1
|
Chen C, Chang ZH, Yao B, Liu XY, Zhang XW, Liang J, Wang JJ, Bao SQ, Chen MM, Zhu P, Li XH. 3D printing of interferon γ-preconditioned NSC-derived exosomes/collagen/chitosan biological scaffolds for neurological recovery after TBI. Bioact Mater 2024; 39:375-391. [PMID: 38846528 PMCID: PMC11153920 DOI: 10.1016/j.bioactmat.2024.05.026] [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: 03/14/2024] [Revised: 05/13/2024] [Accepted: 05/13/2024] [Indexed: 06/09/2024] Open
Abstract
The reconstruction of neural function and recovery of chronic damage following traumatic brain injury (TBI) remain significant clinical challenges. Exosomes derived from neural stem cells (NSCs) offer various benefits in TBI treatment. Numerous studies confirmed that appropriate preconditioning methods enhanced the targeted efficacy of exosome therapy. Interferon-gamma (IFN-γ) possesses immunomodulatory capabilities and is widely involved in neurological disorders. In this study, IFN-γ was employed for preconditioning NSCs to enhance the efficacy of exosome (IFN-Exo, IE) for TBI. miRNA sequencing revealed the potential of IFN-Exo in promoting neural differentiation and modulating inflammatory responses. Through low-temperature 3D printing, IFN-Exo was combined with collagen/chitosan (3D-CC-IE) to preserve the biological activity of the exosome. The delivery of exosomes via biomaterial scaffolds benefited the retention and therapeutic potential of exosomes, ensuring that they could exert long-term effects at the injury site. The 3D-CC-IE scaffold exhibited excellent biocompatibility and mechanical properties. Subsequently, 3D-CC-IE scaffold significantly improved impaired motor and cognitive functions after TBI in rat. Histological results showed that 3D-CC-IE scaffold markedly facilitated the reconstruction of damaged neural tissue and promoted endogenous neurogenesis. Further mechanistic validation suggested that IFN-Exo alleviated neuroinflammation by modulating the MAPK/mTOR signaling pathway. In summary, the results of this study indicated that 3D-CC-IE scaffold engaged in long-term pathophysiological processes, fostering neural function recovery after TBI, offering a promising regenerative therapy avenue.
Collapse
Affiliation(s)
- Chong Chen
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
- Tianjin Key Laboratory of Neurotrauma Repair, Characteristic Medical Center of People's Armed Police Forces, Tianjin, 300162, China
| | - Zhe-Han Chang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Bin Yao
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
| | - Xiao-Yin Liu
- Tianjin Key Laboratory of Neurotrauma Repair, Characteristic Medical Center of People's Armed Police Forces, Tianjin, 300162, China
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xiao-Wang Zhang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Jun Liang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Jing-Jing Wang
- Tianjin Key Laboratory of Neurotrauma Repair, Characteristic Medical Center of People's Armed Police Forces, Tianjin, 300162, China
| | - Shuang-Qing Bao
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Meng-Meng Chen
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
| | - Xiao-Hong Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| |
Collapse
|
2
|
Katzenberger RJ, Ganetzky B, Wassarman DA. Lissencephaly-1 mutations enhance traumatic brain injury outcomes in Drosophila. Genetics 2023; 223:iyad008. [PMID: 36683334 PMCID: PMC9991514 DOI: 10.1093/genetics/iyad008] [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: 11/14/2022] [Revised: 11/14/2022] [Accepted: 01/16/2023] [Indexed: 01/24/2023] Open
Abstract
Traumatic brain injury (TBI) outcomes vary greatly among individuals, but most of the variation remains unexplained. Using a Drosophila melanogaster TBI model and 178 genetically diverse lines from the Drosophila Genetic Reference Panel (DGRP), we investigated the role that genetic variation plays in determining TBI outcomes. Following injury at 20-27 days old, DGRP lines varied considerably in mortality within 24 h ("early mortality"). Additionally, the disparity in early mortality resulting from injury at 20-27 vs 0-7 days old differed among DGRP lines. These data support a polygenic basis for differences in TBI outcomes, where some gene variants elicit their effects by acting on aging-related processes. Our genome-wide association study of DGRP lines identified associations between single nucleotide polymorphisms in Lissencephaly-1 (Lis-1) and Patronin and early mortality following injury at 20-27 days old. Lis-1 regulates dynein, a microtubule motor required for retrograde transport of many cargoes, and Patronin protects microtubule minus ends against depolymerization. While Patronin mutants did not affect early mortality, Lis-1 compound heterozygotes (Lis-1x/Lis-1y) had increased early mortality following injury at 20-27 or 0-7 days old compared with Lis-1 heterozygotes (Lis-1x/+), and flies that survived 24 h after injury had increased neurodegeneration but an unaltered lifespan, indicating that Lis-1 affects TBI outcomes independently of effects on aging. These data suggest that Lis-1 activity is required in the brain to ameliorate TBI outcomes through effects on axonal transport, microtubule stability, and other microtubule proteins, such as tau, implicated in chronic traumatic encephalopathy, a TBI-associated neurodegenerative disease in humans.
Collapse
Affiliation(s)
- Rebeccah J Katzenberger
- Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Barry Ganetzky
- Department of Genetics, College of Agricultural and Life Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - David A Wassarman
- Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
3
|
Apiribu F, Duma SE, Ncama BP. Men's experience of perpetrating intimate partner violence following disclosure of HIV status by their seropositive female intimate partners: a qualitative study. Ann Med 2022; 54:1126-1139. [PMID: 35511257 PMCID: PMC9090398 DOI: 10.1080/07853890.2022.2062444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Gender-Based (GB) intimate partner violence is a social and public health issue globally. Several risks of violence related to male sexual partners' perpetration of intimate partner violence (IPV) following the disclosure of their female intimate partners' HIV + status have been reported. No research has been conducted on male sexual partner's perspectives of perpetrating IPV following their female intimate partners' disclosure of human immunodeficiency virus (HIV) seropositive status as a risk factor for the perpetration of IPV in Ghana. OBJECTIVE The objective of this study is to explore and describe male sexual partners' views or perspectives of perpetrating IPV following their female intimate partners' disclosure of being HIV positive in Ghana. METHODS Interpretive phenomenological approach was used to collect and analyse data from a purposive sample of 18 Male participants whose female intimate relations informed them of being HIV + in Ghana. The sample population was taken from Ghana because such research has been reported elsewhere but none has been done in Ghana. A semi-structured interview guide was used to collect the data. The interview guide covered topics such as background information, participants' reaction to HIV positive disclosure, lived experiences of participants, and Participants' understanding of different forms of IPV. RESULTS The findings of this study reveal five main themes that emerged from the interviews which include views on the perpetration of emotional, psychological, and verbal abuse; views on the perpetration of sexual deprivation; views on the perpetration of social isolation; views on the perpetration of financial abuse and views on escalated perpetration of physical abuse. CONCLUSION From the data, HIV positive status disclosure served as a risk factor for different forms of GB IPV against HIV positive women in Ghana, thus making this group more vulnerable and exposed to more GB IPV. Strategies to prevent the perpetration of IPV against women newly diagnosed as HIV positive are needed. We recommend screening all newly diagnosed HIV-positive women for abuse as an additional prevention strategy for IPV associated with disclosure of positive HIV status. KEY MESSAGESHIV positive status disclosure serves as a risk for the perpetration of IPV.Men are predisposed to violence upon hearing that their female heterosexual intimate partners are HIV positive.HIV infection information is distressful to receive from an intimate partner.
Collapse
Affiliation(s)
- Felix Apiribu
- School of Nursing and Public Health, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa.,Department of Nursing, Faculty of Allied Health Sciences, College of Health Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Sinegugu Evidence Duma
- School of Nursing and Public Health, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Busisiwe Purity Ncama
- School of Nursing and Public Health, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
4
|
Van Wyck D, Kolls BJ, Wang H, Cantillana V, Maughan M, Laskowitz DT. Prophylactic treatment with CN-105 improves functional outcomes in a murine model of closed head injury. Exp Brain Res 2022; 240:2413-2423. [PMID: 35841411 DOI: 10.1007/s00221-022-06417-4] [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: 01/25/2022] [Accepted: 07/04/2022] [Indexed: 11/30/2022]
Abstract
The treatment of traumatic brain injury (TBI) in military populations is hindered by underreporting and underdiagnosis. Clinical symptoms and outcomes may be mitigated with an effective pre-injury prophylaxis. This study evaluates whether CN-105, a 5-amino acid apolipoprotein E (ApoE) mimetic peptide previously shown to modify the post-traumatic neuroinflammatory response, would maintain its neuroprotective effects if administered prior to closed-head injury in a clinically relevant murine model. CN-105 was synthesized by Polypeptide Inc. (San Diego, CA) and administered to C57-BL/6 mice intravenously (IV) and/or by intraperitoneal (IP) injection at various time points prior to injury while vehicle treated animals received IV and/or IP normal saline. Animals were randomized following injury and behavioral observations were conducted by investigators blinded to treatment. Vestibulomotor function was assessed using an automated Rotarod (Ugo Basile, Comerio, Italy), and hippocampal microglial activation was assessed using F4/80 immunohistochemical staining in treated and untreated mice 7 days post-TBI. Separate, in vivo assessments of the pharmacokinetics was performed in healthy CD-1. IV CN-105 administered prior to head injury improved vestibulomotor function compared to vehicle control-treated animals. CN-105 co-administered by IP and IV dosing 6 h prior to injury also improved vestibulomotor function up to 28 days following injury. Microglia counted in CN-105 treated specimens were significantly fewer (P = 0.03) than in vehicle specimens. CN-105 improves functional outcomes and reduces hippocampal microglial activation when administered prior to injury and could be adapted as a pre-injury prophylaxis for soldiers at high risk for TBI.
Collapse
Affiliation(s)
- David Van Wyck
- 3Rd Special Forces Group (A), U.S. Army Special Operations Command, 111 Enduring Freedom Drive (Stop A), Fort Bragg, NC, 28310, USA. .,Department of Neurology, Duke University School of Medicine, Durham, NC, 27710, USA.
| | - Bradley J Kolls
- Department of Neurology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Haichen Wang
- Department of Neurology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Viviana Cantillana
- Department of Neurology, Duke University School of Medicine, Durham, NC, 27710, USA
| | | | - Daniel T Laskowitz
- Department of Neurology, Duke University School of Medicine, Durham, NC, 27710, USA.,Department of Neurobiology, Duke University School of Medicine, Durham, NC, 27710, USA.,Aegis-CN LLC., 701 W Main Street, Durham, NC, 27701, USA
| |
Collapse
|
5
|
Celorrio M, Shumilov K, Payne C, Vadivelu S, Friess SH. Acute minocycline administration reduces brain injury and improves long-term functional outcomes after delayed hypoxemia following traumatic brain injury. Acta Neuropathol Commun 2022; 10:10. [PMID: 35090569 PMCID: PMC8796448 DOI: 10.1186/s40478-022-01310-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 01/08/2022] [Indexed: 11/22/2022] Open
Abstract
Clinical trials of therapeutics for traumatic brain injury (TBI) demonstrating preclinical efficacy for TBI have failed to replicate these results in humans, in part due to the absence of clinically feasible therapeutic windows for administration. Minocycline, an inhibitor of microglial activation, has been shown to be neuroprotective when administered early after experimental TBI but detrimental when administered chronically to human TBI survivors. Rather than focusing on the rescue of primary injury with early administration of therapeutics which may not be clinically feasible, we hypothesized that minocycline administered at a clinically feasible time point (24 h after injury) would be neuroprotective in a model of TBI plus delayed hypoxemia. We first explored several different regimens of minocycline dosing with the initial dose 24 h after injury and 2 h prior to hypoxemia, utilizing short-term neuropathology to select the most promising candidate. We found that a short course of minocycline reduced acute microglial activation, monocyte infiltration and hippocampal neuronal loss at 1 week post injury. We then conducted a preclinical trial to assess the long-term efficacy of a short course of minocycline finding reductions in hippocampal neurodegeneration and synapse loss, preservation of white matter myelination, and improvements in fear memory performance at 6 months after injury. Timing in relation to injury and duration of minocycline treatment and its impact on neuroinflammatory response may be responsible for extensive neuroprotection observed in our studies.
Collapse
|
6
|
Masse I, Moquin L, Bouchard C, Gratton A, De Beaumont L. Efficacy of prophylactic versus therapeutic administration of the NMDA receptor antagonist MK-801 on the acute neurochemical response to a concussion in a rat model combining force and rotation. J Neurosurg 2021:1-10. [PMID: 34653970 DOI: 10.3171/2021.3.jns204163] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 03/30/2021] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Alterations in amino acid concentrations are a major contributor to the persistent neurological and behavioral effects induced by concussions and mild traumatic brain injuries (TBIs). Glutamate, the most abundant excitatory amino acid in the CNS, has a major role in the pathophysiological process of concussion. The indiscriminate liberation of glutamate immediately after a concussion triggers an excitotoxic response that leads to cell death, neuronal damage, and the dysfunction of surviving neurons, largely by overactivation of N-methyl-d-aspartate (NMDA) glutamatergic receptors. The aim of the present study was to investigate the efficacy of prophylactic versus therapeutic administration of MK-801, a promising NMDA receptor antagonist, on the acute changes in amino acid extracellular concentrations involved in excitotoxicity resulting from a concussive trauma. METHODS The immediate neurochemical response to a concussion cannot be characterized in humans. Therefore, the authors used their previously validated combination of a weight-drop concussion rat model and in vivo cerebral microdialysis. The microdialysis probe was inserted inside the hippocampus and left inserted at impact to allow uninterrupted sampling of amino acids of interest immediately after concussion. The primary outcome included amino acid concentrations and the secondary outcome included righting time. Samples were taken in 10-minute increments for 60 minutes before, during, and 60 minutes after impact, and analyzed for glutamate, gamma-aminobutyric acid, taurine, glycine, glutamine, and serine using high-performance liquid chromatography. Righting time was acquired as a neurological restoration indicator. Physiological saline or 10 mg/kg MK-801 was administrated intraperitoneally 60 minutes before or immediately following induction of sham injury or concussion. RESULTS Following induction of concussion, glutamate, taurine, and glycine levels as well as righting times in cases from the MK-801 treatment group were comparable to those of vehicle-treated animals. In contrast, righting times and amino acid concentrations observed within the first 10 minutes after induction of concussion in cases assigned to the MK-801 prophylaxis group were comparable to those of sham-injured animals. CONCLUSIONS These results suggest that presynaptic actions and peak availability of MK-801 following prophylactic administration significantly inhibit the immediate and indiscriminate release of glutamate, taurine, and glycine in extracellular fluid after a concussion.
Collapse
Affiliation(s)
- Ian Masse
- 1Research Center, Hôpital du Sacré-Cœur de Montréal; and
| | - Luc Moquin
- 2Research Center, Douglas Institute, Montreal, Quebec, Canada
| | | | - Alain Gratton
- 2Research Center, Douglas Institute, Montreal, Quebec, Canada
| | | |
Collapse
|
7
|
O'Brien LD, Smith TL, Donvito G, Cravatt BF, Newton J, Spiegel S, Reeves TM, Phillips LL, Lichtman AH. Diacylglycerol Lipase-β Knockout Mice Display a Sex-Dependent Attenuation of Traumatic Brain Injury-Induced Mortality with No Impact on Memory or Other Functional Consequences. Cannabis Cannabinoid Res 2021; 6:508-521. [PMID: 34142866 DOI: 10.1089/can.2020.0175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Background: The endogenous cannabinoid system modulates inflammatory signaling in a variety of pathological states, including traumatic brain injury (TBI). The selective expression of diacylglycerol lipase-β (DAGL-β), the 2-arachidonylglycerol biosynthetic enzyme, on resident immune cells of the brain (microglia) and the role of this pathway in neuroinflammation, suggest that this enzyme may contribute to TBI-induced neuroinflammation. Accordingly, we tested whether DAGL-β-/- mice would show a protective phenotype from the deleterious consequences of TBI on cognitive and neurological motor functions. Materials and Methods: DAGL-β-/- and -β+/+ mice were subjected to the lateral fluid percussion model of TBI and assessed for learning and memory in the Morris water maze (MWM) Fixed Platform (reference memory) and Reversal (cognitive flexibility) tasks, as well as in a cued MWM task to infer potential sensorimotor/motivational deficits. In addition, subjects were assessed for motor behavior (Rotarod and the Neurological Severity Score assays) and in the light/dark box and the elevated plus maze to infer whether these manipulations affected anxiety-like behavior. Finally, we also examined whether brain injury disrupts the ceramide/sphingolipid lipid signaling system and if DAGL-β deletion offers protection. Results: TBI disrupted all measures of neurological motor function and reduced body weight, but did not affect body temperature or performance in common assays used to infer anxiety. TBI also impaired performance in MWM Fixed Platform and Reversal tasks, but did not affect cued MWM performance. Although no differences were found between DAGL-β-/- and -β+/+ mice in any of these measures, male DAGL-β-/- mice displayed an unexpected survival-protective phenotype, which persisted at increased injury severities. In contrast, TBI did not elicit mortality in female mice regardless of genotype. TBI also produced significant changes in sphingolipid profiles (a family of lipids, members of which have been linked to both apoptotic and antiapoptotic pathways), in which DAGL-β deletion modestly altered levels of select species. Conclusions: These findings indicate that although DAGL-β does not play a necessary role in TBI-induced cognitive and neurological function, it appears to contribute to the increased vulnerability of male mice to TBI-induced mortality, whereas female mice show high survival rates irrespective of DAGL-β expression.
Collapse
Affiliation(s)
- Lesley D O'Brien
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Terry L Smith
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Giulia Donvito
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Benjamin F Cravatt
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Jason Newton
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Thomas M Reeves
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Linda L Phillips
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Aron H Lichtman
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA.,Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
8
|
Role of Citicoline in the Management of Traumatic Brain Injury. Pharmaceuticals (Basel) 2021; 14:ph14050410. [PMID: 33926011 PMCID: PMC8146347 DOI: 10.3390/ph14050410] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/20/2021] [Accepted: 04/24/2021] [Indexed: 01/07/2023] Open
Abstract
Head injury is among the most devastating types of injury, specifically called Traumatic Brain Injury (TBI). There is a need to diminish the morbidity related with TBI and to improve the outcome of patients suffering TBI. Among the improvements in the treatment of TBI, neuroprotection is one of the upcoming improvements. Citicoline has been used in the management of brain ischemia related disorders, such as TBI. Citicoline has biochemical, pharmacological, and pharmacokinetic characteristics that make it a potentially useful neuroprotective drug for the management of TBI. A short review of these characteristics is included in this paper. Moreover, a narrative review of almost all the published or communicated studies performed with this drug in the management of patients with head injury is included. Based on the results obtained in these clinical studies, it is possible to conclude that citicoline is able to accelerate the recovery of consciousness and to improve the outcome of this kind of patient, with an excellent safety profile. Thus, citicoline could have a potential role in the management of TBI.
Collapse
|
9
|
McGeown JP, Hume PA, Theadom A, Quarrie KL, Borotkanics R. Nutritional interventions to improve neurophysiological impairments following traumatic brain injury: A systematic review. J Neurosci Res 2020; 99:573-603. [PMID: 33107071 DOI: 10.1002/jnr.24746] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 10/02/2020] [Accepted: 10/07/2020] [Indexed: 12/25/2022]
Abstract
Traumatic brain injury (TBI) accounts for significant global health burden. Effects of TBI can become chronic even following mild injury. There is a need to develop effective therapies to attenuate the damaging effects of TBI and improve recovery outcomes. This literature review using a priori criteria (PROSPERO; CRD42018100623) summarized 43 studies between January 1998 and July 2019 that investigated nutritional interventions (NUT) delivered with the objective of altering neurophysiological (NP) outcomes following TBI. Risk of bias was assessed for included studies, and NP outcomes recorded. The systematic search resulted in 43 of 3,748 identified studies met inclusion criteria. No studies evaluated the effect of a NUT on NP outcomes of TBI in humans. Biomarkers of morphological changes and apoptosis, oxidative stress, and plasticity, neurogenesis, and neurotransmission were the most evaluated NP outcomes across the 43 studies that used 2,897 animals. The risk of bias was unclear in all reviewed studies due to poorly detailed methodology sections. Taking these limitations into account, anti-oxidants, branched chain amino acids, and ω-3 polyunsaturated fatty acids have shown the most promising pre-clinical results for altering NP outcomes following TBI. Refinement of pre-clinical methodologies used to evaluate effects of interventions on secondary damage of TBI would improve the likelihood of translation to clinical populations.
Collapse
Affiliation(s)
- Joshua P McGeown
- Sports Performance Research Institute New Zealand (SPRINZ), Faculty of Health and Environmental Science, Auckland University of Technology, Auckland, New Zealand.,Traumatic Brain Injury Network, Auckland University of Technology, Auckland, New Zealand
| | - Patria A Hume
- Sports Performance Research Institute New Zealand (SPRINZ), Faculty of Health and Environmental Science, Auckland University of Technology, Auckland, New Zealand.,Traumatic Brain Injury Network, Auckland University of Technology, Auckland, New Zealand.,National Institute of Stroke and Applied Neuroscience (NISAN), Faculty of Health and Environmental Science, Auckland University of Technology, Auckland, New Zealand
| | - Alice Theadom
- Traumatic Brain Injury Network, Auckland University of Technology, Auckland, New Zealand.,National Institute of Stroke and Applied Neuroscience (NISAN), Faculty of Health and Environmental Science, Auckland University of Technology, Auckland, New Zealand
| | | | - Robert Borotkanics
- Sports Performance Research Institute New Zealand (SPRINZ), Faculty of Health and Environmental Science, Auckland University of Technology, Auckland, New Zealand
| |
Collapse
|
10
|
Celorrio M, Rhodes J, Vadivelu S, Davies M, Friess SH. N-acetylcysteine reduces brain injury after delayed hypoxemia following traumatic brain injury. Exp Neurol 2020; 335:113507. [PMID: 33065076 DOI: 10.1016/j.expneurol.2020.113507] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 09/10/2020] [Accepted: 10/09/2020] [Indexed: 01/11/2023]
Abstract
Preclinical investigations into neuroprotective agents for traumatic brain injury (TBI) have shown promise when administered before or very early after experimental TBI. However clinical trials of therapeutics demonstrating preclinical efficacy for TBI have failed to replicate these results in humans, a lost in translation phenomenon. N-acetylcysteine (NAC) is a potent anti-oxidant with demonstrated efficacy in pre-clinical TBI when administered early after primary injury. Utilizing our clinically relevant mouse model, we hypothesized that NAC administration in a clinically relevant timeframe could improve the brain's resilience to the secondary insult of hypoxemia. NAC or vehicle administered daily starting 2 h prior to hypoxemia (24 h after controlled cortical impact) for 3 doses in male mice reduced short-term axonal injury and hippocampal neuronal loss. Six month behavioral assessments including novel object recognition, socialization, Barnes maze, and fear conditioning did not reveal performance differences between sham controls and injured mice receiving NAC or saline vehicle. At 7 months after injury, NAC administered mice had reduced hippocampal neuronal loss but no reduction in lesion volume. In summary, our preclinical trial to test the neuroprotective efficacy of NAC against a secondary hypoxic insult after TBI demonstrated short and long-term neuropathological evidence of neuroprotection but a lack of detectable differences in long-term behavioral assessments between sham controls and injured mice limits conclusions on its impact on long-term neurobehavioral outcomes.
Collapse
Affiliation(s)
- Marta Celorrio
- Division of Critical Care Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, One Children's Place, St. Louis, MO 63110, USA
| | - James Rhodes
- Division of Critical Care Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, One Children's Place, St. Louis, MO 63110, USA
| | - Sangeetha Vadivelu
- Division of Critical Care Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, One Children's Place, St. Louis, MO 63110, USA
| | - McKenzie Davies
- Division of Critical Care Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, One Children's Place, St. Louis, MO 63110, USA
| | - Stuart H Friess
- Division of Critical Care Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, One Children's Place, St. Louis, MO 63110, USA.
| |
Collapse
|
11
|
Washington PM, Lee C, Dwyer MKR, Konofagou EE, Kernie SG, Morrison B. Hyaluronidase reduced edema after experimental traumatic brain injury. J Cereb Blood Flow Metab 2020; 40:2026-2037. [PMID: 31648593 PMCID: PMC7786840 DOI: 10.1177/0271678x19882780] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Cerebral edema and the subsequent increased intracranial pressure are associated with mortality and poor outcome following traumatic brain injury. Previous in vitro studies have shown that the Gibbs-Donnan effect, which describes the tendency of a porous, negatively charged matrix to attract positive ions and water, applies to brain tissue and that enzymatic reduction of the fixed charge density can prevent tissue swelling. We tested whether hyaluronidase, an enzyme that degrades the large, negatively charged glycosaminoglycan hyaluronan, could reduce brain edema after traumatic brain injury. In vivo, intracerebroventricular injection of hyaluronidase after controlled cortical impact in mice reduced edema in the ipsilateral hippocampus at 24 h by both the wet-weight/dry-weight method (78.15 ± 0.65% vs. 80.4 ± 0.46%; p < 0.01) and T2-weighted magnetic resonance imaging (13.88 ± 3.09% vs. 29.23 ± 6.14%; p < 0.01). Hyaluronidase did not adversely affect blood-brain-barrier-integrity measured by dynamic contrast-enhanced magnetic resonance imaging, nor did hyaluronidase negatively affect functional recovery after controlled cortical impact measured with the rotarod or Morris water maze tasks. Reduction of fixed charge density by hyaluronidase was confirmed in cortical explants in vitro (5.46 ± 1.15 µg/mg vs. 7.76 ± 1.87 µg/mg; p < 0.05). These data demonstrate that targeting the fixed charge density with hyaluronidase reduced edema in an in vivo mouse model of traumatic brain injury.
Collapse
Affiliation(s)
- Patricia M Washington
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Changhee Lee
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Mary Kate R Dwyer
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Elisa E Konofagou
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Steven G Kernie
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
| | - Barclay Morrison
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Barclay Morrison III, Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, Mail Code 8904, 1210 Amsterdam Avenue, New York, NY 10027, USA.
| |
Collapse
|
12
|
Wagner KE, Binyamin TR, Colley P, Chiluwal AK, Harrop JS, Hawryluk GW, Hickman ZL, Margetis K, Rymarczuk GN, Stippler M, Ullman JS. Trauma. Oper Neurosurg (Hagerstown) 2020; 17:S45-S75. [PMID: 31099847 DOI: 10.1093/ons/opz089] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 02/21/2019] [Indexed: 11/12/2022] Open
Affiliation(s)
| | - Tamar R Binyamin
- Department of Neurosurgery, University of California Davis Medical Center, Sacramento, California
| | - Patrick Colley
- Department of Otolaryngology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Amrit K Chiluwal
- Department of Neurosurgery, Northwell Health, Manhasset, New York
| | - James S Harrop
- Department of Neurosurgery, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | | | - Zachary L Hickman
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Konstantinos Margetis
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, New York
| | - George N Rymarczuk
- Department of Neurosurgery, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania.,Division of Neurosurgery, Walter Reed Medical Center, Bethesda, Maryland
| | - Martina Stippler
- Department of Neurosurgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Jamie S Ullman
- Department of Neurosurgery, Northwell Health, Manhasset, New York
| |
Collapse
|
13
|
Improvement in cognitive dysfunction following blast induced traumatic brain injury by thymosin α1 in rats: Involvement of inhibition of tau phosphorylation at the Thr205 epitope. Brain Res 2020; 1747:147038. [PMID: 32738231 DOI: 10.1016/j.brainres.2020.147038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 07/25/2020] [Accepted: 07/27/2020] [Indexed: 12/13/2022]
Abstract
Cognitive impairment is a significant sequela of traumatic brain injury (TBI) especially blast induced traumatic brain injury (bTBI), which is characterized by rapid impairments of learning and memory ability. Although several neuroprotective agents have been postulated as promising drugs for bTBI in animal studies, very few ideal therapeutic options exist to improve cognitive impairment following bTBI. Thymosin α1(Tα1), a 28-amino-acid protein that possesses immunomodulatory functions, has exhibited beneficial effects in the treatment of infectious diseases, immunodeficiency diseases and cancers. However, it remains unclear whether Tα1 has a therapeutic role in bTBI. Thus, we hypothesized that Tα1 administration could reverse the outcomes of bTBI. The blast induced TBI (bTBI) rat model was established with the compressed gas driven blast injury model system. A consecutive Tα1 therapy (in 1 ml saline, twice a day) at a dose of 200 µg/kg or normal saline (NS) (1 ml, twice a day) for 3 days or 2 weeks was performed. Utilizing our newly designed bTBI model, we investigated the beneficial effects of Tα1 therapy on rats exposed to bTBI including: cognitive functions, general histology, regulatory T (Treg) cells, edema, inflammation reactions and the expression and phosphorylation level of tau via Morris Water Maze test (MWM test), HE staining, flow cytometry, brain water content (BWC) calculation, IL-6 assay and Western blotting, respectively. Tα1 treatment seemed to reduce the 24-hour mortality, albeit with no statistical significance. Moreover, Tα1 treatment markedly improved cognitive dysfunction by decreasing the escape latency in the acquisition phase, and increasing the crossing numbers in the probe phase of MWM test. More interestingly, Tα1 significantly inhibited tau phosphorylation at the Thr205 epitope, but not at the Ser404 and Ser262 epitopes. Tα1 increased the percentage of Treg cells and inhibited plasma IL-6 production on 3d post bTBI. Moreover, Tα1 suppressed brain edema as demonstrated by decrease of BWC. However, there was a lack of obvious change in histopathology in the brain upon Tα1 treatment. This is the first study showing that Tα1 improves neurological deficits after bTBI in rats, which is potentially related to the inhibition of tau phosphorylation at the Thr205 epitope, increased Treg cells and decreased inflammatory reactions and brain edema.
Collapse
|
14
|
Herzog C, Greenald D, Larraz J, Keatinge M, Herrgen L. RNA-seq analysis and compound screening highlight multiple signalling pathways regulating secondary cell death after acute CNS injury in vivo. Biol Open 2020; 9:9/5/bio050260. [PMID: 32366533 PMCID: PMC7225090 DOI: 10.1242/bio.050260] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Understanding the molecular mechanisms that regulate secondary cell death after acute central nervous system (CNS) injury is critical for the development of effective neuroprotective drugs. Previous research has shown that neurotoxic processes including excitotoxicity, oxidative stress and neuroinflammation can cause secondary cell death. Nevertheless, clinical trials targeting these processes have been largely unsuccessful, suggesting that the signalling pathways underlying secondary cell death remain incompletely understood. Due to their suitability for live imaging and their amenability to genetic and pharmacological manipulation, larval zebrafish provide an ideal platform for studying the regulation of secondary cell death in vivo Here, we use RNA-seq gene expression profiling and compound screening to identify signalling pathways that regulate secondary cell death after acute neural injury in larval zebrafish. RNA-seq analysis of genes upregulated in cephalic mpeg1+ macrophage-lineage cells isolated from mpeg1:GFP transgenic larvae after neural injury suggested an involvement of cytokine and polyamine signalling in secondary cell death. Furthermore, screening a library of FDA approved compounds indicated roles for GABA, serotonin and dopamine signalling. Overall, our results highlight multiple signalling pathways that regulate secondary cell death in vivo, and thus provide a starting point for the development of novel neuroprotective treatments for patients with CNS injury.This article has an associated First Person interview with the two first authors of the paper.
Collapse
Affiliation(s)
- Chiara Herzog
- Centre for Discovery Brain Sciences, Deanery of Biomedical Sciences, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - David Greenald
- Centre for Discovery Brain Sciences, Deanery of Biomedical Sciences, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Juan Larraz
- Centre for Discovery Brain Sciences, Deanery of Biomedical Sciences, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Marcus Keatinge
- Centre for Discovery Brain Sciences, Deanery of Biomedical Sciences, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Leah Herrgen
- Centre for Discovery Brain Sciences, Deanery of Biomedical Sciences, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| |
Collapse
|
15
|
Kulesza B, Litak J, Grochowski C, Nogalski A, Rola R. The Initial Factors with Strong Predictive Value in Relation to Six-Month Outcome among Patients Operated due to Extra-Axial Hematomas. Diagnostics (Basel) 2020; 10:diagnostics10030174. [PMID: 32209970 PMCID: PMC7151066 DOI: 10.3390/diagnostics10030174] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 03/18/2020] [Accepted: 03/19/2020] [Indexed: 01/15/2023] Open
Abstract
Introduction: Traumatic brain injuries (TBI) are a real social problem, with an upward trend worldwide. The most frequent consequence of a traumatic brain injury is extra-axial hemorrhage, i.e., an acute subdural (SDH) and epidural hematoma (EDH). Most of the factors affecting the prognosis have been analyzed on a wide group of traumatic brain injuries. Nonetheless, there are few studies analyzing factors influencing the prognosis regarding patients undergoing surgery due to acute subdural and epidural hematoma. The aim of this study was to identify the factors which have the strongest prognostic value in relation to the 6-month outcome of the patients undergoing surgery for SDH and EDH. Patients and methods: The study included a group of 128 patients with isolated craniocerebral injuries. Twenty eight patients were operated upon due to EDH, and a group of 100 patients were operated upon due to SDH. The following factors from the groups were analyzed: demographic data, physiological factors, laboratory factors, computed tomography scan characteristics, and time between the trauma and the surgery. All of these factors were correlated in a multivariate analysis with the six-month outcome in the Glasgow outcome scale. Results: The factors with the strongest prognostic value are GCS score, respiration rate, saturation, glycaemia and systolic blood pressure. Conclusion: Initial GCS score, respiratory rate, saturation, glycaemia and systolic blood pressure were the factors with the strongest prognostic value.
Collapse
Affiliation(s)
- Bartłomiej Kulesza
- Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, Jaczewskiego 8, 20-954 Lublin, Poland; (B.K.); (R.R.)
| | - Jakub Litak
- Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, Jaczewskiego 8, 20-954 Lublin, Poland; (B.K.); (R.R.)
- Department of Immunology, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
- Correspondence:
| | - Cezary Grochowski
- Department of Anatomy, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland;
- Laboratory of Virtual Man, Department of Anatomy, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland
| | - Adam Nogalski
- Department of Trauma Surgery and Emergency Medicine, Medical University of Lublin, Staszica 16, 20-090 Lublin, Poland;
| | - Radosław Rola
- Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, Jaczewskiego 8, 20-954 Lublin, Poland; (B.K.); (R.R.)
| |
Collapse
|
16
|
Willing AE, Das M, Howell M, Mohapatra SS, Mohapatra S. Potential of mesenchymal stem cells alone, or in combination, to treat traumatic brain injury. CNS Neurosci Ther 2020; 26:616-627. [PMID: 32157822 PMCID: PMC7248546 DOI: 10.1111/cns.13300] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 02/17/2020] [Accepted: 02/23/2020] [Indexed: 12/13/2022] Open
Abstract
Traumatic brain injury (TBI) causes death and disability in the United States and around the world. The traumatic insult causes the mechanical injury of the brain and primary cellular death. While a comprehensive pathological mechanism of TBI is still lacking, the focus of the TBI research is concentrated on understanding the pathophysiology and developing suitable therapeutic approaches. Given the complexities in pathophysiology involving interconnected immunologic, inflammatory, and neurological cascades occurring after TBI, the therapies directed to a single mechanism fail in the clinical trials. This has led to the development of the paradigm of a combination therapeutic approach against TBI. While there are no drugs available for the treatment of TBI, stem cell therapy has shown promising results in preclinical studies. But, the success of the therapy depends on the survival of the stem cells, which are limited by several factors including route of administration, health of the administered cells, and inflammatory microenvironment of the injured brain. Reducing the inflammation prior to cell administration may provide a better outcome of cell therapy following TBI. This review is focused on different therapeutic approaches of TBI and the present status of the clinical trials.
Collapse
Affiliation(s)
- Alison E Willing
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Mahasweta Das
- Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, FL, USA.,James A. Haley Veterans Hospital, Tampa, FL, USA
| | - Mark Howell
- Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, FL, USA.,James A. Haley Veterans Hospital, Tampa, FL, USA
| | - Shyam S Mohapatra
- James A. Haley Veterans Hospital, Tampa, FL, USA.,Department of Internal Medicine, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Subhra Mohapatra
- Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, FL, USA.,James A. Haley Veterans Hospital, Tampa, FL, USA
| |
Collapse
|
17
|
Ssebakumba MK, Lule H, Olweny F, Mabweijano J, Kiryabwire J. Thirty-day clinical outcome of traumatic brain injury patients with acute extradural and subdural hematoma: a cohort study at Mulago National Referral Hospital, Uganda. EGYPTIAN JOURNAL OF NEUROSURGERY 2020. [DOI: 10.1186/s41984-020-0073-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Increasing traumatic brain injury (TBI) has paralleled the need for decompression surgery for acute subdural (ASDH) and acute extradural haematoma (AEDH). Knowledge of key determinants of clinical outcomes of such patients is mandatory to guide treatment protocols.
Objective
To determine the 30-day clinical outcomes and predictor variables for patients with extra-axial hematomas at Mulago National Referral Hospital in Uganda.
Methods
Prospective observational cohort study of 109 patients with computed tomography (CT) confirmed extra-axial hematomas. Ethical clearance was obtained from the School of Medicine Research and Ethics Committee of College of Health Sciences, Makerere University (REC REF. 2018-185). Admitted patients were followed-up and reassessed for Glasgow Outcome Scale (GOS) and final disposition. Multivariate regression analysis was performed using Stata 14.0 (StataCorp. 2015) at 95% confidence interval, regarding p < 0.05 as statistically significant.
Results
The overall proportion of favorable outcome was 71.7% (n = 71), with 42.3% (n = 11) and 81.7% (n = 58) for ASDH and AEDH, respectively (p = 0.111). Factors associated with a favorable outcome were admission systolic BP > 90 mmHg [IRR = 0.88 (0.26–0.94) 95%CI, p = 0.032), oxygen saturation > 90% [IRR = 0.5 (0.26–0.94) 95%CI, p = 0.030] and diagnosis AEDH [IRR = 0.53 (0.30–0.92) 95%CI, p = 0.025). Moderate TBI [IRR = 4.57 (1.15–18.06) 95%CI, p = 0.03] and severe TBI [IRR = 6.79 (2.32–19.86) 95%CI, p < 0.001] were significantly associated with unfavorable outcomes.
Conclusion
The study revealed that post resuscitation GCS, systolic BP, oxygen circulation, and diagnosis of AEDH at admission are the most important determinants of outcome for patients with extra-axial intracranial hematomas. These findings are valuable for the triaging teams in resource-constrained settings.
Collapse
|
18
|
Jiang Q, Chen J, Long X, Yao X, Zou X, Yang Y, Huang G, Zhang H. Phillyrin protects mice from traumatic brain injury by inhibiting the inflammation of microglia via PPARγ signaling pathway. Int Immunopharmacol 2020; 79:106083. [PMID: 31923823 DOI: 10.1016/j.intimp.2019.106083] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 11/04/2019] [Accepted: 11/24/2019] [Indexed: 12/29/2022]
Abstract
The neuroinflammatory response induced by microglia plays a vital role in causing secondary brain damage after traumatic brain injury (TBI). Previous studies have found that the improved regulation of activated microglia could reduce neurological damage post-TBI. Phillyrin (Phi) is one of the main active ingredients extracted from the fruits of the medicinal plant Forsythia suspensa (Thunb.) with anti-inflammatory effects. Our study attempted to investigate the effects of phillyrin on microglial activation and neuron damage after TBI. The TBI model was applied to induce brain injury in mice, and neurological scores, brain water content, hematoxylin and eosin staining and Nissl staining were employed to determine the neuroprotective effects of phillyrin. Immunofluorescent staining and western blot analysis were used to detect nuclear factor-kappa B (NF-κB) and peroxisome proliferator-activated receptor gamma (PPARγ) expression and nuclear translocation, and the inflammation-related proteins and mRNAs were assessed by western blot analysis and quantitative real-time PCR. The results revealed that phillyrin not only inhibited the proinflammatory response induced by activated microglia but also attenuated neurological impairment and brain edema in vivo in a mouse TBI model. Additionally, phillyrin suppressed the phosphorylation of NF-κB in microglia after TBI insult. These effects of phillyrin were mostly abolished by the antagonist of PPARγ. Our results reveal that phillyrin could prominently inhibit the inflammation of microglia via the PPARγ signaling pathway, thus leading to potential neuroprotective treatment after traumatic brain injury.
Collapse
Affiliation(s)
- Qian Jiang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong, University of Science and Technology, Wuhan 430030, PR China
| | - Jun Chen
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong, University of Science and Technology, Wuhan 430030, PR China
| | - Xiaobing Long
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong, University of Science and Technology, Wuhan 430030, PR China
| | - Xiaolong Yao
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong, University of Science and Technology, Wuhan 430030, PR China; Department of Neurosurgery, Taikang Tongji Hospital, Wuhan 430050, PR China
| | - Xin Zou
- Department of Traditional Chinese Medicine, Tongji Hospital, Tongji Medical College, Huazhong, University of Science and Technology, Wuhan 430030, PR China
| | - Yiping Yang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong, University of Science and Technology, Wuhan 430030, PR China
| | - Guangying Huang
- Department of Traditional Chinese Medicine, Tongji Hospital, Tongji Medical College, Huazhong, University of Science and Technology, Wuhan 430030, PR China.
| | - Huaqiu Zhang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong, University of Science and Technology, Wuhan 430030, PR China.
| |
Collapse
|
19
|
Shan D, Ma C, Yang J. Enabling biodegradable functional biomaterials for the management of neurological disorders. Adv Drug Deliv Rev 2019; 148:219-238. [PMID: 31228483 PMCID: PMC6888967 DOI: 10.1016/j.addr.2019.06.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 06/05/2019] [Accepted: 06/17/2019] [Indexed: 02/07/2023]
Abstract
An increasing number of patients are being diagnosed with neurological diseases, but are rarely cured because of the lack of curative therapeutic approaches. This situation creates an urgent clinical need to develop effective diagnosis and treatment strategies for repair and regeneration of injured or diseased neural tissues. In this regard, biodegradable functional biomaterials provide promising solutions to meet this demand owing to their unique responsiveness to external stimulation fields, which enable neuro-imaging, neuro-sensing, specific targeting, hyperthermia treatment, controlled drug delivery, and nerve regeneration. This review discusses recent progress in the research and development of biodegradable functional biomaterials including electroactive biomaterials, magnetic materials and photoactive biomaterials for the management of neurological disorders with emphasis on their applications in bioimaging (photoacoustic imaging, MRI and fluorescence imaging), biosensing (electrochemical sensing, magnetic sensing and opical sensing), and therapy strategies (drug delivery, hyperthermia treatment, and tissue engineering). It is expected that this review will provide an insightful discussion on the roles of biodegradable functional biomaterials in the diagnosis and treatment of neurological diseases, and lead to innovations for the design and development of the next generation biodegradable functional biomaterials.
Collapse
Affiliation(s)
- Dingying Shan
- Department of Biomedical Engineering, Materials Research Institute, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Chuying Ma
- Department of Biomedical Engineering, Materials Research Institute, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Jian Yang
- Department of Biomedical Engineering, Materials Research Institute, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA.
| |
Collapse
|
20
|
Longitudinal Molecular Magnetic Resonance Imaging of Endothelial Activation after Severe Traumatic Brain Injury. J Clin Med 2019; 8:jcm8081134. [PMID: 31366109 PMCID: PMC6722937 DOI: 10.3390/jcm8081134] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 07/26/2019] [Accepted: 07/29/2019] [Indexed: 12/31/2022] Open
Abstract
Traumatic brain injury (TBI) is a major cause of death and disability. Despite progress in neurosurgery and critical care, patients still lack a form of neuroprotective treatment that can counteract or attenuate injury progression. Inflammation after TBI is a key modulator of injury progression and neurodegeneration, but its spatiotemporal dissemination is only partially known. In vivo approaches to study post-traumatic inflammation longitudinally are pivotal for monitoring injury progression/recovery and the effectiveness of therapeutic approaches. Here, we provide a minimally invasive, highly sensitive in vivo molecular magnetic resonance imaging (MRI) characterization of endothelial activation associated to neuroinflammatory response after severe TBI in mice, using microparticles of iron oxide targeting P-selectin (MPIOs-α-P-selectin). Strong endothelial activation was detected from 24 h in perilesional regions, including the cortex and hippocampus, and peaked in intensity and diffusion at two days, then partially decreased but persisted up to seven days and was back to baseline 15 days after injury. There was a close correspondence between MPIOs-α-P-selectin signal voids and the P-selectin stained area, confirming maximal endothelial activation at two days. Molecular MRI markers of inflammation may thus represent a useful tool to evaluate in vivo endothelial activation in TBI and monitoring the responses to therapeutic agents targeting vascular activation and permeability.
Collapse
|
21
|
Herzog C, Pons Garcia L, Keatinge M, Greenald D, Moritz C, Peri F, Herrgen L. Rapid clearance of cellular debris by microglia limits secondary neuronal cell death after brain injury in vivo. Development 2019; 146:146/9/dev174698. [PMID: 31076485 DOI: 10.1242/dev.174698] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 04/11/2019] [Indexed: 12/25/2022]
Abstract
Moderate or severe traumatic brain injury (TBI) causes widespread neuronal cell death. Microglia, the resident macrophages of the brain, react to injury by migrating to the lesion site, where they phagocytose cellular debris. Microglial phagocytosis can have both beneficial (e.g. debris clearance) and detrimental (e.g. respiratory burst, phagoptosis) consequences. Hence, whether the overall effect of microglial phagocytosis after brain injury in vivo is neuroprotective or neurotoxic is not known. Here, we establish a system with which to carry out dynamic real-time analyses of the mechanisms regulating cell death after brain injury in vivo We show that mechanical injury to the larval zebrafish brain induces distinct phases of primary and secondary cell death. Excitotoxicity contributes to secondary cell death in zebrafish, reflecting findings from mammals. Microglia arrive at the lesion site within minutes of injury, where they rapidly engulf dead cells. Importantly, the rate of secondary cell death is increased when the rapid removal of cellular debris by microglia is reduced pharmacologically or genetically. In summary, our results provide evidence that microglial debris clearance is neuroprotective after brain injury in vivo.
Collapse
Affiliation(s)
- Chiara Herzog
- Centre for Discovery Brain Sciences, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Laura Pons Garcia
- Centre for Discovery Brain Sciences, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Marcus Keatinge
- Centre for Discovery Brain Sciences, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - David Greenald
- Centre for Discovery Brain Sciences, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | | | - Francesca Peri
- Institute of Molecular Life Sciences, University of Zürich, Winterthurerstraße 190, 8057 Zürich, Switzerland
| | - Leah Herrgen
- Centre for Discovery Brain Sciences, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| |
Collapse
|
22
|
Continuous remote ischemic conditioning attenuates cognitive and motor deficits from moderate traumatic brain injury. J Trauma Acute Care Surg 2019; 85:48-53. [PMID: 29443855 DOI: 10.1097/ta.0000000000001835] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND While studies show that single-dose remote ischemic conditioning (RIC) improves outcomes, the effect of continuous (daily) RIC is unknown. Thus, we aimed to investigate the role of continuous RIC on cognitive and motor function following traumatic brain injury (TBI). METHODS We subjected 24 male C57BL mice to a cortical-controlled TBI. Two hours after TBI, the animals were randomly allocated to the RIC group (n = 12) or the sham group (n = 12). Remote ischemic conditioning was induced by noninvasive external compression of the hind limb using an occlusive band (six 4-minute cycles/24 hours) for six consecutive days. Before TBI, a baseline rotarod test and novel object recognition were performed. Post-TBI rotarod and novel object recognition tests were performed on Days 1 to 5, 7, 14, and 21. After the animals were sacrificed on Day 21, brain sections were analyzed using hematoxylin and eosin and glial fibrillary acidic protein staining to evaluate the hippocampal CA1 area for neuronal injury. RESULTS Both the RIC and sham groups had lower latency to fall compared with the baseline post-TBI. The RIC animals had a higher latency to fall compared with the sham animals at all time points, statistically significant after Day 3, until Day 21 post-TBI. Both the RIC and sham groups had lower recognition index compared with the baseline post-TBI. The RIC animals had a significantly higher recognition index than the sham animals after Day 1, until Day 21 post-TBI. Hematoxylin and eosin and glial fibrillary acidic protein staining of the brain samples of the sham group revealed that more neurons in the hippocampal CA1 area appeared shrunken with eosinophilic cytoplasm and pyknotic nuclei compared with the brain samples of the RIC group. CONCLUSION Postinjury continuous RIC resulted in improved cognitive functions and motor coordination in a mouse model of moderate TBI. Further studies are required to determine optimum dosage and frequency of this novel therapy to maximize its beneficial effects following TBI.
Collapse
|
23
|
Kassi AAY, Mahavadi AK, Clavijo A, Caliz D, Lee SW, Ahmed AI, Yokobori S, Hu Z, Spurlock MS, Wasserman JM, Rivera KN, Nodal S, Powell HR, Di L, Torres R, Leung LY, Rubiano AM, Bullock RM, Gajavelli S. Enduring Neuroprotective Effect of Subacute Neural Stem Cell Transplantation After Penetrating TBI. Front Neurol 2019; 9:1097. [PMID: 30719019 PMCID: PMC6348935 DOI: 10.3389/fneur.2018.01097] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 12/03/2018] [Indexed: 12/13/2022] Open
Abstract
Traumatic brain injury (TBI) is the largest cause of death and disability of persons under 45 years old, worldwide. Independent of the distribution, outcomes such as disability are associated with huge societal costs. The heterogeneity of TBI and its complicated biological response have helped clarify the limitations of current pharmacological approaches to TBI management. Five decades of effort have made some strides in reducing TBI mortality but little progress has been made to mitigate TBI-induced disability. Lessons learned from the failure of numerous randomized clinical trials and the inability to scale up results from single center clinical trials with neuroprotective agents led to the formation of organizations such as the Neurological Emergencies Treatment Trials (NETT) Network, and international collaborative comparative effectiveness research (CER) to re-orient TBI clinical research. With initiatives such as TRACK-TBI, generating rich and comprehensive human datasets with demographic, clinical, genomic, proteomic, imaging, and detailed outcome data across multiple time points has become the focus of the field in the United States (US). In addition, government institutions such as the US Department of Defense are investing in groups such as Operation Brain Trauma Therapy (OBTT), a multicenter, pre-clinical drug-screening consortium to address the barriers in translation. The consensus from such efforts including "The Lancet Neurology Commission" and current literature is that unmitigated cell death processes, incomplete debris clearance, aberrant neurotoxic immune, and glia cell response induce progressive tissue loss and spatiotemporal magnification of primary TBI. Our analysis suggests that the focus of neuroprotection research needs to shift from protecting dying and injured neurons at acute time points to modulating the aberrant glial response in sub-acute and chronic time points. One unexpected agent with neuroprotective properties that shows promise is transplantation of neural stem cells. In this review we present (i) a short survey of TBI epidemiology and summary of current care, (ii) findings of past neuroprotective clinical trials and possible reasons for failure based upon insights from human and preclinical TBI pathophysiology studies, including our group's inflammation-centered approach, (iii) the unmet need of TBI and unproven treatments and lastly, (iv) present evidence to support the rationale for sub-acute neural stem cell therapy to mediate enduring neuroprotection.
Collapse
Affiliation(s)
- Anelia A. Y. Kassi
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Anil K. Mahavadi
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Angelica Clavijo
- Neurosurgery Service, INUB-MEDITECH Research Group, El Bosque University, Bogotá, CO, United States
| | - Daniela Caliz
- Neurosurgery Service, INUB-MEDITECH Research Group, El Bosque University, Bogotá, CO, United States
| | - Stephanie W. Lee
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Aminul I. Ahmed
- Wessex Neurological Centre, University Hospitals Southampton, Southampton, United Kingdom
| | - Shoji Yokobori
- Department of Emergency and Critical Care Medicine, Nippon Medical School, Tokyo, Japan
| | - Zhen Hu
- Department of Neurosurgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Markus S. Spurlock
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Joseph M Wasserman
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Karla N. Rivera
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Samuel Nodal
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Henry R. Powell
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Long Di
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Rolando Torres
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Lai Yee Leung
- Branch of Brain Trauma Neuroprotection and Neurorestoration, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
- Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Andres Mariano Rubiano
- Neurosurgery Service, INUB-MEDITECH Research Group, El Bosque University, Bogotá, CO, United States
| | - Ross M. Bullock
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Shyam Gajavelli
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
24
|
Kusmenkov T, Braunstein M, Schneider HJ, Bidlingmaier M, Prall WC, Flatz W, Boecker W, Bogner V. Initial free cortisol dynamics following blunt multiple trauma and traumatic brain injury: A clinical study. J Int Med Res 2019; 47:1185-1194. [PMID: 30616490 PMCID: PMC6421390 DOI: 10.1177/0300060518819603] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Objective To determine free and total cortisol serum concentrations in the first 24 h
after trauma and to evaluate the influence of traumatic brain injury (TBI)
on their dynamics. Methods This prospective cohort study enrolled patients who had experienced multiple
trauma and were admitted to a level 1 trauma centre. The patients were
divided in two groups based on the presence of TBI according to clinical and
radiological findings. Blood was collected initially as well as at 12 h and
24 h after the traumatic injury. Total cortisol, corticosteroid binding
globulin (CBG) and free cortisol levels were determined. Results The study analysed data from 49 patients (36 males and 13 females) with a
mean ± SD age of 45.0 ± 16.0 years. Of these, 36 presented with TBI and 13
had multiple injuries without TBI. Patients with TBI showed significantly
lower concentrations of total cortisol and free cortisol compared with
patients without TBI. Repeated measures analysis revealed different
concentration dynamics in patients with TBI, with no increase in cortisol
after trauma. Conclusion Multiple trauma patients with TBI are at risk of acute impaired cortisol
secretion and show an attenuated stress response as early as 12 h after
injury.
Collapse
Affiliation(s)
- T Kusmenkov
- 1 Department of Trauma Surgery, Ludwig Maximilians University Munich, Munich, Germany
| | - M Braunstein
- 1 Department of Trauma Surgery, Ludwig Maximilians University Munich, Munich, Germany
| | - H J Schneider
- 2 Department of Internal Medicine IV: Endocrinology, Ludwig Maximilians University Munich, Munich, Germany
| | - M Bidlingmaier
- 2 Department of Internal Medicine IV: Endocrinology, Ludwig Maximilians University Munich, Munich, Germany
| | - W C Prall
- 1 Department of Trauma Surgery, Ludwig Maximilians University Munich, Munich, Germany
| | - W Flatz
- 3 Department of Radiology, Ludwig Maximilians University Munich, Munich, Germany
| | - W Boecker
- 1 Department of Trauma Surgery, Ludwig Maximilians University Munich, Munich, Germany
| | - V Bogner
- 1 Department of Trauma Surgery, Ludwig Maximilians University Munich, Munich, Germany
| |
Collapse
|
25
|
Chiu LS, Anderton RS, Cross JL, Clark VW, Knuckey NW, Meloni BP. Poly-arginine Peptide R18D Reduces Neuroinflammation and Functional Deficits Following Traumatic Brain Injury in the Long-Evans Rat. Int J Pept Res Ther 2019. [DOI: 10.1007/s10989-018-09799-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
26
|
Scaled traumatic brain injury results in unique metabolomic signatures between gray matter, white matter, and serum in a piglet model. PLoS One 2018; 13:e0206481. [PMID: 30379914 PMCID: PMC6209298 DOI: 10.1371/journal.pone.0206481] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 10/12/2018] [Indexed: 01/08/2023] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death and long-term disability in the United States. The heterogeneity of the disease coupled with the lack of comprehensive, standardized scales to adequately characterize multiple types of TBI remain to be major challenges facing effective therapeutic development. A systems level approach to TBI diagnosis through the use of metabolomics could lead to a better understanding of cellular changes post-TBI and potential therapeutic targets. In the current study, we utilize a GC-MS untargeted metabolomics approach to demonstrate altered metabolism in response to TBI in a translational pig model, which possesses many neuroanatomical and pathophysiologic similarities to humans. TBI was produced by controlled cortical impact (CCI) in Landrace piglets with impact velocity and depth of depression set to 2m/s;6mm, 4m/s;6mm, 4m/s;12mm, or 4m/s;15mm resulting in graded neural injury. Serum samples were collected pre-TBI, 24 hours post-TBI, and 7 days post-TBI. Partial least squares discriminant analysis (PLS-DA) revealed that each impact parameter uniquely influenced the metabolomic profile after TBI, and gray and white matter responds differently to TBI on the biochemical level with evidence of white matter displaying greater metabolic change. Furthermore, pathway analysis revealed unique metabolic signatures that were dependent on injury severity and brain tissue type. Metabolomic signatures were also detected in serum samples which potentially captures both time after injury and injury severity. These findings provide a platform for the development of a more accurate TBI classification scale based unique metabolomic signatures.
Collapse
|
27
|
Fan XX, Hao YY, Guo SW, Zhao XP, Xiang Y, Feng FX, Liang GT, Dong YW. Knockdown of RTN1-C attenuates traumatic neuronal injury through regulating intracellular Ca 2+ homeostasis. Neurochem Int 2018; 121:19-25. [PMID: 30352262 DOI: 10.1016/j.neuint.2018.10.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 09/28/2018] [Accepted: 10/18/2018] [Indexed: 11/24/2022]
Abstract
Reticulons (RTNs) are a family of membrane-bound proteins that are dominantly localized to the endoplasmic reticulum (ER) membrane. RTN1-C is one member of RTNs abundantly expressed in the brain and has been shown to mediate neuronal injury in cerebral ischemia models. In the present study, we investigated the role of RTN1-C in an in vitro brain trauma model mimicked by traumatic neuronal injury (TNI) in primary cultured cortical neurons. TNI increased the expression of RTN1-C in cortical neurons but had no effect on RTN1-A and RTN1-B. Knockdown of RTN1-C with specific siRNA (Si-RTN1-C) significantly decreased cytotoxicity and apoptosis after TNI. The results of Ca2+ imaging showed that intracellular Ca2+ overload induced by TNI was attenuated by RTN1-C knockdown. Furthermore, the activation of metabotropic glutamate receptor 1 (mGluR1)-induced Ca2+ response was partially prevented by Si-RTN1-C transfection. We also evaluated the role of RTN1-C in store-operated Ca2+ entry (SOCE) in cortical neurons using the ER Ca2+ inducer thapsigargin (Tg). The results showed that knockdown of RTN1-C alleviated the SOCE-mediated Ca2+ influx and decreased the expression of stromal interactive molecule 1 (STIM1). In summary, the present study found that knockdown of RTN1-C protected neurons against TNI via preservation of intracellular Ca2+ homeostasis, which was associated with the inhibition of mGluR1-mediated ER Ca2+ release and suppression of STIM1-related SOCE. Thus, RTN1-C might represent a therapeutic target for traumatic brain injury (TBI) research.
Collapse
Affiliation(s)
- Xiao-Xuan Fan
- Neurosurgery Department, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China; Neurosurgery Department, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, 712000, Shaanxi, China
| | - Yu-Ying Hao
- Neurosurgery Department, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, 712000, Shaanxi, China
| | - Shi-Wen Guo
- Neurosurgery Department, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| | - Xiao-Ping Zhao
- Neurosurgery Department, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, 712000, Shaanxi, China
| | - Yi Xiang
- Neurosurgery Department, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Fei-Xue Feng
- Medical Inspection Center, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, 712000, Shaanxi, China
| | - Ge-Ting Liang
- Neurosurgery Department, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, 712000, Shaanxi, China
| | - Yu-Wei Dong
- Neurosurgery Department, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| |
Collapse
|
28
|
Wu H, Li J, Xu D, Zhang Q, Cui T. Growth Differentiation Factor 5 Improves Neurogenesis and Functional Recovery in Adult Mouse Hippocampus Following Traumatic Brain Injury. Front Neurol 2018; 9:592. [PMID: 30083129 PMCID: PMC6064945 DOI: 10.3389/fneur.2018.00592] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 07/02/2018] [Indexed: 01/01/2023] Open
Abstract
The aim of this study was to investigate the therapeutic effect of growth differentiation factor 5 (GDF-5) on traumatic brain injury (TBI) in mice. We utilized a controlled cortical impact to establish a mouse TBI model, and then stereotaxically administered 25 or 100 ng GDF-5 into the bilateral hippocampal dentate gyrus (DG) of each of the animals. Seven days after the injury, some of the animals were sacrificed for immunohistochemical and immunofluorescence examination of 5-bromo-2'-deoxyuridine (BrdU), Sox-2, doublecortin (DCX) and phosphorylated cAMP response element binding protein (p-CREB). Dendrite quantification was also performed using DCX positive cells. Activation of newborn neurons was assessed 35 days after the injury. The remaining animals were subjected to open field, Y maze and contextual fear conditioning tests 2 months after TBI. As a result, we found that post-injury stereotaxical administration of GDF-5 can improve neural stem cell proliferation and differentiation in the DG of the hippocampus, evidenced by the increase in BrdU, Sox-2, and DCX-labeled cells, as well as the improvement in dendrite arborization and newborn neuron activation in response to GDF-5 treatment. Mechanistically, these effects of GDF-5 may be mediated by the CREB pathway, manifested by the recovery of TBI-induced dephosphorylation of CREB upon GDF-5 administration. Behavioral tests further verified the effects of GDF-5 on improving cognitive and behavioral dysfunction after TBI. Collectively, these results reveal that direct injection of GDF-5 into the hippocampus can stimulate neurogenesis and improve functional recovery in a mouse TBI model, indicating the potential therapeutic effects of GDF-5 on TBI.
Collapse
Affiliation(s)
- Hongjie Wu
- Department of Neurosurgery The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Jing Li
- Department of Neurosurgery The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Dongxiao Xu
- Department of Neurosurgery The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Qiansheng Zhang
- Department of Neurosurgery The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Tao Cui
- Department of Neurosurgery The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
29
|
Puntambekar SS, Saber M, Lamb BT, Kokiko-Cochran ON. Cellular players that shape evolving pathology and neurodegeneration following traumatic brain injury. Brain Behav Immun 2018; 71:9-17. [PMID: 29601944 DOI: 10.1016/j.bbi.2018.03.033] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 03/16/2018] [Accepted: 03/26/2018] [Indexed: 11/28/2022] Open
Abstract
Traumatic brain injury (TBI) is one of the leading causes of death and disability worldwide, and has emerged as a critical risk factor for multiple neurodegenerative diseases, particularly Alzheimer's disease (AD). How the inflammatory cascade resulting from mechanical stress, axonal shearing and the loss of neurons and glia following initial impact in TBI, contributes to the development of AD-like disease is unclear. Neuroinflammation, characterized by blood-brain barrier (BBB) dysfunction and activation of brain-resident microglia and astrocytes, resulting in secretion of inflammatory mediators and subsequent recruitment of peripheral immune cells has been the focus of extensive research in attempts to identify drug-targets towards improving functional outcomes post TBI. While knowledge of intricate cellular interactions that shape lesion pathophysiology is incomplete, a major limitation in the field is the lack of understanding of how distinct cell types differentially alter TBI pathology. The aim of this review is to highlight functional differences between populations of bone marrow derived, infiltrating monocytes/macrophages and brain-resident microglia based on differential expression of the chemokine receptors CCR2 and CX3CR1. This review will focus on how unique subsets of mononuclear phagocytes shape TBI pathophysiology, neurotoxicity and BBB function, in a disease-stage dependent manner. Additionally, this review summarizes the role of multiple microglia and macrophage receptors, namely CCR2, CX3CR1 and Triggering Receptor Expressed on Myeloid Cells-2 (TREM2) in pathological neuroinflammation and neurodegeneration vs. recovery following TBI. TREM2 has been implicated in mediating AD-related pathology, and variants in TREM2 are particularly important due to their correlation with exacerbated neurodegeneration. Finally, this review highlights behavioral outcomes associated with microglial vs. macrophage variances, the need for novel treatment strategies that target unique subpopulations of peripheral macrophages, and the importance of development of therapeutics to modulate inflammatory functions of brain-resident microglia at specific stages of TBI.
Collapse
Affiliation(s)
- Shweta S Puntambekar
- Stark Neuroscience Research Institute, Indiana University-Purdue University, Indianapolis, IN 46202, USA.
| | - Maha Saber
- Barrows Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ 85016, USA.
| | - Bruce T Lamb
- Stark Neuroscience Research Institute, Indiana University-Purdue University, Indianapolis, IN 46202, USA.
| | - Olga N Kokiko-Cochran
- Department of Neuroscience, The Ohio State University, Institute for Behavioral Medicine Research, Columbus, OH, 43210, USA.
| |
Collapse
|
30
|
Xiong Y, Mahmood A, Chopp M. Current understanding of neuroinflammation after traumatic brain injury and cell-based therapeutic opportunities. Chin J Traumatol 2018; 21:137-151. [PMID: 29764704 PMCID: PMC6034172 DOI: 10.1016/j.cjtee.2018.02.003] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 03/02/2018] [Accepted: 03/05/2018] [Indexed: 02/04/2023] Open
Abstract
Traumatic brain injury (TBI) remains a major cause of death and disability worldwide. Increasing evidence indicates that TBI is an important risk factor for neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, and chronic traumatic encephalopathy. Despite improved supportive and rehabilitative care of TBI patients, unfortunately, all late phase clinical trials in TBI have yet to yield a safe and effective neuroprotective treatment. The disappointing clinical trials may be attributed to variability in treatment approaches and heterogeneity of the population of TBI patients as well as a race against time to prevent or reduce inexorable cell death. TBI is not just an acute event but a chronic disease. Among many mechanisms involved in secondary injury after TBI, emerging preclinical studies indicate that posttraumatic prolonged and progressive neuroinflammation is associated with neurodegeneration which may be treatable long after the initiating brain injury. This review provides an overview of recent understanding of neuroinflammation in TBI and preclinical cell-based therapies that target neuroinflammation and promote functional recovery after TBI.
Collapse
Affiliation(s)
- Ye Xiong
- Department of Neurosurgery Henry Ford Health System, 2799 West Grand Boulevard, Detroit, MI, 48202, USA.
| | - Asim Mahmood
- Department of Neurosurgery Henry Ford Health System, 2799 West Grand Boulevard, Detroit, MI, 48202, USA
| | - Michael Chopp
- Department of Neurology, Henry Ford Health System, 2799 West Grand Boulevard, Detroit, MI, 48202, USA; Department of Physics, Oakland University, Rochester, MI, 48309, USA
| |
Collapse
|
31
|
Therapeutic potential of vitamin E and its derivatives in traumatic brain injury-associated dementia. Neurol Sci 2018; 39:989-998. [DOI: 10.1007/s10072-018-3398-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 03/28/2018] [Indexed: 01/01/2023]
|
32
|
Skrifvars MB, French C, Bailey M, Presneill J, Nichol A, Little L, Durantea J, Huet O, Haddad S, Arabi Y, McArthur C, Cooper DJ, Bellomo R, for the EPO-TBI Investigators and t. Cause and Timing of Death and Subgroup Differential Effects of Erythropoietin in the EPO-TBI Study. J Neurotrauma 2018; 35:333-340. [DOI: 10.1089/neu.2017.5135] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Markus B. Skrifvars
- Australian and New Zealand Intensive Care Research Centre, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
- Division of Intensive Care, Department of Anaesthesiology, Intensive Care and Pain Medicine, Helsinki University and Helsinki University Hospital, Helsinki, Finland
| | - Craig French
- Department of Intensive Care, Western Health, Melbourne, Victoria, Australia
- North West Academic Centre, University of Melbourne, Melbourne, Victoria, Australia
| | - Michael Bailey
- Australian and New Zealand Intensive Care Research Centre, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Jeffrey Presneill
- Australian and New Zealand Intensive Care Research Centre, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
- Department of Intensive Care, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Alistair Nichol
- Australian and New Zealand Intensive Care Research Centre, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
- School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
- St Vincent's University Hospital, Dublin, Ireland
- Department of Intensive Care and Hyperbaric Medicine, The Alfred, Melbourne, Victoria, Australia
| | - Lorraine Little
- Australian and New Zealand Intensive Care Research Centre, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Jacques Durantea
- Département d'Anesthésie-Réanimation, Hôpital de Bicêtre, Assistance Publique des Hopitaux de Paris, Hôpitaux Universitaires Paris-Sud, Université Paris-Sud, Paris, France
| | - Olivier Huet
- Department of Anaesthesiology and Intensive Care Medicine, CHU La Cavale Blanche, Brest, France
| | - Samir Haddad
- King Saud Bin Abdulaziz University for Health Sciences and King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Yaseen Arabi
- King Saud Bin Abdulaziz University for Health Sciences and King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | | | - D. James Cooper
- Australian and New Zealand Intensive Care Research Centre, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
- Department of Intensive Care and Hyperbaric Medicine, The Alfred, Melbourne, Victoria, Australia
| | - Rinaldo Bellomo
- Australian and New Zealand Intensive Care Research Centre, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
- Department of Intensive Care, Austin Health, Melbourne, Victoria, Australia
| | | |
Collapse
|
33
|
Neuroimmunology of Traumatic Brain Injury: Time for a Paradigm Shift. Neuron 2017; 95:1246-1265. [PMID: 28910616 DOI: 10.1016/j.neuron.2017.07.010] [Citation(s) in RCA: 496] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 07/07/2017] [Accepted: 07/10/2017] [Indexed: 12/14/2022]
Abstract
Traumatic brain injury (TBI) is a leading cause of morbidity and disability, with a considerable socioeconomic burden. Heterogeneity of pathoanatomical subtypes and diversity in the pathogenesis and extent of injury contribute to differences in the course and outcome of TBI. Following the primary injury, extensive and lasting damage is sustained through a complex cascade of events referred to as "secondary injury." Neuroinflammation is proposed as an important manipulable aspect of secondary injury in animal and human studies. Because neuroinflammation can be detrimental or beneficial, before developing immunomodulatory therapies, it is necessary to better understand the timing and complexity of the immune responses that follow TBI. With a rapidly increasing body of literature, there is a need for a clear summary of TBI neuroimmunology. This review presents our current understanding of the immune response to TBI in a chronological and compartment-based manner, highlighting early changes in gene expression and initial signaling pathways that lead to activation of innate and adaptive immunity. Based on recent advances in our understanding of innate immune cell activation, we propose a new paradigm to study innate immune cells following TBI that moves away from the existing M1/M2 classification of activation states toward a stimulus- and disease-specific understanding of polarization state based on transcriptomic and proteomic profiling.
Collapse
|
34
|
Paterno R, Folweiler KA, Cohen AS. Pathophysiology and Treatment of Memory Dysfunction After Traumatic Brain Injury. Curr Neurol Neurosci Rep 2017; 17:52. [PMID: 28500417 PMCID: PMC5861722 DOI: 10.1007/s11910-017-0762-x] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Memory is fundamental to everyday life, and cognitive impairments resulting from traumatic brain injury (TBI) have devastating effects on TBI survivors. A contributing component to memory impairments caused by TBI is alteration in the neural circuits associated with memory function. In this review, we aim to bring together experimental findings that characterize behavioral memory deficits and the underlying pathophysiology of memory-involved circuits after TBI. While there is little doubt that TBI causes memory and cognitive dysfunction, it is difficult to conclude which memory phase, i.e., encoding, maintenance, or retrieval, is specifically altered by TBI. This is most likely due to variation in behavioral protocols and experimental models. Additionally, we review a selection of experimental treatments that hold translational potential to mitigate memory dysfunction following injury.
Collapse
Affiliation(s)
- Rosalia Paterno
- Center for Sleep and Circadian Neurobiology, Perelman School of Medicine, University of Pennsylvania, 3615 Civic Center Boulevard, Abramson Research Center, Rm. 816-h, Philadelphia, PA, 19104, USA.
| | - Kaitlin A Folweiler
- Department of Anesthesiology and Critical Care Medicine, Joseph Stokes, Jr. Research Institute, Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Abramson Research Center, Rm. 816-h, Philadelphia, PA, 19104, USA
| | - Akiva S Cohen
- Department of Anesthesiology and Critical Care Medicine, Joseph Stokes, Jr. Research Institute, Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Abramson Research Center, Rm. 816-h, Philadelphia, PA, 19104, USA
- Department of Anesthesiology and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, 3615 Civic Center Boulevard, Abramson Research Center, Rm. 816-h, Philadelphia, PA, 19104, USA
| |
Collapse
|
35
|
Mirzaei S, Reinig AM, Berlau DJ. Translational obstacles with off-label drug use in acute traumatic brain injury. FUTURE NEUROLOGY 2017. [DOI: 10.2217/fnl-2016-0032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Traumatic brain injury results in significant morbidity and mortality, and there is an urgent need for neuroprotective medications that can prevent the persisting symptoms and disabilities following injury. Several existing pharmacotherapies have been targeted for off-label benefit in traumatic brain injury, as these agents are well characterized and commercially available, easing the process of clinical trial development. Despite promising results in animal models, clinical trials have demonstrated minimal benefit. One possible reason for these failed translations could be that drug selection, characterization and dosing are not routinely established in the appropriate early phase trials before larger scale testing. Examining how recent trials may have bypassed these steps may help future trials to more definitively determine the efficacy of potential therapeutics.
Collapse
Affiliation(s)
- Sara Mirzaei
- Rueckert-Hartman College for Health Professions, Regis University, 3333 Regis Blvd, H-28, Denver, CO 80221, USA
| | - Andrea M Reinig
- Rueckert-Hartman College for Health Professions, Regis University, 3333 Regis Blvd, H-28, Denver, CO 80221, USA
| | - Daniel J Berlau
- Rueckert-Hartman College for Health Professions, Regis University, 3333 Regis Blvd, H-28, Denver, CO 80221, USA
| |
Collapse
|
36
|
Venegoni W, Shen Q, Thimmesch AR, Bell M, Hiebert JB, Pierce JD. The use of antioxidants in the treatment of traumatic brain injury. J Adv Nurs 2017; 73:1331-1338. [PMID: 28103389 DOI: 10.1111/jan.13259] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2017] [Indexed: 11/26/2022]
Abstract
AIMS The aim of this study was to discuss secondary traumatic brain injury, the mitochondria and the use of antioxidants as a treatment. BACKGROUND One of the leading causes of death globally is traumatic brain injury, affecting individuals in all demographics. Traumatic brain injury is produced by an external blunt force or penetration resulting in alterations in brain function or pathology. Often, with a traumatic brain injury, secondary injury causes additional damage to the brain tissue that can have further impact on recovery and the quality of life. Secondary injury occurs when metabolic and physiologic processes alter after initial injury and includes increased release of toxic free radicals that cause damage to adjacent tissues and can eventually lead to neuronal necrosis. Although antioxidants in the tissues can reduce free radical damage, the magnitude of increased free radicals overwhelms the body's reduced defence mechanisms. Supplementing the body's natural supply of antioxidants, such as coenzyme Q10, can attenuate oxidative damage caused by reactive oxygen species. DESIGN Discussion paper. DATA SOURCES Research literature published from 2011-2016 in PubMed, CINAHL and Cochrane. IMPLICATIONS FOR NURSING Prompt and accurate assessment of patients with traumatic brain injury by nurses is important to ensure optimal recovery and reduced lasting disability. Thus, it is imperative that nurses be knowledgeable about the secondary injury that occurs after a traumatic brain injury and aware of possible antioxidant treatments. CONCLUSION The use of antioxidants has potential to reduce the magnitude of secondary injury in patients who experience a traumatic brain injury.
Collapse
Affiliation(s)
| | - Qiuhua Shen
- School of Nursing, University of Kansas, Kansas, USA
| | | | - Meredith Bell
- School of Nursing, University of Kansas, Kansas, USA
| | | | | |
Collapse
|
37
|
Corrigan F, Arulsamy A, Teng J, Collins-Praino LE. Pumping the Brakes: Neurotrophic Factors for the Prevention of Cognitive Impairment and Dementia after Traumatic Brain Injury. J Neurotrauma 2016; 34:971-986. [PMID: 27630018 DOI: 10.1089/neu.2016.4589] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) is the leading cause of disability and death worldwide, affecting as many as 54,000,000-60,000,000 people annually. TBI is associated with significant impairments in brain function, impacting cognitive, emotional, behavioral, and physical functioning. Although much previous research has focused on the impairment immediately following injury, TBI may have much longer-lasting consequences, including neuropsychiatric disorders and cognitive impairment. TBI, even mild brain injury, has also been recognized as a significant risk factor for the later development of dementia and Alzheimer's disease. Although the link between TBI and dementia is currently unknown, several proposed mechanisms have been put forward, including alterations in glucose metabolism, excitotoxicity, calcium influx, mitochondrial dysfunction, oxidative stress, and neuroinflammation. A treatment for the devastating long-term consequences of TBI is desperately needed. Unfortunately, however, no such treatment is currently available, making this a major area of unmet medical need. Increasing the level of neurotrophic factor expression in key brain areas may be one potential therapeutic strategy. Of the neurotrophic factors, granulocyte-colony stimulating factor (G-CSF) may be particularly effective for preventing the emergence of long-term complications of TBI, including dementia, because of its ability to reduce apoptosis, stimulate neurogenesis, and increase neuroplasticity.
Collapse
Affiliation(s)
- Frances Corrigan
- Translational Neuropathology Lab, Discipline of Anatomy and Pathology, School of Medicine, University of Adelaide , Adelaide, Australia
| | - Alina Arulsamy
- Translational Neuropathology Lab, Discipline of Anatomy and Pathology, School of Medicine, University of Adelaide , Adelaide, Australia
| | - Jason Teng
- Translational Neuropathology Lab, Discipline of Anatomy and Pathology, School of Medicine, University of Adelaide , Adelaide, Australia
| | - Lyndsey E Collins-Praino
- Translational Neuropathology Lab, Discipline of Anatomy and Pathology, School of Medicine, University of Adelaide , Adelaide, Australia
| |
Collapse
|