1
|
Campbell A, Lai T, Wahba AE, Boison D, Gebril HM. Enhancing neurogenesis after traumatic brain injury: The role of adenosine kinase inhibition in promoting neuronal survival and differentiation. Exp Neurol 2024; 381:114930. [PMID: 39173898 DOI: 10.1016/j.expneurol.2024.114930] [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: 05/20/2024] [Revised: 07/30/2024] [Accepted: 08/16/2024] [Indexed: 08/24/2024]
Abstract
Traumatic brain injury (TBI) presents a significant public health challenge, necessitating innovative interventions for effective treatment. Recent studies have challenged conventional perspectives on neurogenesis, unveiling endogenous repair mechanisms within the adult brain following injury. However, the intricate mechanisms governing post-TBI neurogenesis remain unclear. The microenvironment of an injured brain, characterized by astrogliosis, neuroinflammation, and excessive cell death, significantly influences the fate of newly generated neurons. Adenosine kinase (ADK), the key metabolic regulator of adenosine, emerges as a crucial factor in brain development and cell proliferation after TBI. This study investigates the hypothesis that targeting ADK could enhance brain repair, promote neuronal survival, and facilitate differentiation. In a TBI model induced by controlled cortical impact, C57BL/6 male mice received intraperitoneal injections of the small molecule ADK inhibitor 5-iodotubercidin (ITU) for three days following TBI. To trace the fate of TBI-associated proliferative cells, animals received intraperitoneal injections of BrdU for seven days, beginning immediately after TBI. Our results show that ADK inhibition by ITU improved brain repair 14 days after injury as evidenced by a diminished injury size. Additionally, the number of mature neurons generated after TBI was increased in ITU-treated mice. Remarkably, the TBI-associated pathological events including astrogliosis, neuroinflammation, and cell death were arrested in ITU-treated mice. Finally, ADK inhibition modulated cell death by regulating the PERK signaling pathway. Together, these findings demonstrate a novel therapeutic approach to target multiple pathological mechanisms involved in TBI. This research contributes valuable insights into the intricate molecular mechanisms underlying neurogenesis and gliosis after TBT.
Collapse
Affiliation(s)
- Andrea Campbell
- Departement of Neuroscience, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14620, USA; Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - Tho Lai
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - Amir E Wahba
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA; Chemistry Department, Faculty of Science, Damietta University, New Damietta City 34518, Egypt
| | - Detlev Boison
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - Hoda M Gebril
- Departement of Biomedical Engineering, School of Engineering, Rutgers University, Piscataway, NJ 08854, USA.
| |
Collapse
|
2
|
Yadav P, Nasir F, Sivanandam TM. Neuroprotective effect of vitamin B 12 supplementation on cognitive functions and neuronal morphology at different time intervals after traumatic brain injury in male Swiss albino mice. Neurochem Int 2024; 180:105869. [PMID: 39332530 DOI: 10.1016/j.neuint.2024.105869] [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: 07/15/2024] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 09/29/2024]
Abstract
Traumatic brain injury is a highly irreversible process that consists of primary as well as secondary injury which develops and progresses over months to years, leading to cognitive dysfunctions. Vitamin B12 received considerable interest due to its potential therapeutic properties. The pathways of vitamin B12 are closely related to neuronal survival but its effects on the pathophysiology of injury with respect to cognition is a relatively unexplored area of research. In this study, we investigated, the effect of vitamin B12 and its involvement in neuroprotection on TBI-induced pathophysiology in male Swiss albino mice. Our findings suggested that vitamin B12 supplementation improves TBI-mediated neurological impairments, spatial and recognition memory, and anxiety-like behavior. Furthermore, the oxidative stress was reduced by declined homocysteine level with vitamin B12 supplementation validating declined expression of astrocytes and TBI biomarkers. The studies on neuronal morphology revealed that vitamin B12 supplementation increases the dendritic arborization and density of mushroom and filopodia-shaped spines and further increases the expression of synaptic plasticity-related genes and proteins. Taken together, our findings reveal that, supplementation of vitamin B12 restored the TBI-induced downregulation of dendritic arborization, and spine density which ultimately increases synaptic plasticity, cell survival, and recovery of cognitive dysfunctions.
Collapse
Affiliation(s)
- Priyanka Yadav
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| | - Farheen Nasir
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| | - Thamil Mani Sivanandam
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
3
|
Wang J, Gong R, Heidari S, Rogers M, Tani T, Abe H, Ichinohe N, Woodward A, Delmas PJ. A Deep Learning-based Pipeline for Segmenting the Cerebral Cortex Laminar Structure in Histology Images. Neuroinformatics 2024:10.1007/s12021-024-09688-0. [PMID: 39417954 DOI: 10.1007/s12021-024-09688-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2024] [Indexed: 10/19/2024]
Abstract
Characterizing the anatomical structure and connectivity between cortical regions is a critical step towards understanding the information processing properties of the brain and will help provide insight into the nature of neurological disorders. A key feature of the mammalian cerebral cortex is its laminar structure. Identifying these layers in neuroimaging data is important for understanding their global structure and to help understand the connectivity patterns of neurons in the brain. We studied Nissl-stained and myelin-stained slice images of the brain of the common marmoset (Callithrix jacchus), which is a new world monkey that is becoming increasingly popular in the neuroscience community as an object of study. We present a novel computational framework that first acquired the cortical labels using AI-based tools followed by a trained deep learning model to segment cerebral cortical layers. We obtained a Euclidean distance of 1274.750 ± 156.400 μ m for the cortical labels acquisition, which was in the acceptable range by computing the half Euclidean distance of the average cortex thickness ( 1800.630 μ m ). We compared our cortical layer segmentation pipeline with the pipeline proposed by Wagstyl et al. (PLoS biology, 18(4), e3000678 2020) adapted to 2D data. We obtained a better mean95 th percentile Hausdorff distance (95HD) of 92.150 μ m . Whereas a mean 95HD of 94.170 μ m was obtained from Wagstyl et al. We also compared our pipeline's performance against theirs using their dataset (the BigBrain dataset). The results also showed better segmentation quality, 85.318 % Jaccard Index acquired from our pipeline, while 83.000 % was stated in their paper.
Collapse
Affiliation(s)
- Jiaxuan Wang
- Intelligent Vision Systems Lab, The University of Auckland, Auckland, New Zealand
| | - Rui Gong
- Theoretical Biology Group, Department of Creative Research, Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, Aichi, 444-8787, Japan
| | - Shahrokh Heidari
- Intelligent Vision Systems Lab, The University of Auckland, Auckland, New Zealand
| | - Mitchell Rogers
- Intelligent Vision Systems Lab, The University of Auckland, Auckland, New Zealand
| | - Toshiki Tani
- Laboratory for Molecular Analysis of Higher Brain Function, RIKEN Center for Brain Science, Wako, Japan
| | - Hiroshi Abe
- Laboratory for Molecular Analysis of Higher Brain Function, RIKEN Center for Brain Science, Wako, Japan
| | - Noritaka Ichinohe
- Department of Ultrastructural Research, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Alexander Woodward
- Intelligent Vision Systems Lab, The University of Auckland, Auckland, New Zealand
| | - Patrice J Delmas
- Intelligent Vision Systems Lab, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
4
|
Boese M, Berman RY, Qiu J, Spencer HF, Radford KD, Choi KH. Effects of Mild Closed-Head Injury and Subanesthetic Ketamine Infusion on Microglia, Axonal Injury, and Synaptic Density in Sprague-Dawley Rats. Int J Mol Sci 2024; 25:4287. [PMID: 38673871 PMCID: PMC11050690 DOI: 10.3390/ijms25084287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/06/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Mild traumatic brain injury (mTBI) affects millions of people in the U.S. Approximately 20-30% of those individuals develop adverse symptoms lasting at least 3 months. In a rat mTBI study, the closed-head impact model of engineered rotational acceleration (CHIMERA) produced significant axonal injury in the optic tract (OT), indicating white-matter damage. Because retinal ganglion cells project to the lateral geniculate nucleus (LGN) in the thalamus through the OT, we hypothesized that synaptic density may be reduced in the LGN of rats following CHIMERA injury. A modified SEQUIN (synaptic evaluation and quantification by imaging nanostructure) method, combined with immunofluorescent double-labeling of pre-synaptic (synapsin) and post-synaptic (PSD-95) markers, was used to quantify synaptic density in the LGN. Microglial activation at the CHIMERA injury site was determined using Iba-1 immunohistochemistry. Additionally, the effects of ketamine, a potential neuroprotective drug, were evaluated in CHIMERA-induced mTBI. A single-session repetitive (ssr-) CHIMERA (3 impacts, 1.5 joule/impact) produced mild effects on microglial activation at the injury site, which was significantly enhanced by post-injury intravenous ketamine (10 mg/kg) infusion. However, ssr-CHIMERA did not alter synaptic density in the LGN, although ketamine produced a trend of reduction in synaptic density at post-injury day 4. Further research is necessary to characterize the effects of ssr-CHIMERA and subanesthetic doses of intravenous ketamine on different brain regions and multiple time points post-injury. The current study demonstrates the utility of the ssr-CHIMERA as a rodent model of mTBI, which researchers can use to identify biological mechanisms of mTBI and to develop improved treatment strategies for individuals suffering from head trauma.
Collapse
Affiliation(s)
- Martin Boese
- Daniel K. Inouye Graduate School of Nursing, Uniformed Services University, Bethesda, MD 20814, USA; (M.B.); (K.D.R.)
| | - Rina Y. Berman
- Center for the Study of Traumatic Stress, Uniformed Services University, Bethesda, MD 20814, USA;
| | - Jennifer Qiu
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742, USA;
| | - Haley F. Spencer
- Program in Neuroscience, Uniformed Services University, Bethesda, MD 20814, USA;
| | - Kennett D. Radford
- Daniel K. Inouye Graduate School of Nursing, Uniformed Services University, Bethesda, MD 20814, USA; (M.B.); (K.D.R.)
| | - Kwang H. Choi
- Daniel K. Inouye Graduate School of Nursing, Uniformed Services University, Bethesda, MD 20814, USA; (M.B.); (K.D.R.)
- Center for the Study of Traumatic Stress, Uniformed Services University, Bethesda, MD 20814, USA;
- Program in Neuroscience, Uniformed Services University, Bethesda, MD 20814, USA;
- Department of Psychiatry, F. E. Hébert School of Medicine, Uniformed Services University, Bethesda, MD 20814, USA
| |
Collapse
|
5
|
Tian Z, Cao Z, Yang E, Li J, Liao D, Wang F, Wang T, Zhang Z, Zhang H, Jiang X, Li X, Luo P. Quantitative proteomic and phosphoproteomic analyses of the hippocampus reveal the involvement of NMDAR1 signaling in repetitive mild traumatic brain injury. Neural Regen Res 2023; 18:2711-2719. [PMID: 37449635 DOI: 10.4103/1673-5374.374654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023] Open
Abstract
The cumulative damage caused by repetitive mild traumatic brain injury can cause long-term neurodegeneration leading to cognitive impairment. This cognitive impairment is thought to result specifically from damage to the hippocampus. In this study, we detected cognitive impairment in mice 6 weeks after repetitive mild traumatic brain injury using the novel object recognition test and the Morris water maze test. Immunofluorescence staining showed that p-tau expression was increased in the hippocampus after repetitive mild traumatic brain injury. Golgi staining showed a significant decrease in the total density of neuronal dendritic spines in the hippocampus, as well as in the density of mature dendritic spines. To investigate the specific molecular mechanisms underlying cognitive impairment due to hippocampal damage, we performed proteomic and phosphoproteomic analyses of the hippocampus with and without repetitive mild traumatic brain injury. The differentially expressed proteins were mainly enriched in inflammation, immunity, and coagulation, suggesting that non-neuronal cells are involved in the pathological changes that occur in the hippocampus in the chronic stage after repetitive mild traumatic brain injury. In contrast, differentially expressed phosphorylated proteins were mainly enriched in pathways related to neuronal function and structure, which is more consistent with neurodegeneration. We identified N-methyl-D-aspartate receptor 1 as a hub molecule involved in the response to repetitive mild traumatic brain injury , and western blotting showed that, while N-methyl-D-aspartate receptor 1 expression was not altered in the hippocampus after repetitive mild traumatic brain injury, its phosphorylation level was significantly increased, which is consistent with the omics results. Administration of GRP78608, an N-methyl-D-aspartate receptor 1 antagonist, to the hippocampus markedly improved repetitive mild traumatic brain injury-induced cognitive impairment. In conclusion, our findings suggest that N-methyl-D-aspartate receptor 1 signaling in the hippocampus is involved in cognitive impairment in the chronic stage after repetitive mild traumatic brain injury and may be a potential target for intervention and treatment.
Collapse
Affiliation(s)
- Zhicheng Tian
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Zixuan Cao
- The Sixth Regiment, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Erwan Yang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Juan Li
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Dan Liao
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Fei Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an; Medical Experiment Center, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Taozhi Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi Province; Department of Anesthesiology, The Second Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| | - Zhuoyuan Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University; School of Life Science, Northwest University, Xi'an, Shaanxi Province, China
| | - Haofuzi Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Xiaofan Jiang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Xin Li
- Department of Anesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Peng Luo
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| |
Collapse
|
6
|
Hennigan K, Lavik E. Nature vs. Manmade: Comparing Exosomes and Liposomes for Traumatic Brain Injury. AAPS J 2023; 25:83. [PMID: 37610471 DOI: 10.1208/s12248-023-00849-8] [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: 03/30/2023] [Accepted: 08/08/2023] [Indexed: 08/24/2023] Open
Abstract
Traumatic brain injury (TBI) of all severities is a significant public health burden, causing a range of effects that can lead to death or a diminished quality of life. Liposomes and mesenchymal stem cell-derived exosomes are two drug delivery agents with potential to be leveraged in the treatment of TBI by increasing the efficacy of drug therapies as well as having additional therapeutic effects. They exhibit several physical similarities, but key differences affect their performances as nanocarriers. Liposomes can be produced commercially at scale, and liposomes achieve higher encapsulation efficiency. Meanwhile, the intrinsic cargo and targeting moieties of exosomes, which liposomes lack, give exosomes a greater ability to facilitate neural regeneration, and exosomes do not trigger the infusion reactions that liposomes can. However, there are concerns about both exosomes and liposomes regarding interactions with tumors. The same routes of administration can be used for both exosomes and liposomes, resulting in somewhat different distribution throughout the body. While the effect of the nanocarrier type on accumulation in the brain is not concrete, targeting leads to increased accumulation of both exosomes and liposomes in the brain, upon which on-demand release can be used for both drug deliverers. Although neither have been applied to TBI in humans, preclinical trials have shown their immense potential, as have clinical trials pertaining to other brain injuries and conditions. While questions remain, research thus far shows that the various differences make exosomes a better choice of nanocarrier for TBI.
Collapse
Affiliation(s)
- Kate Hennigan
- Marriotts Ridge High School, Ellicott City, Maryland, 21042, USA
| | - Erin Lavik
- University of Maryland, Baltimore County, 1000 Hilltop Circle, Baltimore, Maryland, 21250, USA.
| |
Collapse
|
7
|
Nelles DG, Hazrati LN. The pathological potential of ependymal cells in mild traumatic brain injury. Front Cell Neurosci 2023; 17:1216420. [PMID: 37396927 PMCID: PMC10312375 DOI: 10.3389/fncel.2023.1216420] [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: 05/03/2023] [Accepted: 06/05/2023] [Indexed: 07/04/2023] Open
Abstract
Mild traumatic brain injury (mTBI) is a common neurological condition affecting millions of individuals worldwide. Although the pathology of mTBI is not fully understood, ependymal cells present a promising approach for studying the pathogenesis of mTBI. Previous studies have revealed that DNA damage in the form of γH2AX accumulates in ependymal cells following mTBI, with evidence of widespread cellular senescence in the brain. Ependymal ciliary dysfunction has also been observed, leading to altered cerebrospinal fluid homeostasis. Even though ependymal cells have not been extensively studied in the context of mTBI, these observations reflect the pathological potential of ependymal cells that may underlie the neuropathological and clinical presentations of mTBI. This mini review explores the molecular and structural alterations that have been reported in ependymal cells following mTBI, as well as the potential pathological mechanisms mediated by ependymal cells that may contribute to overall dysfunction of the brain post-mTBI. Specifically, we address the topics of DNA damage-induced cellular senescence, dysregulation of cerebrospinal fluid homeostasis, and the consequences of impaired ependymal cell barriers. Moreover, we highlight potential ependymal cell-based therapies for the treatment of mTBI, with a focus on neurogenesis, ependymal cell repair, and modulation of senescence signaling pathways. Further insight and research in this field will help to establish the role of ependymal cells in the pathogenesis of mTBI and may lead to improved treatments that leverage ependymal cells to target the origins of mTBI pathology.
Collapse
Affiliation(s)
- Diana G. Nelles
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- The Hospital for Sick Children, Toronto, ON, Canada
| | - Lili-Naz Hazrati
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- The Hospital for Sick Children, Toronto, ON, Canada
| |
Collapse
|
8
|
Paul MM, Mieden HJ, Lefering R, Kupczyk EK, Jordan MC, Gilbert F, Meffert RH, Sirén AL, Hoelscher-Doht S. Impact of a Femoral Fracture on Outcome after Traumatic Brain Injury-A Matched-Pair Analysis of the TraumaRegister DGU ®. J Clin Med 2023; 12:jcm12113802. [PMID: 37297997 DOI: 10.3390/jcm12113802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/28/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
Traumatic brain injury (TBI) is the leading cause of death and disability in polytrauma and is often accompanied by concomitant injuries. We conducted a retrospective matched-pair analysis of data from a 10-year period from the multicenter database TraumaRegister DGU® to analyze the impact of a concomitant femoral fracture on the outcome of TBI patients. A total of 4508 patients with moderate to critical TBI were included and matched by severity of TBI, American Society of Anesthesiologists (ASA) risk classification, initial Glasgow Coma Scale (GCS), age, and sex. Patients who suffered combined TBI and femoral fracture showed increased mortality and worse outcome at the time of discharge, a higher chance of multi-organ failure, and a rate of neurosurgical intervention. Especially those with moderate TBI showed enhanced in-hospital mortality when presenting with a concomitant femoral fracture (p = 0.037). The choice of fracture treatment (damage control orthopedics vs. early total care) did not impact mortality. In summary, patients with combined TBI and femoral fracture have higher mortality, more in-hospital complications, an increased need for neurosurgical intervention, and inferior outcome compared to patients with TBI solely. More investigations are needed to decipher the pathophysiological consequences of a long-bone fracture on the outcome after TBI.
Collapse
Affiliation(s)
- Mila M Paul
- Department of Orthopedic Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital of Würzburg, 97080 Würzburg, Germany
- Department of Neurophysiology, Institute for Physiology, Julius-Maximilians-University Würzburg, 97070 Würzburg, Germany
- Department of Neurosurgery, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Hannah J Mieden
- Department of Orthopedic Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Rolf Lefering
- Institute for Research in Operative Medicine (IFOM), University of Witten/Herdecke, 51109 Cologne, Germany
| | - Eva K Kupczyk
- Department of Orthopedic Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Martin C Jordan
- Department of Orthopedic Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Fabian Gilbert
- LMU Klinikum Campus Innenstadt, University of München, 80336 Munich, Germany
| | - Rainer H Meffert
- Department of Orthopedic Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Anna-Leena Sirén
- Department of Neurophysiology, Institute for Physiology, Julius-Maximilians-University Würzburg, 97070 Würzburg, Germany
- Department of Neurosurgery, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Stefanie Hoelscher-Doht
- Department of Orthopedic Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital of Würzburg, 97080 Würzburg, Germany
| |
Collapse
|
9
|
Xu H, Xu C, Gu P, Hu Y, Guo Y, Bai G. Neuroanatomical restoration of salience network links reduced headache impact to cognitive function improvement in mild traumatic brain injury with posttraumatic headache. J Headache Pain 2023; 24:43. [PMID: 37081382 PMCID: PMC10120179 DOI: 10.1186/s10194-023-01579-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 04/14/2023] [Indexed: 04/22/2023] Open
Abstract
BACKGROUND Neuroanatomical alterations have been associated with cognitive deficits in mild traumatic brain injury (MTBI). However, most studies have focused on the abnormal gray matter volume in widespread brain regions using a cross-sectional design in MTBI. This study investigated the neuroanatomical restoration of key regions in salience network and the outcomes in MTBI. METHODS Thirty-six MTBI patients with posttraumatic headache (PTH) and 34 matched healthy controls were enrolled in this study. All participants underwent magnetic resonance imaging scans and were assessed with clinical measures during the acute and subacute phases. Surface-based morphometry was conducted to get cortical thickness (CT) and cortical surface area (CSA) of neuroanatomical regions which were defined by the Desikan atlas. Then mixed analysis of variance models were performed to examine CT and CSA restoration in patients from acute to subacute phase related to controls. Finally, mediation effects models were built to explore the relationships between neuroanatomical restoration and symptomatic improvement in patients. RESULTS MTBI patients with PTH showed reduced headache impact and improved cognitive function from the acute to subacute phase. Moreover, patients experienced restoration of CT of the left caudal anterior cingulate cortex (ACC) and left insula and cortical surface area of the right superior frontal gyrus from acute to subacute phase. Further mediation analysis found that CT restoration of the ACC and insula mediated the relationship between reduced headache impact and improved cognitive function in patients. CONCLUSIONS These results showed that neuroanatomical restoration of key regions in salience network correlated reduced headache impact with cognitive function improvement in MTBI with PTH, which further substantiated the vital role of salience network and provided an alternative clinical target for cognitive improvement in MTBI patients with PTH.
Collapse
Affiliation(s)
- Hui Xu
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.
- Peter Boris Centre for Addictions Research, St. Joseph's Healthcare Hamilton/McMaster University, 100 West 5Th Street, Hamilton, ON, L8P 3R2, Canada.
| | - Cheng Xu
- Peter Boris Centre for Addictions Research, St. Joseph's Healthcare Hamilton/McMaster University, 100 West 5Th Street, Hamilton, ON, L8P 3R2, Canada
- School of Psychology and Cognitive Science, East China Normal University, Shanghai, 200062, China
| | - Pengpeng Gu
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Yike Hu
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Yunyu Guo
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Guanghui Bai
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.
| |
Collapse
|
10
|
The Dialogue Between Neuroinflammation and Adult Neurogenesis: Mechanisms Involved and Alterations in Neurological Diseases. Mol Neurobiol 2023; 60:923-959. [PMID: 36383328 DOI: 10.1007/s12035-022-03102-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 10/23/2022] [Indexed: 11/18/2022]
Abstract
Adult neurogenesis occurs mainly in the subgranular zone of the hippocampal dentate gyrus and the subventricular zone of the lateral ventricles. Evidence supports the critical role of adult neurogenesis in various conditions, including cognitive dysfunction, Alzheimer's disease (AD), and Parkinson's disease (PD). Several factors can alter adult neurogenesis, including genetic, epigenetic, age, physical activity, diet, sleep status, sex hormones, and central nervous system (CNS) disorders, exerting either pro-neurogenic or anti-neurogenic effects. Compelling evidence suggests that any insult or injury to the CNS, such as traumatic brain injury (TBI), infectious diseases, or neurodegenerative disorders, can provoke an inflammatory response in the CNS. This inflammation could either promote or inhibit neurogenesis, depending on various factors, such as chronicity and severity of the inflammation and underlying neurological disorders. Notably, neuroinflammation, driven by different immune components such as activated glia, cytokines, chemokines, and reactive oxygen species, can regulate every step of adult neurogenesis, including cell proliferation, differentiation, migration, survival of newborn neurons, maturation, synaptogenesis, and neuritogenesis. Therefore, this review aims to present recent findings regarding the effects of various components of the immune system on adult neurogenesis and to provide a better understanding of the role of neuroinflammation and neurogenesis in the context of neurological disorders, including AD, PD, ischemic stroke (IS), seizure/epilepsy, TBI, sleep deprivation, cognitive impairment, and anxiety- and depressive-like behaviors. For each disorder, some of the most recent therapeutic candidates, such as curcumin, ginseng, astragaloside, boswellic acids, andrographolide, caffeine, royal jelly, estrogen, metformin, and minocycline, have been discussed based on the available preclinical and clinical evidence.
Collapse
|
11
|
Kim JT, Kim TY, Youn DH, Han SW, Park CH, Lee Y, Jung H, Rhim JK, Park JJ, Ahn JH, Kim HC, Cho SM, Jeon JP. Human embryonic stem cell-derived cerebral organoids for treatment of mild traumatic brain injury in a mouse model. Biochem Biophys Res Commun 2022; 635:169-178. [PMID: 36274367 DOI: 10.1016/j.bbrc.2022.10.045] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 09/28/2022] [Accepted: 10/12/2022] [Indexed: 11/24/2022]
Abstract
OBJECTIVE There are no effective treatments for relieving neuronal dysfunction after mild traumatic brain injury (TBI). Here, we evaluated therapeutic efficacy of human embryonic stem cell-derived cerebral organoids (hCOs) in a mild TBI model, in terms of repair of damaged cortical regions, neurogenesis, and improved cognitive function. METHODS Male C57BL/6 J mice were randomly divided into sham-operated, mild TBI, and mild TBI with hCO groups. hCOs cultured at 8 weeks were used for transplantation. Mice were sacrificed at 7 and 14 days after transplantation followed by immunofluorescence staining, cytokine profile microarray, and novel object recognition test. RESULTS 8W-hCOs transplantation significantly reduced neuronal cell death, recovered microvessel density, and promoted neurogenesis in the ipsilateral subventricular zone and dentate gyrus of hippocampus after mild TBI. In addition, increased angiogenesis into the engrafted hCOs was observed. Microarray results of hCOs revealed neuronal differentiation potential and higher expression of early brain development proteins associated with neurogenesis, angiogenesis and extracellular matrix remodeling. Ultimately, 8W-hCO transplantation resulted in reconstruction of damaged cortex and improvement in cognitive function after mild TBI. CONCLUSION hCO transplantation may be feasible for treating mild TBI-related neuronal dysfunction via reconstruction of damaged cortex and neurogenesis in the hippocampus.
Collapse
Affiliation(s)
- Jong-Tae Kim
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon, South Korea
| | - Tae Yeon Kim
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon, South Korea
| | - Dong Hyuk Youn
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon, South Korea
| | - Sung Woo Han
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon, South Korea
| | - Chan Hum Park
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon, South Korea
| | - Younghyurk Lee
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon, South Korea
| | - Harry Jung
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon, South Korea
| | - Jong Kook Rhim
- Department of Neurosurgery, Jeju National University College of Medicine, Jeju, South Korea
| | - Jeong Jin Park
- Department of Neurology, Konkuk University Medical Center, Seoul, South Korea
| | - Jun Hyong Ahn
- Department of Neurosurgery, Kangwon National University Hospital, Chuncheon, South Korea
| | - Heung Cheol Kim
- Department of Radiology, Hallym University College of Medicine, Chuncheon, South Korea
| | - Sung Min Cho
- Department of Neurosurgery, Yonsei University Wonju College of Medicine, Wonju, South Korea.
| | - Jin Pyeong Jeon
- Department of Neurosurgery, Hallym University College of Medicine, Chuncheon, South Korea.
| |
Collapse
|
12
|
Kaya D, Micili SC, Kizmazoglu C, Mucuoglu AO, Buyukcoban S, Ersoy N, Yilmaz O, Isik AT. Allopurinol attenuates repeated traumatic brain injury in old rats: A preliminary report. Exp Neurol 2022; 357:114196. [PMID: 35931122 DOI: 10.1016/j.expneurol.2022.114196] [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: 04/17/2022] [Revised: 07/13/2022] [Accepted: 07/31/2022] [Indexed: 12/01/2022]
Abstract
Traumatic brain injury (TBI) is an overlooked cause of morbidity, which was shown to accelerate inflammation, oxidative stress, and neuronal cell loss and is associated with spatial learning and memory impairments and some psychiatric disturbances in older adults. However, there is no effective treatment in order to offer a favorable outcome encompassing a good recovery after TBI in older adults. Hence, the present study aimed to investigate the histological and neurobehavioral effects of Allopurinol (ALL) in older rats that received repeated TBI (rTBI). For this purpose, a weight-drop rTBI model was used on old male Wistar rats. Rats received 5 repeated TBI/sham injuries 24 h apart and were treated with saline or Allopurinol 100 mg/kg, i.p. each time. They were randomly assigned to three groups: control group (no injury); rTBI group (received 5 rTBI and treated with saline); rTBI+ALL group (received 5 rTBI and treated with Allopurinol). Then, half of the animals from each group were sacrificed on day 6 and the remaining animals were assessed with Open field, Elevated plus maze and Morris Water Maze test. Basic neurological tasks were evaluated with neurological assessment protocol every other day until after the 19th day from the last injury. Brain sections were processed for neuronal cell count in the hippocampus (CA1), dentate gyrus (DG), and prefrontal cortex (PC). Also, an immunohistochemical assay was performed to determine NeuN, iNOS, and TNFα levels in the brain regions. The number of neurons was markedly reduced in CA1, GD, and PC in rats receiving saline compared to those receiving allopurinol treatment. Immunohistochemical analysis showed marked induction of iNOS and TNFα expression in the brain tissues which were reduced after allopurinol at 6 and 19 days post-injury. Also, ALL-treated rats demonstrated a remarkable induce in NeuN expression, indicating a reduction in rTBI-induced neuronal cell death. In neurobehavioral analyses, time spent in closed arms, in the corner of the open field, swimming latency, and distance were impaired in injured rats; however, all of them were significantly improved by allopurinol therapy. To sum up, this study demonstrated that ALL may mitigate rTBI-induced damage in aged rats, which suggests ALL as a potential therapeutic strategy for the treatment of recurrent TBI.
Collapse
Affiliation(s)
- Derya Kaya
- Dokuz Eylul University Faculty of Medicine, Department of Geriatric Medicine, Unit for Brain Aging and Dementia, Izmir, Turkey; Geriatric Science Association, Izmir, Turkey.
| | - Serap Cilaker Micili
- Dokuz Eylul University Faculty of Medicine, Department of Histology and Embryology, Izmir, Turkey
| | - Ceren Kizmazoglu
- Dokuz Eylul University Faculty of Medicine, Department of Neurosurgery, Izmir, Turkey
| | - Ali Osman Mucuoglu
- Dokuz Eylul University Faculty of Medicine, Department of Neurosurgery, Izmir, Turkey
| | - Sibel Buyukcoban
- Dokuz Eylul University Faculty of Medicine, Department of Anaesthesiology and Reanimation, İzmir, Turkey
| | - Nevin Ersoy
- Dokuz Eylul University Faculty of Medicine, Department of Histology and Embryology, Izmir, Turkey
| | - Osman Yilmaz
- Dokuz Eylul University Health Sciences Institute, Department of Laboratory Animal Science, Izmir, Turkey
| | - Ahmet Turan Isik
- Dokuz Eylul University Faculty of Medicine, Department of Geriatric Medicine, Unit for Brain Aging and Dementia, Izmir, Turkey; Geriatric Science Association, Izmir, Turkey
| |
Collapse
|
13
|
Eyolfson E, Carr T, Fraunberger E, Khan A, Clark I, Mychasiuk R, Lohman AW. Repeated mild traumatic brain injuries in mice cause age- and sex-specific alterations in dendritic spine density. Exp Neurol 2022; 357:114172. [PMID: 35863503 DOI: 10.1016/j.expneurol.2022.114172] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/08/2022] [Accepted: 07/14/2022] [Indexed: 11/25/2022]
Abstract
Mild traumatic brain injuries (mTBI) plague the human population and their prevalence is increasing annually. More so, repeated mTBIs (RmTBI) are known to manifest and compound neurological deficits in vulnerable populations. Age at injury and sex are two important factors influencing RmTBI pathophysiology, but we continue to know little about the specific effects of RmTBI in youth and females. In this study, we directly quantified the effects of RmTBI on adolescent and adult, male and female mice, with a closed-head lateral impact model. We report age- and sex-specific neurobehavioural deficits in motor function and working memory, microglia responses to injury, and the subsequent changes in dendritic spine density in select brain regions. Specifically, RmTBI caused increased footslips in adult male mice as assessed in a beam walk assay and significantly reduced the time spent with a novel object in adolescent male and female mice. RmTBIs caused a significant reduction in microglia density in male mice in the motor cortex, but not female mice. Finally, RmTBI significantly reduced dendritic spine density in the agranular insular cortex (a region of the prefrontal cortex in mice) and increased dendritic spine density in the adolescent male motor cortex. Together, the data provided in this study sheds new light on the heterogeneity in RmTBI-induced behavioural, glial, and neuronal architecture changes dependent on age and sex.
Collapse
Affiliation(s)
- Eric Eyolfson
- Department of Psychology, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute (ACHRI), Calgary, AB, Canada.
| | - Thomas Carr
- Alberta Children's Hospital Research Institute (ACHRI), Calgary, AB, Canada; Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada.
| | - Erik Fraunberger
- Alberta Children's Hospital Research Institute (ACHRI), Calgary, AB, Canada; Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada.
| | - Asher Khan
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada.
| | - Isabel Clark
- Alberta Children's Hospital Research Institute (ACHRI), Calgary, AB, Canada; Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada.
| | - Richelle Mychasiuk
- Department of Psychology, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute (ACHRI), Calgary, AB, Canada; Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada; Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia.
| | - Alexander W Lohman
- Alberta Children's Hospital Research Institute (ACHRI), Calgary, AB, Canada; Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
14
|
Tu X, Li X, Zhu H, Kuang X, Si X, Zou S, Hao S, Huang Y, Xiao J. Unilateral cerebral ischemia induces morphological changes in the layer V projection neurons of the contralateral hemisphere. Neurosci Res 2022; 182:41-51. [PMID: 35777459 DOI: 10.1016/j.neures.2022.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 06/22/2022] [Accepted: 06/26/2022] [Indexed: 10/17/2022]
Abstract
Decreased blood flow to the brain causes stroke and damage to neuronal networks. Neuronal damage occurs not only in the infarct core but also in areas away from the infarcts. This study was aimed to assess alterations of the cortical projection neurons that were distantly connected with the infarcts. Unilateral cortical ischemia was generated by middle cerebral artery occlusion in the right somatosensory cortex. Pre-labeled thalamocortical neurons disappeared, whereas contralateral callosal projection neurons survived 48 h post-ischemia. The unilateral ischemia increased the total length, segment length and the spine volume of dendrites from layer V callosal neurons in the homotopic cortex of the contralateral hemisphere. The morphological remolding of the contralateral cortical neurons cannot be reproduced by the spinal cord hemisection that cuts axons of corticospinal projection neurons of layer V. The data suggest that the retrograde degeneration of axons may not account for the early morphological changes in the contralateral cortex. We hypothesize that the loss of innervations from the ischemic cortex may bring in adaptive changes to the connected neurons, and adult cortical neurons can adjust their morphology to meet the reduction of synaptic inputs. This study may improve our understanding of the re-organization of cortical circuits following focal cerebral ischemia and help the development of new treatments designed to minimize the disability associated with stroke.
Collapse
Affiliation(s)
- XiaoMeng Tu
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, PR China; State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou, Zhejiang, PR China
| | - Xue Li
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, PR China; State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou, Zhejiang, PR China
| | - Hao Zhu
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, PR China; State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou, Zhejiang, PR China
| | - Xiuli Kuang
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, PR China; State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou, Zhejiang, PR China
| | - Xiang Si
- Department of Ophthalmology, Anhui Provincial Hospital, Hefei, Anhui, PR China
| | - Shimin Zou
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, PR China; State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou, Zhejiang, PR China
| | - Shishuai Hao
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, PR China; State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou, Zhejiang, PR China
| | - Yang Huang
- Department of Orthopedics, Taizhou Municipal Hospital, Taizhou, Zhejiang, PRChina.
| | - Jian Xiao
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, PR China; State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou, Zhejiang, PR China.
| |
Collapse
|
15
|
Urrutia-Ruiz C, Rombach D, Cursano S, Gerlach-Arbeiter S, Schoen M, Bockmann J, Demestre M, Boeckers TM. Deletion of the Autism-Associated Protein SHANK3 Abolishes Structural Synaptic Plasticity after Brain Trauma. Int J Mol Sci 2022; 23:ijms23116081. [PMID: 35682760 PMCID: PMC9181590 DOI: 10.3390/ijms23116081] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 12/14/2022] Open
Abstract
Autism spectrum disorders (ASDs) are characterized by repetitive behaviors and impairments of sociability and communication. About 1% of ASD cases are caused by mutations of SHANK3, a major scaffolding protein of the postsynaptic density. We studied the role of SHANK3 in plastic changes of excitatory synapses within the central nervous system by employing mild traumatic brain injury (mTBI) in WT and Shank3 knockout mice. In WT mice, mTBI triggered ipsi- and contralateral loss of hippocampal dendritic spines and excitatory synapses with a partial recovery over time. In contrast, no significant synaptic alterations were detected in Shank3∆11−/− mice, which showed fewer dendritic spines and excitatory synapses at baseline. In line, mTBI induced the upregulation of synaptic plasticity-related proteins Arc and p-cofilin only in WT mice. Interestingly, microglia proliferation was observed in WT mice after mTBI but not in Shank3∆11−/− mice. Finally, we detected TBI-induced increased fear memory at the behavioral level, whereas in Shank3∆11−/− animals, the already-enhanced fear memory levels increased only slightly after mTBI. Our data show the lack of structural synaptic plasticity in Shank3 knockout mice that might explain at least in part the rigidity of behaviors, problems in adjusting to new situations and cognitive deficits seen in ASDs.
Collapse
Affiliation(s)
- Carolina Urrutia-Ruiz
- Institute for Anatomy and Cell Biology, Albert Einstein Allee 11, 89081 Ulm, Germany; (C.U.-R.); (D.R.); (S.C.); (S.G.-A.); (M.S.); (J.B.); (M.D.)
| | - Daniel Rombach
- Institute for Anatomy and Cell Biology, Albert Einstein Allee 11, 89081 Ulm, Germany; (C.U.-R.); (D.R.); (S.C.); (S.G.-A.); (M.S.); (J.B.); (M.D.)
| | - Silvia Cursano
- Institute for Anatomy and Cell Biology, Albert Einstein Allee 11, 89081 Ulm, Germany; (C.U.-R.); (D.R.); (S.C.); (S.G.-A.); (M.S.); (J.B.); (M.D.)
| | - Susanne Gerlach-Arbeiter
- Institute for Anatomy and Cell Biology, Albert Einstein Allee 11, 89081 Ulm, Germany; (C.U.-R.); (D.R.); (S.C.); (S.G.-A.); (M.S.); (J.B.); (M.D.)
| | - Michael Schoen
- Institute for Anatomy and Cell Biology, Albert Einstein Allee 11, 89081 Ulm, Germany; (C.U.-R.); (D.R.); (S.C.); (S.G.-A.); (M.S.); (J.B.); (M.D.)
| | - Juergen Bockmann
- Institute for Anatomy and Cell Biology, Albert Einstein Allee 11, 89081 Ulm, Germany; (C.U.-R.); (D.R.); (S.C.); (S.G.-A.); (M.S.); (J.B.); (M.D.)
| | - Maria Demestre
- Institute for Anatomy and Cell Biology, Albert Einstein Allee 11, 89081 Ulm, Germany; (C.U.-R.); (D.R.); (S.C.); (S.G.-A.); (M.S.); (J.B.); (M.D.)
| | - Tobias M. Boeckers
- Institute for Anatomy and Cell Biology, Albert Einstein Allee 11, 89081 Ulm, Germany; (C.U.-R.); (D.R.); (S.C.); (S.G.-A.); (M.S.); (J.B.); (M.D.)
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Translational Biochemistry, 89081 Ulm, Germany
- Correspondence: ; Tel.: +49-731-5002-3220
| |
Collapse
|
16
|
Characterisation of Severe Traumatic Brain Injury Severity from Fresh Cerebral Biopsy of Living Patients: An Immunohistochemical Study. Biomedicines 2022; 10:biomedicines10030518. [PMID: 35327320 PMCID: PMC8945429 DOI: 10.3390/biomedicines10030518] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 02/12/2022] [Accepted: 02/15/2022] [Indexed: 12/21/2022] Open
Abstract
Traumatic brain injury (TBI) is an extremely complex disease and current systems classifying TBI as mild, moderate, and severe often fail to capture this complexity. Neuroimaging cannot resolve the cellular and molecular changes due to lack of resolution, and post-mortem tissue examination may not adequately represent acute disease. Therefore, we examined the cellular and molecular sequelae of TBI in fresh brain samples and related these to clinical outcomes. Brain biopsies, obtained shortly after injury from 25 living adult patients suffering severe TBI, underwent immunohistochemical analysis. There were no adverse events. Immunostaining revealed various qualitative cellular and biomolecular changes relating to neuronal injury, dendritic injury, neurovascular injury, and neuroinflammation, which we classified into 4 subgroups for each injury type using the newly devised Yip, Hasan and Uff (YHU) grading system. Based on the Glasgow Outcome Scale-Extended, a total YHU grade of ≤8 or ≥11 had a favourable and unfavourable outcome, respectively. Biomolecular changes observed in fresh brain samples enabled classification of this heterogeneous patient population into various injury severity categories based on the cellular and molecular pathophysiology according to the YHU grading system, which correlated with outcome. This is the first study investigating the acute biomolecular response to TBI.
Collapse
|
17
|
Chang S, Varadarajan D, Yang J, Chen IA, Kura S, Magnain C, Augustinack JC, Fischl B, Greve DN, Boas DA, Wang H. Scalable mapping of myelin and neuron density in the human brain with micrometer resolution. Sci Rep 2022; 12:363. [PMID: 35013441 PMCID: PMC8748995 DOI: 10.1038/s41598-021-04093-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 12/03/2021] [Indexed: 12/23/2022] Open
Abstract
Optical coherence tomography (OCT) is an emerging 3D imaging technique that allows quantification of intrinsic optical properties such as scattering coefficient and back-scattering coefficient, and has proved useful in distinguishing delicate microstructures in the human brain. The origins of scattering in brain tissues are contributed by the myelin content, neuron size and density primarily; however, no quantitative relationships between them have been reported, which hampers the use of OCT in fundamental studies of architectonic areas in the human brain and the pathological evaluations of diseases. Here, we built a generalized linear model based on Mie scattering theory that quantitatively links tissue scattering to myelin content and neuron density in the human brain. We report a strong linear relationship between scattering coefficient and the myelin content that is retained across different regions of the brain. Neuronal cell body turns out to be a secondary contribution to the overall scattering. The optical property of OCT provides a label-free solution for quantifying volumetric myelin content and neuron cells in the human brain.
Collapse
Affiliation(s)
- Shuaibin Chang
- Department of Electrical and Computer Engineering, Boston University, 8 St Mary's St, Boston, 02215, USA
| | - Divya Varadarajan
- Department of Radiology, Massachusetts General Hospital, A.A. Martinos Center for Biomedical Imaging, 13th Street, Boston, 02129, USA
| | - Jiarui Yang
- Department of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston, 02215, USA
| | - Ichun Anderson Chen
- Department of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston, 02215, USA
| | - Sreekanth Kura
- Department of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston, 02215, USA
| | - Caroline Magnain
- Department of Radiology, Massachusetts General Hospital, A.A. Martinos Center for Biomedical Imaging, 13th Street, Boston, 02129, USA
| | - Jean C Augustinack
- Department of Radiology, Massachusetts General Hospital, A.A. Martinos Center for Biomedical Imaging, 13th Street, Boston, 02129, USA
| | - Bruce Fischl
- Department of Radiology, Massachusetts General Hospital, A.A. Martinos Center for Biomedical Imaging, 13th Street, Boston, 02129, USA
| | - Douglas N Greve
- Department of Radiology, Massachusetts General Hospital, A.A. Martinos Center for Biomedical Imaging, 13th Street, Boston, 02129, USA
| | - David A Boas
- Department of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston, 02215, USA
- Department of Electrical and Computer Engineering, Boston University, 8 St Mary's St, Boston, 02215, USA
| | - Hui Wang
- Department of Radiology, Massachusetts General Hospital, A.A. Martinos Center for Biomedical Imaging, 13th Street, Boston, 02129, USA.
| |
Collapse
|
18
|
Selvaraj P, Tanaka M, Wen J, Zhang Y. The Novel Monoacylglycerol Lipase Inhibitor MJN110 Suppresses Neuroinflammation, Normalizes Synaptic Composition and Improves Behavioral Performance in the Repetitive Traumatic Brain Injury Mouse Model. Cells 2021; 10:cells10123454. [PMID: 34943962 PMCID: PMC8700188 DOI: 10.3390/cells10123454] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 12/22/2022] Open
Abstract
Modulation of the endocannabinoid system has emerged as an effective approach for the treatment of many neurodegenerative and neuropsychological diseases. However, the underlying mechanisms are still uncertain. Using a repetitive mild traumatic brain injury (mTBI) mouse model, we found that there was an impairment in locomotor function and working memory within two weeks post-injury, and that treatment with MJN110, a novel inhibitor of the principal 2-arachidononyl glycerol (2-AG) hydrolytic enzyme monoacylglycerol lipase dose-dependently ameliorated those behavioral changes. Spatial learning and memory deficits examined by Morris water maze between three and four weeks post-TBI were also reversed in the drug treated animals. Administration of MJN110 selectively elevated the levels of 2-AG and reduced the production of arachidonic acid (AA) and prostaglandin E2 (PGE2) in the TBI mouse brain. The increased production of proinflammatory cytokines, accumulation of astrocytes and microglia in the TBI mouse ipsilateral cerebral cortex and hippocampus were significantly reduced by MJN110 treatment. Neuronal cell death was also attenuated in the drug treated animals. MJN110 treatment normalized the expression of the NMDA receptor subunits NR2A and NR2B, the AMPA receptor subunits GluR1 and GluR2, and the GABAA receptor subunits α1, β2,3 and γ2, which were all reduced at 1, 2 and 4 weeks post-injury. The reduced inflammatory response and restored glutamate and GABA receptor expression likely contribute to the improved motor function, learning and memory in the MJN110 treated animals. The therapeutic effects of MJN110 were partially mediated by activation of CB1 and CB2 cannabinoid receptors and were eliminated when it was co-administered with DO34, a novel inhibitor of the 2-AG biosynthetic enzymes. Our results suggest that augmentation of the endogenous levels of 2-AG can be therapeutically useful in the treatment of TBI by suppressing neuroinflammation and maintaining the balance between excitatory and inhibitory neurotransmission.
Collapse
Affiliation(s)
- Prabhuanand Selvaraj
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA; (P.S.); (M.T.); (J.W.)
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Mikiei Tanaka
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA; (P.S.); (M.T.); (J.W.)
| | - Jie Wen
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA; (P.S.); (M.T.); (J.W.)
| | - Yumin Zhang
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA; (P.S.); (M.T.); (J.W.)
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
- Correspondence: ; Tel.: +1-301-295-3212
| |
Collapse
|
19
|
Raikwar SP, Thangavel R, Ahmed ME, Selvakumar GP, Kempuraj D, Wu K, Khan O, Bazley K, Bussinger B, Kukulka K, Zaheer S, Iyer SS, Govindarajan R, Burton C, James D, Zaheer A. Real-Time Noninvasive Bioluminescence, Ultrasound and Photoacoustic Imaging in NFκB-RE-Luc Transgenic Mice Reveal Glia Maturation Factor-Mediated Immediate and Sustained Spatio-Temporal Activation of NFκB Signaling Post-Traumatic Brain Injury in a Gender-Specific Manner. Cell Mol Neurobiol 2021; 41:1687-1706. [PMID: 32785863 PMCID: PMC8188847 DOI: 10.1007/s10571-020-00937-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 08/05/2020] [Indexed: 12/15/2022]
Abstract
Neurotrauma especially traumatic brain injury (TBI) is the leading cause of death and disability worldwide. To improve upon the early diagnosis and develop precision-targeted therapies for TBI, it is critical to understand the underlying molecular mechanisms and signaling pathways. The transcription factor, nuclear factor kappa B (NFκB), which is ubiquitously expressed, plays a crucial role in the normal cell survival, proliferation, differentiation, function, as well as in disease states like neuroinflammation and neurodegeneration. Here, we hypothesized that real-time noninvasive bioluminescence molecular imaging allows rapid and precise monitoring of TBI-induced immediate and rapid spatio-temporal activation of NFκB signaling pathway in response to Glia maturation factor (GMF) upregulation which in turn leads to neuroinflammation and neurodegeneration post-TBI. To test and validate our hypothesis and to gain novel mechanistic insights, we subjected NFκB-RE-Luc transgenic male and female mice to TBI and performed real-time noninvasive bioluminescence imaging (BLI) as well as photoacoustic and ultrasound imaging (PAI). Our BLI data revealed that TBI leads to an immediate and sustained activation of NFκB signaling. Further, our BLI data suggest that especially in male NFκB-RE-Luc transgenic mice subjected to TBI, in addition to brain, there is widespread activation of NFκB signaling in multiple organs. However, in the case of the female NFκB-RE-Luc transgenic mice, TBI induces a very specific and localized activation of NFκB signaling in the brain. Further, our microRNA data suggest that TBI induces significant upregulation of mir-9-5p, mir-21a-5p, mir-34a-5p, mir-16-3p, as well as mir-155-5p within 24 h and these microRNAs can be successfully used as TBI-specific biomarkers. To the best of our knowledge, this is one of the first and unique study of its kind to report immediate and sustained activation of NFκB signaling post-TBI in a gender-specific manner by utilizing real-time non-invasive BLI and PAI in NFκB-RE-Luc transgenic mice. Our study will prove immensely beneficial to gain novel mechanistic insights underlying TBI, unravel novel therapeutic targets, as well as enable us to monitor in real-time the response to innovative TBI-specific precision-targeted gene and stem cell-based precision medicine.
Collapse
Affiliation(s)
- Sudhanshu P Raikwar
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, USA.
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA.
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA.
| | - Ramasamy Thangavel
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA
| | - Mohammad Ejaz Ahmed
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA
| | - Govindhasamy Pushpavathi Selvakumar
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA
| | - Duraisamy Kempuraj
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA
| | - Kristopher Wu
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Osaid Khan
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Kieran Bazley
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Bret Bussinger
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Klaudia Kukulka
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Smita Zaheer
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Shankar S Iyer
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA
| | - Raghav Govindarajan
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, USA
| | | | | | - Asgar Zaheer
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, USA.
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA.
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA.
| |
Collapse
|
20
|
de Fraga LS, Tassinari ID, Jantsch J, Guedes RP, Bambini-Junior V. 'A picture is worth a thousand words': The use of microscopy for imaging neuroinflammation. Clin Exp Immunol 2021; 206:325-345. [PMID: 34596237 DOI: 10.1111/cei.13669] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 01/08/2023] Open
Abstract
Since the first studies of the nervous system by the Nobel laureates Camillo Golgi and Santiago Ramon y Cajal using simple dyes and conventional light microscopes, microscopy has come a long way to the most recent techniques that make it possible to perform images in live cells and animals in health and disease. Many pathological conditions of the central nervous system have already been linked to inflammatory responses. In this scenario, several available markers and techniques can help imaging and unveil the neuroinflammatory process. Moreover, microscopy imaging techniques have become even more necessary to validate the large quantity of data generated in the era of 'omics'. This review aims to highlight how to assess neuroinflammation by using microscopy as a tool to provide specific details about the cell's architecture during neuroinflammatory conditions. First, we describe specific markers that have been used in light microscopy studies and that are widely applied to unravel and describe neuroinflammatory mechanisms in distinct conditions. Then, we discuss some important methodologies that facilitate the imaging of these markers, such as immunohistochemistry and immunofluorescence techniques. Emphasis will be given to studies using two-photon microscopy, an approach that revolutionized the real-time assessment of neuroinflammatory processes. Finally, some studies integrating omics with microscopy will be presented. The fusion of these techniques is developing, but the high amount of data generated from these applications will certainly improve comprehension of the molecular mechanisms involved in neuroinflammation.
Collapse
Affiliation(s)
- Luciano Stürmer de Fraga
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Isadora D'Ávila Tassinari
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Jeferson Jantsch
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Renata Padilha Guedes
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Victorio Bambini-Junior
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire (UCLan), Preston, UK
| |
Collapse
|
21
|
Whitney K, Nikulina E, Rahman SN, Alexis A, Bergold PJ. Delayed dosing of minocycline plus N-acetylcysteine reduces neurodegeneration in distal brain regions and restores spatial memory after experimental traumatic brain injury. Exp Neurol 2021; 345:113816. [PMID: 34310944 DOI: 10.1016/j.expneurol.2021.113816] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 07/07/2021] [Accepted: 07/20/2021] [Indexed: 11/16/2022]
Abstract
Multiple drugs to treat traumatic brain injury (TBI) have failed clinical trials. Most drugs lose efficacy as the time interval increases between injury and treatment onset. Insufficient therapeutic time window is a major reason underlying failure in clinical trials. Few drugs have been developed with therapeutic time windows sufficiently long enough to treat TBI because little is known about which brain functions can be targeted if therapy is delayed hours to days after injury. We identified multiple injury parameters that are improved by first initiating treatment with the drug combination minocycline (MINO) plus N-acetylcysteine (NAC) at 72 h after injury (MN72) in a mouse closed head injury (CHI) experimental TBI model. CHI produces spatial memory deficits resulting in impaired performance on Barnes maze, hippocampal neuronal loss, and bilateral damage to hippocampal neurons, dendrites, spines and synapses. MN72 treatment restores Barnes maze acquisition and retention, protects against hippocampal neuronal loss, limits damage to dendrites, spines and synapses, and accelerates recovery of microtubule associated protein 2 (MAP2) expression, a key protein in maintaining proper dendritic architecture and synapse density. These data show that in addition to the structural integrity of the dendritic arbor, spine and synapse density can be successfully targeted with drugs first dosed days after injury. Retention of substantial drug efficacy even when first dosed 72 h after injury makes MINO plus NAC a promising candidate to treat clinical TBI.
Collapse
Affiliation(s)
- Kristen Whitney
- Department of Physiology and Pharmacology, State University of New York-Downstate Health Sciences University, Brooklyn, NY 11215, United States of America; Program in Neural and Behavioral Science, School of Graduate Studies, State University of New York-Downstate Health Sciences University, Brooklyn, NY 11215, United States of America
| | - Elena Nikulina
- Department of Physiology and Pharmacology, State University of New York-Downstate Health Sciences University, Brooklyn, NY 11215, United States of America
| | - Syed N Rahman
- Department of Physiology and Pharmacology, State University of New York-Downstate Health Sciences University, Brooklyn, NY 11215, United States of America
| | - Alisia Alexis
- Department of Physiology and Pharmacology, State University of New York-Downstate Health Sciences University, Brooklyn, NY 11215, United States of America
| | - Peter J Bergold
- Department of Physiology and Pharmacology, State University of New York-Downstate Health Sciences University, Brooklyn, NY 11215, United States of America; Program in Neural and Behavioral Science, School of Graduate Studies, State University of New York-Downstate Health Sciences University, Brooklyn, NY 11215, United States of America.
| |
Collapse
|
22
|
Mester JR, Bazzigaluppi P, Dorr A, Beckett T, Burke M, McLaurin J, Sled JG, Stefanovic B. Attenuation of tonic inhibition prevents chronic neurovascular impairments in a Thy1-ChR2 mouse model of repeated, mild traumatic brain injury. Am J Cancer Res 2021; 11:7685-7699. [PMID: 34335958 PMCID: PMC8315057 DOI: 10.7150/thno.60190] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 06/04/2021] [Indexed: 12/24/2022] Open
Abstract
Rationale: Mild traumatic brain injury (mTBI), the most common type of brain trauma, frequently leads to chronic cognitive and neurobehavioral deficits. Intervening effectively is impeded by our poor understanding of its pathophysiological sequelae. Methods: To elucidate the long-term neurovascular sequelae of mTBI, we combined optogenetics, two-photon fluorescence microscopy, and intracortical electrophysiological recordings in mice to selectively stimulate peri-contusional neurons weeks following repeated closed-head injury and probe individual vessel's function and local neuronal reactivity. Results: Compared to sham-operated animals, mTBI mice showed doubled cortical venular speeds (115 ± 25%) and strongly elevated cortical venular reactivity (53 ± 17%). Concomitantly, the pericontusional neurons exhibited attenuated spontaneous activity (-57 ± 79%) and decreased reactivity (-47 ± 28%). Post-mortem immunofluorescence revealed signs of peri-contusional senescence and DNA damage, in the absence of neuronal loss or gliosis. Alteration of neuronal and vascular functioning was largely prevented by chronic, low dose, systemic administration of a GABA-A receptor inverse agonist (L-655,708), commencing 3 days following the third impact. Conclusions: Our findings indicate that repeated mTBI leads to dramatic changes in the neurovascular unit function and that attenuation of tonic inhibition can prevent these alterations. The sustained disruption of the neurovascular function may underlie the concussed brain's long-term susceptibility to injury, and calls for development of better functional assays as well as of neurovascularly targeted interventions.
Collapse
|
23
|
Wiegand TLT, Sollmann N, Bonke EM, Umeasalugo KE, Sobolewski KR, Plesnila N, Shenton ME, Lin AP, Koerte IK. Translational neuroimaging in mild traumatic brain injury. J Neurosci Res 2021; 100:1201-1217. [PMID: 33789358 DOI: 10.1002/jnr.24840] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/09/2021] [Accepted: 03/17/2021] [Indexed: 01/26/2023]
Abstract
Traumatic brain injuries (TBIs) are common with an estimated 27.1 million cases per year. Approximately 80% of TBIs are categorized as mild TBI (mTBI) based on initial symptom presentation. While in most individuals, symptoms resolve within days to weeks, in some, symptoms become chronic. Advanced neuroimaging has the potential to characterize brain morphometric, microstructural, biochemical, and metabolic abnormalities following mTBI. However, translational studies are needed for the interpretation of neuroimaging findings in humans with respect to the underlying pathophysiological processes, and, ultimately, for developing novel and more targeted treatment options. In this review, we introduce the most commonly used animal models for the study of mTBI. We then summarize the neuroimaging findings in humans and animals after mTBI and, wherever applicable, the translational aspects of studies available today. Finally, we highlight the importance of translational approaches and outline future perspectives in the field of translational neuroimaging in mTBI.
Collapse
Affiliation(s)
- Tim L T Wiegand
- cBRAIN, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Ludwig-Maximilians-Universität, Munich, Germany
| | - Nico Sollmann
- cBRAIN, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Ludwig-Maximilians-Universität, Munich, Germany
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- TUM-Neuroimaging Center, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Department of Diagnostic and Interventional Radiology, University Hospital Ulm, Ulm, Germany
| | - Elena M Bonke
- cBRAIN, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Ludwig-Maximilians-Universität, Munich, Germany
- Graduate School of Systemic Neurosciences, Ludwig-Maximilians-Universität, Munich, Germany
| | - Kosisochukwu E Umeasalugo
- cBRAIN, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Ludwig-Maximilians-Universität, Munich, Germany
- Graduate School of Systemic Neurosciences, Ludwig-Maximilians-Universität, Munich, Germany
- Institute for Stroke and Dementia Research, Ludwig-Maximilians-Universität, Munich, Germany
| | - Kristen R Sobolewski
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Center for Clinical Spectroscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research, Ludwig-Maximilians-Universität, Munich, Germany
- Munich Cluster for Systems Neurology (Synergy), Munich, Germany
| | - Martha E Shenton
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Psychiatry Neuroimaging Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Alexander P Lin
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Center for Clinical Spectroscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Psychiatry Neuroimaging Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Inga K Koerte
- cBRAIN, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Ludwig-Maximilians-Universität, Munich, Germany
- Psychiatry Neuroimaging Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
24
|
Ghosh KK, Padmanabhan P, Yang CT, Wang Z, Palanivel M, Ng KC, Lu J, Carlstedt-Duke J, Halldin C, Gulyás B. An In Vivo Study of a Rat Fluid-Percussion-Induced Traumatic Brain Injury Model with [ 11C]PBR28 and [ 18F]flumazenil PET Imaging. Int J Mol Sci 2021; 22:ijms22020951. [PMID: 33477960 PMCID: PMC7835883 DOI: 10.3390/ijms22020951] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 12/29/2022] Open
Abstract
Traumatic brain injury (TBI) modelled by lateral fluid percussion-induction (LFPI) in rats is a widely used experimental rodent model to explore and understand the underlying cellular and molecular alterations in the brain caused by TBI in humans. Current improvements in imaging with positron emission tomography (PET) have made it possible to map certain features of TBI-induced cellular and molecular changes equally in humans and animals. The PET imaging technique is an apt supplement to nanotheranostic-based treatment alternatives that are emerging to tackle TBI. The present study aims to investigate whether the two radioligands, [11C]PBR28 and [18F]flumazenil, are able to accurately quantify in vivo molecular-cellular changes in a rodent TBI-model for two different biochemical targets of the processes. In addition, it serves to observe any palpable variations associated with primary and secondary injury sites, and in the affected versus the contralateral hemispheres. As [11C]PBR28 is a radioligand of the 18 kD translocator protein, the up-regulation of which is coupled to the level of neuroinflammation in the brain, and [18F]flumazenil is a radioligand for GABAA-benzodiazepine receptors, whose level mirrors interneuronal activity and eventually cell death, the use of the two radioligands may reveal two critical features of TBI. An up-regulation in the [11C]PBR28 uptake triggered by the LFP in the injured (right) hemisphere was noted on day 14, while the uptake of [18F]flumazenil was down-regulated on day 14. When comparing the left (contralateral) and right (LFPI) hemispheres, the differences between the two in neuroinflammation were obvious. Our results demonstrate a potential way to measure the molecular alterations in a rodent-based TBI model using PET imaging with [11C]PBR28 and [18F]flumazenil. These radioligands are promising options that can be eventually used in exploring the complex in vivo pharmacokinetics and delivery mechanisms of nanoparticles in TBI treatment.
Collapse
Affiliation(s)
- Krishna Kanta Ghosh
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore; (K.K.G.); (C.-T.Y.); (Z.W.); (M.P.); (C.H.)
| | - Parasuraman Padmanabhan
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore; (K.K.G.); (C.-T.Y.); (Z.W.); (M.P.); (C.H.)
- Cognitive Neuroimaging Centre, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore
- Correspondence: (P.P.); (B.G.); Tel.:+65-69041186 (P.P.)
| | - Chang-Tong Yang
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore; (K.K.G.); (C.-T.Y.); (Z.W.); (M.P.); (C.H.)
- Department of Nuclear Medicine and Molecular Imaging, Radiological Sciences Division, Singapore General Hospital, Outram Road, Singapore 169608, Singapore
- Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Zhimin Wang
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore; (K.K.G.); (C.-T.Y.); (Z.W.); (M.P.); (C.H.)
| | - Mathangi Palanivel
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore; (K.K.G.); (C.-T.Y.); (Z.W.); (M.P.); (C.H.)
| | - Kian Chye Ng
- DSO National Laboratories (Kent Ridge), 27 Medical Drive, Singapore 117510, Singapore; (K.C.N.); (J.L.)
| | - Jia Lu
- DSO National Laboratories (Kent Ridge), 27 Medical Drive, Singapore 117510, Singapore; (K.C.N.); (J.L.)
| | - Jan Carlstedt-Duke
- President’s Office, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore;
| | - Christer Halldin
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore; (K.K.G.); (C.-T.Y.); (Z.W.); (M.P.); (C.H.)
- Department of Clinical Neuroscience, Karolinska Institute, S-171 76 Stockholm, Sweden
| | - Balázs Gulyás
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore; (K.K.G.); (C.-T.Y.); (Z.W.); (M.P.); (C.H.)
- Cognitive Neuroimaging Centre, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore
- Department of Clinical Neuroscience, Karolinska Institute, S-171 76 Stockholm, Sweden
- Correspondence: (P.P.); (B.G.); Tel.:+65-69041186 (P.P.)
| |
Collapse
|
25
|
miR-499-5p suppresses C-reactive protein and provides neuroprotection in hypoxic-ischemic encephalopathy in neonatal rat. Neurosci Res 2020; 161:44-50. [DOI: 10.1016/j.neures.2019.12.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 11/26/2019] [Accepted: 12/03/2019] [Indexed: 12/16/2022]
|
26
|
An update on the association between traumatic brain injury and Alzheimer's disease: Focus on Tau pathology and synaptic dysfunction. Neurosci Biobehav Rev 2020; 120:372-386. [PMID: 33171143 DOI: 10.1016/j.neubiorev.2020.10.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/09/2020] [Accepted: 10/19/2020] [Indexed: 02/06/2023]
Abstract
L.P. Li, J.W. Liang and H.J. Fu. An update on the association between traumatic brain injury and Alzheimer's disease: Focus on Tau pathology and synaptic dysfunction. NEUROSCI BIOBEHAV REVXXX-XXX,2020.-Traumatic brain injury (TBI) and Alzheimer's disease (AD) are devastating conditions that have long-term consequences on individual's cognitive functions. Although TBI has been considered a risk factor for the development of AD, the link between TBI and AD is still in debate. Aggregation of hyperphosphorylated tau and intercorrelated synaptic dysfunction, two key pathological elements in both TBI and AD, play a pivotal role in mediating neurodegeneration and cognitive deficits, providing a mechanistic link between these two diseases. In the first part of this review, we analyze the experimental literatures on tau pathology in various TBI models and review the distribution, biological features and mechanisms of tau pathology following TBI with implications in AD pathogenesis. In the second part, we review evidences of TBI-mediated structural and functional impairments in synapses, with a focus on the overlapped mechanisms underlying synaptic abnormalities in both TBI and AD. Finally, future perspectives are proposed for uncovering the complex relationship between TBI and neurodegeneration, and developing potential therapeutic avenues for alleviating cognitive deficits after TBI.
Collapse
|
27
|
Glycyrrhizin Blocks the Detrimental Effects of HMGB1 on Cortical Neurogenesis After Traumatic Neuronal Injury. Brain Sci 2020; 10:brainsci10100760. [PMID: 33096930 PMCID: PMC7593920 DOI: 10.3390/brainsci10100760] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 10/17/2020] [Accepted: 10/19/2020] [Indexed: 12/20/2022] Open
Abstract
Despite medical advances, neurological recovery after severe traumatic brain injury (TBI) remains poor. Elevated levels of high mobility group box protein-1 (HMGB1) are associated with poor outcomes; likely via interaction with receptors for advanced-glycation-end-products (RAGE). We examined the hypothesis that HMGB1 post-TBI is anti-neurogenic and whether this is pharmacologically reversible. Post-natal rat cortical mixed neuro-glial cell cultures were subjected to needle-scratch injury and examined for HMGB1-activation/neuroinflammation. HMGB1-related genes/networks were examined using genome-wide RNA-seq studies in cortical perilesional tissue samples from adult mice. Post-natal rat cortical neural stem/progenitor cell cultures were generated to quantify effects of injury-condition medium (ICM) on neurogenesis with/without RAGE antagonist glycyrrhizin. Needle-injury upregulated TNF-α/NOS-2 mRNA-expressions at 6 h, increased proportions of activated microglia, and caused neuronal loss at 24 h. Transcriptome analysis revealed activation of HMGB1 pathway genes/canonical pathways in vivo at 24 h. A 50% increase in HMGB1 protein expression, and nuclear-to-cytoplasmic translocation of HMGB1 in neurons and microglia at 24 h post-injury was demonstrated in vitro. ICM reduced total numbers/proportions of neuronal cells, but reversed by 0.5 μM glycyrrhizin. HMGB1 is activated following in vivo post mechanical injury, and glycyrrhizin alleviates detrimental effects of ICM on cortical neurogenesis. Our findings highlight glycyrrhizin as a potential therapeutic agent post-TBI.
Collapse
|
28
|
Makarevich O, Sabirzhanov B, Aubrecht TG, Glaser EP, Polster BM, Henry RJ, Faden AI, Stoica BA. Mithramycin selectively attenuates DNA-damage-induced neuronal cell death. Cell Death Dis 2020; 11:587. [PMID: 32719328 PMCID: PMC7385624 DOI: 10.1038/s41419-020-02774-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/06/2020] [Accepted: 07/10/2020] [Indexed: 12/15/2022]
Abstract
DNA damage triggers cell death mechanisms contributing to neuronal loss and cognitive decline in neurological disorders, including traumatic brain injury (TBI), and as a side effect of chemotherapy. Mithramycin, which competitively targets chromatin-binding sites of specificity protein 1 (Sp1), was used to examine previously unexplored neuronal cell death regulatory mechanisms via rat primary neurons in vitro and after TBI in mice (males). In primary neurons exposed to DNA-damage-inducing chemotherapy drugs in vitro we showed that DNA breaks sequentially initiate DNA-damage responses, including phosphorylation of ATM, H2AX and tumor protein 53 (p53), transcriptional activation of pro-apoptotic BH3-only proteins, and mitochondrial outer membrane permeabilization (MOMP), activating caspase-dependent and caspase-independent intrinsic apoptosis. Mithramycin was highly neuroprotective in DNA-damage-dependent neuronal cell death, inhibiting chemotherapeutic-induced cell death cascades downstream of ATM and p53 phosphorylation/activation but upstream of p53-induced expression of pro-apoptotic molecules. Mithramycin reduced neuronal upregulation of BH3-only proteins and mitochondrial dysfunction, attenuated caspase-3/7 activation and caspase substrates' cleavage, and limited c-Jun activation. Chromatin immunoprecipitation indicated that mithramycin attenuates Sp1 binding to pro-apoptotic gene promoters without altering p53 binding suggesting it acts by removing cofactors required for p53 transactivation. In contrast, the DNA-damage-independent neuronal death models displayed caspase initiation in the absence of p53/BH3 activation and were not protected even when mithramycin reduced caspase activation. Interestingly, experimental TBI triggers a multiplicity of neuronal death mechanisms. Although markers of DNA-damage/p53-dependent intrinsic apoptosis are detected acutely in the injured cortex and are attenuated by mithramycin, these processes may play a reduced role in early neuronal death after TBI, as caspase-dependent mechanisms are repressed in mature neurons while other, mithramycin-resistant mechanisms are active. Our data suggest that Sp1 is required for p53-mediated transactivation of neuronal pro-apoptotic molecules and that mithramycin may attenuate neuronal cell death in conditions predominantly involving DNA-damage-induced p53-dependent intrinsic apoptosis.
Collapse
Affiliation(s)
- Oleg Makarevich
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Boris Sabirzhanov
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Taryn G Aubrecht
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Ethan P Glaser
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Brian M Polster
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Rebecca J Henry
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Alan I Faden
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Bogdan A Stoica
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
29
|
Shorey M, Stone MC, Mandel J, Rolls MM. Neurons survive simultaneous injury to axons and dendrites and regrow both types of processes in vivo. Dev Biol 2020; 465:108-118. [PMID: 32687893 DOI: 10.1016/j.ydbio.2020.07.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 12/11/2022]
Abstract
Neurons extend dendrites and axons to receive and send signals. If either type of process is removed, the cell cannot function. Rather than undergoing cell death, some neurons can regrow axons and dendrites. Axon and dendrite regeneration have been examined separately and require sensing the injury and reinitiating the correct growth program. Whether neurons in vivo can sense and respond to simultaneous axon and dendrite injury with polarized regeneration has not been explored. To investigate the outcome of simultaneous axon and dendrite damage, we used a Drosophila model system in which neuronal polarity, axon regeneration, and dendrite regeneration have been characterized. After removal of the axon and all but one dendrite, the remaining dendrite was converted to a process that had a long unbranched region that extended over long distances and a region where shorter branched processes were added. These observations suggested axons and dendrites could regrow at the same time. To further test the capacity of neurons to implement polarized regeneration after axon and dendrite damage, we removed all neurites from mature neurons. In this case a long unbranched neurite and short branched neurites were regrown from the stripped cell body. Moreover, the long neurite had axonal plus-end-out microtubule polarity and the shorter neurites had mixed polarity consistent with dendrite identity. The long process also accumulated endoplasmic reticulum at its tip like regenerating axons. We conclude that neurons in vivo can respond to simultaneous axon and dendrite injury by initiating growth of a new axon and new dendrites.
Collapse
Affiliation(s)
- Matthew Shorey
- Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Michelle C Stone
- Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Jenna Mandel
- Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Melissa M Rolls
- Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
30
|
Zhu Z, Chuckowree JA, Musgrove R, Dickson TC, Blizzard CA. The pathologic outcomes and efficacy of epothilone treatment following traumatic brain injury is determined by age. Neurobiol Aging 2020; 93:85-96. [PMID: 32480164 DOI: 10.1016/j.neurobiolaging.2020.03.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/27/2020] [Accepted: 03/28/2020] [Indexed: 02/05/2023]
Abstract
Traumatic brain injury (TBI) can affect individuals at any age, with the potential of causing lasting neurologic consequences. The lack of effective therapeutic solutions and recommendations for patients that acquire a TBI can be attributed, at least in part, to an inability to confidently predict long-term outcomes following TBI, and how the response of the brain differs across the life span. The purpose of this study was to determine how age specifically affects TBI outcomes in a preclinical model. Male Thy1-YFPH mice, that express yellow fluorescent protein in the cytosol of a subset of Layer V pyramidal neurons in the neocortex, were subjected to a lateral fluid percussion injury over the right parietal cortex at distinct time points throughout the life span (1.5, 3, and 12 months of age). We found that the degree of neuronal injury, astrogliosis, and microglial activation differed depending on the age of the animal when the injury occurred. Furthermore, age affected the initial injury response and how it resolved over time. Using the microtubule stabilizing agent Epothilone D, to potentially protect against these pathologic outcomes, we found that the neuronal response was different depending on age. This study clearly shows that age must be taken into account in neurologic studies and preclinical trials involving TBI, and that future therapeutic interventions must be tailored to age.
Collapse
Affiliation(s)
- Zhendan Zhu
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Jyoti A Chuckowree
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Ruth Musgrove
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Tracey C Dickson
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Catherine A Blizzard
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia.
| |
Collapse
|
31
|
Nye DMR, Albertson RM, Weiner AT, Hertzler JI, Shorey M, Goberdhan DCI, Wilson C, Janes KA, Rolls MM. The receptor tyrosine kinase Ror is required for dendrite regeneration in Drosophila neurons. PLoS Biol 2020; 18:e3000657. [PMID: 32163406 PMCID: PMC7067388 DOI: 10.1371/journal.pbio.3000657] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 02/07/2020] [Indexed: 12/13/2022] Open
Abstract
While many regulators of axon regeneration have been identified, very little is known about mechanisms that allow dendrites to regenerate after injury. Using a Drosophila model of dendrite regeneration, we performed a candidate screen of receptor tyrosine kinases (RTKs) and found a requirement for RTK-like orphan receptor (Ror). We confirmed that Ror was required for regeneration in two different neuron types using RNA interference (RNAi) and mutants. Ror was not required for axon regeneration or normal dendrite development, suggesting a specific role in dendrite regeneration. Ror can act as a Wnt coreceptor with frizzleds (fzs) in other contexts, so we tested the involvement of Wnt signaling proteins in dendrite regeneration. We found that knockdown of fz, dishevelled (dsh), Axin, and gilgamesh (gish) also reduced dendrite regeneration. Moreover, Ror was required to position dsh and Axin in dendrites. We recently found that Wnt signaling proteins, including dsh and Axin, localize microtubule nucleation machinery in dendrites. We therefore hypothesized that Ror may act by regulating microtubule nucleation at baseline and during dendrite regeneration. Consistent with this hypothesis, localization of the core nucleation protein γTubulin was reduced in Ror RNAi neurons, and this effect was strongest during dendrite regeneration. In addition, dendrite regeneration was sensitive to partial reduction of γTubulin. We conclude that Ror promotes dendrite regeneration as part of a Wnt signaling pathway that regulates dendritic microtubule nucleation.
Collapse
Affiliation(s)
- Derek M. R. Nye
- Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- MSTP Program, Milton S. Hershey College of Medicine, Hershey, Pennsylvania, United States of America
| | - Richard M. Albertson
- Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- MSTP Program, Milton S. Hershey College of Medicine, Hershey, Pennsylvania, United States of America
| | - Alexis T. Weiner
- Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - J. Ian Hertzler
- Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Matthew Shorey
- Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | | | - Clive Wilson
- Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Kevin A. Janes
- Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Melissa M. Rolls
- Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
32
|
Eyolfson E, Yamakawa GR, Griep Y, Collins R, Carr T, Wang M, Lohman AW, Mychasiuk R. Examining the Progressive Behavior and Neuropathological Outcomes Associated with Chronic Repetitive Mild Traumatic Brain Injury in Rats. Cereb Cortex Commun 2020; 1:tgaa002. [PMID: 34296084 PMCID: PMC8152839 DOI: 10.1093/texcom/tgaa002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/24/2020] [Accepted: 01/30/2020] [Indexed: 02/07/2023] Open
Abstract
While the physical and behavioral symptomologies associated with a single mild traumatic brain injury (mTBI) are typically transient, repetitive mTBIs (RmTBI) have been associated with persisting neurological deficits. Therefore, this study examined the progressive changes in behavior and the neuropathological outcomes associated with chronic RmTBI through adolescence and adulthood in male and female Sprague Dawley rats. Rats experienced 2 mTBIs/week for 15 weeks and were periodically tested for changes in motor behavior, cognitive function, emotional disturbances, and aggression. Brain tissue was examined for neuropathological changes in ventricle size and presentation of Iba1 and GFAP. We did not see progressively worse behavioral impairments with the accumulation of injuries or time, but did find evidence for neurological and functional change (motor disturbance, reduced exploration, reduced aggression, alteration in depressive-like behavior, deficits in short-term working memory). Neuropathological assessment of RmTBI animals identified an increase in ventricle size, prolonged changes in GFAP, and sex differences in Iba1, in the corpus callosum, thalamus, and medial prefrontal cortex. Telomere length reduced exponentially as the injury load increased. Overall, chronic RmTBI did not result in accumulating behavioral impairment, and there is a need to further investigate progressive behavioral changes associated with repeated injuries in adolescence and young adulthood.
Collapse
Affiliation(s)
- Eric Eyolfson
- Department of Psychology, Alberta Children’s Hospital Research Institute, The University of Calgary, Calgary, AB, T2N 1N4, Canada
- Department of Psychology, Hotchkiss Brain Institute, The University of Calgary, Calgary, AB, T2N 1N4, Canada
| | - Glenn R Yamakawa
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Yannick Griep
- Department of Psychology, Alberta Children’s Hospital Research Institute, The University of Calgary, Calgary, AB, T2N 1N4, Canada
- Department of Psychology, Hotchkiss Brain Institute, The University of Calgary, Calgary, AB, T2N 1N4, Canada
- Division of Epidemiology, Stress Research Institute, Stockholm University, 106 91 Stockholm, Sweden
- Behavioral Science Institute, Radbound University, 9104, 6500 HE, Nijmegen, The Netherlands
| | - Reid Collins
- Department of Psychology, Alberta Children’s Hospital Research Institute, The University of Calgary, Calgary, AB, T2N 1N4, Canada
- Department of Psychology, Hotchkiss Brain Institute, The University of Calgary, Calgary, AB, T2N 1N4, Canada
| | - Thomas Carr
- Department of Cell Biology and Anatomy, Alberta Children’s Hospital Research Institute, The University of Calgary, Calgary, AB, T2N 1N4, Canada
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, The University of Calgary, Calgary, AB, T2N 1N4, Canada
| | - Melinda Wang
- Department of Psychology, Alberta Children’s Hospital Research Institute, The University of Calgary, Calgary, AB, T2N 1N4, Canada
- Department of Psychology, Hotchkiss Brain Institute, The University of Calgary, Calgary, AB, T2N 1N4, Canada
| | - Alexander W Lohman
- Department of Cell Biology and Anatomy, Alberta Children’s Hospital Research Institute, The University of Calgary, Calgary, AB, T2N 1N4, Canada
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, The University of Calgary, Calgary, AB, T2N 1N4, Canada
| | - Richelle Mychasiuk
- Department of Psychology, Alberta Children’s Hospital Research Institute, The University of Calgary, Calgary, AB, T2N 1N4, Canada
- Department of Psychology, Hotchkiss Brain Institute, The University of Calgary, Calgary, AB, T2N 1N4, Canada
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| |
Collapse
|
33
|
Effect of mild blast-induced TBI on dendritic architecture of the cortex and hippocampus in the mouse. Sci Rep 2020; 10:2206. [PMID: 32042033 PMCID: PMC7010659 DOI: 10.1038/s41598-020-59252-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 01/20/2020] [Indexed: 11/09/2022] Open
Abstract
Traumatic brain injury (TBI) has been designated as a signature injury of modern military conflicts. Blast trauma, in particular, has come to make up a significant portion of the TBIs which are sustained in warzones. Though most TBIs are mild, even mild TBI can induce long term effects, including cognitive and memory deficits. In our study, we utilized a mouse model of mild blast-related TBI (bTBI) to investigate TBI-induced changes within the cortex and hippocampus. We performed rapid Golgi staining on the layer IV and V pyramidal neurons of the parietal cortex and the CA1 basilar tree of the hippocampus and quantified dendritic branching and distribution. We found decreased dendritic branching within both the cortex and hippocampus in injured mice. Within parietal cortex, this decreased branching was most evident within the middle region, while outer and inner regions resembled that of control mice. This study provides important knowledge in the study of how the shockwave associated with a blast explosion impacts different brain regions.
Collapse
|
34
|
Mulherkar S, Tolias KF. RhoA-ROCK Signaling as a Therapeutic Target in Traumatic Brain Injury. Cells 2020; 9:E245. [PMID: 31963704 PMCID: PMC7016605 DOI: 10.3390/cells9010245] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/09/2020] [Accepted: 01/16/2020] [Indexed: 12/19/2022] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability worldwide. TBIs, which range in severity from mild to severe, occur when a traumatic event, such as a fall, a traffic accident, or a blow, causes the brain to move rapidly within the skull, resulting in damage. Long-term consequences of TBI can include motor and cognitive deficits and emotional disturbances that result in a reduced quality of life and work productivity. Recovery from TBI can be challenging due to a lack of effective treatment options for repairing TBI-induced neural damage and alleviating functional impairments. Central nervous system (CNS) injury and disease are known to induce the activation of the small GTPase RhoA and its downstream effector Rho kinase (ROCK). Activation of this signaling pathway promotes cell death and the retraction and loss of neural processes and synapses, which mediate information flow and storage in the brain. Thus, inhibiting RhoA-ROCK signaling has emerged as a promising approach for treating CNS disorders. In this review, we discuss targeting the RhoA-ROCK pathway as a therapeutic strategy for treating TBI and summarize the recent advances in the development of RhoA-ROCK inhibitors.
Collapse
Affiliation(s)
- Shalaka Mulherkar
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Kimberley F. Tolias
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA;
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
35
|
Lv X, Tang R, Gao Z, Hu D, Li G, Lang Y, He J. Activation of the primary motor cortex using fully-implanted electrical sciatic nerve stimulation. Exp Ther Med 2019; 18:3357-3364. [PMID: 31602209 PMCID: PMC6777333 DOI: 10.3892/etm.2019.7993] [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: 03/06/2019] [Accepted: 07/12/2019] [Indexed: 11/30/2022] Open
Abstract
Functional degradation of the motor cortex usually results from brain injury, stroke, limb amputation, aging or other diseases. Currently, there are no ideal means of treatment, other than medication and sports rehabilitation. The present study investigated whether electrical stimulation of the sciatic nerve can activate the motor-related area of the brain. The study is based on a self-developed fully implantable nerve electrical stimulator and a self-developed multi-channel electroencephalogram (EEG) electrode array. The sciatic nerves of Sprague-Dawley rats (sorted into old and young groups) were stimulated by the electrical stimulator under anesthesia, and the EEG signal was recorded simultaneously. The relationship between sciatic nerve stimulation and brain activity was analyzed. The results showed that when the sciatic nerve was stimulated by the implanted electrical stimulator, motor-related channels were activated, causing contraction of the left leg. It was found that at the frequency band of 8-16 Hz, the EEG signal in the right motor area was higher than at other frequency bands. This phenomenon was identical in both young and old rats. The results indicated that electrical stimulation of the sciatic nerve can activate the motor region of the rat brain, and provided evidence that stimulation of the sciatic nerve could be a method of preventing motor cortex degeneration.
Collapse
Affiliation(s)
- Xiaodong Lv
- Beijing Advanced Innovation Center for Intelligent Robot and System, Beijing Institute of Technology, Beijing 100081, P.R. China
| | - Rongyu Tang
- Beijing Advanced Innovation Center for Intelligent Robot and System, Beijing Institute of Technology, Beijing 100081, P.R. China
| | - Zhaolong Gao
- Neural Interface and Rehabilitation Technology Research Center, School of Automation, Huazhong University of Science and Technology, Wuhan, Hubei 430074, P.R. China
| | - Dingyin Hu
- Beijing Advanced Innovation Center for Intelligent Robot and System, Beijing Institute of Technology, Beijing 100081, P.R. China
| | - Guanghui Li
- Beijing Advanced Innovation Center for Intelligent Robot and System, Beijing Institute of Technology, Beijing 100081, P.R. China
| | - Yiran Lang
- Beijing Advanced Innovation Center for Intelligent Robot and System, Beijing Institute of Technology, Beijing 100081, P.R. China
| | - Jiping He
- Beijing Advanced Innovation Center for Intelligent Robot and System, Beijing Institute of Technology, Beijing 100081, P.R. China
| |
Collapse
|
36
|
Temporal Sequences of Synapse Disintegration Triggered by Afferent Axon Transection, Time-Lapse Imaging Study of Presynaptic and Postsynaptic Molecules. eNeuro 2019; 6:ENEURO.0459-18.2019. [PMID: 31515235 PMCID: PMC6785539 DOI: 10.1523/eneuro.0459-18.2019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 07/22/2019] [Accepted: 07/30/2019] [Indexed: 11/22/2022] Open
Abstract
Traumatic brain injury (TBI) is one of the major causes of death and disability. Multiple animal models have been developed to explore therapeutic targets for TBI. However, heterogeneity of pathophysiology obstructs discovery of therapeutic targets. To facilitate understanding of TBI pathophysiology, each element of neuronal and glial responses should be studied separately. We focused on synapse remodeling which plays an important role in recovery from TBI and developed a new method, afferent elimination, for analyzing synapse remodeling after selective damage to presynaptic axons by mechanical transection in culture of mouse hippocampal neurons. Afferent elimination can induce various events related to synapse remodeling and we could determine their temporal orders and find relationships between them. Specifically, loss of presynaptic sites preceded loss of postsynaptic sites and spines. Some of the postsynaptic sites initially located inside spines showed translocation toward dendritic shafts. These translocation events started after the loss of contacting presynaptic sites. Also, these events could be blocked or delayed by NMDA receptor inhibition. Taken together, these findings suggest that postsynaptic changes occur in afferent elimination are NMDA dependent and imply that these NMDA-dependent events underlie synaptic remodeling of TBI.
Collapse
|
37
|
MMP-9 Contributes to Dendritic Spine Remodeling Following Traumatic Brain Injury. Neural Plast 2019; 2019:3259295. [PMID: 31198417 PMCID: PMC6526556 DOI: 10.1155/2019/3259295] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 04/03/2019] [Indexed: 01/30/2023] Open
Abstract
Traumatic brain injury (TBI) occurs when a blow to the head causes brain damage. Apart from physical trauma, it causes a wide range of cognitive, behavioral, and emotional deficits including impairments in learning and memory. On neuronal level, TBI may lead to circuitry remodeling and in effect imbalance between excitatory and inhibitory neurotransmissions. Such change in brain homeostasis may often lead to brain disorders. The basic units of neuronal connectivity are dendritic spines that are tiny protrusions forming synapses between two cells in a network. Spines are dynamic structures that undergo morphological transformation throughout life. Their shape is strictly related to an on/off state of synapse and the strength of synaptic transmission. Matrix metalloproteinase-9 (MMP-9) is an extrasynaptically operating enzyme that plays a role in spine remodeling and has been reported to be activated upon TBI. The aim of the present study was to evaluate the influence of MMP-9 on dendritic spine density and morphology following controlled cortical impact (CCI) as animal model of TBI. We examined spine density and dendritic spine shape in the cerebral cortex and the hippocampus. CCI caused a marked decrease in spine density as well as spine shrinkage in the cerebral cortex ipsilateral to the injury, when compared to sham animals and contralateral side both 1 day and 1 week after the insult. Decreased spine density was also observed in the dentate gyrus of the hippocampus; however, in contrast to the cerebral cortex, spines in the DG became more filopodia-like. In mice lacking MMP-9, no effects of TBI on spine density and morphology were observed.
Collapse
|
38
|
Adamson SXF, Shen X, Jiang W, Lai V, Wang X, Shannahan JH, Cannon JR, Chen J, Zheng W. Subchronic Manganese Exposure Impairs Neurogenesis in the Adult Rat Hippocampus. Toxicol Sci 2019; 163:592-608. [PMID: 29579278 DOI: 10.1093/toxsci/kfy062] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Adult neurogenesis takes place in the brain subventricular zone (SVZ) in the lateral walls of lateral ventricles and subgranular zone (SGZ) in the hippocampal dentate gyrus (HDG), and functions to supply newborn neurons for normal brain functionality. Subchronic Mn exposure is known to disrupt adult neurogenesis in the SVZ. This study was designed to determine whether Mn exposure disturbed neurogenesis within the adult HDG. Adult rats (10 weeks old) received a single dose of bromodeoxyuridine (BrdU) at the end of 4-week Mn exposure to label the proliferating cells. Immunostaining and cell counting data showed that BrdU(+) cells in Mn-exposed HDG were about 37% lower than that in the control (p < .05). The majority of BrdU(+) cells were identified as Sox2(+) cells. Another set of adult rats received BrdU injections for 3 consecutive days followed by 2- or 4-week Mn exposure to trace the fate of BrdU-labeled cells in the HDG. The time course studies indicated that Mn exposure significantly reduced the survival rate (54% at 2 weeks and 33% at 4 weeks), as compared with that in the control (80% at 2 weeks and 51% at 4 weeks) (p < .01). A significant time-dependent migration of newborn cells from the SGZ toward the granule cell layer was also observed in both control and Mn-exposed HDG. Triple-stained neuroblasts and mature neurons further revealed that Mn exposure significantly inhibited the differentiation of immature neuroblasts into mature neurons in the HDG. Taken together, these observations suggest that subchronic Mn exposure results in a reduced cell proliferation, diminished survival of adult-born neurons, and inhibited overall neurogenesis in the adult HDG. Impaired adult neurogenesis is likely one of the mechanisms contribute to Mn-induced Parkinsonian disorder.
Collapse
Affiliation(s)
| | | | | | | | - Xiaoting Wang
- Spinal Cord and Brain Injury Research Group, Stark Neuroscience Research Institute.,Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202
| | | | - Jason R Cannon
- School of Health Sciences.,Purdue Institute for Integrative Neurosciences, Purdue University, West Lafayette, IN 47907
| | - Jinhui Chen
- Spinal Cord and Brain Injury Research Group, Stark Neuroscience Research Institute.,Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Wei Zheng
- School of Health Sciences.,Purdue Institute for Integrative Neurosciences, Purdue University, West Lafayette, IN 47907
| |
Collapse
|
39
|
Arulsamy A, Corrigan F, Collins-Praino LE. Age, but not severity of injury, mediates decline in executive function: Validation of the rodent touchscreen paradigm for preclinical models of traumatic brain injury. Behav Brain Res 2019; 368:111912. [PMID: 30998995 DOI: 10.1016/j.bbr.2019.111912] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/12/2019] [Accepted: 04/13/2019] [Indexed: 12/14/2022]
Abstract
Increasingly, it is being recognised that traumatic brain injury (TBI) is not just an acute event but instead results in ongoing neuronal injury that may lead to chronic impairments in multiple cognitive domains. Of these, deficits in executive function are one of the more common changes reported following TBI, and are a major predictor of well-being, social function and quality of life in individuals with a history of TBI. In order to fully understand the relationship between TBI and executive dysfunction, including brain mechanisms that may account for this, experimental models are clearly needed. However, to date, there have been a lack of preclinical studies systematically comparing the effect of injury severity on executive function, particularly at long-term timepoints post-injury. Furthermore, many previous studies have not used behavioural measures that are sensitive to the full range of executive function impairments that may manifest after injury, particularly in models of diffuse axonal injury (Lv et al.). The current study aimed to investigate the temporal profile, up to 12 months post-injury, of the evolution of executive dysfunction following different severities of injury in an experimental model of DAI. In order to do so, we utilised a rodent touchscreen paradigm to administer the 5 Choice- Continuous Performance Task (5C-CPT), an extension of the 5-choice serial reaction time task (5CSRT). Interestingly, there were no differences in learning, motivation, attention, response time or impulsivity at 1 month, 6 months or 12 months post-injury in any of the TBI groups compared to sham, regardless of the initial severity of the injury. Instead, most of the effects on executive function seen at the 12 month timepoint appeared to be a result of ageing, not injury. As even the 12-month timepoint represents middle age in the rat, future studies will be needed to further probe these effects, in order to determine whether DAI may influence the presentation of executive dysfunction in older age.
Collapse
Affiliation(s)
- Alina Arulsamy
- Cognition, Ageing and Neurodegenerative Disease Lab, Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005 Australia
| | | | - Lyndsey E Collins-Praino
- Cognition, Ageing and Neurodegenerative Disease Lab, Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005 Australia.
| |
Collapse
|
40
|
Spader HS, Dean DC, LaFrance WC, Raukar NP, Cosgrove GR, Eyerly-Webb SA, Ellermeier A, Correia S, Deoni SCL, Rogg J. Prospective study of myelin water fraction changes after mild traumatic brain injury in collegiate contact sports. J Neurosurg 2019; 130:1321-1329. [PMID: 29712487 PMCID: PMC6541528 DOI: 10.3171/2017.12.jns171597] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 12/05/2017] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Mild traumatic brain injury (mTBI) in athletes, including concussion, is increasingly being found to have long-term sequelae. Current imaging techniques have not been able to identify early damage caused by mTBI that is predictive of long-term symptoms or chronic traumatic encephalopathy. In this preliminary feasibility study, the authors investigated the use of an emerging magnetic resonance imaging (MRI) technique, multicomponent driven equilibrium single pulse observation of T1 and T2 (mcDESPOT), in visualizing acute and chronic white matter changes after mTBI in collegiate football and rugby players. METHODS This study was a nonrandomized, nonblinded prospective trial designed to quantify changes in the myelin water fraction (MWF), used as a surrogate MRI measure of myelin content, in a group of male collegiate football and rugby players, classified here as a contact sport player (CSP) cohort, at the time of mTBI diagnosis and 3 months after injury when the acute symptoms of the injury had resolved. In addition, differences in the MWF between the CSP cohort and a control cohort of noncontact sport players (NCSPs) were quantified. T-tests and a threshold-free cluster enhancement (TFCE) statistical analysis technique were used to identify brain structures with significant changes in the MWF between the CSP and NCSP cohorts and between immediately postinjury and follow-up images obtained in the CSP cohort. RESULTS Brain MR images of 12 right-handed male CSPs were analyzed and compared with brain images of 10 right-handed male NCSPs from the same institution. A comparison of CSP and NCSP baseline images using TFCE showed significantly higher MWFs in the bilateral basal ganglia, anterior and posterior corpora callosa, left corticospinal tract, and left anterior and superior temporal lobe (p < 0.05). At the 3-month follow-up examination, images from the CSP cohort still showed significantly higher MWFs than those identified on baseline images from the NCSP cohort in the bilateral basal ganglia, anterior and posterior corpora callosa, and left anterior temporal lobe, and also in the bilateral corticospinal tracts, parahippocampal gyrus, and bilateral juxtapositional (previously known as supplemental motor) areas (p < 0.05). In the CSP cohort, a t-test comparing the MWF at the time of injury and 3 months later showed a significant increase in the overall MWF at follow-up (p < 0.005). These increases were greatest in the bilateral basal ganglia and deep white matter. MWF decreases were seen in more superficial white matter (p < 0.005). CONCLUSIONS In this preliminary study, MWF was found to be increased in the brains of CSPs compared with the brains of controls, suggesting acute/chronic MWF alterations in CSPs from previous injuries. Increases in the MWF were also demonstrated in the brains of CSPs 3 months after the players sustained an mTBI. The full clinical significance of an increased MWF and whether this reflects axon neuropathology or disorderly remyelination leading to hypermyelination has yet to be determined.
Collapse
Affiliation(s)
- Heather S Spader
- 1Division of Pediatric Neurosurgery, Joe DiMaggio Children's Hospital, and
| | - Douglas C Dean
- 2Waisman Center, University of Wisconsin-Madison, Wisconsin
| | - W Curt LaFrance
- 3Division of Neuropsychiatry and Behavioral Neurology
- 4Department of Psychiatry and Human Behavior
- 5Department of Neurology, and
| | | | - G Rees Cosgrove
- 10Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | | | - Stephen Correia
- 4Department of Psychiatry and Human Behavior
- 9Providence VA Medical Center, Providence; and
| | - Sean C L Deoni
- 11Advanced Baby Imaging Lab, School of Engineering, Brown University; and
- 12Department of Pediatrics, Memorial Hospital of Rhode Island, Pawtucket, Rhode Island; and
| | - Jeffrey Rogg
- 7Department of Diagnostic Imaging, Rhode Island Hospital
| |
Collapse
|
41
|
Cortical thinning in military blast compared to non-blast persistent mild traumatic brain injuries. NEUROIMAGE-CLINICAL 2019; 22:101793. [PMID: 30939340 PMCID: PMC6446073 DOI: 10.1016/j.nicl.2019.101793] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 01/28/2019] [Accepted: 03/24/2019] [Indexed: 01/07/2023]
Abstract
In the military, explosive blasts are a significant cause of mild traumatic brain injuries (mTBIs). The symptoms associated with blast mTBIs causes significant economic burdens and a diminished quality of life for many service members. At present, the distinction of the injury mechanism (blast versus non-blast) may not influence TBI diagnosis. However, using noninvasive imaging, this study reveals significant distinctions between the blast and non-blast TBI mechanisms. A cortical whole-brain thickness analysis was performed using structural high-resolution T1-weighted MRI to identify the effects of blasts in persistent mTBI (pmTBI) subjects. A total of 41 blast pmTBI subjects were individually age- and gender-matched to 41 non-blast pmTBI subjects. Using FreeSurfer, cortical thickness was quantified for the blast group, relative to the non-blast group. Cortical thinning was identified within the blast mTBI group, in two clusters bilaterally. In the left hemisphere, the cluster overlapped with the lateral orbitofrontal, rostral middle frontal, medial orbitofrontal, superior frontal, rostral anterior cingulate and frontal pole cortices (p < 0.02, two-tailed, size = 1680 mm2). In the right hemisphere, the cluster overlapped with the lateral orbitofrontal, rostral middle frontal, medial orbitofrontal, pars orbitalis, pars triangularis and insula cortices (p < 0.002, two-tailed, cluster size = 2453 mm2). Self-report assessments suggest significant differences in the Post-Traumatic Stress Disorder Checklist-Civilian Version (p < 0.05, Bonferroni-corrected) and the Neurobehavioral Symptom Inventory (p < 0.01, uncorrected) between the blast and non-blast mTBI groups. These results suggest that blast may cause a unique injury pattern related to a reduction in cortical thickness within specific brain regions which could affect symptoms. No other study has found cortical thickness difference between blast and non-blast mTBI groups and further replication is needed to confirm these initial observations.
Collapse
|
42
|
Parent M, Li Y, Santhakumar V, Hyder F, Sanganahalli BG, Kannurpatti SS. Alterations of Parenchymal Microstructure, Neuronal Connectivity, and Cerebrovascular Resistance at Adolescence after Mild-to-Moderate Traumatic Brain Injury in Early Development. J Neurotrauma 2019; 36:601-608. [PMID: 29855211 PMCID: PMC6354598 DOI: 10.1089/neu.2018.5741] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of morbidity in children. To investigate outcome of early developmental TBI during adolescence, a rat model of fluid percussion injury was developed, where previous work reported deficits in sensorimotor behavior and cortical blood flow at adolescence.1 Based on the nonlocalized outcome, we hypothesized that multiple neurophysiological components of brain function, namely neuronal connectivity, synapse/axonal microstructural integrity, and neurovascular function, are altered and magnetic resonance imaging (MRI) methods could be used to determine regional alterations. Adolescent outcomes of developmental TBI were studied 2 months after injury, using functional MRI (fMRI) and diffusion tensor imaging (DTI). fMRI-based resting-state functional connectivity (RSFC), representing neural connectivity, was significantly altered between sham and TBI. RSFC strength decreased in the cortex, hippocampus, and thalamus, accompanied by decrease in spatial extent of their corresponding RSFC networks and interhemispheric asymmetry. Cerebrovascular reactivity to arterial CO2 changes diminished after TBI across both hemispheres, with a more pronounced decrease in the ipsilateral hippocampus, thalamus, and motor cortex. DTI measures of fractional anisotropy and apparent diffusion coefficient, reporting on axonal and microstructural integrity of the brain, indicated similar interhemispheric asymmetry, with highest change in the ipsilateral hippocampus and regions adjoining the ipsilateral thalamus, hypothalamus, and amygdala. TBI-induced corpus callosal microstructural alterations indicated measurable changes in interhemispheric structural connectivity. Hippocampus, thalamus, and select cortical regions were most consistently affected in multiple imaging markers. The multi-modal MRI results demonstrate cortical and subcortical alterations in neural connectivity, cerebrovascular resistance, and parenchymal microstructure in the adolescent brain, indicating the highly diffuse and persistent nature of the lateral fluid percussion TBI early in development.
Collapse
Affiliation(s)
- Maxime Parent
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | - Ying Li
- Department of Pharmacology, Physiology and Neuroscience, Rutgers Biomedical and Health Sciences–New Jersey Medical School, Newark, New Jersey
| | - Vijayalakshmi Santhakumar
- Department of Pharmacology, Physiology and Neuroscience, Rutgers Biomedical and Health Sciences–New Jersey Medical School, Newark, New Jersey
- Department of Molecular, Cell and Systems Neuroscience, University of California at Riverside, Riverside, California
| | - Fahmeed Hyder
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Basavaraju G. Sanganahalli
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | - Sridhar S. Kannurpatti
- Department of Radiology, Rutgers Biomedical and Health Sciences–New Jersey Medical School, Newark, New Jersey
| |
Collapse
|
43
|
Neuroanatomical and functional alterations of insula in mild traumatic brain injury patients at the acute stage. Brain Imaging Behav 2019; 14:907-916. [PMID: 30734204 DOI: 10.1007/s11682-019-00053-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cognitive impairment is a major cause of disability and decline in quality of life in mild traumatic brain injury (mTBI) survivors, but the underlying pathophysiology is still poorly understood. The insula has extensive connections to other cortex and is believed to responsible for integrating external and internal processes and controlling cognitive functions. To explore this hypothesis, we investigated early alterations in the gray matter volume (GMV) and brain functional connectivity (FC) of insula in mTBI patients within 7 days after injury and any possible correlations with cognitive function. A total of 58 mTBI patients at the acute stage and 32 matched healthy controls were recruited and underwentT1-weighted magnetic resonance imaging (MRI)andresting-state functional MRI scans within 7 days of injury. FC was characterized using seed-based region of interest analysis method. The patients' cognitive function was evaluated with Montreal Cognitive Assessment (MoCA) score. The resulting of GMV and FC of insula were correlated with cognitive alterations. We found that the GMV was significantly reduced only in the right insula in mTBI patients and no significant GMV increase was observed in either hemisphere. mTBI patients demonstrated decreased FC in the right parahippocampal gyrus and increased FC in the right supramargianl gyrus. In addition, compared to the healthy controls, the mTBI patients in the acute stage presented a decline in the visuospatial/executive (p = 0.013) and attention (p = 0.038) subcategories. In the mTBI group, the changes in GMV in the right insula were positively correlated with poor attention performance (r = 0.316, p = 0.016). Our data demonstrated alterations of the GMV and resting-stateFC of the right insula in mTBI patients at the acute stage. These early changes in GMV and resting-state FC perhaps serve as a potential biomarker for improving the understanding of cognitive decline for mTBI in the acute setting.
Collapse
|
44
|
Abstract
The brain undergoes several changes at structural, molecular, and cellular levels leading to alteration in its functions and these processes are primarily maintained by proteostasis in cells. However, an imbalance in proteostasis due to the abnormal accumulation of protein aggregates induces endoplasmic reticulum (ER) stress. This event, in turn, activate the unfolded protein response; however, in most neurodegenerative conditions and brain injury, an uncontrolled unfolded protein response elicits memory dysfunction. Although the underlying signaling mechanism for impairment of memory function following induction of ER stress remains elusive, recent studies have highlighted that inactivation of a transcription factor, CREB, which is essential for synaptic function and memory formation, plays an essential role for ER stress-induced synaptic and memory dysfunction. In this review, current studies and most updated view on how ER stress affects memory function in both physiological and pathological conditions will be highlighted.
Collapse
Affiliation(s)
- Nilkantha Sen
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
45
|
Zhang Y, Chopp M, Gang Zhang Z, Zhang Y, Zhang L, Lu M, Zhang T, Winter S, Brandstätter H, Mahmood A, Xiong Y. Prospective, randomized, blinded, and placebo-controlled study of Cerebrolysin dose-response effects on long-term functional outcomes in a rat model of mild traumatic brain injury. J Neurosurg 2018; 129:1295-1304. [PMID: 29303438 DOI: 10.3171/2017.6.jns171007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 06/20/2017] [Indexed: 01/30/2023]
Abstract
Using a prospective, randomized, blinded, placebo-controlled protocol, the authors demonstrated that Cerebrolysin at doses of 0.8-7.5 ml/kg, administered 4 hours after injury and then once daily for a total of 10 consecutive days, improves long-term functional outcomes in a rat model of mild closed head injury; a 2.5-ml/kg dose was identified as optimal. These findings suggest that Cerebrolysin has the potential to treat mild traumatic brain injury, the incidence of which is high without effective treatments.
Collapse
Affiliation(s)
| | - Michael Chopp
- 2Neurology, and
- 3Department of Physics, Oakland University, Rochester, Michigan; and
| | | | | | | | - Mei Lu
- 4Biostatistics and Research Epidemiology, Henry Ford Hospital, Detroit, Michigan
| | - Talan Zhang
- 4Biostatistics and Research Epidemiology, Henry Ford Hospital, Detroit, Michigan
| | | | | | | | | |
Collapse
|
46
|
Moquin A, Ji J, Neibert K, Winnik F, Maysinger D. Encapsulation and Delivery of Neutrophic Proteins and Hydrophobic Agents Using PMOXA-PDMS-PMOXA Triblock Polymersomes. ACS OMEGA 2018; 3:13882-13893. [PMID: 30411053 PMCID: PMC6217674 DOI: 10.1021/acsomega.8b02311] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 10/11/2018] [Indexed: 06/08/2023]
Abstract
Polymersomes are attractive nanocarriers for hydrophilic and lipophilic drugs; they are more stable than liposomes, tunable, and relatively easy to prepare. The copolymer composition and molar mass are critical features that determine the physicochemical properties of the polymersomes including the rate of drug release. We used the triblock-copolymer, poly(2-methyl-2-oxazoline)-block-poly-(dimethysiloxane)-block-poly(2-methyl-2-oxazoline) (PMOXA-PDMS-PMOXA), to form amphipathic polymersomes capable of loading proteins and small hydrophobic agents. The selected agents were unstable neurotrophins (nerve growth factor and brain-derived neurotrophic factor), a large protein CD109, and the fluorescent drug curcumin. We prepared, characterized, and tested polymersomes loaded with selected agents in 2D and 3D biological models. Curcumin-loaded and rhodamine-bound PMOXA-PDMS-PMOXA polymersomes were used to visualize them inside cells. N-Methyl-d-aspartate receptor (NMDAR) agonists and antagonists were also covalently attached to the surface of polymersomes for targeting neurons. Labeled and unlabeled polymersomes with or without loaded agents were characterized using dynamic light scattering (DLS), UV-vis fluorescence spectroscopy, and asymmetrical flow field-flow fractionation (AF4). Polymersomes were imaged and tested for biological activity in human and murine fibroblasts, murine macrophages, primary murine dorsal root ganglia, and murine hippocampal cultures. Polymersomes were rapidly internalized and there was a clear intracellular co-localization of the fluorescent drug (curcumin) with the fluorescent rhodamine-labeled polymersomes. Polymersomes containing CD109, a glycosylphosphatidylinositol-anchored protein, promoted cell migration in the model of wound healing. Nerve growth factor-loaded polymersomes effectively enhanced neurite outgrowth in dissociated and explanted dorsal root ganglia. Brain-derived neurotrophic factor increased dendritic spine density in serum-deprived hippocampal slice cultures. NMDAR agonist- and antagonist-functionalized polymersomes targeted selectively neurons over glial cells in mixed cultures. Collectively, the study reveals the successful incorporation into polymersomes of biologically active trophic factors and small hydrophilic agents that retain their biological activity in vitro, as demonstrated in selected central and peripheral tissue models.
Collapse
Affiliation(s)
- Alexandre Moquin
- Department
of Pharmacology and Therapeutics, Faculty of Medicine, McGill University, 3655 Promenade Sir-William-Osler, H3G
1Y6 Montreal, Québec, Canada
| | - Jeff Ji
- Department
of Pharmacology and Therapeutics, Faculty of Medicine, McGill University, 3655 Promenade Sir-William-Osler, H3G
1Y6 Montreal, Québec, Canada
| | - Kevin Neibert
- Department
of Pharmacology and Therapeutics, Faculty of Medicine, McGill University, 3655 Promenade Sir-William-Osler, H3G
1Y6 Montreal, Québec, Canada
| | - Françoise
M. Winnik
- Département
de Chimie, Université de Montréal, CP 6128 Succursale Centre-Ville, H3C 3J7 Montréal, Québec, Canada
- International
Center for Materials Nanoarchitectonics (MANA), National Institute for Materials Science (NIMS), 1-1 Namiki, 305-0044 Tsukuba, Ibaraki, Japan
- Department
of Chemistry, University of Helsinki, FI-00014 Helsinki, Finland
| | - Dusica Maysinger
- Department
of Pharmacology and Therapeutics, Faculty of Medicine, McGill University, 3655 Promenade Sir-William-Osler, H3G
1Y6 Montreal, Québec, Canada
| |
Collapse
|
47
|
Zhang Y, Chopp M, Rex CS, Simmon VF, Sarraf ST, Zhang ZG, Mahmood A, Xiong Y. A Small Molecule Spinogenic Compound Enhances Functional Outcome and Dendritic Spine Plasticity in a Rat Model of Traumatic Brain Injury. J Neurotrauma 2018; 36:589-600. [PMID: 30014757 DOI: 10.1089/neu.2018.5790] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The tetra (ethylene glycol) derivative of benzothiazole aniline (SPG101) has been shown to improve dendritic spine density and cognitive memory in the triple transgenic mouse model of Alzheimer disease (AD) when administered intraperitoneally. The present study was designed to investigate the therapeutic effects of SPG101 on dendritic spine density and morphology and sensorimotor and cognitive functional recovery in a rat model of traumatic brain injury (TBI) induced by controlled cortical impact (CCI). Young adult male Wistar rats with CCI were randomly divided into the following two groups (n = 7/group): (1) Vehicle, and (2) SPG101. SPG101 (30 mg/kg) dissolved in vehicle (1% dimethyl sulfoxide in phosphate buffered saline) or Vehicle were intraperitoneally administered starting at 1 h post-injury and once daily for the next 34 days. Sensorimotor deficits were assessed using a modified neurological severity score and adhesive removal and foot fault tests. Cognitive function was measured by Morris water maze, novel object recognition (NOR), and three-chamber social recognition tests. The animals were sacrificed 35 days after injury, and their brains were processed for measurement of dendritic spine density and morphology using ballistic dye labeling. Compared with the vehicle treatment, SPG101 treatment initiated 1 h post-injury significantly improved sensorimotor functional recovery (days 7-35, p < 0.0001), spatial learning (days 32-35, p < 0.0001), NOR (days 14 and 35, p < 0.0001), social recognition (days 14 and 35, p < 0.0001). Further, treatment significantly increased dendritic spine density in the injured cortex (p < 0.05), decreased heterogeneous distribution of spine lengths in the injured cortex and hippocampus (p < 0.0001), modifications that are associated with the promotion of spine maturation in these brain regions. In summary, treatment with SPG101 initiated 1 h post-injury and continued for an additional 34 days improves both sensorimotor and cognitive functional recovery, indicating that SPG101 acts as a spinogenic agent and may have potential as a novel treatment of TBI.
Collapse
Affiliation(s)
- Yanlu Zhang
- 1 Department of Neurosurgery, Henry Ford Hospital , Detroit, Michigan
| | - Michael Chopp
- 2 Department of Neurology, Henry Ford Hospital , Detroit, Michigan.,3 Department of Physics, Oakland University , Rochester, Michigan
| | | | | | | | - Zheng Gang Zhang
- 2 Department of Neurology, Henry Ford Hospital , Detroit, Michigan
| | - Asim Mahmood
- 1 Department of Neurosurgery, Henry Ford Hospital , Detroit, Michigan
| | - Ye Xiong
- 1 Department of Neurosurgery, Henry Ford Hospital , Detroit, Michigan
| |
Collapse
|
48
|
Arulsamy A, Teng J, Colton H, Corrigan F, Collins-Praino L. Evaluation of early chronic functional outcomes and their relationship to pre-frontal cortex and hippocampal pathology following moderate-severe traumatic brain injury. Behav Brain Res 2018; 348:127-138. [DOI: 10.1016/j.bbr.2018.04.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 03/20/2018] [Accepted: 04/06/2018] [Indexed: 01/02/2023]
|
49
|
Chuckowree JA, Zhu Z, Brizuela M, Lee KM, Blizzard CA, Dickson TC. The Microtubule-Modulating Drug Epothilone D Alters Dendritic Spine Morphology in a Mouse Model of Mild Traumatic Brain Injury. Front Cell Neurosci 2018; 12:223. [PMID: 30104961 PMCID: PMC6077201 DOI: 10.3389/fncel.2018.00223] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 07/09/2018] [Indexed: 12/27/2022] Open
Abstract
Microtubule dynamics underpin a plethora of roles involved in the intricate development, structure, function, and maintenance of the central nervous system. Within the injured brain, microtubules are vulnerable to misalignment and dissolution in neurons and have been implicated in injury-induced glial responses and adaptive neuroplasticity in the aftermath of injury. Unfortunately, there is a current lack of therapeutic options for treating traumatic brain injury (TBI). Thus, using a clinically relevant model of mild TBI, lateral fluid percussion injury (FPI) in adult male Thy1-YFPH mice, we investigated the potential therapeutic effects of the brain-penetrant microtubule-stabilizing agent, epothilone D. At 7 days following a single mild lateral FPI the ipsilateral hemisphere was characterized by mild astroglial activation and a stereotypical and widespread pattern of axonal damage in the internal and external capsule white matter tracts. These alterations occurred in the absence of other overt signs of trauma: there were no alterations in cortical thickness or in the number of cortical projection neurons, axons or dendrites expressing YFP. Interestingly, a single low dose of epothilone D administered immediately following FPI (and sham-operation) caused significant alterations in the dendritic spines of layer 5 cortical projection neurons, while the astroglial response and axonal pathology were unaffected. Specifically, spine length was significantly decreased, whereas the density of mushroom spines was significantly increased following epothilone D treatment. Together, these findings have implications for the use of microtubule stabilizing agents in manipulating injury-induced synaptic plasticity and indicate that further study into the viability of microtubule stabilization as a therapeutic strategy in combating TBI is warranted.
Collapse
Affiliation(s)
- Jyoti A. Chuckowree
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Zhendan Zhu
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Mariana Brizuela
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
- Centre for Neuroscience, School of Medicine, Flinders University, Adelaide, SA, Australia
| | - Ka M. Lee
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Catherine A. Blizzard
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Tracey C. Dickson
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| |
Collapse
|
50
|
Thau-Zuchman O, Gomes RN, Dyall SC, Davies M, Priestley JV, Groenendijk M, De Wilde MC, Tremoleda JL, Michael-Titus AT. Brain Phospholipid Precursors Administered Post-Injury Reduce Tissue Damage and Improve Neurological Outcome in Experimental Traumatic Brain Injury. J Neurotrauma 2018; 36:25-42. [PMID: 29768974 PMCID: PMC6306688 DOI: 10.1089/neu.2017.5579] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Traumatic brain injury (TBI) leads to cellular loss, destabilization of membranes, disruption of synapses and altered brain connectivity, and increased risk of neurodegenerative disease. A significant and long-lasting decrease in phospholipids (PLs), essential membrane constituents, has recently been reported in plasma and brain tissue, in human and experimental TBI. We hypothesized that supporting PL synthesis post-injury could improve outcome post-TBI. We tested this hypothesis using a multi-nutrient combination designed to support the biosynthesis of PLs and available for clinical use. The multi-nutrient, Fortasyn® Connect (FC), contains polyunsaturated omega-3 fatty acids, choline, uridine, vitamins, cofactors required for PL biosynthesis, and has been shown to have significant beneficial effects in early Alzheimer's disease. Male C57BL/6 mice received a controlled cortical impact injury and then were fed a control diet or a diet enriched with FC for 70 days. FC led to a significantly improved sensorimotor outcome and cognition, reduced lesion size and oligodendrocyte loss, and it restored myelin. It reversed the loss of the synaptic protein synaptophysin and decreased levels of the axon growth inhibitor, Nogo-A, thus creating a permissive environment. It decreased microglia activation and the rise in ß-amyloid precursor protein and restored the depressed neurogenesis. The effects of this medical multi-nutrient suggest that support of PL biosynthesis post-TBI, a new treatment paradigm, has significant therapeutic potential in this neurological condition for which there is no satisfactory treatment. The multi-nutrient tested has been used in dementia patients and is safe and well tolerated, which would enable rapid clinical exploration in TBI.
Collapse
Affiliation(s)
- Orli Thau-Zuchman
- 1 Centre for Neuroscience and Trauma, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Rita N Gomes
- 1 Centre for Neuroscience and Trauma, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Simon C Dyall
- 3 Bournemouth University, Royal London House, Bournemouth, United Kingdom
| | - Meirion Davies
- 1 Centre for Neuroscience and Trauma, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - John V Priestley
- 1 Centre for Neuroscience and Trauma, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Martine Groenendijk
- 2 Nutricia Research-Nutricia Advanced Medical Nutrition, Utrecht, The Netherlands
| | - Martijn C De Wilde
- 2 Nutricia Research-Nutricia Advanced Medical Nutrition, Utrecht, The Netherlands
| | - Jordi L Tremoleda
- 1 Centre for Neuroscience and Trauma, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Adina T Michael-Titus
- 1 Centre for Neuroscience and Trauma, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|