western blot, and immunohistochemistry analysis to evaluate the cellular and molecular effects of EA at LI4 and LI11 in both weight drop-impact acceleration (WD)- and controlled cortical impact (CCI)-induced TBI models. Results Both WD- and CCI-induced TBI caused behavioral dysfunction, increased cortical levels of HDAC1 and HDAC3 isoforms, activated microglia and astrocytes, and decreased cortical levels of BDNF as well as its downstream mediators phosphorylated-Akt and phosphorylated-GSK-3β. Application of EA reversed motor, sensorimotor, and learning/memory deficits. EA also restored overexpression of HDAC1 and HDAC3, and recovered downregulation of BDNF-associated signaling in the cortex of TBI mice. Conclusion The results strongly suggest that acupuncture has multiple benefits against TBI-associated adverse behavioral and biochemical effects and that the underlying mechanisms are likely mediated by targeting HDAC overexpression and aberrant BDNF-associated Akt/GSK-3 signaling.
Collapse
Affiliation(s)
- Shih-Ya Hung
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan
- Division of Colorectal Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Hsin-Yi Chung
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan
| | - Sih-Ting Luo
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan
| | - Yu-Ting Chu
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan
| | - Yu-Hsin Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Iona J. MacDonald
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan
| | - Szu-Yu Chien
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan
| | - Peddanna Kotha
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan
| | - Liang-Yo Yang
- Department of Physiology, School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
- Laboratory for Neural Repair, China Medical University Hospital, Taichung, Taiwan
| | - Ling-Ling Hwang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Nae J. Dun
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA, United States
| | - De-Maw Chuang
- Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Yi-Hung Chen
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
- Department of Photonics and Communication Engineering, Asia University, Taichung, Taiwan
- *Correspondence: Yi-Hung Chen,
| |
Collapse
|
10
|
Foecking EM, Segismundo AB, Lotesto KM, Westfall EJ, Bolduan AJ, Peter TK, Wallace DG, Kozlowski DA, Stubbs EB, Marzo SJ, Byram SC. Testosterone treatment restores vestibular function by enhancing neuronal survival in an experimental closed-head repetitive mild traumatic brain injury model. Behav Brain Res 2022; 433:113998. [PMID: 35809692 DOI: 10.1016/j.bbr.2022.113998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 06/22/2022] [Accepted: 07/05/2022] [Indexed: 11/28/2022]
Abstract
Repetitive mild traumatic brain injury (rmTBI) results in a myriad of symptoms, including vestibular impairment. The mechanisms underlying vestibular dysfunction in rmTBI patients remain poorly understood. Concomitantly, acute hypogonadism occurs following TBI and can persist chronically in many patients. Using a repetitive mild closed-head animal model of TBI, the role of testosterone on vestibular function was tested. Male Long Evans Hooded rats were randomly divided into sham or rmTBI groups. Significant vestibular deficits were observed both acutely and chronically in the rmTBI groups. Systemic testosterone was administered after the development of chronic vestibular dysfunction. rmTBI animals given testosterone showed improved vestibular function that was sustained for 175 days post-rmTBI. Significant vestibular neuronal cell loss was, however, observed in the rmTBI animals compared to Sham animals at 175 days post-rmTBI and testosterone treatment significantly improved vestibular neuronal survival. Taken together, these data demonstrate a critical restorative role of testosterone in vestibular function following rmTBI. This study has important clinical implications because it identifies testosterone treatment as a viable therapeutic strategy for the long-term recovery of vestibular function following TBI.
Collapse
Affiliation(s)
- Eileen M Foecking
- Loyola University Chicago, Department of Otolaryngology, Burn Shock Trauma Research Institute, Loyola University Chicago, 2160 South 1st Avenue, Maywood, IL 60153, the United States of America; Burn Shock Trauma Research Institute, Loyola University Chicago, 2160 South 1st Avenue, Maywood, IL 60153, the United States of America; Edward Hines Jr. VA Hospital Research Service, Hines, IL 60141, the United States of America.
| | - Arthur B Segismundo
- Loyola University of Chicago, Biomedical Graduate School, 2160 South 1st Avenue, Maywood, IL 60153, the United States of America.
| | - Krista M Lotesto
- Burn Shock Trauma Research Institute, Loyola University Chicago, 2160 South 1st Avenue, Maywood, IL 60153, the United States of America.
| | - Edward J Westfall
- Loyola University Medical Center, Department of Otolaryngology, 2160 South 1st Avenue, Maywood, IL 60153, the United States of America.
| | - Alyssa J Bolduan
- Loyola University Medical Center, Department of Otolaryngology, 2160 South 1st Avenue, Maywood, IL 60153, the United States of America.
| | - Tony K Peter
- Loyola University Medical Center, Department of Otolaryngology, 2160 South 1st Avenue, Maywood, IL 60153, the United States of America.
| | - Douglas G Wallace
- Northern Illinois University, Department of Psychology, 1425 Lincoln Hwy, DeKalb, IL 60115, the United States of America.
| | - Dorothy A Kozlowski
- DePaul University, Department of Biological Sciences and Neuroscience Program, 2325 N., Chicago, IL 60604, the United States of America.
| | - Evan B Stubbs
- Edward Hines Jr. VA Research Service, Hines, IL 60141, the United States of America; Loyola University Medical Center, Department of Otolaryngology, 2160 South 1st Avenue, Maywood, IL 60153, the United States of America.
| | - Sam J Marzo
- Loyola University Medical Center, Department of Otolaryngology, 2160 South 1st Avenue, Maywood, IL 60153, the United States of America.
| | - Susanna C Byram
- Loyola University Medical Center, Department of Anesthesiology and Perioperative Medicine, 2160 South 1st Avenue, Maywood, IL 60153, the United States of America; Edward Hines Jr. VA Hospital Research Service, Hines, IL 60141, the United States of America.
| |
Collapse
|
11
|
Hiskens MI. Targets of neuroprotection and review of pharmacological interventions in traumatic brain injury. J Pharmacol Exp Ther 2022; 382:149-166. [DOI: 10.1124/jpet.121.001023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 05/06/2022] [Indexed: 11/22/2022] Open
|
12
|
Andersen MS, Güler DB, Larsen J, Rich KK, Svenningsen ÅF, Zhang M. The Development of Hindlimb Postural Asymmetry Induced by Focal Traumatic Brain Injury Is Not Related to Serotonin 2A/C Receptor Expression in the Spinal Cord. Int J Mol Sci 2022; 23:ijms23105358. [PMID: 35628167 PMCID: PMC9140651 DOI: 10.3390/ijms23105358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/05/2022] [Accepted: 05/07/2022] [Indexed: 11/16/2022] Open
Abstract
Brain injury and stroke are leading causes of adult disability. Motor deficits are common problems, and their underlying pathological mechanisms remain poorly understood. The serotoninergic system is implicated in both functional recovery from and the occurrence of spasticity after injuries to the central nervous system. This study, which was conducted on rats, investigated the development of limb postural changes and their relationship to the expression of serotonin (5-HT) 2A and 2C receptors in the spinal cord in the 4 weeks after focal traumatic brain injury (TBI) to the right hindlimb sensorimotor cortex. The limb motor deficits were assessed by measuring gait pattern changes during walking and hindlimb postural asymmetry at different time intervals (3−28 days) after surgery. The expressions of the 5-HT2A and 2C receptors in the lumbar spinal cord were investigated using immunohistochemistry. The results showed that all the rats with TBI, independently of the duration of the interval, displayed postural asymmetry with flexion on the contralateral (left) side (>2 mm), while the sham-operated rats showed no apparent postural asymmetry. The TBI rats also had longer stride lengths during walking in both their hindlimbs and their forelimbs compared with the sham rats. For both the TBI and the sham rats, the hind-paw placement angles were larger on the contralateral side in some of the groups. Compared to the sham-operated rats, the 5-HT2A and 2C receptor expression did not significantly change on either side of the lumbar spinal cords of the TBI rats in any of the groups. These results suggest that focal TBI can induce motor deficits lasting a relatively long time, and that these deficits are not related to the expression of the 5-HT2A and 2C receptors in the spinal cord.
Collapse
Affiliation(s)
- Marlene Storm Andersen
- Department of Molecular Medicine, University of Southern Denmark, DK-5000 Odense, Denmark; (M.S.A.); (D.B.G.); (J.L.); (K.K.R.); (Å.F.S.)
| | - Dilârâ Bedriye Güler
- Department of Molecular Medicine, University of Southern Denmark, DK-5000 Odense, Denmark; (M.S.A.); (D.B.G.); (J.L.); (K.K.R.); (Å.F.S.)
| | - Jonas Larsen
- Department of Molecular Medicine, University of Southern Denmark, DK-5000 Odense, Denmark; (M.S.A.); (D.B.G.); (J.L.); (K.K.R.); (Å.F.S.)
| | - Karen Kalhøj Rich
- Department of Molecular Medicine, University of Southern Denmark, DK-5000 Odense, Denmark; (M.S.A.); (D.B.G.); (J.L.); (K.K.R.); (Å.F.S.)
| | - Åsa Fex Svenningsen
- Department of Molecular Medicine, University of Southern Denmark, DK-5000 Odense, Denmark; (M.S.A.); (D.B.G.); (J.L.); (K.K.R.); (Å.F.S.)
- BRIDGE, University of Southern Denmark, DK-5000 Odense, Denmark
| | - Mengliang Zhang
- Department of Molecular Medicine, University of Southern Denmark, DK-5000 Odense, Denmark; (M.S.A.); (D.B.G.); (J.L.); (K.K.R.); (Å.F.S.)
- BRIDGE, University of Southern Denmark, DK-5000 Odense, Denmark
- Correspondence:
| |
Collapse
|
13
|
Golub VM, Reddy DS. Post-Traumatic Epilepsy and Comorbidities: Advanced Models, Molecular Mechanisms, Biomarkers, and Novel Therapeutic Interventions. Pharmacol Rev 2022; 74:387-438. [PMID: 35302046 PMCID: PMC8973512 DOI: 10.1124/pharmrev.121.000375] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Post-traumatic epilepsy (PTE) is one of the most devastating long-term, network consequences of traumatic brain injury (TBI). There is currently no approved treatment that can prevent onset of spontaneous seizures associated with brain injury, and many cases of PTE are refractory to antiseizure medications. Post-traumatic epileptogenesis is an enduring process by which a normal brain exhibits hypersynchronous excitability after a head injury incident. Understanding the neural networks and molecular pathologies involved in epileptogenesis are key to preventing its development or modifying disease progression. In this article, we describe a critical appraisal of the current state of PTE research with an emphasis on experimental models, molecular mechanisms of post-traumatic epileptogenesis, potential biomarkers, and the burden of PTE-associated comorbidities. The goal of epilepsy research is to identify new therapeutic strategies that can prevent PTE development or interrupt the epileptogenic process and relieve associated neuropsychiatric comorbidities. Therefore, we also describe current preclinical and clinical data on the treatment of PTE sequelae. Differences in injury patterns, latency period, and biomarkers are outlined in the context of animal model validation, pathophysiology, seizure frequency, and behavior. Improving TBI recovery and preventing seizure onset are complex and challenging tasks; however, much progress has been made within this decade demonstrating disease modifying, anti-inflammatory, and neuroprotective strategies, suggesting this goal is pragmatic. Our understanding of PTE is continuously evolving, and improved preclinical models allow for accelerated testing of critically needed novel therapeutic interventions in military and civilian persons at high risk for PTE and its devastating comorbidities.
Collapse
Affiliation(s)
- Victoria M Golub
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas
| | - Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas
| |
Collapse
|
14
|
Zhang ZW, Wei P, Zhang GJ, Yan JX, Zhang S, Liang J, Wang XL. Intravenous infusion of the exosomes derived from human umbilical cord mesenchymal stem cells enhance neurological recovery after traumatic brain injury via suppressing the NF-κB pathway. Open Life Sci 2022; 17:189-201. [PMID: 35415238 PMCID: PMC8932398 DOI: 10.1515/biol-2022-0022] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 12/13/2021] [Accepted: 01/03/2022] [Indexed: 02/05/2023] Open
Abstract
Abstract
Traumatic brain injury (TBI) is a predominant cause of death and permanent disability globally. In recent years, much emphasis has been laid on treatments for TBI. Increasing evidence suggests that human umbilical cord mesenchymal stem cells (HUCMSCs) can improve neurological repair after TBI. However, the clinical use of HUCMSCs transplantation in TBI has been limited by immunological rejection, ethical issues, and the risk of tumorigenicity. Many studies have shown that HUCMSCs-derived exosomes may be an alternative approach for HUCMSCs transplantation. We hypothesized that exosomes derived from HUCMSCs could inhibit apoptosis after TBI, reduce neuroinflammation, and promote neurogenesis. A rat model of TBI was established to investigate the efficiency of neurological recovery with exosome therapy. We found that exosomes derived from HUCMSCs significantly ameliorated sensorimotor function and spatial learning in rats after TBI. Moreover, HUCMSCs-derived exosomes significantly reduced proinflammatory cytokine expression by suppressing the NF-κB signaling pathway. Furthermore, we found that HUCMSC-derived exosomes inhibited neuronal apoptosis, reduced inflammation, and promoted neuron regeneration in the injured cortex of rats after TBI. These results indicate that HUCMSCs-derived exosomes may be a promising therapeutic strategy for TBI.
Collapse
Affiliation(s)
- Zhen-Wen Zhang
- Department of Encephalopathy, Affiliated Hospital of Gansu University of Chinese Medicine , Lanzhou 730000 , Gansu , China
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of PAPF , Tianjin 300162 , China
- College of Integrated Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine , Lanzhou 730000 , Gansu , China
| | - Pan Wei
- Department of Neurosurgery, The First People’s Hospital of Long Quan Yi District , Cheng Du 610000 , Si Chuan , China
| | - Gui-Jun Zhang
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University , Chengdu 610041 , Sichuan , China
| | - Jing-Xing Yan
- Department of Encephalopathy, Affiliated Hospital of Gansu University of Chinese Medicine , Lanzhou 730000 , Gansu , China
- College of Integrated Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine , Lanzhou 730000 , Gansu , China
| | - Sai Zhang
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of PAPF , Tianjin 300162 , China
| | - Jin Liang
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of PAPF , Tianjin 300162 , China
| | - Xiao-Li Wang
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of PAPF , Tianjin 300162 , China
| |
Collapse
|
15
|
White M, Duquette-Laplante F, Jutras B, Bursch C, Koravand A. A Retrospective Study of the Effects of Traumatic Brain Injury on Auditory Function: From a Clinical Perspective. NEUROSCI 2022; 3:52-62. [PMID: 39484668 PMCID: PMC11523697 DOI: 10.3390/neurosci3010004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 01/10/2022] [Indexed: 11/03/2024] Open
Abstract
PURPOSE The main purpose of this retrospective study was to identify auditory dysfunctions related to traumatic brain injury (TBI) in individuals evaluated in an Audiology clinic. METHOD Peripheral and central auditory evaluations were performed from March 2014 to June 2018 in 26 patients (14 males) with TBI. The age of the participants ranged from 9 to 59 years old (34.24 ± 15.21). Six participants had blast-related TBI and 20 had blunt force TBI. Sixteen experienced a single TBI event whereas ten experienced several. Correlation analyses were performed to verify the relationship, if any, between the number of auditory tests failed and the number, type, and severity of TBIs. RESULT All participants failed at least one auditory test. Nearly 60% had abnormal results on degraded speech tests (compressed and echoed, filtered or in background noise) and 25% had a high frequency hearing loss. There was no statistically significant correlation between the number of auditory tests failed and the number, type, and severity of TBIs. CONCLUSION Results indicated negative and heterogenous effects of TBI on peripheral and central auditory function and highlighted the need for a more extensive auditory assessment in individuals with TBI.
Collapse
Affiliation(s)
- Mira White
- Audiology and Speech Pathology Program, School of Rehabilitation Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (M.W.); (F.D.-L.)
| | - Fauve Duquette-Laplante
- Audiology and Speech Pathology Program, School of Rehabilitation Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (M.W.); (F.D.-L.)
| | - Benoît Jutras
- School of Speech-Language Pathology and Audiology, Faculty of Medicine, Université de Montréal, Montreal, QC H3N 1X7, Canada;
| | - Caryn Bursch
- APD Ottawa, Audiology Private Practice, Ottawa, ON K2V 5G7, Canada;
| | - Amineh Koravand
- Audiology and Speech Pathology Program, School of Rehabilitation Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (M.W.); (F.D.-L.)
| |
Collapse
|
16
|
Varlow C, Knight AC, McQuade P, Vasdev N. Characterization of neuroinflammatory positron emission tomography biomarkers in chronic traumatic encephalopathy. Brain Commun 2022; 4:fcac019. [PMID: 35198978 PMCID: PMC8856182 DOI: 10.1093/braincomms/fcac019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/10/2021] [Accepted: 02/01/2022] [Indexed: 11/12/2022] Open
Abstract
Chronic traumatic encephalopathy is a neurological disorder associated with head trauma and is confirmed upon autopsy. PET imaging of chronic traumatic encephalopathy may provide a means to move towards ante-mortem diagnosis and therapeutic intervention following brain injuries. Characterization of the neuroinflammatory PET biomarkers, 18 kDa translocator protein and monoamine oxidase-B was conducted using [3H]PBR-28 and [3H]L-deprenyl, respectively, in post-mortem chronic traumatic encephalopathy brain tissue. [3H]PBR-28 displayed high specific binding in both chronic traumatic encephalopathy (95.40 ± 1.87%; n = 11 cases) and healthy controls (89.89 ± 8.52%, n = 3 cases). Cell-type expression of the 18 kDa translocator protein was confirmed by immunofluorescence to microglia, astrocyte and macrophage markers. [3H]L-deprenyl also displayed high specific binding in chronic traumatic encephalopathy (96.95 ± 1.43%; n = 12 cases) and healthy controls (93.24 ± 0.43%; n = 2 cases), with the distribution co-localized to astrocytes by immunofluorescence. Saturation analysis was performed to quantify the target density of the 18 kDa translocator protein and monoamine oxidase-B in both chronic traumatic encephalopathy and healthy control tissue. Using [3H]PBR-28, the target density of the 18 kDa translocator protein in healthy controls was 177.91 ± 56.96 nM (n = 7 cases; mean ± standard deviation); however, a highly variable target density (345.84 ± 372.42 nM; n = 11 cases; mean ± standard deviation) was measured in chronic traumatic encephalopathy. [3H]L-deprenyl quantified a monoamine oxidase-B target density of 304.23 ± 115.93 nM (n = 8 cases; mean ± standard deviation) in healthy control tissue and is similar to the target density in chronic traumatic encephalopathy tissues (365.80 ± 128.55 nM; n = 12 cases; mean ± standard deviation). A two-sample t-test determined no significant difference in the target density values of the 18 kDa translocator protein and monoamine oxidase-B between healthy controls and chronic traumatic encephalopathy (P > 0.05), albeit a trend towards increased expression of both targets was observed in chronic traumatic encephalopathy. To our knowledge, this work represents the first in vitro characterization of 18 kDa translocator protein and monoamine oxidase-B in chronic traumatic encephalopathy and reveals the variability in neuroinflammatory pathology following brain injuries. These preliminary findings will be considered when designing PET imaging studies after brain injury and for the ultimate goal of imaging chronic traumatic encephalopathy in vivo.
Collapse
Affiliation(s)
- Cassis Varlow
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada M5T 1R8
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada M5S 1A8
| | - Ashley C. Knight
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada M5T 1R8
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada M5S 1A8
| | - Paul McQuade
- Takeda Pharmaceutical Company, Cambridge, MA 02139, USA
| | - Neil Vasdev
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada M5T 1R8
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada M5S 1A8
| |
Collapse
|
17
|
Tucker LB, McCabe JT. Measuring Anxiety-Like Behaviors in Rodent Models of Traumatic Brain Injury. Front Behav Neurosci 2021; 15:682935. [PMID: 34776887 PMCID: PMC8586518 DOI: 10.3389/fnbeh.2021.682935] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 10/06/2021] [Indexed: 12/31/2022] Open
Abstract
Anxiety is a common complaint following acquired traumatic brain injury (TBI). However, the measurement of dysfunctional anxiety behavioral states following experimental TBI in rodents is complex. Some studies report increased anxiety after TBI, whereas others find a decreased anxiety-like state, often described as increased risk-taking behavior or impulsivity. These inconsistencies may reflect a lack of standardization of experimental injury models or of behavioral testing techniques. Here, we review the most commonly employed unconditioned tests of anxiety and discuss them in a context of experimental TBI. Special attention is given to the effects of repeated testing, and consideration of potential sensory and motor confounds in injured rodents. The use of multiple tests and alternative data analysis methods are discussed, as well as the potential for the application of common data elements (CDEs) as a means of providing a format for documentation of experimental details and procedures of each published research report. CDEs may improve the rigor, reproducibility, as well as endpoint for better relating findings with clinical TBI phenotypes and the final goal of translation. While this may not resolve all incongruities in findings across laboratories, it is seen as a way forward for standardized and universal data collection for improvement of data quality and sharing, and advance therapies for neuropsychiatric symptoms that often present for decades following TBI.
Collapse
Affiliation(s)
- Laura B Tucker
- Preclinical Behavior and Models Core, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States.,Department of Anatomy, Physiology and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Joseph T McCabe
- Preclinical Behavior and Models Core, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States.,Department of Anatomy, Physiology and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
18
|
Chen YH, Chen YC, Hwang LL, Yang LY, Lu DY. Deficiency in Androgen Receptor Aggravates Traumatic Brain Injury-Induced Pathophysiology and Motor Deficits in Mice. Molecules 2021; 26:6250. [PMID: 34684832 PMCID: PMC8537172 DOI: 10.3390/molecules26206250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/12/2021] [Accepted: 10/13/2021] [Indexed: 11/16/2022] Open
Abstract
Androgens have been shown to have a beneficial effect on brain injury and lower reactive astrocyte expression after TBI. Androgen receptors (ARs) are known to mediate the neuroprotective effects of androgens. However, whether ARs play a crucial role in TBI remains unknown. In this study, we investigated the role of ARs in TBI pathophysiology, using AR knockout (ARKO) mice. We used the controlled cortical impact model to produce primary and mechanical brain injuries and assessed motor function and brain-lesion volume. In addition, the AR knockout effects on necrosis and autophagy were evaluated after TBI. AR knockout significantly increased TBI-induced expression of the necrosis marker alpha-II-spectrin breakdown product 150 and astrogliosis marker glial fibrillary acidic protein. In addition, the TBI-induced astrogliosis increase in ARKO mice lasted for three weeks after a TBI. The autophagy marker Beclin-1 was also enhanced in ARKO mice compared with wild-type mice after TBI. Our results also indicated that ARKO mice showed a more unsatisfactory performance than wild-type mice in a motor function test following TBI. Further, they were observed to have more severe lesions than wild-type mice after injury. These findings strongly suggest that ARs play a role in TBI.
Collapse
Affiliation(s)
- Yu-Hsin Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-H.C.); (Y.-C.C.); (L.-L.H.)
| | - Yen-Chou Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-H.C.); (Y.-C.C.); (L.-L.H.)
| | - Ling-Ling Hwang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-H.C.); (Y.-C.C.); (L.-L.H.)
| | - Liang-Yo Yang
- Department of Physiology, School of Medicine, College of Medicine, China Medical University, Taichung 40402, Taiwan
- Laboratory of Neural Repair, Department of Medical Research, China Medical University Hospital, Taichung 40447, Taiwan
| | - Dah-Yuu Lu
- Department of Pharmacology, School of Medicine, China Medical University, Taichung 404333, Taiwan
- Department of Photonics and Communication Engineering, Asia University, Taichung 404333, Taiwan
| |
Collapse
|
19
|
Traumatic Brain Injury: An Age-Dependent View of Post-Traumatic Neuroinflammation and Its Treatment. Pharmaceutics 2021; 13:pharmaceutics13101624. [PMID: 34683918 PMCID: PMC8537402 DOI: 10.3390/pharmaceutics13101624] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/24/2021] [Accepted: 09/26/2021] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability all over the world. TBI leads to (1) an inflammatory response, (2) white matter injuries and (3) neurodegenerative pathologies in the long term. In humans, TBI occurs most often in children and adolescents or in the elderly, and it is well known that immune responses and the neuroregenerative capacities of the brain, among other factors, vary over a lifetime. Thus, age-at-injury can influence the consequences of TBI. Furthermore, age-at-injury also influences the pharmacological effects of drugs. However, the post-TBI inflammatory, neuronal and functional consequences have been mostly studied in experimental young adult animal models. The specificity and the mechanisms underlying the consequences of TBI and pharmacological responses are poorly understood in extreme ages. In this review, we detail the variations of these age-dependent inflammatory responses and consequences after TBI, from an experimental point of view. We investigate the evolution of microglial, astrocyte and other immune cells responses, and the consequences in terms of neuronal death and functional deficits in neonates, juvenile, adolescent and aged male animals, following a single TBI. We also describe the pharmacological responses to anti-inflammatory or neuroprotective agents, highlighting the need for an age-specific approach to the development of therapies of TBI.
Collapse
|
20
|
Ackermans NL, Varghese M, Wicinski B, Torres J, De Gasperi R, Pryor D, Elder GA, Gama Sosa MA, Reidenberg JS, Williams TM, Hof PR. Unconventional animal models for traumatic brain injury and chronic traumatic encephalopathy. J Neurosci Res 2021; 99:2463-2477. [PMID: 34255876 PMCID: PMC8596618 DOI: 10.1002/jnr.24920] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/09/2021] [Accepted: 06/24/2021] [Indexed: 12/11/2022]
Abstract
Traumatic brain injury (TBI) is one of the main causes of death worldwide. It is a complex injury that influences cellular physiology, causes neuronal cell death, and affects molecular pathways in the brain. This in turn can result in sensory, motor, and behavioral alterations that deeply impact the quality of life. Repetitive mild TBI can progress into chronic traumatic encephalopathy (CTE), a neurodegenerative condition linked to severe behavioral changes. While current animal models of TBI and CTE such as rodents, are useful to explore affected pathways, clinical findings therein have rarely translated into clinical applications, possibly because of the many morphofunctional differences between the model animals and humans. It is therefore important to complement these studies with alternative animal models that may better replicate the individuality of human TBI. Comparative studies in animals with naturally evolved brain protection such as bighorn sheep, woodpeckers, and whales, may provide preventive applications in humans. The advantages of an in-depth study of these unconventional animals are threefold. First, to increase knowledge of the often-understudied species in question; second, to improve common animal models based on the study of their extreme counterparts; and finally, to tap into a source of biological inspiration for comparative studies and translational applications in humans.
Collapse
Affiliation(s)
- Nicole L Ackermans
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Anatomy and Functional Morphology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Merina Varghese
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bridget Wicinski
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joshua Torres
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rita De Gasperi
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- General Medical Research Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY, USA
| | - Dylan Pryor
- General Medical Research Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY, USA
| | - Gregory A Elder
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Neurology Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY, USA
| | - Miguel A Gama Sosa
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- General Medical Research Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY, USA
| | - Joy S Reidenberg
- Center for Anatomy and Functional Morphology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Terrie M Williams
- Department of Ecology and Evolutionary Biology, University of California, Santa Cruz, CA, USA
| | - Patrick R Hof
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
21
|
Semple BD, Raghupathi R. A Pro-social Pill? The Potential of Pharmacological Treatments to Improve Social Outcomes After Pediatric Traumatic Brain Injury. Front Neurol 2021; 12:714253. [PMID: 34489853 PMCID: PMC8417315 DOI: 10.3389/fneur.2021.714253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/30/2021] [Indexed: 11/13/2022] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of injury-induced disability in young children worldwide, and social behavior impairments in this population are a significant challenge for affected patients and their families. The protracted trajectory of secondary injury processes triggered by a TBI during early life-alongside ongoing developmental maturation-offers an extended time window when therapeutic interventions may yield functional benefits. This mini-review explores the scarce but promising pre-clinical literature to date demonstrating that social behavior impairments after early life brain injuries can be modified by drug therapies. Compounds that provide broad neuroprotection, such as those targeting neuroinflammation, oxidative stress, axonal injury and/or myelination, may prevent social behavior impairments by reducing secondary neuropathology. Alternatively, targeted treatments that promote affiliative behaviors, exemplified by the neuropeptide oxytocin, may reduce the impact of social dysfunction after pediatric TBI. Complementary literature from other early life neurodevelopmental conditions such as hypoxic ischemic encephalopathy also provides avenues for future research in neurotrauma. Knowledge gaps in this emerging field are highlighted throughout, toward the goal of accelerating translational research to support optimal social functioning after a TBI during early childhood.
Collapse
Affiliation(s)
- Bridgette D Semple
- Department of Neuroscience, Monash University, Prahran, VIC, Australia.,Department of Neurology, Alfred Health, Prahran, VIC, Australia.,Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Ramesh Raghupathi
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences and Professional Studies, Drexel University College of Medicine, Philadelphia, PA, United States.,Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
22
|
Zamani A, Ryan NP, Wright DK, Caeyenberghs K, Semple BD. The Impact of Traumatic Injury to the Immature Human Brain: A Scoping Review with Insights from Advanced Structural Neuroimaging. J Neurotrauma 2021; 37:724-738. [PMID: 32037951 DOI: 10.1089/neu.2019.6895] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Traumatic brain injury (TBI) during critical periods of early-life brain development can affect the normal formation of brain networks responsible for a range of complex social behaviors. Because of the protracted nature of brain and behavioral development, deficits in cognitive and socioaffective behaviors may not become evident until late adolescence and early adulthood, when such skills are expected to reach maturity. In addition, multiple pre- and post-injury factors can interact with the effects of early brain insult to influence long-term outcomes. In recent years, with advancements in magnetic-resonance-based neuroimaging techniques and analysis, studies of the pediatric population have revealed a link between neurobehavioral deficits, such as social dysfunction, with white matter damage. In this review, in which we focus on contributions from Australian researchers to the field, we have highlighted pioneering longitudinal studies in pediatric TBI, in relation to social deficits specifically. We also discuss the use of advanced neuroimaging and novel behavioral assays in animal models of TBI in the immature brain. Together, this research aims to understand the relationship between injury consequences and ongoing brain development after pediatric TBI, which promises to improve prediction of the behavioral deficits that emerge in the years subsequent to early-life injury.
Collapse
Affiliation(s)
- Akram Zamani
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Nicholas P Ryan
- Cognitive Neuroscience Unit, School of Psychology, Deakin University, Melbourne, Victoria, Australia.,Brain & Mind Research, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - David K Wright
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Karen Caeyenberghs
- Cognitive Neuroscience Unit, School of Psychology, Deakin University, Melbourne, Victoria, Australia
| | - Bridgette D Semple
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia.,Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
23
|
Morin A, Mouzon B, Ferguson S, Paris D, Saltiel N, Browning M, Mullan M, Crawford F. A 3-month-delayed treatment with anatabine improves chronic outcomes in two different models of repetitive mild traumatic brain injury in hTau mice. Sci Rep 2021; 11:7900. [PMID: 33846461 PMCID: PMC8041866 DOI: 10.1038/s41598-021-87161-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 03/24/2021] [Indexed: 02/01/2023] Open
Abstract
To date, an overwhelming number of preclinical studies have addressed acute treatment in mild TBI (mTBI) and repetitive mTBI (r-mTBI), whereas, in humans, there often exists a significant time gap between the injury and the first medical intervention. Our study focused on a delayed treatment with anatabine, an anti-inflammatory compound, in hTau mice using two different models of r-mTBI. The rationale for using two models of the same impact but different frequencies (5 hit mTBI over 9 days and 24 hit mTBI over 90 days) was chosen to address the heterogeneity of r-mTBI in clinical population. Following the last injury in each model, three months elapsed before the initiation of treatment. Anatabine was administered in drinking water for 3 months thereafter. Our data demonstrated that a 3-month delayed treatment with anatabine mitigated astrogliosis in both TBI paradigms but improved cognitive functions only in more-frequently-injured mice (24 hit mTBI). We also found that anatabine decreased the phosphorylation of tau protein and NFκB, which were increased after r-mTBI in both models. The ability of anatabine to suppress these mechanisms suggests that delayed treatment can be effective for clinical population of r-mTBI. The discrepancy between the two models with regard to changes in cognitive performance suggests that r-mTBI heterogeneity may influence treatment efficiency and should be considered in therapeutic development.
Collapse
Affiliation(s)
- Alexander Morin
- The Roskamp Institute, 2040 Whitfield Ave., Sarasota, FL, 34243, USA.
- The Open University, Milton-Keynes, UK.
- James A Haley Veterans Administration, Tampa, FL, USA.
| | - Benoit Mouzon
- The Roskamp Institute, 2040 Whitfield Ave., Sarasota, FL, 34243, USA
- The Open University, Milton-Keynes, UK
- James A Haley Veterans Administration, Tampa, FL, USA
| | - Scott Ferguson
- The Roskamp Institute, 2040 Whitfield Ave., Sarasota, FL, 34243, USA
- The Open University, Milton-Keynes, UK
- James A Haley Veterans Administration, Tampa, FL, USA
| | - Daniel Paris
- The Roskamp Institute, 2040 Whitfield Ave., Sarasota, FL, 34243, USA
- The Open University, Milton-Keynes, UK
- James A Haley Veterans Administration, Tampa, FL, USA
| | - Nicole Saltiel
- The Roskamp Institute, 2040 Whitfield Ave., Sarasota, FL, 34243, USA
- James A Haley Veterans Administration, Tampa, FL, USA
| | | | - Mike Mullan
- The Roskamp Institute, 2040 Whitfield Ave., Sarasota, FL, 34243, USA
- The Open University, Milton-Keynes, UK
| | - Fiona Crawford
- The Roskamp Institute, 2040 Whitfield Ave., Sarasota, FL, 34243, USA
- The Open University, Milton-Keynes, UK
- James A Haley Veterans Administration, Tampa, FL, USA
| |
Collapse
|
24
|
McNamara EH, Grillakis AA, Tucker LB, McCabe JT. The closed-head impact model of engineered rotational acceleration (CHIMERA) as an application for traumatic brain injury pre-clinical research: A status report. Exp Neurol 2020; 333:113409. [PMID: 32692987 DOI: 10.1016/j.expneurol.2020.113409] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 06/18/2020] [Accepted: 07/14/2020] [Indexed: 02/06/2023]
Abstract
Closed-head traumatic brain injury (TBI) is a worldwide concern with increasing prevalence and cost to society. Rotational acceleration is a primary mechanism in TBI that results from tissue strains that give rise to diffuse axonal injury. The Closed-Head Impact Model of Engineered Rotational Acceleration (CHIMERA) was recently introduced as a method for the study of impact acceleration effects in pre-clinical TBI research. This review provides a survey of the published literature implementing the CHIMERA device and describes pathological, imaging, neurophysiological, and behavioral findings. Findings show CHIMERA inflicts damage in white matter tracts as a key area of injury. Behaviorally, repeated studies have shown motor deficits and more chronic cognitive effects after CHIMERA injury. Good progress with model application has been accomplished by investigators attending to what is required for model validation. However, the majority of CHIMERA studies only utilize adult male mice. To further establish this model, more work with female animals and various age groups need to be performed, as well as studies to further establish and standardize methodologies for validation of the models for clinical relevance. Common data elements to standardize the reporting methodology for the CHIMERA literature are suggested.
Collapse
Affiliation(s)
- Eileen H McNamara
- Neuroscience Graduate Program, F.E. Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20817-4799, USA; Department of Anatomy, Physiology & Genetics, F.E. Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20817-4799, USA
| | - Antigone A Grillakis
- Neuroscience Graduate Program, F.E. Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20817-4799, USA; Department of Anatomy, Physiology & Genetics, F.E. Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20817-4799, USA
| | - Laura B Tucker
- Department of Anatomy, Physiology & Genetics, F.E. Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20817-4799, USA; Pre-Clinical Studies Core, Center for Neuroscience and Regenerative Medicine, F.E. Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20817-4799, USA
| | - Joseph T McCabe
- Neuroscience Graduate Program, F.E. Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20817-4799, USA; Department of Anatomy, Physiology & Genetics, F.E. Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20817-4799, USA; Pre-Clinical Studies Core, Center for Neuroscience and Regenerative Medicine, F.E. Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20817-4799, USA.
| |
Collapse
|
25
|
Yu J, Zhang Y, Ma H, Zeng R, Liu R, Wang P, Jin X, Zhao Y. Epitranscriptomic profiling of N6-methyladenosine-related RNA methylation in rat cerebral cortex following traumatic brain injury. Mol Brain 2020; 13:11. [PMID: 31992337 PMCID: PMC6986156 DOI: 10.1186/s13041-020-0554-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 01/19/2020] [Indexed: 12/31/2022] Open
Abstract
Background N6-methyladenosine (m6A) is the most prevalent post-transcriptional modification of eukaryotic mRNA. It has been reported that there is a stimulus-dependent regulation of m6A in the mammalian central nervous system in response to sensory experience, learning, and injury. The mRNA m6A methylation pattern in rat cortex after traumatic brain injury (TBI) has not been investigated. Results In this study, we conducted a genome-wide profiling of mRNA m6A methylation in rat cortex via methylated RNA immunoprecipitation sequencing (MeRIP-Seq). After TBI, the expressions of METTL14 and FTO were significantly down-regulated in rat cerebral cortex. Using MeRIP-Seq, we identified a total of 2165 significantly changed peaks, of which 1062 were significantly up-regulated and 1103 peaks were significantly down-regulated. These m6A peaks were located across 1850 genes. The analysis of both m6A peaks and mRNA expression revealed that there were 175 mRNA significantly altered methylation and expression levels after TBI. Moreover, it was found that functional FTO is necessary to repair neurological damage caused by TBI but has no effect on the spatial learning and memory abilities of TBI rats by using FTO inhibitor FB23–2. Conclusion This study explored the m6A methylation pattern of mRNA after TBI in rat cortex and identified FTO as possible intervention targets in the epigenetic modification of TBI.
Collapse
Affiliation(s)
- Jiangtao Yu
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yuxian Zhang
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Haoli Ma
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Rong Zeng
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Ruining Liu
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Pengcheng Wang
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Xiaoqing Jin
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.,Hubei Clinical Research Center for Emergency and Resuscitation, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yan Zhao
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China. .,Hubei Clinical Research Center for Emergency and Resuscitation, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
26
|
Shahror RA, Linares GR, Wang Y, Hsueh SC, Wu CC, Chuang DM, Chiang YH, Chen KY. Transplantation of Mesenchymal Stem Cells Overexpressing Fibroblast Growth Factor 21 Facilitates Cognitive Recovery and Enhances Neurogenesis in a Mouse Model of Traumatic Brain Injury. J Neurotrauma 2019; 37:14-26. [PMID: 31298621 DOI: 10.1089/neu.2019.6422] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Traumatic brain injury (TBI) is a progressive and complex pathological condition that results in multiple adverse consequences, including impaired learning and memory. Transplantation of mesenchymal stem cells (MSCs) has produced limited benefits in experimental TBI models. Fibroblast growth factor 21 (FGF21) is a novel metabolic regulator that has neuroprotective effects, promotes remyelination, enhances angiogenesis, and elongates astrocytic processes. In this study, MSCs were genetically engineered to overexpress FGF21 in order to improve their efficacy in TBI. MSCs overexpressing FGF21 (MSC-FGF21) were transplanted to mouse brain by intracerebroventricular injection 24 h after TBI was induced by controlled cortical impact (CCI). Hippocampus-dependent spatial learning and memory, assessed by the Morris water maze test, was markedly decreased 3-4 weeks after TBI, a deficit that was robustly recovered by treatment with MSC-FGF21, but not MSC-mCherry control. Hippocampus-independent learning and memory, assessed by the novel object recognition test, was also impaired; these effects were blocked by treatment with both MSC-FGF21 and MSC-mCherry control. FGF21 protein levels in the ipsilateral hippocampus were drastically reduced 4 weeks post-TBI, a loss that was restored by treatment with MSC-FGF21, but not MSC-mCherry. MSC-FGF21 treatment also partially restored TBI-induced deficits in neurogenesis and maturation of immature hippocampal neurons, whereas MSC-mCherry was less effective. Finally, MSC-FGF21 treatment also normalized TBI-induced impairments in dendritic arborization of hippocampal neurons. Taken together, the results indicate that MSC-FGF21 treatment significantly improved TBI-induced spatial memory deficits, impaired hippocampal neurogenesis, and abnormal dendritic morphology. Future clinical investigations using MSC-FGF21 to improve post-TBI outcomes are warranted.
Collapse
Affiliation(s)
- Rami Ahmad Shahror
- Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei, Taiwan.,TMU Neuroscience Research Center, Taipei Medical University, Taipei, Taiwan
| | - Gabriel R Linares
- Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland.,Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Yun Wang
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan
| | - Shih-Chang Hsueh
- Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei, Taiwan.,TMU Neuroscience Research Center, Taipei Medical University, Taipei, Taiwan
| | - Chung-Che Wu
- TMU Neuroscience Research Center, Taipei Medical University, Taipei, Taiwan.,Department of Neurosurgery, Taipei Medical University Hospital, Taipei, Taiwan.,Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - De-Maw Chuang
- Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei, Taiwan.,Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Yung-Hsiao Chiang
- Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei, Taiwan.,TMU Neuroscience Research Center, Taipei Medical University, Taipei, Taiwan.,Department of Neurosurgery, Taipei Medical University Hospital, Taipei, Taiwan.,Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kai-Yun Chen
- Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei, Taiwan.,TMU Neuroscience Research Center, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
27
|
Li L, Chopp M, Ding G, Davoodi-Bojd E, Li Q, Mahmood A, Xiong Y, Jiang Q. Diffuse white matter response in trauma-injured brain to bone marrow stromal cell treatment detected by diffusional kurtosis imaging. Brain Res 2019; 1717:127-135. [PMID: 31009610 PMCID: PMC6571170 DOI: 10.1016/j.brainres.2019.04.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 03/25/2019] [Accepted: 04/18/2019] [Indexed: 12/14/2022]
Abstract
Diffuse white matter (WM) response to traumatic brain injury (TBI) and transplantation of human bone marrow stromal cells (hMSCs) after the injury were non-invasively and dynamically investigated. Male Wistar rats (300-350 g) subjected to TBI were intravenously injected with 1 ml of saline (n = 10) or with hMSCs in suspension (∼3 × 106 hMSCs, n = 10) 1-week post-TBI. MRI measurements of T2-weighted imaging and diffusional kurtosis imaging (DKI) were acquired on all animals at multiple time points up to 3-months post-injury. Functional outcome was assessed using the Morris water maze test. DKI-derived metrics of fractional anisotropy (FA), axonal water fraction (AWF) and radial kurtosis (RK) longitudinally reveal an evolving pattern of structural alteration post-TBI occurring in the brain region remote from primary impact site. The progressive structural change is characterized by gradual disruption of WM integrity at an early stage (weeks post-TBI), followed by spontaneous recovery at a later stage (months post-TBI). Transplantation of hMSCs post-TBI promotes this structural plasticity as indicated by significantly increased FA and AWF in conjunction with substantially elevated RK at the later stage. Our long-term imaging data demonstrate that hMSC therapy leads to modified temporal profiles of these metrics, inducing an earlier presence of enhanced structural remodeling, which may contribute to improved functional recovery.
Collapse
Affiliation(s)
- Lian Li
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA.
| | - Michael Chopp
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; Department of Physics, Oakland University, Rochester, MI 48309, USA.
| | - Guangliang Ding
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA.
| | | | - Qingjiang Li
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA.
| | - Asim Mahmood
- Department of Neurosurgery, Henry Ford Health System, Detroit, MI 48208, USA.
| | - Ye Xiong
- Department of Neurosurgery, Henry Ford Health System, Detroit, MI 48208, USA.
| | - Quan Jiang
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA.
| |
Collapse
|
28
|
Zhou Y, Shao A, Xu W, Wu H, Deng Y. Advance of Stem Cell Treatment for Traumatic Brain Injury. Front Cell Neurosci 2019; 13:301. [PMID: 31456663 PMCID: PMC6700304 DOI: 10.3389/fncel.2019.00301] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 06/19/2019] [Indexed: 01/01/2023] Open
Abstract
Traumatic brain injury (TBI) is an important cause of human mortality and morbidity, which can induce serious neurological damage. At present, clinical treatments for neurological dysfunction after TBI include hyperbaric oxygen, brain stimulation and behavioral therapy, but the therapeutic effect is not satisfactory. Recent studies have found that exogenous stem cells can migrate to damaged brain tissue, then participate in the repair of damaged brain tissue by further differentiation to replace damaged cells, while releasing anti-inflammatory factors and growth factors, thereby significantly improving neurological function. This article will mainly review the effects, deficiencies and related mechanisms of different types of stem cells in TBI.
Collapse
Affiliation(s)
- Yunxiang Zhou
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Weilin Xu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Haijian Wu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yongchuan Deng
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
29
|
Yip PK, Chapman GE, Sillito RR, Ip THR, Akhigbe G, Becker SC, Price AW, Michael-Titus AT, Armstrong JD, Tremoleda JL. Studies on long term behavioural changes in group-housed rat models of brain and spinal cord injury using an automated home cage recording system. J Neurosci Methods 2019; 321:49-63. [PMID: 30991030 DOI: 10.1016/j.jneumeth.2019.04.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 04/10/2019] [Accepted: 04/12/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND Neurotrauma patients face major neurological sequelae. The failure in the preclinical-to-clinical translation of candidate therapies could be due to poor evaluation of rodent behaviours after neurotrauma. NEW METHOD A home cage automated system was used to study the long term behaviour of individual rats with traumatic brain injury (TBI), spinal cord injury (SCI) and non-CNS injured controls, whilst group-housed in their home cages. Naïve rats were used as baseline controls. Automated locomotor activity and body temperature recordings were carried out 24 h /day for 3 days/week during 12 weeks post-injury. Behavioural patterns, including aggression, rearing, grooming, feeding and drinking were analysed from automated video recordings during week 1, 6 and 12. RESULTS SCI animals showed a lower locomotor activity compared to TBI or control animals during light and dark phases. TBI animals showed a higher aggression during the dark phase in the first week post-injury compared to SCI or control animals. Individual grooming and rearing were reduced in SCI animals compared to TBI and control animals in the first week post-injury during the dark phase. No differences in drinking or feeding were detected between groups. Locomotor activity did not differ between naïve male and female rats, but body temperature differ between light and dark phases for both. STANDARD METHODS Injury severity was compared to standard SCI and TBI behaviour scores (BBB and mNSS, respectively) and histological analysis. CONCLUSIONS This study demonstrates the practical benefits of using a non-intrusive automated home cage recording system to observe long term individual behaviour of group-housed SCI and TBI rats.
Collapse
Affiliation(s)
- Ping K Yip
- Centre for Neuroscience, Surgery and Trauma, Centre for Trauma Sciences, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - George E Chapman
- Centre for Neuroscience, Surgery and Trauma, Centre for Trauma Sciences, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | | | - T H Richard Ip
- Centre for Neuroscience, Surgery and Trauma, Centre for Trauma Sciences, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Georgia Akhigbe
- Centre for Neuroscience, Surgery and Trauma, Centre for Trauma Sciences, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Stephanie C Becker
- Centre for Neuroscience, Surgery and Trauma, Centre for Trauma Sciences, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Anthony W Price
- Biological Services, Queen Mary University of London, London, United Kingdom
| | - Adina T Michael-Titus
- Centre for Neuroscience, Surgery and Trauma, Centre for Trauma Sciences, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - J Douglas Armstrong
- Actual Analytics Ltd, Edinburgh, United Kingdom; School of Informatics, Institute for Adaptive and Neural Computation. University of Edinburgh, Edinburgh, United Kingdom
| | - Jordi L Tremoleda
- Centre for Neuroscience, Surgery and Trauma, Centre for Trauma Sciences, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom; Biological Services, Queen Mary University of London, London, United Kingdom.
| |
Collapse
|
30
|
Ma X, Aravind A, Pfister BJ, Chandra N, Haorah J. Animal Models of Traumatic Brain Injury and Assessment of Injury Severity. Mol Neurobiol 2019; 56:5332-5345. [DOI: 10.1007/s12035-018-1454-5] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 12/07/2018] [Indexed: 10/27/2022]
|
31
|
Shaver TK, Ozga JE, Zhu B, Anderson KG, Martens KM, Vonder Haar C. Long-term deficits in risky decision-making after traumatic brain injury on a rat analog of the Iowa gambling task. Brain Res 2018; 1704:103-113. [PMID: 30296430 DOI: 10.1016/j.brainres.2018.10.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 08/28/2018] [Accepted: 10/04/2018] [Indexed: 12/20/2022]
Abstract
Traumatic brain injury (TBI) affects 2.8 million people annually in the United States, with significant populations suffering from ongoing cognitive dysfunction. Impairments in decision-making can have major implications for patients and their caregivers, often enduring for years to decades, yet are rarely explored in experimental TBI. In the current study, the Rodent Gambling Task (RGT), an Iowa Gambling Task analog, was used to assess risk-based decision-making and motor impulsivity after TBI. During testing, rats chose between options associated with different probabilities of reinforcement (sucrose) or punishment (timeout). To determine effects of TBI on learned behaviors versus the learning process, rats were trained either before, or after, a bilateral frontal controlled cortical impact TBI, and then assessed for 12 weeks. To evaluate the degree to which monoamine systems, such as dopamine, were affected by TBI, rats were given an amphetamine challenge, and behavior recorded. Injury immediately and chronically decreased optimal decision-making, and biased rats towards both riskier, and safer (but suboptimal) choices, regardless of prior learning history. TBI also increased motor impulsivity across time, reflecting ongoing neural changes. Despite these similarities in trained and acquisition rats, those that learned the task after injury demonstrated reduced effects of amphetamine on optimal decision-making, suggesting a lesser role of monoamines in post-injury learning. Amphetamine also dose-dependently reduced motor impulsivity in injured rats. This study opens up the investigation of psychiatric-like dysfunction in animal models of TBI and tasks such as the RGT will be useful in identifying therapeutics for the chronic post-injury period.
Collapse
Affiliation(s)
- Trinity K Shaver
- Department of Psychology, West Virginia University, Morgantown, WV, USA
| | - Jenny E Ozga
- Department of Psychology, West Virginia University, Morgantown, WV, USA
| | - Binxing Zhu
- Department of Psychology, West Virginia University, Morgantown, WV, USA
| | - Karen G Anderson
- Department of Psychology, West Virginia University, Morgantown, WV, USA
| | - Kris M Martens
- Department of Psychology, West Virginia University, Morgantown, WV, USA
| | - Cole Vonder Haar
- Department of Psychology, West Virginia University, Morgantown, WV, USA.
| |
Collapse
|
32
|
Ikonomovic MD, Abrahamson EE, Carlson SW, Graham SH, Dixon CE. Novel therapies for combating chronic neuropathological sequelae of TBI. Neuropharmacology 2018; 145:160-176. [PMID: 29933008 DOI: 10.1016/j.neuropharm.2018.06.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 06/13/2018] [Accepted: 06/15/2018] [Indexed: 02/06/2023]
Abstract
Traumatic brain injury (TBI) is a risk factor for development of chronic neurodegenerative disorders later in life. This review summarizes the current knowledge and concepts regarding the connection between long-term consequences of TBI and aging-associated neurodegenerative disorders including Alzheimer's disease (AD), chronic traumatic encephalopathy (CTE), and Parkinsonism, with implications for novel therapy targets. Several aggregation-prone proteins such as the amyloid-beta (Aβ) peptides, tau proteins, and α-synuclein protein are involved in secondary pathogenic cascades initiated by a TBI and are also major building blocks of the hallmark pathological lesions in chronic human neurodegenerative diseases with dementia. Impaired metabolism and degradation pathways of aggregation-prone proteins are discussed as potentially critical links between the long-term aftermath of TBI and chronic neurodegeneration. Utility and limitations of previous and current preclinical TBI models designed to study the link between TBI and chronic neurodegeneration, and promising intervention pharmacotherapies and non-pharmacologic strategies to break this link, are also summarized. Complexity of long-term neuropathological consequences of TBI is discussed, with a goal of guiding future preclinical studies and accelerating implementation of promising therapeutics into clinical trials. This article is part of the Special Issue entitled "Novel Treatments for Traumatic Brain Injury".
Collapse
Affiliation(s)
- Milos D Ikonomovic
- Geriatric Research Education and Clinical Center, Pittsburgh VA Healthcare System, Pittsburgh, PA, USA; Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Eric E Abrahamson
- Geriatric Research Education and Clinical Center, Pittsburgh VA Healthcare System, Pittsburgh, PA, USA; Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Shaun W Carlson
- Geriatric Research Education and Clinical Center, Pittsburgh VA Healthcare System, Pittsburgh, PA, USA; Department of Neurosurgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Steven H Graham
- Geriatric Research Education and Clinical Center, Pittsburgh VA Healthcare System, Pittsburgh, PA, USA; Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - C Edward Dixon
- Geriatric Research Education and Clinical Center, Pittsburgh VA Healthcare System, Pittsburgh, PA, USA; Department of Neurosurgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
33
|
Abstract
Mild traumatic brain injury (mTBI) represents a significant public healthcare concern, accounting for the majority of all head injuries. While symptoms are generally transient, some patients go on to experience long-term cognitive impairments and additional mild impacts can result in exacerbated and persisting negative outcomes. To date, studies using a range of experimental models have reported chronic behavioral deficits in the presence of axonal injury and inflammation following repeated mTBI; assessments of oxidative stress and myelin pathology have thus far been limited. However, some models employed induced acute focal damage more suggestive of moderate–severe brain injury and are therefore not relevant to repeated mTBI. Given that the nature of mechanical loading in TBI is implicated in downstream pathophysiological changes, the mechanisms of damage and chronic consequences of single and repeated closed-head mTBI remain to be fully elucidated. This review covers literature on potential mechanisms of damage following repeated mTBI, integrating known mechanisms of pathology underlying moderate–severe TBIs, with recent studies on adult rodent models relevant to direct impact injuries rather than blast-induced damage. Pathology associated with excitotoxicity and cerebral blood flow-metabolism uncoupling, oxidative stress, cell death, blood-brain barrier dysfunction, astrocyte reactivity, microglial activation, diffuse axonal injury, and dysmyelination is discussed, followed by a summary of functional deficits and preclinical assessments of therapeutic strategies. Comprehensive characterization of the pathology underlying delayed and persisting deficits following repeated mTBI is likely to facilitate further development of therapeutic strategies to limit long-term sequelae.
Collapse
Affiliation(s)
- Brooke Fehily
- 1 Experimental and Regenerative Neurosciences, School of Biological sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Melinda Fitzgerald
- 1 Experimental and Regenerative Neurosciences, School of Biological sciences, The University of Western Australia, Perth, Western Australia, Australia.,2 Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia.,3 Perron Institute for Neurological and Translational Science, Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia
| |
Collapse
|
34
|
Nichols JN, Hagan KL, Floyd CL. Evaluation of Touchscreen Chambers To Assess Cognition in Adult Mice: Effect of Training and Mild Traumatic Brain Injury. J Neurotrauma 2018; 34:2481-2494. [PMID: 28558476 DOI: 10.1089/neu.2017.4998] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Cognitive impairments are often experienced after a mild traumatic brain injury (mTBI). In the clinical arena, neuropsychological assessments are used frequently to detect cognitive deficits. Animal models of mTBI, however, rely on an assortment of behavioral tasks to assess cognitive outcome. Computer-based touchscreen systems have been developed for rodents and are hypothesized to offer a translational approach to evaluate cognitive function because of the similarities of tasks performed in rodents to those implemented in humans. While these touchscreen systems have been used in pre-clinical models of neurodegenerative diseases and psychiatric disorders, their use in assessing cognitive impairment after mTBI has not been investigated. We hypothesized that mTBI would result in impaired cognitive performance on touchscreen tasks, particularly those with hippocampal-based learning components, including the paired associate learning (PAL) task and the location discrimination (LD) task. Adult male, C57BL/6 mice received a single impact-acceleration mTBI. We found that training mice before injury to perform to criteria is arduous and that performance is sensitive to many environmental variables. Despite extensive optimization and training, mice failed to perform better than chance in the PAL paradigm. Alternatively, mice demonstrated some capacity to learn in the LD paradigm, but only with the easier stages of the task. The mTBI did not affect performance in the LD paradigm, however. Thus, we concluded that under the conditions presented here, the PAL and LD touchscreen tasks are not robust outcome measures for the evaluation of cognitive performance in C57BL/6 mice after a single impact-acceleration mTBI.
Collapse
Affiliation(s)
- Jessica N Nichols
- 1 Department of Physical Medicine and Rehabilitation, University of Alabama at Birmingham , Birmingham, Alabama
| | - Kenton L Hagan
- 2 Department of Physical Medicine and Rehabilitation, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Candace L Floyd
- 1 Department of Physical Medicine and Rehabilitation, University of Alabama at Birmingham , Birmingham, Alabama
| |
Collapse
|
35
|
Hausser N, Johnson K, Parsley MA, Guptarak J, Spratt H, Sell SL. Detecting Behavioral Deficits in Rats After Traumatic Brain Injury. J Vis Exp 2018. [PMID: 29443022 PMCID: PMC5912256 DOI: 10.3791/56044] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
With the increasing incidence of traumatic brain injury (TBI) in both civilian and military populations, TBI is now considered a chronic disease; however, few studies have investigated the long-term effects of injury in rodent models of TBI. Shown here are behavioral measures that are well-established in TBI research for times early after injury, such as two weeks, until two months. Some of these methods have previously been used at later times after injury, up to one year, but by very few laboratories. The methods demonstrated here are a short neurological assessment to test reflexes, a Beam-Balance to test balance, a Beam-Walk to test balance and motor coordination, and a working memory version of the Morris water maze that can be sensitive to deficits in reference memory. Male rats were handled and pre-trained to neurological, balance, and motor coordination tests prior to receiving parasagittal fluid percussion injury (FPI) or sham injury. Rats can be tested on the short neurological assessment (neuroscore), the beam-balance, and the Beam-Walk multiple times, while testing on the water maze can only be done once. This difference is because rats can remember the task, thus confounding the results if repeated testing is attempted in the same animal. When testing from one to three days after injury, significant differences are detected in all three non-cognitive tasks. However, differences in the Beam-Walk task were not detectable at later time points (after 3 months). Deficits were detected at 3 months in the Beam-Balance and at 6 months in the neuroscore. Deficits in working memory were detected out to 12 months after injury, and a deficit in a reference memory first appeared at 12 months. Thus, standard behavioral tests can be useful measures of persistent behavioral deficits after FPI.
Collapse
Affiliation(s)
- Nicole Hausser
- Department of Anesthesiology, University of Texas Medical Branch
| | - Kathia Johnson
- Department of Anesthesiology, University of Texas Medical Branch
| | | | - Jutatip Guptarak
- Department of Anesthesiology, University of Texas Medical Branch
| | - Heidi Spratt
- Department of Anesthesiology, University of Texas Medical Branch
| | - Stacy L Sell
- Department of Anesthesiology, University of Texas Medical Branch;
| |
Collapse
|
36
|
Cheng SX, Xu ZW, Yi TL, Sun HT, Yang C, Yu ZQ, Yang XS, Jin XH, Tu Y, Zhang S. iTRAQ-Based Quantitative Proteomics Reveals the New Evidence Base for Traumatic Brain Injury Treated with Targeted Temperature Management. Neurotherapeutics 2018; 15:216-232. [PMID: 29247448 PMCID: PMC5794703 DOI: 10.1007/s13311-017-0591-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
This study aimed to investigate the effects of targeted temperature management (TTM) modulation on traumatic brain injury (TBI) and the involved mechanisms using quantitative proteomics technology. SH-SY5Y and HT-22 cells were subjected to moderate stretch injury using the cell injury controller (CIC), followed by incubation at TTM (mild hypothermia, 32°C), or normothermia (37°C). The real-time morphological changes, cell cycle phase distribution, death, and cell viability were evaluated. Moderate TBI was produced by the controlled cortical impactor (CCI), and the effects of TTM on the neurological damage, neurodegeneration, cerebrovascular histopathology, and behavioral outcome were determined in vivo. Results showed that TTM treatment prevented TBI-induced neuronal necrosis in the brain, achieved a substantial reduction in neuronal death both in vitro and in vivo, reduced cortical lesion volume and neuronal loss, attenuated cerebrovascular histopathological damage, brain edema, and improved behavioral outcome. Using an iTRAQ proteomics approach, proteins that were significantly associated with TTM in experimental TBI were identified. Importantly, changes in four candidate molecules (plasminogen [PLG], antithrombin III [AT III], fibrinogen gamma chain [FGG], transthyretin [TTR]) were verified using TBI rat brain tissues and TBI human cerebrospinal fluid (CSF) samples. This study is one of the first to investigate the neuroprotective effects of TTM on the proteome of human and experimental models of TBI, providing an overall landscape of the TBI brain proteome and a scientific foundation for further assessment of candidate molecules associated with TTM for the promotion of reparative strategies post-TBI.
Collapse
Affiliation(s)
- Shi-Xiang Cheng
- Tianjin Key Laboratory of Neurotrauma Repair, Institute of Traumatic Brain Injury and Neuroscience, Center for Neurology and Neurosurgery of Affiliated Hospital, Logistics University of Chinese People's Armed Police Force (PAP), Tianjin, China.
| | - Zhong-Wei Xu
- Central Laboratory of Logistics University of Chinese People's Armed Police Force (PAP), Tianjin, China
| | - Tai-Long Yi
- Tianjin Key Laboratory of Neurotrauma Repair, Institute of Traumatic Brain Injury and Neuroscience, Center for Neurology and Neurosurgery of Affiliated Hospital, Logistics University of Chinese People's Armed Police Force (PAP), Tianjin, China
| | - Hong-Tao Sun
- Tianjin Key Laboratory of Neurotrauma Repair, Institute of Traumatic Brain Injury and Neuroscience, Center for Neurology and Neurosurgery of Affiliated Hospital, Logistics University of Chinese People's Armed Police Force (PAP), Tianjin, China
| | - Cheng Yang
- Tianjin Key Laboratory of Neurotrauma Repair, Institute of Traumatic Brain Injury and Neuroscience, Center for Neurology and Neurosurgery of Affiliated Hospital, Logistics University of Chinese People's Armed Police Force (PAP), Tianjin, China
| | - Ze-Qi Yu
- Tianjin Key Laboratory of Neurotrauma Repair, Institute of Traumatic Brain Injury and Neuroscience, Center for Neurology and Neurosurgery of Affiliated Hospital, Logistics University of Chinese People's Armed Police Force (PAP), Tianjin, China
| | - Xiao-Sa Yang
- Tianjin Key Laboratory of Neurotrauma Repair, Institute of Traumatic Brain Injury and Neuroscience, Center for Neurology and Neurosurgery of Affiliated Hospital, Logistics University of Chinese People's Armed Police Force (PAP), Tianjin, China
| | - Xiao-Han Jin
- Central Laboratory of Logistics University of Chinese People's Armed Police Force (PAP), Tianjin, China
| | - Yue Tu
- Tianjin Key Laboratory of Neurotrauma Repair, Institute of Traumatic Brain Injury and Neuroscience, Center for Neurology and Neurosurgery of Affiliated Hospital, Logistics University of Chinese People's Armed Police Force (PAP), Tianjin, China.
| | - Sai Zhang
- Tianjin Key Laboratory of Neurotrauma Repair, Institute of Traumatic Brain Injury and Neuroscience, Center for Neurology and Neurosurgery of Affiliated Hospital, Logistics University of Chinese People's Armed Police Force (PAP), Tianjin, China.
| |
Collapse
|
37
|
Gold EM, Vasilevko V, Hasselmann J, Tiefenthaler C, Hoa D, Ranawaka K, Cribbs DH, Cummings BJ. Repeated Mild Closed Head Injuries Induce Long-Term White Matter Pathology and Neuronal Loss That Are Correlated With Behavioral Deficits. ASN Neuro 2018; 10:1759091418781921. [PMID: 29932344 PMCID: PMC6050992 DOI: 10.1177/1759091418781921] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 04/29/2018] [Accepted: 05/12/2018] [Indexed: 11/16/2022] Open
Abstract
An estimated 5.3 million Americans are living with a disability from a traumatic brain injury (TBI). There is emerging evidence of the detrimental effects from repeated mild TBIs (rmTBIs). rmTBI manifests its own unique set of behavioral and neuropathological changes. A subset of individuals exposed to rmTBI develop permanent behavioral and pathological consequences, defined postmortem as chronic traumatic encephalopathy. We have combined components of two classic rodent models of TBI, the controlled cortical impact model and the weight drop model, to develop a repeated mild closed head injury (rmCHI) that produces long-term deficits in several behaviors that correlate with neuropathological changes. Mice receiving rmCHI performed differently from 1-hit or sham controls on the elevated plus maze; these deficits persist up to 6 months postinjury (MPI). rmCHI mice performed worse than 1-hit and control sham mice at 2 MPI and 6 MPI on the Morris water maze. Mice receiving rmCHI exhibited significant atrophy of the corpus callosum at both 2 MPI and 6 MPI, as assessed by stereological volume analysis. Stereological analysis also revealed significant loss of cortical neurons in comparison with 1-hit and controls. Moreover, both of these pathological changes correlated with behavioral impairments. In human tau transgenic mice, rmCHI induced increases in hyperphosphorylated paired helical filament 1 tau in the hippocampus. This suggests that strategies to restore myelination or reduce neuronal loss may ameliorate the behavioral deficits observed following rmCHI and that rmCHI may model chronic traumatic encephalopathy in human tau mice.
Collapse
Affiliation(s)
- Eric M. Gold
- Department of Anatomy and Neurobiology,
University
of California-Irvine, CA, USA
- Sue and Bill Gross Stem Cell Center,
University
of California-Irvine, CA, USA
| | - Vitaly Vasilevko
- UCI Institute for Memory Impairments and Neurological Disorders,
University
of California-Irvine, CA, USA
| | - Jonathan Hasselmann
- Department of Anatomy and Neurobiology,
University
of California-Irvine, CA, USA
- UCI Institute for Memory Impairments and Neurological Disorders,
University
of California-Irvine, CA, USA
| | - Casey Tiefenthaler
- Department of Anatomy and Neurobiology,
University
of California-Irvine, CA, USA
- Sue and Bill Gross Stem Cell Center,
University
of California-Irvine, CA, USA
| | - Danny Hoa
- Department of Anatomy and Neurobiology,
University
of California-Irvine, CA, USA
- Sue and Bill Gross Stem Cell Center,
University
of California-Irvine, CA, USA
| | - Kasuni Ranawaka
- Department of Anatomy and Neurobiology,
University
of California-Irvine, CA, USA
- Sue and Bill Gross Stem Cell Center,
University
of California-Irvine, CA, USA
| | - David H. Cribbs
- UCI Institute for Memory Impairments and Neurological Disorders,
University
of California-Irvine, CA, USA
| | - Brian J. Cummings
- Department of Anatomy and Neurobiology,
University
of California-Irvine, CA, USA
- Sue and Bill Gross Stem Cell Center,
University
of California-Irvine, CA, USA
- UCI Institute for Memory Impairments and Neurological Disorders,
University
of California-Irvine, CA, USA
- Department of Physical Medicine and Rehabilitation,
University
of California-Irvine, CA, USA
| |
Collapse
|
38
|
Pischiutta F, Micotti E, Hay JR, Marongiu I, Sammali E, Tolomeo D, Vegliante G, Stocchetti N, Forloni G, De Simoni MG, Stewart W, Zanier ER. Single severe traumatic brain injury produces progressive pathology with ongoing contralateral white matter damage one year after injury. Exp Neurol 2017; 300:167-178. [PMID: 29126888 DOI: 10.1016/j.expneurol.2017.11.003] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 10/27/2017] [Accepted: 11/06/2017] [Indexed: 01/29/2023]
Abstract
There is increasing recognition that traumatic brain injury (TBI) may initiate long-term neurodegenerative processes, particularly chronic traumatic encephalopathy. However, insight into the mechanisms transforming an initial biomechanical injury into a neurodegenerative process remain elusive, partly as a consequence of the paucity of informative pre-clinical models. This study shows the functional, whole brain imaging and neuropathological consequences at up to one year survival from single severe TBI by controlled cortical impact in mice. TBI mice displayed persistent sensorimotor and cognitive deficits. Longitudinal T2 weighted magnetic resonance imaging (MRI) showed progressive ipsilateral (il) cortical, hippocampal and striatal volume loss, with diffusion tensor imaging demonstrating decreased fractional anisotropy (FA) at up to one year in the il-corpus callosum (CC: -30%) and external capsule (EC: -21%). Parallel neuropathological studies indicated reduction in neuronal density, with evidence of microgliosis and astrogliosis in the il-cortex, with further evidence of microgliosis and astrogliosis in the il-thalamus. One year after TBI there was also a decrease in FA in the contralateral (cl) CC (-17%) and EC (-13%), corresponding to histopathological evidence of white matter loss (cl-CC: -68%; cl-EC: -30%) associated with ongoing microgliosis and astrogliosis. These findings indicate that a single severe TBI induces bilateral, long-term and progressive neuropathology at up to one year after injury. These observations support this model as a suitable platform for exploring the mechanistic link between acute brain injury and late and persistent neurodegeneration.
Collapse
Affiliation(s)
- Francesca Pischiutta
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Edoardo Micotti
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Jennifer R Hay
- Institute of Neuroscience and Psychology, University of Glasgow, UK; Department of Laboratory Medicine, Queen Elizabeth University Hospital, Glasgow, UK
| | - Ines Marongiu
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Eliana Sammali
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy; Department of Cerebrovascular Diseases, Fondazione IRCCS - Istituto Neurologico Carlo Besta, Milan, Italy
| | - Daniele Tolomeo
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Gloria Vegliante
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Nino Stocchetti
- Department of Physiopathology and Transplantation, Milan University, Milan, Italy; ICU Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Gianluigi Forloni
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Maria-Grazia De Simoni
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - William Stewart
- Institute of Neuroscience and Psychology, University of Glasgow, UK; Department of Laboratory Medicine, Queen Elizabeth University Hospital, Glasgow, UK
| | - Elisa R Zanier
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy.
| |
Collapse
|
39
|
Dean DD, Frank JA, Turtzo LC. Controlled Cortical Impact in the Rat. ACTA ACUST UNITED AC 2017; 81:9.62.1-9.62.12. [PMID: 29058772 DOI: 10.1002/cpns.37] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Traumatic brain injury (TBI) is a major cause of death and disability world-wide. Following initial injury, TBI patients can face long-term disability in the form of cognitive, physical, and psychological deficits, depending on the severity and location of injury. This results in an economic burden in the United States estimated to be $60 billion due to health-care costs and loss of productivity. TBI is a significant area of active research interest for both military and civilian medicine. Numerous pre-clinical animal models of TBI are used to characterize the anatomical and physiological pathways involved and to evaluate therapeutic interventions. Due to its flexibility and scalability, controlled cortical impact (CCI) is one of the most commonly used preclinical TBI models. This unit provides a basic CCI protocol performed in the rat. © 2017 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Dana D Dean
- Neuroscience Institute, Baylor Scott & White Health, Temple, Texas
| | - Joseph A Frank
- Frank Laboratory, National Institutes of Health, Clinical Center and National Institutes of Biomedical Imaging and Bioengineering, Bethesda, Maryland
| | - L Christine Turtzo
- National Institute of Neurological Disease and Stroke, Bethesda, Maryland
| |
Collapse
|
40
|
Beretta S, Cunningham KM, Haus DL, Gold EM, Perez H, López-Velázquez L, Cummings BJ. Effects of Human ES-Derived Neural Stem Cell Transplantation and Kindling in a Rat Model of Traumatic Brain Injury. Cell Transplant 2017; 26:1247-1261. [PMID: 28933218 PMCID: PMC5657732 DOI: 10.1177/0963689717714107] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 10/21/2016] [Accepted: 10/26/2016] [Indexed: 11/15/2022] Open
Abstract
Traumatic brain injury (TBI) is one of the leading causes of death and disability in the population worldwide, with a broad spectrum of symptoms and disabilities. Posttraumatic hyperexcitability is one of the most common neurological disorders that affect people after a head injury. A reliable animal model of posttraumatic hyperexcitability induced by TBI which allows one to test effective treatment strategies is yet to be developed. To address these issues, in the present study, we tested human embryonic stem cell-derived neural stem cell (NSC) transplantation in an animal model of posttraumatic hyperexcitability in which the brain injury was produced in one hemisphere of immunodeficient athymic nude rats by controlled cortical impact, and spontaneous seizures were produced by repeated electrical stimulation (kindling) in the contralateral hemisphere. At 14 wk posttransplantation, we report human NSC (hNSC) survival and differentiation into all 3 neural lineages in both sham and injured animals. We observed twice as many surviving hNSCs in the injured versus sham brain, and worse survival on the kindled side in both groups, indicating that kindling/seizures are detrimental to survival or proliferation of hNSCs. We also replicated our previous finding that hNSCs can ameliorate deficits on the novel place recognition task,33 but such improvements are abolished following kindling. We found no significant differences pre- or post-kindling on the elevated plus maze. No significant correlations were observed between hNSC survival and cognitive performance on either task. Together these findings suggest that Shef6-derived hNSCs may be beneficial as a therapy for TBI, but not in animals or patients with posttraumatic hyperexcitability.
Collapse
Affiliation(s)
- Stefania Beretta
- Dipartimento di Biotecnologie e Bioscienze, University of Milano-Bicocca, Milano, Italy
| | - Kelly M. Cunningham
- UCI Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, CA, USA
| | - Daniel L. Haus
- Sue & Bill Gross Cell Center, University of California, Irvine, CA, USA
- Department of Anatomy & Neurobiology, University of California, Irvine, CA, USA
| | - Eric M. Gold
- Sue & Bill Gross Cell Center, University of California, Irvine, CA, USA
- Department of Anatomy & Neurobiology, University of California, Irvine, CA, USA
| | - Harvey Perez
- UCI Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, CA, USA
| | - Luci López-Velázquez
- UCI Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, CA, USA
| | - Brian J. Cummings
- UCI Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, CA, USA
- Sue & Bill Gross Cell Center, University of California, Irvine, CA, USA
- Department of Anatomy & Neurobiology, University of California, Irvine, CA, USA
- Physical and Medical Rehabilitation, University of California, Irvine, CA, USA
| |
Collapse
|
41
|
Pöttker B, Stöber F, Hummel R, Angenstein F, Radyushkin K, Goldschmidt J, Schäfer MKE. Traumatic brain injury causes long-term behavioral changes related to region-specific increases of cerebral blood flow. Brain Struct Funct 2017; 222:4005-4021. [DOI: 10.1007/s00429-017-1452-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 05/27/2017] [Indexed: 12/19/2022]
|
42
|
Spurlock MS, Ahmed AI, Rivera KN, Yokobori S, Lee SW, Sam PN, Shear DA, Hefferan MP, Hazel TG, Johe KK, Gajavelli S, Tortella FC, Bullock RM. Amelioration of Penetrating Ballistic-Like Brain Injury Induced Cognitive Deficits after Neuronal Differentiation of Transplanted Human Neural Stem Cells. J Neurotrauma 2017; 34:1981-1995. [PMID: 28249550 PMCID: PMC6913783 DOI: 10.1089/neu.2016.4602] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Penetrating traumatic brain injury (PTBI) is one of the major cause of death and disability worldwide. Previous studies with penetrating ballistic-like brain injury (PBBI), a PTBI rat model revealed widespread perilesional neurodegeneration, similar to that seen in humans following gunshot wound to the head, which is unmitigated by any available therapies to date. Therefore, we evaluated human neural stem cell (hNSC) engraftment to putatively exploit the potential of cell therapy that has been seen in other central nervous system injury models. Toward this objective, green fluorescent protein (GFP) labeled hNSC (400,000 per animal) were transplanted in immunosuppressed Sprague-Dawley (SD), Fisher, and athymic (ATN) PBBI rats 1 week after injury. Tacrolimus (3 mg/kg 2 days prior to transplantation, then 1 mg/kg/day), methylprednisolone (10 mg/kg on the day of transplant, 1 mg/kg/week thereafter), and mycophenolate mofetil (30 mg/kg/day) for 7 days following transplantation were used to confer immunosuppression. Engraftment in SD and ATN was comparable at 8 weeks post-transplantation. Evaluation of hNSC differentiation and distribution revealed increased neuronal differentiation of transplanted cells with time. At 16 weeks post-transplantation, neither cell proliferation nor glial lineage markers were detected. Transplanted cell morphology was similar to that of neighboring host neurons, and there was relatively little migration of cells from the peritransplant site. By 16 weeks, GFP-positive processes extended both rostrocaudally and bilaterally into parenchyma, spreading along host white matter tracts, traversing the internal capsule, and extending ∼13 mm caudally from transplantation site reaching into the brainstem. In a Morris water maze test at 8 weeks post-transplantation, animals with transplants had shorter latency to platform than vehicle-treated animals. However, weak injury-induced cognitive deficits in the control group at the delayed time point confounded benefits of durable engraftment and neuronal differentiation. Therefore, these results justify further studies to progress towards clinical translation of hNSC therapy for PTBI.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Deborah A. Shear
- Branch of Brain Trauma Neuroprotection and Neurorestoration, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland
| | | | | | | | | | - Frank C. Tortella
- Branch of Brain Trauma Neuroprotection and Neurorestoration, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland
| | | |
Collapse
|
43
|
Osier ND, Pham L, Pugh BJ, Puccio A, Ren D, Conley YP, Alexander S, Dixon CE. Brain injury results in lower levels of melatonin receptors subtypes MT1 and MT2. Neurosci Lett 2017; 650:18-24. [PMID: 28377323 DOI: 10.1016/j.neulet.2017.03.053] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 03/29/2017] [Accepted: 03/31/2017] [Indexed: 02/09/2023]
Abstract
BACKGROUND Traumatic brain injury (TBI) is a devastating and costly acquired condition that affects individuals of all ages, races, and geographies via a number of mechanisms. The effects of TBI on melatonin receptors remain unknown. PURPOSE The purpose of this study is to explore whether endogenous changes in two melatonin receptor subtypes (MT1 and MT2) occur after experimental TBI. SAMPLE A total of 25 adult male Sprague Dawley rats were used with 6 or 7 rats per group. METHODS Rats were randomly assigned to receive either TBI modeled using controlled cortical impact or sham surgery and to be sacrificed at either 6- or 24-h post-operatively. Brains were harvested, dissected, and flash frozen until whole cell lysates were prepared, and the supernatant fluid aliquoted and used for western blotting. Primary antibodies were used to probe for melatonin receptors (MT1 and MT2), and beta actin, used for a loading control. ImageJ and Image Lab software were used to quantify the data which was analyzed using t-tests to compare means. RESULTS Melatonin receptor levels were reduced in a brain region- and time point- dependent manner. Both MT1 and MT2 were reduced in the frontal cortex at 24h and in the hippocampus at both 6h and 24h. DISCUSSION MT1 and MT2 are less abundant after injury, which may alter response to MEL therapy. Studies characterizing MT1 and MT2 after TBI are needed, including exploration of the time course and regional patterns, replication in diverse samples, and use of additional variables, especially sleep-related outcomes. CONCLUSION TBI in rats resulted in lower levels of MT1 and MT2; replication of these findings is necessary as is evaluation of the consequences of lower receptor levels.
Collapse
Affiliation(s)
- Nicole D Osier
- University of Pittsburgh School of Nursing, Victoria Building, 3500 Victoria Street, Pittsburgh, PA, 15213, USA; Safar Center for Resuscitation Research, Children's Hospital of Pittsburgh of UPMC, John G. Rangos Research, Center - 6th floor, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA.
| | - Lan Pham
- University of Pittsburgh School of Nursing, Victoria Building, 3500 Victoria Street, Pittsburgh, PA, 15213, USA.
| | - Bunny J Pugh
- Safar Center for Resuscitation Research, Children's Hospital of Pittsburgh of UPMC, John G. Rangos Research, Center - 6th floor, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA.
| | - Ava Puccio
- University of Pittsburgh School of Nursing, Victoria Building, 3500 Victoria Street, Pittsburgh, PA, 15213, USA; University of Pittsburgh Department of Neurological Surgery, Brain Trauma Research Center, UPMC Presbyterian, Suite B-400, 200 Lothrop Street, Pittsburgh, PA, 15213, USA.
| | - Dianxu Ren
- University of Pittsburgh School of Nursing, Victoria Building, 3500 Victoria Street, Pittsburgh, PA, 15213, USA.
| | - Yvette P Conley
- University of Pittsburgh School of Nursing, Victoria Building, 3500 Victoria Street, Pittsburgh, PA, 15213, USA; University of Pittsburgh Department of Human Genetics, Crabtree Hall, 130 De Soto Street, Pittsburgh, PA, 15261, USA.
| | - Sheila Alexander
- University of Pittsburgh School of Nursing, Victoria Building, 3500 Victoria Street, Pittsburgh, PA, 15213, USA; University of Pittsburgh School of Medicine, M240 Scaife Hall, 3550 Terrace Street, Pittsburgh, PA, 15261, USA.
| | - C Edward Dixon
- Safar Center for Resuscitation Research, Children's Hospital of Pittsburgh of UPMC, John G. Rangos Research, Center - 6th floor, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA; University of Pittsburgh Department of Neurological Surgery, Brain Trauma Research Center, UPMC Presbyterian, Suite B-400, 200 Lothrop Street, Pittsburgh, PA, 15213, USA; VA Pittsburgh Healthcare System, 4100 Allequippa St, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
44
|
Pinkernell S, Becker K, Lindauer U. Severity assessment and scoring for neurosurgical models in rodents. Lab Anim 2016; 50:442-452. [DOI: 10.1177/0023677216675010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The most important acute neurological diseases seen at neurosurgery departments are traumatic brain injuries (TBI) and subarachnoid hemorrhages (SAH). In both diseases the pathophysiological sequela are complex and have not been fully understood up to now, and rodent models using rats and mice are most suitable for the investigation of the pathophysiological details. In both models, surgery is performed under anesthesia, followed by assessment of their functional outcome and behavioral testing before brain tissue analysis after euthanasia. Postoperative analgesia is mandatory, and supplementary care is highly recommended for refinement purposes. Pain and stress assessment is mainly based on clinical and behavioral signs, and further research is needed to improve the evaluation of severity in these models.
Collapse
Affiliation(s)
- Sarah Pinkernell
- Translational Neurosurgery, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Katrin Becker
- Translational Neurosurgery, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Ute Lindauer
- Translational Neurosurgery, Medical Faculty, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
45
|
Sell SL, Johnson K, DeWitt DS, Prough DS. Persistent Behavioral Deficits in Rats after Parasagittal Fluid Percussion Injury. J Neurotrauma 2016; 34:1086-1096. [PMID: 27650266 DOI: 10.1089/neu.2016.4616] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Although traumatic brain injury (TBI) is now considered a chronic disease, few studies have investigated the long-term behavioral deficits elicited by a well-established rodent model of injury. Here we evaluate behavioral measures, commonly used in TBI research, to determine which tests are useful for studying long-term effects of brain injury in rats. Male Sprague-Dawley rats were handled and pre-trained to neurological, balance, and motor coordination tests prior to receiving parasagittal fluid-percussion injury (FPI), sham injury, or maintenance as naïve cohorts. Rats underwent neuroscore, beam-balance, and beam-walk tests for 3 days after injury. Subsequently, in separate groups at 3, 6, or 12 months, they were re-tested on the same tasks followed by a working memory version of the Morris water maze. On post-injury days (PIDs) 1-3, significant effects of injury on neuroscore, beam-balance, and beam-walk were observed. Differences in the beam-walk task were not detectable at any of the later time-points. However, deficits persisted in beam-balance out to 3 months and neuroscore out to 6 months. Working memory deficits persisted out to 12 months, at which time a reference memory deficit was also evident. These data suggest that balance and motor coordination recovered more quickly than neurological deficits. Furthermore, while deficits in working memory remained stable over the 12-month period, the late onset of the reference memory deficit points to the progressive nature of the injury, or an age/TBI interaction. In conclusion, standard behavioral tests are useful measures of persistent behavioral deficits after parasagittal FPI and provide evidence that TBI is a chronic condition that can change over time and worsen with age.
Collapse
Affiliation(s)
- Stacy L Sell
- Department of Anesthesiology, University of Texas Medical Branch , Galveston, Texas
| | - Kathia Johnson
- Department of Anesthesiology, University of Texas Medical Branch , Galveston, Texas
| | - Douglas S DeWitt
- Department of Anesthesiology, University of Texas Medical Branch , Galveston, Texas
| | - Donald S Prough
- Department of Anesthesiology, University of Texas Medical Branch , Galveston, Texas
| |
Collapse
|
46
|
Yang Z, Lin F, Weissman AS, Jaalouk E, Xue QS, Wang KKW. A Repetitive Concussive Head Injury Model in Mice. J Vis Exp 2016. [PMID: 27768069 DOI: 10.3791/54530] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Despite the concussion/ mild traumatic brain injury (mTBI) being the most frequent occurrence of traumatic brain injury, there is still a lack of knowledge on the injury and its effects. To develop a better understanding of concussions, animals are often used because they provide a controlled, rigorous, and efficient model. Studies have adapted traditional animal models to perform mTBI to stimulate mild injury severity by changing the injury parameters. These models have been used because they can produce morphologically similar brain injuries to the clinical condition and provide a spectrum of injury severities. However, they are limited in their ability to present the identical features of injuries in patients. Using a traditional impact system, a repetitive concussive injury (rCHI) model can induce mild to moderate human-like concussion. The injury degree can be determined by measuring the period of loss of consciousness (LOC) with a sign of a transient termination of breathing. The rCHI model is beneficial to use for its accuracy and simplicity in determining mTBI effects and potential treatments.
Collapse
Affiliation(s)
- Zhihui Yang
- Program for Neurotrauma, Neuroproteomics & Biomarkers Research, University of Florida; Department of Psychiatry, University of Florida;
| | - Fan Lin
- Program for Neurotrauma, Neuroproteomics & Biomarkers Research, University of Florida; Department of Psychiatry, University of Florida
| | - Amanda S Weissman
- Program for Neurotrauma, Neuroproteomics & Biomarkers Research, University of Florida; Department of Psychiatry, University of Florida
| | - Emily Jaalouk
- Program for Neurotrauma, Neuroproteomics & Biomarkers Research, University of Florida; Department of Psychiatry, University of Florida
| | | | - Kevin K W Wang
- Program for Neurotrauma, Neuroproteomics & Biomarkers Research, University of Florida; Department of Psychiatry, University of Florida
| |
Collapse
|
47
|
Ryan NP, Catroppa C, Godfrey C, Noble-Haeusslein LJ, Shultz SR, O'Brien TJ, Anderson V, Semple BD. Social dysfunction after pediatric traumatic brain injury: A translational perspective. Neurosci Biobehav Rev 2016; 64:196-214. [PMID: 26949224 PMCID: PMC5627971 DOI: 10.1016/j.neubiorev.2016.02.020] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Revised: 02/24/2016] [Accepted: 02/24/2016] [Indexed: 12/21/2022]
Abstract
Social dysfunction is common after traumatic brain injury (TBI), contributing to reduced quality of life for survivors. Factors which influence the development or persistence of social deficits after injury remain poorly understood, particularly in the context of ongoing brain maturation during childhood and adolescence. Aberrant social interactions have recently been modeled in adult and juvenile rodents after experimental TBI, providing an opportunity to gain new insights into the underlying neurobiology of these behaviors. Here, we review our current understanding of social dysfunction in both humans and rodent models of TBI, with a focus on brain injuries acquired during early development. Modulators of social outcomes are discussed, including injury-related and environmental risk and resilience factors. Disruption of social brain network connectivity and aberrant neuroendocrine function are identified as potential mechanisms of social impairments after pediatric TBI. Throughout, we highlight the overlap and disparities between outcome measures and findings from clinical and experimental approaches, and explore the translational potential of future research to prevent or ameliorate social dysfunction after childhood TBI.
Collapse
Affiliation(s)
- Nicholas P Ryan
- Australian Centre for Child Neuropsychological Studies, Murdoch Childrens Research Institute, Parkville, VIC, Australia; Melbourne School of Psychological Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, Australia.
| | - Cathy Catroppa
- Australian Centre for Child Neuropsychological Studies, Murdoch Childrens Research Institute, Parkville, VIC, Australia; Melbourne School of Psychological Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, Australia; Department of Psychology, Royal Children's Hospital, Parkville, VIC, Australia.
| | - Celia Godfrey
- Australian Centre for Child Neuropsychological Studies, Murdoch Childrens Research Institute, Parkville, VIC, Australia.
| | - Linda J Noble-Haeusslein
- Departments of Neurological Surgery and Physical Therapy and Rehabilitation Science, University of California, San Francisco, San Francisco, CA, USA.
| | - Sandy R Shultz
- Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, VIC, Australia.
| | - Terence J O'Brien
- Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, VIC, Australia.
| | - Vicki Anderson
- Australian Centre for Child Neuropsychological Studies, Murdoch Childrens Research Institute, Parkville, VIC, Australia; Melbourne School of Psychological Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, Australia; Department of Psychology, Royal Children's Hospital, Parkville, VIC, Australia.
| | - Bridgette D Semple
- Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
48
|
Lamprecht MR, Elkin BS, Kesavabhotla K, Crary JF, Hammers JL, Huh JW, Raghupathi R, Morrison B. Strong Correlation of Genome-Wide Expression after Traumatic Brain Injury In Vitro and In Vivo Implicates a Role for SORLA. J Neurotrauma 2016; 34:97-108. [PMID: 26919808 DOI: 10.1089/neu.2015.4306] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The utility of in vitro models of traumatic brain injury (TBI) depends on their ability to recapitulate the in vivo TBI cascade. In this study, we used a genome-wide approach to compare changes in gene expression at several time points post-injury in both an in vitro model and an in vivo model of TBI. We found a total of 2073 differentially expressed genes in our in vitro model and 877 differentially expressed genes in our in vivo model when compared to noninjured controls. We found a strong correlation in gene expression changes between the two models (r = 0.69), providing confidence that the in vitro model represented at least part of the in vivo injury cascade. From these data, we searched for genes with significant changes in expression over time (analysis of covariance) and identified sorting protein-related receptor with A-type repeats (SORLA). SORLA directs amyloid precursor protein to the recycling pathway by direct binding and away from amyloid-beta producing enzymes. Mutations of SORLA have been linked to Alzheimer's disease (AD). We confirmed downregulation of SORLA expression in organotypic hippocampal slice cultures by immunohistochemistry and Western blotting and present preliminary data from human tissue that is consistent with these experimental results. Together, these data suggest that the in vitro model of TBI used in this study strongly recapitulates the in vivo TBI pathobiology and is well suited for future mechanistic or therapeutic studies. The data also suggest the possible involvement of SORLA in the post-traumatic cascade linking TBI to AD.
Collapse
Affiliation(s)
- Michael R Lamprecht
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - Benjamin S Elkin
- 1 Department of Biomedical Engineering, Columbia University , New York, New York.,2 MEA Forensic Engineers & Scientists , Mississauga, Ontario, Canada
| | - Kartik Kesavabhotla
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - John F Crary
- 3 Department of Pathology, Fishberg Department of Neuroscience, Friedman Brain Institute , and the Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jennifer L Hammers
- 4 Office of Chief Medical Examiner , City of New York, New York, New York
| | - Jimmy W Huh
- 5 Department of Anesthesia and Critical Care, Children's Hospital of Philadelphia , Philadelphia, Pennsylvania
| | - Ramesh Raghupathi
- 6 Department of Neurobiology and Anatomy, Drexel University College of Medicine , Philadelphia, Pennsylvania
| | - Barclay Morrison
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| |
Collapse
|
49
|
Haus DL, López-Velázquez L, Gold EM, Cunningham KM, Perez H, Anderson AJ, Cummings BJ. Transplantation of human neural stem cells restores cognition in an immunodeficient rodent model of traumatic brain injury. Exp Neurol 2016; 281:1-16. [PMID: 27079998 DOI: 10.1016/j.expneurol.2016.04.008] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 03/15/2016] [Accepted: 04/07/2016] [Indexed: 12/11/2022]
Abstract
Traumatic brain injury (TBI) in humans can result in permanent tissue damage and has been linked to cognitive impairment that lasts years beyond the initial insult. Clinically effective treatment strategies have yet to be developed. Transplantation of human neural stem cells (hNSCs) has the potential to restore cognition lost due to injury, however, the vast majority of rodent TBI/hNSC studies to date have evaluated cognition only at early time points, typically <1month post-injury and cell transplantation. Additionally, human cell engraftment and long-term survival in rodent models of TBI has been difficult to achieve due to host immunorejection of the transplanted human cells, which confounds conclusions pertaining to transplant-mediated behavioral improvement. To overcome these shortfalls, we have developed a novel TBI xenotransplantation model that utilizes immunodeficient athymic nude (ATN) rats as the host recipient for the post-TBI transplantation of human embryonic stem cell (hESC) derived NSCs and have evaluated cognition in these animals at long-term (≥2months) time points post-injury. We report that immunodeficient ATN rats demonstrate hippocampal-dependent spatial memory deficits (Novel Place, Morris Water Maze), but not non-spatial (Novel Object) or emotional/anxiety-related (Elevated Plus Maze, Conditioned Taste Aversion) deficits, at 2-3months post-TBI, confirming that ATN rats recapitulate some of the cognitive deficits found in immunosufficient animal strains. Approximately 9-25% of transplanted hNSCs survived for at least 5months post-transplantation and differentiated into mature neurons (NeuN, 18-38%), astrocytes (GFAP, 13-16%), and oligodendrocytes (Olig2, 11-13%). Furthermore, while this model of TBI (cortical impact) targets primarily cortex and the underlying hippocampus and generates a large lesion cavity, hNSC transplantation facilitated cognitive recovery without affecting either lesion volume or total spared cortical or hippocampal tissue volume. Instead, we have found an overall increase in host hippocampal neuron survival in hNSC transplanted animals and demonstrate that a correlation exists between hippocampal neuron survival and cognitive performance. Together, these findings support the use of immunodeficient rodents in models of TBI that involve the transplantation of human cells, and suggest that hNSC transplantation may be a viable, long-term therapy to restore cognition after brain injury.
Collapse
Affiliation(s)
- Daniel L Haus
- Sue & Bill Gross Stem Cell Center, University of California, Irvine,CA 92697-1750, USA; Anatomy & Neurobiology, University of California, Irvine,CA 92697-1750, USA
| | - Luci López-Velázquez
- UCI Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine,CA 92697-1750, USA
| | - Eric M Gold
- Sue & Bill Gross Stem Cell Center, University of California, Irvine,CA 92697-1750, USA; Anatomy & Neurobiology, University of California, Irvine,CA 92697-1750, USA
| | - Kelly M Cunningham
- UCI Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine,CA 92697-1750, USA
| | - Harvey Perez
- UCI Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine,CA 92697-1750, USA
| | - Aileen J Anderson
- Sue & Bill Gross Stem Cell Center, University of California, Irvine,CA 92697-1750, USA; Anatomy & Neurobiology, University of California, Irvine,CA 92697-1750, USA; Physical and Medical Rehabilitation, University of California, Irvine,CA 92697-1750, USA; UCI Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine,CA 92697-1750, USA
| | - Brian J Cummings
- Sue & Bill Gross Stem Cell Center, University of California, Irvine,CA 92697-1750, USA; Anatomy & Neurobiology, University of California, Irvine,CA 92697-1750, USA; Physical and Medical Rehabilitation, University of California, Irvine,CA 92697-1750, USA; UCI Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine,CA 92697-1750, USA.
| |
Collapse
|
50
|
Lee SA, Holly KS, Voziyanov V, Villalba SL, Tong R, Grigsby HE, Glasscock E, Szele FG, Vlachos I, Murray TA. Gradient Index Microlens Implanted in Prefrontal Cortex of Mouse Does Not Affect Behavioral Test Performance over Time. PLoS One 2016; 11:e0146533. [PMID: 26799938 PMCID: PMC4723314 DOI: 10.1371/journal.pone.0146533] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 12/19/2015] [Indexed: 12/11/2022] Open
Abstract
Implanted gradient index lenses have extended the reach of standard multiphoton microscopy from the upper layers of the mouse cortex to the lower cortical layers and even subcortical regions. These lenses have the clarity to visualize dynamic activities, such as calcium transients, with subcellular and millisecond resolution and the stability to facilitate repeated imaging over weeks and months. In addition, behavioral tests can be used to correlate performance with observed changes in network function and structure that occur over time. Yet, this raises the questions, does an implanted microlens have an effect on behavioral tests, and if so, what is the extent of the effect? To answer these questions, we compared the performance of three groups of mice in three common behavioral tests. A gradient index lens was implanted in the prefrontal cortex of experimental mice. We compared their performance with mice that had either a cranial window or a sham surgery. Three presurgical and five postsurgical sets of behavioral tests were performed over seven weeks. Behavioral tests included rotarod, foot fault, and Morris water maze. No significant differences were found between the three groups, suggesting that microlens implantation did not affect performance. The results for the current study clear the way for combining behavioral studies with gradient index lens imaging in the prefrontal cortex, and potentially other regions of the mouse brain, to study structural, functional, and behavioral relationships in the brain.
Collapse
Affiliation(s)
- Seon A. Lee
- Center for Biomedical Research and Rehabilitation Sciences, Louisiana Tech University, Ruston, Louisiana, United States of America
| | - Kevin S. Holly
- Center for Biomedical Research and Rehabilitation Sciences, Louisiana Tech University, Ruston, Louisiana, United States of America
| | - Vladislav Voziyanov
- Center for Biomedical Research and Rehabilitation Sciences, Louisiana Tech University, Ruston, Louisiana, United States of America
| | - Stephanie L. Villalba
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA, United States of America
| | - Rudi Tong
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Holly E. Grigsby
- Center for Biomedical Research and Rehabilitation Sciences, Louisiana Tech University, Ruston, Louisiana, United States of America
| | - Edward Glasscock
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA, United States of America
| | - Francis G. Szele
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Ioannis Vlachos
- Center for Biomedical Research and Rehabilitation Sciences, Louisiana Tech University, Ruston, Louisiana, United States of America
| | - Teresa A. Murray
- Center for Biomedical Research and Rehabilitation Sciences, Louisiana Tech University, Ruston, Louisiana, United States of America
| |
Collapse
|