1
|
Grandizoli Saletti P, Casillas-Espinosa PM, Panagiotis Lisgaras C, Bi Mowrey W, Li Q, Liu W, Brady RD, Ali I, Silva J, Yamakawa G, Hudson M, Li C, Braine EL, Coles L, Cloyd JC, Jones NC, Shultz SR, Moshé SL, O'Brien TJ, Galanopoulou AS. Tau Phosphorylation Patterns in the Rat Cerebral Cortex After Traumatic Brain Injury and Sodium Selenate Effects: An Epibios4rx Project 2 Study. J Neurotrauma 2024; 41:222-243. [PMID: 36950806 PMCID: PMC11079442 DOI: 10.1089/neu.2022.0219] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2023] Open
Abstract
Sodium selenate (SS) activates protein phosphatase 2 (PP2A) and reduces phosphorylated tau (pTAU) and late post-traumatic seizures after lateral fluid percussion injury (LFPI). In EpiBioS4Rx Project 2, a multi-center international study for post-traumatic targets, biomarkers, and treatments, we tested the target relevance and modification by SS of pTAU forms and PP2A and in the LFPI model, at two sites: Einstein and Melbourne. In Experiment 1, adult male rats were assigned to LFPI and sham (both sites) and naïve controls (Einstein). Motor function was monitored by neuroscores. Brains were studied with immunohistochemistry (IHC), Western blots (WBs), or PP2A activity assay, from 2 days to 8 weeks post-operatively. In Experiment 2, LFPI rats received SS for 7 days (SS0.33: 0.33 mg/kg/day; SS1: 1 mg/kg/day, subcutaneously) or vehicle (Veh) post-LFPI and pTAU, PR55 expression, or PP2A activity were studied at 2 days and 1 week (on treatment), or 2 weeks (1 week off treatment). Plasma selenium and SS levels were measured. In Experiment 1 IHC, LFPI rats had higher cortical pTAU-Ser202/Thr205-immunoreactivity (AT8-ir) and pTAU-Ser199/202-ir at 2 days, and pTAU-Thr231-ir (AT180-ir) at 2 days, 2 weeks, and 8 weeks, ipsilaterally to LFPI, than controls. LFPI-2d rats also had higher AT8/total-TAU5-ir in cortical extracts ipsilateral to the lesion (WB). PP2A (PR55-ir) showed time- and region-dependent changes in IHC, but not in WB. PP2A activity was lower in LFPI-1wk than in sham rats. In Experiment 2, SS did not affect neuroscores or cellular AT8-ir, AT180-ir, or PR55-ir in IHC. In WB, total cortical AT8/total-TAU-ir was lower in SS0.33 and SS1 LFPI rats than in Veh rats (2 days, 1 week); total cortical PR55-ir (WB) and PP2A activity were higher in SS1 than Veh rats (2 days). SS dose dependently increased plasma selenium and SS levels. Concordant across-sites data confirm time and pTAU form-specific cortical increases ipsilateral to LFPI. The discordant SS effects may either suggest SS-induced reduction in the numbers of cells with increased pTAU-ir, need for longer treatment, or the involvement of other mechanisms of action.
Collapse
Affiliation(s)
- Patricia Grandizoli Saletti
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx New York, USA
| | - Pablo M. Casillas-Espinosa
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
- Department of Neurology, Alfred Health, Melbourne, Australia
| | - Christos Panagiotis Lisgaras
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx New York, USA
| | - Wenzhu Bi Mowrey
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx New York, USA
| | - Qianyun Li
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx New York, USA
| | - Wei Liu
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx New York, USA
| | - Rhys D. Brady
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
| | - Idrish Ali
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
| | - Juliana Silva
- Department of Neuroscience, Monash University, Melbourne, Australia
| | - Glenn Yamakawa
- Department of Medicine, The University of Melbourne, Parkville, Australia
| | - Matt Hudson
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
| | - Crystal Li
- Department of Neuroscience, Monash University, Melbourne, Australia
| | - Emma L. Braine
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
| | - Lisa Coles
- University of Minnesota Twin Cities, Minneapolis, Minnesota, USA
| | - James C. Cloyd
- University of Minnesota Twin Cities, Minneapolis, Minnesota, USA
| | - Nigel C. Jones
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
- Department of Neurology, Alfred Health, Melbourne, Australia
| | - Sandy R. Shultz
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
- Department of Neurology, Alfred Health, Melbourne, Australia
| | - Solomon L. Moshé
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx New York, USA
- Isabelle Rapin Division of Child Neurology, Albert Einstein College of Medicine, Bronx New York, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx New York, USA
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx New York, USA
| | - Terence J. O'Brien
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
- Department of Neurology, Alfred Health, Melbourne, Australia
| | - Aristea S. Galanopoulou
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx New York, USA
- Isabelle Rapin Division of Child Neurology, Albert Einstein College of Medicine, Bronx New York, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx New York, USA
| |
Collapse
|
2
|
Wong KR, Wright DK, Sgro M, Salberg S, Bain J, Li C, Sun M, McDonald SJ, Mychasiuk R, Brady RD, Shultz SR. Persistent Changes in Mechanical Nociception in Rats With Traumatic Brain Injury Involving Polytrauma. THE JOURNAL OF PAIN 2023; 24:1383-1395. [PMID: 36958460 DOI: 10.1016/j.jpain.2023.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 03/12/2023] [Accepted: 03/15/2023] [Indexed: 03/25/2023]
Abstract
Traumatic brain injury (TBI) survivors often experience debilitating consequences. Due to the high impact nature of TBI, patients often experience concomitant peripheral injuries (ie, polytrauma). A common, yet often overlooked, comorbidity of TBI is chronic pain. Therefore, this study investigated how common concomitant peripheral injuries (ie, femoral fracture and muscle crush) can affect long-term behavioral and structural TBI outcomes with a particular focus on nociception. Rats were randomly assigned to 1 of 4 groups: polytrauma (POLY; ie, fracture + muscle crush + TBI), peripheral injury (PERI; ie, fracture + muscle crush + sham TBI), TBI (ie, sham fracture + sham muscle crush + TBI), and sham-injured (SHAM; ie, sham fracture + sham muscle crush + sham TBI). Rats underwent behavioral testing at 3-, 6-, and 11-weeks postinjury, and were then euthanized for postmortem magnetic resonance imaging (MRI). POLY rats had a persisting increase in pain sensitivity compared to all groups on the von Frey test. MRI revealed that POLY rats also had abnormalities in the cortical and subcortical brain structures involved in nociceptive processing. These findings have important implications and provide a foundation for future studies to determine the underlying mechanisms and potential treatment strategies for chronic pain in TBI survivors. PERSPECTIVE: Rats with TBI and concomitant peripheral trauma displayed chronic nociceptive pain and MRI images also revealed damaged brain structures/pathways that are involved in chronic pain development. This study highlights the importance of polytrauma and the affected brain regions for developing chronic pain.
Collapse
Affiliation(s)
- Ker Rui Wong
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - David K Wright
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Marissa Sgro
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Sabrina Salberg
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Jesse Bain
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Crystal Li
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Mujun Sun
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Stuart J McDonald
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia; Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Bundoora, VIC, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Rhys D Brady
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia; Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia; Department of Medicine, The University of Melbourne, Parkville, VIC, Australia; Department of Nursing, Health and Human Services, Vancouver Island University, Nanaimo, BC, Canada.
| |
Collapse
|
3
|
Sherawat K, Mehan S. Tanshinone-IIA mediated neuroprotection by modulating neuronal pathways. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:1647-1667. [PMID: 37010572 DOI: 10.1007/s00210-023-02476-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 03/23/2023] [Indexed: 04/04/2023]
Abstract
The progression of neurological diseases is mainly attributed to oxidative stress, apoptosis, inflammation, and trauma, making them a primary public concern. Since no drugs can stop these neurological disorders from happening, active phytochemical intervention has been suggested as a possible treatment. Among the several phytochemicals being studied for their potential health advantages, tanshinone-IIA (Tan-IIA ) stands out due to its wide range of therapeutic effects. Tan-IIA, derived from the Salvia miltiorrhiza plant, is a phenanthrenequinone. The pharmacological characteristics of Tan-IIAagainst various neurodegenerative and neuropsychiatric illnesses have led researchers to believe that the compound possesses neuroprotective potential. Tan-IIA has therapeutic potential in treating neurological diseases due to its capacity to cross the blood-brain barrier and its broad range of activities. In treating neurological disorders, Tan-IIA has been shown to have neuroprotective effects such as anti-apoptotic, anti-inflammatory, BBB protectant, and antioxidant properties. This article concisely summarises the latest scientific findings about the cellular and molecular aspects of Tan-IIA neuroprotection in relation to various neurological diseases. The results of preclinical studies on Tan-IIA provide insight into its potential application in future therapeutic development. This molecule rapidly establishes as a prominent bioactive compound for clinical research.
Collapse
Affiliation(s)
- Kajal Sherawat
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India.
| |
Collapse
|
4
|
Brady RD, Bird S, Sun M, Yamakawa GR, Major BP, Mychasiuk R, O'Brien TJ, McDonald SJ, Shultz SR. Activation of the Protein Kinase R-Like Endoplasmic Reticulum Kinase (PERK) Pathway of the Unfolded Protein Response after Experimental Traumatic Brain Injury and Treatment with a PERK Inhibitor. Neurotrauma Rep 2021; 2:330-342. [PMID: 34318301 PMCID: PMC8310749 DOI: 10.1089/neur.2021.0001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Neurodegeneration after traumatic brain injury (TBI) is increasingly recognized as a key factor contributing to poor chronic outcomes. Activation (i.e., phosphorylation) of the protein kinase R-like endoplasmic reticulum kinase (PERK) pathway has been implicated in neurodegenerative conditions with pathological similarities to TBI and may be a potential target to improve TBI outcomes. Here, we aimed to determine whether a moderate TBI would induce activation of the PERK pathway and whether treatment with the PERK inhibitor, GSK2606414, would improve TBI recovery. Male mice were administered a lateral fluid percussion injury (FPI) or sham injury and were euthanized at either 2 h, 24 h, or 1 week post-injury (n = 5 per injury group and time point) to assess changes in the PERK pathway. In the injured cortex, there was increased phosphorylated-PERK at 2 h post-FPI and increased phosphorylation of eukaryotic translation initiation factor α at 24 h post-FPI. We next examined the effect of acute treatment with GSK2606414 on pathological and behavioral outcomes at 4 weeks post-injury. Thus, there were a total of four groups: sham + VEH (n = 9); sham + GSK4606414 (n = 10); FPI + VEH (n = 9); and FPI + GSK2606414 (n = 9). GSK2606414 (50 mg/kg) or vehicle treatment was delivered by oral gavage beginning at 30 min post-injury, followed by two further treatments at 12-h increments. There were no significant effects of GSK2606414 on any of the outcomes assessed, which could be attributable to several reasons. For example, activation of PERK may not be a significant contributor to the neurological consequences 4 weeks post-FPI in mice. Further research is required to elucidate the role of the PERK pathway in TBI and whether interventions that target this pathway are beneficial.
Collapse
Affiliation(s)
- Rhys D Brady
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Stefanie Bird
- Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Mujun Sun
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Glenn R Yamakawa
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Brendan P Major
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Stuart J McDonald
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Physiology, Anatomy, and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
5
|
Deng Y, Jiang X, Deng X, Chen H, Xu J, Zhang Z, Liu G, Yong Z, Yuan C, Sun X, Wang C. Pioglitazone ameliorates neuronal damage after traumatic brain injury via the PPARγ/NF-κB/IL-6 signaling pathway. Genes Dis 2020; 7:253-265. [PMID: 32215295 PMCID: PMC7083749 DOI: 10.1016/j.gendis.2019.05.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 05/12/2019] [Accepted: 05/20/2019] [Indexed: 02/06/2023] Open
Abstract
Traumatic brain injury (TBI) is the major cause of high mortality and disability rates worldwide. Pioglitazone is an activator of peroxisome proliferator-activated receptor-gamma (PPARγ) that can reduce inflammation following TBI. Clinically, neuroinflammation after TBI lacks effective treatment. Although there are many studies on PPARγ in TBI animals, only few could be converted into clinical, since TBI mechanisms in humans and animals are not completely consistent. The present study, provided a potential theoretical basis and therapeutic target for neuroinflammation treatment after TBI. First, we detected interleukin-6 (IL-6), nitric oxide (NO) and Caspase-3 in TBI clinical specimens, confirming a presence of a high expression of inflammatory factors. Western blot (WB), quantitative real-time PCR (qRT-PCR) and immunohistochemistry (IHC) were used to detect PPARγ, IL-6, and p-NF-κB to identify the mechanisms of neuroinflammation. Then, in the rat TBI model, neurobehavioral and cerebral edema levels were investigated after intervention with pioglitazone (PPARγ activator) or T0070907 (PPARγ inhibitor), and PPARγ, IL-6 and p-NF-κB were detected again by qRT-PCR, WB and immunofluorescence (IF). The obtained results revealed that: 1) increased expression of IL-6, NO and Caspase-3 in serum and cerebrospinal fluid in patients after TBI, and decreased PPARγ in brain tissue; 2) pioglitazone could improve neurobehavioral and reduce brain edema in rats after TBI; 3) the protective effect of pioglitazone was achieved by activating PPARγ and reducing NF-κB and IL-6. The neuroprotective effect of pioglitazone on TBI was mediated through the PPARγ/NF-κB/IL-6 pathway.
Collapse
Affiliation(s)
- Yongbing Deng
- Department of Neurosurgery of the First Affiliated Hospital of Chongqing Medical University, Yixueyuan Road #1, Chongqing, 400016, PR China
- Department of Neurosurgery of the Chongqing Emergency Medical Center, Jiankang Road #1, Chongqing, 400014, PR China
| | - Xue Jiang
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Yixueyuan Road #1, Chongqing, 400016, PR China
| | - Xiaoyan Deng
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Yixueyuan Road #1, Chongqing, 400016, PR China
| | - Hong Chen
- Department of Neurosurgery of the First Affiliated Hospital of Chongqing Medical University, Yixueyuan Road #1, Chongqing, 400016, PR China
| | - Jie Xu
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Yixueyuan Road #1, Chongqing, 400016, PR China
| | - Zhaosi Zhang
- Department of Neurosurgery of the First Affiliated Hospital of Chongqing Medical University, Yixueyuan Road #1, Chongqing, 400016, PR China
| | - Geli Liu
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Yixueyuan Road #1, Chongqing, 400016, PR China
| | - Zhu Yong
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Chengfu Yuan
- College of Medical Science, China Three Gorges University, Yichang, Hubei, 443002, PR China
| | - Xiaochuan Sun
- Department of Neurosurgery of the First Affiliated Hospital of Chongqing Medical University, Yixueyuan Road #1, Chongqing, 400016, PR China
| | - Changdong Wang
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Yixueyuan Road #1, Chongqing, 400016, PR China
| |
Collapse
|
6
|
The Attenuation of Traumatic Brain Injury via Inhibition of Oxidative Stress and Apoptosis by Tanshinone IIA. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:4170156. [PMID: 32454938 PMCID: PMC7218958 DOI: 10.1155/2020/4170156] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 10/31/2019] [Accepted: 11/28/2019] [Indexed: 12/17/2022]
Abstract
Traumatic brain injury (TBI) is a major source of mortality and long-term disability worldwide. The mechanisms associated with TBI development are poorly understood, and little progress has been made in the treatment of TBI. Tanshinone IIA is an effective agent to treat a variety of disorders; however, the mechanisms of Tanshinone IIA on TBI remain unclear. The aim of the present study was to investigate the therapeutic potential of Tanshinone IIA on TBI and its underlying molecular mechanisms. Changes in microvascular permeability were examined to determine the extent of TBI with Evans blue dye. Brain edema was assessed by measuring the wet weight to dry weight ratio. The expression levels of CD11, interleukin- (IL-) 1β, and tumor necrosis factor- (TNF-) α mRNA were determined by reverse transcription-quantitative PCR. Aquaporin-4 (AQP4), glial fibrillary acidic protein (GFAP), and p47phox protein expression levels were detected by western blotting. Superoxide dismutase (SOD), catalase and glutathione peroxidase (GSH-PX) activities, and malondialdehyde (MDA) content were determined using commercial kits. Cell apoptosis was detected by western blotting and TUNEL staining. Tanshinone IIA (10 mg/kg/day, intraperitoneal administration) significantly reduced brain water content and vascular permeability at 12, 24, 48, and 72 h after TBI. Tanshinone IIA downregulated the mRNA expression levels of various factors induced by TBI, including CD11, IL-1β, and TNF-α. Notably, CD11 mRNA downregulation suggested that Tanshinone IIA inhibited microglia activation. Further results showed that Tanshinone IIA treatment significantly downregulated AQP4 and GFAP expression. TBI-induced oxidative stress and apoptosis were markedly reversed by Tanshinone IIA, with an increase in SOD and GSH-PX activities and a decrease in the MDA content. Moreover, Tanshinone IIA decreased TBI-induced NADPH oxidase activation via the inhibition of p47phox. Tanshinone IIA attenuated TBI, and its mechanism of action may involve the inhibition of oxidative stress and apoptosis.
Collapse
|
7
|
Anderson J, Patel M, Forenzo D, Ai X, Cai C, Wade Q, Risman R, Cai L. A novel mouse model for the study of endogenous neural stem and progenitor cells after traumatic brain injury. Exp Neurol 2020; 325:113119. [PMID: 31751572 PMCID: PMC10885014 DOI: 10.1016/j.expneurol.2019.113119] [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] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 10/23/2019] [Accepted: 11/16/2019] [Indexed: 11/29/2022]
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability in the US. Neural stem/progenitor cells (NSPCs) persist in the adult brain and represent a potential cell source for tissue regeneration and wound healing after injury. The Notch signaling pathway is critical for embryonic development and adult brain injury response. However, the specific role of Notch signaling in the injured brain is not well characterized. Our previous study has established a Notch1CR2-GFP reporter mouse line in which the Notch1CR2 enhancer directs GFP expression in NSPCs and their progeny. In this study, we performed closed head injury (CHI) in the Notch1CR2-GFP mice to study the response of injury-activated NSPCs. We show that CHI induces neuroinflammation, cell death, and the expression of typical TBI markers (e.g., ApoE, Il1b, and Tau), validating the animal model. In addition, CHI induces cell proliferation in GFP+ cells expressing NSPC markers, e.g., Notch1 and Nestin. A significant higher percentage of GFP+ astrocytes and GABAergic neurons was observed in the injured brain, with no significant change in oligodendrocyte lineage between the CHI and sham animal groups. Since injury is known to activate astrogliosis, our results suggest that injury-induced GFP+ NSPCs preferentially differentiate into GABAergic neurons. Our study establishes that Notch1CR2-GFP transgenic mouse is a useful tool for the study of NSPC behavior in vivo after TBI. Unveiling the potential of NSPCs response to TBI (e.g., proliferation and differentiation) will identify new therapeutic strategy for the treatment of brain trauma.
Collapse
Affiliation(s)
- Jeremy Anderson
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, United States of America
| | - Misaal Patel
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, United States of America
| | - Dylan Forenzo
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, United States of America
| | - Xin Ai
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, United States of America
| | - Catherine Cai
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, United States of America
| | - Quinn Wade
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, United States of America
| | - Rebecca Risman
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, United States of America
| | - Li Cai
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, United States of America.
| |
Collapse
|
8
|
Zvejniece L, Stelfa G, Vavers E, Kupats E, Kuka J, Svalbe B, Zvejniece B, Albert-Weissenberger C, Sirén AL, Plesnila N, Dambrova M. Skull Fractures Induce Neuroinflammation and Worsen Outcomes after Closed Head Injury in Mice. J Neurotrauma 2019; 37:295-304. [PMID: 31441378 PMCID: PMC6964812 DOI: 10.1089/neu.2019.6524] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The weight-drop model is used widely to replicate closed-head injuries in mice; however, the histopathological and functional outcomes may vary significantly between laboratories. Because skull fractures are reported to occur in this model, we aimed to evaluate whether these breaks may influence the variability of the weight-drop (WD) model. Male Swiss Webster mice underwent WD injury with either a 2 or 5 mm cone tip, and behavior was assessed at 2 h and 24 h thereafter using the neurological severity score. The expression of interleukin (IL)-6, IL-1β, tumor necrosis factor-α, matrix metalloproteinase-9, and tissue inhibitor of metalloproteinase-1 genes was measured at 12 h and 1, 3, and 14 days after injury. Before the injury, micro-computed tomography (micro-CT) was performed to quantify skull thickness at the impact site. With a conventional tip diameter of 2 mm, 33% of mice showed fractures of the parietal bone; the 5 mm tip produced only 10% fractures. Compared with mice without fractures, mice with fractures had a severity-dependent worse functional outcome and a more pronounced upregulation of inflammatory genes in the brain. Older mice were associated with thicker parietal bones and were less prone to skull fractures. In addition, mice that underwent traumatic brain injury (TBI) with skull fracture had macroscopic brain damage because of skull depression. Skull fractures explain a considerable proportion of the variability observed in the WD model in mice—i.e., mice with skull fractures have a much stronger inflammatory response than do mice without fractures. Using older mice with thicker skull bones and an impact cone with a larger diameter reduces the rate of skull fractures and the variability in this very useful closed-head TBI model.
Collapse
Affiliation(s)
- Liga Zvejniece
- Latvian Institute of Organic Synthesis, Riga, Latvia
- Address correspondence to: Liga Zvejniece, MD, PhD, Latvian Institute of Organic Synthesis, Aizkraukles 21, LV-1006, Riga, Latvia
| | - Gundega Stelfa
- Latvian Institute of Organic Synthesis, Riga, Latvia
- Latvia University of Life Sciences and Technologies, Jelgava, Latvia
| | - Edijs Vavers
- Latvian Institute of Organic Synthesis, Riga, Latvia
| | - Einars Kupats
- Latvian Institute of Organic Synthesis, Riga, Latvia
- Riga Stradins University, Riga, Latvia
| | - Janis Kuka
- Latvian Institute of Organic Synthesis, Riga, Latvia
| | - Baiba Svalbe
- Latvian Institute of Organic Synthesis, Riga, Latvia
| | - Baiba Zvejniece
- Latvian Institute of Organic Synthesis, Riga, Latvia
- University of Latvia, Riga, Latvia
| | | | - Anna-Leena Sirén
- Department of Neurosurgery, University Hospital Würzburg, Würzburg, Germany
| | - Nikolaus Plesnila
- University of Munich, Institute for Stroke and Dementia Research, Munich, Germany
| | - Maija Dambrova
- Latvian Institute of Organic Synthesis, Riga, Latvia
- Riga Stradins University, Riga, Latvia
| |
Collapse
|
9
|
Sribnick EA, Weber MD, Hall MW. Innate immune suppression after traumatic brain injury and hemorrhage in a juvenile rat model of polytrauma. J Neuroimmunol 2019; 337:577073. [PMID: 31670063 DOI: 10.1016/j.jneuroim.2019.577073] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 09/16/2019] [Accepted: 09/22/2019] [Indexed: 12/20/2022]
Abstract
Traumatic injury in children is known to cause immune suppression. Polytrauma involving a traumatic brain injury (TBI) may increase this degree of immune suppression, which increases the risk of developing nosocomial infections, potentially causing secondary brain injury and worsening patient outcomes. Despite the high prevalence of polytrauma with TBI in children, mechanisms of immune suppression following such injuries remain poorly understood. Here, we used a combined animal injury model of TBI and hemorrhage to assess immune function after polytrauma. Pre-pubescent rats were injured using a prefrontal controlled cortical impact method and a controlled hemorrhage by femoral arteriotomy. Immune function was measured by whole blood ex-vivo tumor necrosis factor alpha production capacity following incubation with lipopolysaccharide, measuring the percentage of monocytes by flow cytometry, and by examining concentrations of plasma cytokines. The degree of brain injury was sufficient to produce deficits in spatial memory testing (Barnes maze). Both hemorrhage and TBI with hemorrhage (combined injury) reduced several of the measured plasma cytokines, as compared with TBI alone. The combined injury correlated with reduced concentration of monocytes and reduced tumor necrosis factor alpha production capacity at post-injury day 1. These results demonstrate that this animal model can be used to study post-injury immune suppression.
Collapse
Affiliation(s)
- Eric A Sribnick
- Department of Surgery, Division of Neurosurgery, Nationwide Children's Hospital, Columbus, OH, USA; Department of Neurosurgery, The Ohio State University College of Medicine, Columbus, OH, USA; Center for Clinical and Translation Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.
| | - Michael D Weber
- Center for Clinical and Translation Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.
| | - Mark W Hall
- Center for Clinical and Translation Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA; Department of Pediatrics, Division of Critical Care, Nationwide Children's Hospital, Columbus, OH, USA.
| |
Collapse
|