1
|
Essex CA, Merenstein JL, Overson DK, Truong TK, Madden DJ, Bedggood MJ, Murray H, Holdsworth SJ, Stewart AW, Morgan C, Faull RLM, Hume P, Theadom A, Pedersen M. Characterizing positive and negative quantitative susceptibility values in the cortex following mild traumatic brain injury: a depth- and curvature-based study. Cereb Cortex 2025; 35:bhaf059. [PMID: 40099836 PMCID: PMC11915090 DOI: 10.1093/cercor/bhaf059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 02/17/2025] [Accepted: 02/19/2025] [Indexed: 03/20/2025] Open
Abstract
Evidence has linked head trauma to increased risk factors for neuropathology, including mechanical deformation of the sulcal fundus and, later, perivascular accumulation of hyperphosphorylated tau adjacent to these spaces related to chronic traumatic encephalopathy. However, little is known about microstructural abnormalities and cellular dyshomeostasis in acute mild traumatic brain injury in humans, particularly in the cortex. To address this gap, we designed the first architectonically motivated quantitative susceptibility mapping study to assess regional patterns of net positive (iron-related) and net negative (myelin-, calcium-, and protein-related) magnetic susceptibility across 34 cortical regions of interest following mild traumatic brain injury. Bilateral, between-group analyses sensitive to cortical depth and curvature were conducted between 25 males with acute (<14 d) sports-related mild traumatic brain injury and 25 age-matched male controls. Results suggest a trauma-induced increase in net positive susceptibility focal to superficial, perivascular-adjacent spaces in the parahippocampal sulcus. Decreases in net negative susceptibility values in distinct voxel populations within the same region indicate a potential dual pathology of neural substrates. These mild traumatic brain injury-related patterns were distinct from age-related processes revealed by correlation analyses. Our findings suggest depth- and curvature-specific deposition of biological substrates in cortical tissue convergent with features of misfolded proteins in trauma-related neurodegeneration.
Collapse
Affiliation(s)
- Christi A Essex
- Department of Psychology and Neuroscience, Auckland University of Technology, 90 Akoranga Drive, Northcote, Auckland 0627, New Zealand
| | - Jenna L Merenstein
- Brain Imaging and Analysis Center, Duke University Medical Center, 40 Duke Medicine Cir #414, Durham, NC 27710, United States
| | - Devon K Overson
- Brain Imaging and Analysis Center, Duke University Medical Center, 40 Duke Medicine Cir #414, Durham, NC 27710, United States
| | - Trong-Kha Truong
- Brain Imaging and Analysis Center, Duke University Medical Center, 40 Duke Medicine Cir #414, Durham, NC 27710, United States
| | - David J Madden
- Brain Imaging and Analysis Center, Duke University Medical Center, 40 Duke Medicine Cir #414, Durham, NC 27710, United States
| | - Mayan J Bedggood
- Department of Psychology and Neuroscience, Auckland University of Technology, 90 Akoranga Drive, Northcote, Auckland 0627, New Zealand
| | - Helen Murray
- Center for Brain Research, The University of Auckland, 85 Park Road, Grafton, Auckland 1023, New Zealand
| | - Samantha J Holdsworth
- Mātai Medical Research Institute, 466 Childers Road, Te Hapara, Gisborne 4010, New Zealand
| | - Ashley W Stewart
- Center for Advanced Imaging, The University of Queensland, Building 57 of, University Dr, St Lucia QLD 4067, Australia
| | - Catherine Morgan
- Center for Advanced MRI, The University of Auckland, 85 Park Road, Grafton, Auckland 1023, New Zealand
| | - Richard L M Faull
- Center for Brain Research, The University of Auckland, 85 Park Road, Grafton, Auckland 1023, New Zealand
| | - Patria Hume
- Sports Performance Research Institute New Zealand, Auckland University of Technology, 17 Antares Place, Rosedale, Auckland 0632, New Zealand
| | - Alice Theadom
- Department of Psychology and Neuroscience, Auckland University of Technology, 90 Akoranga Drive, Northcote, Auckland 0627, New Zealand
| | - Mangor Pedersen
- Department of Psychology and Neuroscience, Auckland University of Technology, 90 Akoranga Drive, Northcote, Auckland 0627, New Zealand
| |
Collapse
|
2
|
Liang Y, Wang Y, Sun C, Xiang Y, Deng Y. Deferoxamine reduces endothelial ferroptosis and protects cerebrovascular function after experimental traumatic brain injury. Brain Res Bull 2024; 207:110878. [PMID: 38218407 DOI: 10.1016/j.brainresbull.2024.110878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/20/2023] [Accepted: 01/09/2024] [Indexed: 01/15/2024]
Abstract
Cerebrovascular dysfunction resulting from traumatic brain injury (TBI) significantly contributes to poor patient outcomes. Recent studies revealed the involvement of iron metabolism in neuronal survival, yet its effect on vasculature remains unclear. This study aims to explore the impact of endothelial ferroptosis on cerebrovascular function in TBI. A Controlled Cortical Impact (CCI) model was established in mice, resulting in a significant increase in iron-related proteins such as TfR1, FPN1, and FTH, as well as oxidative stress biomarker 4HNE. This was accompanied by a decline in expression of the ferroptosis inhibitor GPX4. Moreover, Perls' staining and nonhemin iron content assay showed iron overload in brain microvascular endothelial cells (BMECs) and the ipsilateral cortex. Immunofluorescence staining revealed more FTH-positive cerebral endothelial cells, consistent with impaired perfusion vessel density and cerebral blood flow. As a specific iron chelator, deferoxamine (DFO) treatment inhibited such ferroptotic proteins expression and the accumulation of lipid-reactive oxygen species following CCI, enhancing glutathione peroxidase (GPx) activity. DFO treatment significantly reduced iron deposition in BMECs and brain tissue, and increased density of the cerebral capillaries as well. Consequently, DFO treatment led to improvements in cerebral blood flow (as measured by laser speckle imaging) and behavioral performance (as measured by the neurological severity scores, rotarod test, and Morris water maze test). Taken together, our results indicated that TBI induces remarkable iron disorder and endothelial ferroptosis, and DFO treatment may help maintain iron homeostasis and protect vascular function. This may provide a novel therapeutic strategy to prevent cerebrovascular dysfunction following TBI.
Collapse
Affiliation(s)
- Yidan Liang
- Department of Neurosurgery, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing, China; Chongqing Key Laboratory of Emergency Medicine, Chongqing, China
| | - Yanglingxi Wang
- Department of Neurosurgery, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing, China; Chongqing Key Laboratory of Emergency Medicine, Chongqing, China
| | - Chao Sun
- Department of Neurosurgery, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing, China; Chongqing Key Laboratory of Emergency Medicine, Chongqing, China
| | - Yi Xiang
- Department of Neurosurgery, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing, China; Chongqing Key Laboratory of Emergency Medicine, Chongqing, China
| | - Yongbing Deng
- Department of Neurosurgery, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing, China; Chongqing Key Laboratory of Emergency Medicine, Chongqing, China.
| |
Collapse
|
3
|
Jia H, Liu X, Cao Y, Niu H, Li R, Li F, Sun D, Shi M, Wa L, Liu X, Yang G, Chen F, Zhang S, Zhang J. Deferoxamine ameliorates neurological dysfunction by inhibiting ferroptosis and neuroinflammation after traumatic brain injury. Brain Res 2023; 1812:148383. [PMID: 37149247 DOI: 10.1016/j.brainres.2023.148383] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 04/26/2023] [Accepted: 04/30/2023] [Indexed: 05/08/2023]
Abstract
Traumatic brain injury (TBI) is an important reason of neurological damage and has high morbidity and mortality rates. The secondary damage caused by TBI leads to a poor clinical prognosis. According to the literature, TBI leads to ferrous iron aggregation at the site of trauma and may be a key factor in secondary injury. Deferoxamine (DFO), which is an iron chelator, has been shown to inhibit neuron degeneration; however, the role of DFO in TBI is unclear. The purpose of this study was to explore whether DFO can ameliorate TBI by inhibiting ferroptosis and neuroinflammation. Here, our findings suggest that DFO can reduce the accumulation of iron, lipid peroxides, and reactive oxygen species (ROS) and modulate the expression of ferroptosis-related indicators. Moreover, DFO may reduce NLRP3 activation via the ROS/NF-κB pathway, modulate microglial polarization, reduce neutrophil and macrophage infiltration, and inhibit the release of inflammatory factors after TBI. Additionally, DFO may reduce the activation of neurotoxic responsive astrocytes. Finally, we demonstrated that DFO can protect motor memory function, reduce edema and improve peripheral blood perfusion at the site of trauma in mice with TBI, as shown by behavioral experiments such as the Morris water maze test, cortical blood perfusion assessment and animal MRI. In conclusion, DFO ameliorates TBI by reducing iron accumulation to alleviate ferroptosis and neuroinflammation, and these findings provide a new therapeutic perspective for TBI.
Collapse
Affiliation(s)
- Haoran Jia
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Repair and Regeneration in Central Nervous System, Tianjin, China; Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China; Graduate School, Tianjin Medical University, Tianjin, China
| | - Xilei Liu
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Repair and Regeneration in Central Nervous System, Tianjin, China; Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yiyao Cao
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Repair and Regeneration in Central Nervous System, Tianjin, China; Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China; Graduate School, Tianjin Medical University, Tianjin, China
| | - Hanhong Niu
- Graduate School, Tianjin Medical University, Tianjin, China; Department of Radiotherapy, Tianjin Medical University General Hospital, Tianjin, China
| | - RuiJun Li
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Fanjian Li
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Repair and Regeneration in Central Nervous System, Tianjin, China; Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China; Graduate School, Tianjin Medical University, Tianjin, China
| | - Dongdong Sun
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin, China
| | - Mingming Shi
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Liang Wa
- Department of Urology, General Hospital of Tianjin Medical University
| | - Xiao Liu
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Guili Yang
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Fanglian Chen
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Shu Zhang
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Repair and Regeneration in Central Nervous System, Tianjin, China.
| | - Jianning Zhang
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Repair and Regeneration in Central Nervous System, Tianjin, China; Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
4
|
Lynch DG, Narayan RK, Li C. Multi-Mechanistic Approaches to the Treatment of Traumatic Brain Injury: A Review. J Clin Med 2023; 12:jcm12062179. [PMID: 36983181 PMCID: PMC10052098 DOI: 10.3390/jcm12062179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 03/18/2023] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability worldwide. Despite extensive research efforts, the majority of trialed monotherapies to date have failed to demonstrate significant benefit. It has been suggested that this is due to the complex pathophysiology of TBI, which may possibly be addressed by a combination of therapeutic interventions. In this article, we have reviewed combinations of different pharmacologic treatments, combinations of non-pharmacologic interventions, and combined pharmacologic and non-pharmacologic interventions for TBI. Both preclinical and clinical studies have been included. While promising results have been found in animal models, clinical trials of combination therapies have not yet shown clear benefit. This may possibly be due to their application without consideration of the evolving pathophysiology of TBI. Improvements of this paradigm may come from novel interventions guided by multimodal neuromonitoring and multimodal imaging techniques, as well as the application of multi-targeted non-pharmacologic and endogenous therapies. There also needs to be a greater representation of female subjects in preclinical and clinical studies.
Collapse
Affiliation(s)
- Daniel G. Lynch
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Zucker School of Medicine at Hofstra/Northwell Health, Hempstead, NY 11549, USA
| | - Raj K. Narayan
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Department of Neurosurgery, St. Francis Hospital, Roslyn, NY 11576, USA
| | - Chunyan Li
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Zucker School of Medicine at Hofstra/Northwell Health, Hempstead, NY 11549, USA
- Department of Neurosurgery, Northwell Health, Manhasset, NY 11030, USA
- Correspondence:
| |
Collapse
|
5
|
Femi-Akinlosotu OM, Olopade FE, Obiako J, Olopade JO, Shokunbi MT. Vanadium improves memory and spatial learning and protects the pyramidal cells of the hippocampus in juvenile hydrocephalic mice. Front Neurol 2023; 14:1116727. [PMID: 36846142 PMCID: PMC9947794 DOI: 10.3389/fneur.2023.1116727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/23/2023] [Indexed: 02/11/2023] Open
Abstract
Background Hydrocephalus is a neurological condition known to cause learning and memory disabilities due to its damaging effect on the hippocampal neurons, especially pyramidal neurons. Vanadium at low doses has been observed to improve learning and memory abilities in neurological disorders but it is uncertain whether such protection will be provided in hydrocephalus. We investigated the morphology of hippocampal pyramidal neurons and neurobehavior in vanadium-treated and control juvenile hydrocephalic mice. Methods Hydrocephalus was induced by intra-cisternal injection of sterile-kaolin into juvenile mice which were then allocated into 4 groups of 10 pups each, with one group serving as an untreated hydrocephalic control while others were treated with 0.15, 0.3 and 3 mg/kg i.p of vanadium compound respectively, starting 7 days post-induction for 28 days. Non-hydrocephalic sham controls (n = 10) were sham operated without any treatment. Mice were weighed before dosing and sacrifice. Y-maze, Morris Water Maze and Novel Object Recognition tests were carried out before the sacrifice, the brains harvested, and processed for Cresyl Violet and immunohistochemistry for neurons (NeuN) and astrocytes (GFAP). The pyramidal neurons of the CA1 and CA3 regions of the hippocampus were assessed qualitatively and quantitatively. Data were analyzed using GraphPad prism 8. Results Escape latencies of vanadium-treated groups were significantly shorter (45.30 ± 26.30 s, 46.50 ± 26.35 s, 42.99 ± 18.44 s) than untreated group (62.06 ± 24.02 s) suggesting improvements in learning abilities. Time spent in the correct quadrant was significantly shorter in the untreated group (21.19 ± 4.15 s) compared to control (34.15 ± 9.44 s) and 3 mg/kg vanadium-treated group (34.35 ± 9.74 s). Recognition index and mean % alternation were lowest in untreated group (p = 0.0431, p=0.0158) suggesting memory impairments, with insignificant improvements in vanadium-treated groups. NeuN immuno-stained CA1 revealed loss of apical dendrites of the pyramidal cells in untreated hydrocephalus group relative to control and a gradual reversal attempt in the vanadium-treated groups. Astrocytic activation (GFAP stain) in the untreated hydrocephalus group were attenuated in the vanadium-treated groups under the GFAP stain. Pyknotic index in CA1 pyramidal layer of untreated (18.82 ± 2.59) and 0.15mg/kg vanadium-treated groups (18.14 ± 5.92) were significantly higher than control (11.11 ± 0.93; p = 0.0205, p = 0.0373) while there was no significant difference in CA3 pyknotic index across all groups. Conclusion Our results suggest that vanadium has a dose-dependent protective effect on the pyramidal cells of the hippocampus and on memory and spatial learning functions in juvenile hydrocephalic mice.
Collapse
Affiliation(s)
| | - Funmilayo Eniola Olopade
- Developmental Neurobiology Laboratory, Department of Anatomy, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Jane Obiako
- Developmental Neurobiology Laboratory, Department of Anatomy, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - James Olukayode Olopade
- Neuroscience Unit, Department of Veterinary Anatomy, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Matthew Temitayo Shokunbi
- Developmental Neurobiology Laboratory, Department of Anatomy, College of Medicine, University of Ibadan, Ibadan, Nigeria,Division of Neurological Surgery, Department of Surgery, University of Ibadan, Ibadan, Nigeria,*Correspondence: Matthew Temitayo Shokunbi ✉
| |
Collapse
|
6
|
To XV, Mohamed AZ, Cumming P, Nasrallah FA. Association of sub-acute changes in plasma amino acid levels with long-term brain pathologies in a rat model of moderate-severe traumatic brain injury. Front Neurosci 2023; 16:1014081. [PMID: 36685246 PMCID: PMC9853432 DOI: 10.3389/fnins.2022.1014081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 12/12/2022] [Indexed: 01/09/2023] Open
Abstract
Introduction Traumatic brain injury (TBI) induces a cascade of cellular alterations that are responsible for evolving secondary brain injuries. Changes in brain structure and function after TBI may occur in concert with dysbiosis and altered amino acid fermentation in the gut. Therefore, we hypothesized that subacute plasma amino acid levels could predict long-term microstructural outcomes as quantified using neurite orientation dispersion and density imaging (NODDI). Methods Fourteen 8-10-week-old male rats were randomly assigned either to sham (n = 6) or a single moderate-severe TBI (n = 8) procedure targeting the primary somatosensory cortex. Venous blood samples were collected at days one, three, seven, and 60 post-procedure and NODDI imaging were carried out at day 60. Principal Component Regression analysis was used to identify time dependent plasma amino acid concentrations after in the subacute phase post-injury that predicted NODDI metric outcomes at day 60. Results The TBI group had significantly increased plasma levels of glutamine, arginine, alanine, proline, tyrosine, valine, isoleucine, leucine, and phenylalanine at days three-seven post-injury. Higher levels of several neuroprotective amino acids, especially the branched-chain amino acids (valine, isoleucine, leucine) and phenylalanine, as well as serine, arginine, and asparagine at days three-seven post-injury were also associated with lower isotropic diffusion volume fraction measures in the ventricles and thus lesser ventricular dilation at day 60. Discussion In the first such study, we examined the relationship between the long-term post-TBI microstructural outcomes across whole brain and the subacute changes in plasma amino acid concentrations. At days three to seven post-injury, we observed that increased plasma levels of several amino acids, particularly the branched-chain amino acids and phenylalanine, were associated with lesser degrees of ventriculomegaly and hydrocephalus TBI neuropathology at day 60 post-injury. The results imply that altered amino acid fermentation in the gut may mediate neuroprotection in the aftermath of TBI.
Collapse
Affiliation(s)
- Xuan Vinh To
- The Queensland Brain Institute, The University of Queensland, Saint Lucia, QLD, Australia
| | - Abdalla Z. Mohamed
- The Queensland Brain Institute, The University of Queensland, Saint Lucia, QLD, Australia,Thompson Institute, University of the Sunshine Coast, Sunshine Coast, QLD, Australia
| | - Paul Cumming
- Department of Nuclear Medicine, Bern University Hospital, Bern, Switzerland,School of Psychology and Counselling, Queensland University of Technology, Brisbane, QLD, Australia
| | - Fatima A. Nasrallah
- The Queensland Brain Institute, The University of Queensland, Saint Lucia, QLD, Australia,Centre for Advanced Imaging, The University of Queensland, Saint Lucia, QLD, Australia,*Correspondence: Fatima A. Nasrallah,
| |
Collapse
|
7
|
Cheng Y, Gao Y, Li J, Rui T, Li Q, Chen H, Jia B, Song Y, Gu Z, Wang T, Gao C, Wang Y, Wang Z, Wang F, Tao L, Luo C. TrkB agonist N-acetyl serotonin promotes functional recovery after traumatic brain injury by suppressing ferroptosis via the PI3K/Akt/Nrf2/Ferritin H pathway. Free Radic Biol Med 2023; 194:184-198. [PMID: 36493983 DOI: 10.1016/j.freeradbiomed.2022.12.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/19/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022]
Abstract
Ferroptosis is a form of regulated cell death that is mainly triggered by iron-dependent lipid peroxidation. A growing body of evidence suggests that ferroptosis is involved in the pathophysiology of traumatic brain injury (TBI), and tropomyosin-related kinase B (TrkB) deficiency would mediate TBI pathologies. As an agonist of TrkB and an immediate precursor of melatonin, N-acetyl serotonin (NAS) exerts several beneficial effects on TBI, but there is no information regarding the role of NAS in ferroptosis after TBI. Here, we examined the effect of NAS treatment on TBI-induced functional outcomes and ferroptosis. Remarkably, the administration of NAS alleviated TBI-induced neurobehavioral deficits, lesion volume, and neurodegeneration. NAS also rescued TBI-induced mitochondrial shrinkage, the changes in ferroptosis-related molecule expression, and iron accumulation in the ipsilateral cortex. Similar results were obtained with a well-established ferroptosis inhibitor, liproxstatin-1. Furthermore, NAS activated the TrkB/PI3K/Akt/Nrf2 pathway in the mouse model of TBI, while inhibition of PI3K and Nrf2 weakened the protection of NAS against ferroptosis both in vitro and in vivo, suggesting that a possible pathway linking NAS to the action of anti-ferroptosis was TrkB/PI3K/Akt/Nrf2. Given that ferritin H (Fth) is a known transcription target of Nrf2, we then investigated the effects of NAS on neuron-specific Fth knockout (Fth-KO) mice. Strikingly, Fth deletion almost abolished the protective effects of NAS against TBI-induced ferroptosis and synaptic damage, although Fth deletion-induced susceptibility toward ferroptosis after TBI was reversed by an iron chelator, deferoxamine. Taken together, these data indicate that the TrkB agonist NAS treatment appears to improve brain function after TBI by suppressing ferroptosis, at least in part, through activation of the PI3K/Akt/Nrf2/Fth pathway, providing evidence that NAS is likely to be a promising anti-ferroptosis agent for further treatment for TBI.
Collapse
Affiliation(s)
- Ying Cheng
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, 215123, China
| | - Yuan Gao
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, 215123, China
| | - Jing Li
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, 215123, China
| | - Tongyu Rui
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, 215123, China
| | - Qianqian Li
- School of Forensic Medicine, Wannan Medical College, Wuhu, 241002, China
| | - Huan Chen
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, 215123, China
| | - Bowen Jia
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, 215123, China
| | - Yiting Song
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, 215123, China
| | - Zhiya Gu
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, 215123, China
| | - Tao Wang
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, 215123, China
| | - Cheng Gao
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, 215123, China
| | - Ying Wang
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, 215123, China
| | - Zufeng Wang
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, 215123, China
| | - Fudi Wang
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Luyang Tao
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, 215123, China.
| | - Chengliang Luo
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
8
|
Daglas M, Truong PH, Miles LQ, Juan SMA, Rao SS, Adlard PA. Deferiprone attenuates neuropathology and improves outcome following traumatic brain injury. Br J Pharmacol 2023; 180:214-234. [PMID: 36102035 DOI: 10.1111/bph.15950] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 08/27/2022] [Accepted: 09/08/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Traumatic brain injury (TBI) remains a leading cause of mortality and morbidity in young adults. The role of iron in potentiating neurodegeneration following TBI has gained recent interest as iron deposition has been detected in the injured brain in the weeks to months post-TBI, in both the preclinical and clinical setting. A failure in iron homeostasis can lead to oxidative stress, inflammation and excitotoxicity; and whether this is a cause or consequence of the long-term effects of TBI remains unknown. EXPERIMENTAL APPROACH We investigated the role of iron and the effect of therapeutic intervention using a brain-permeable iron chelator, deferiprone, in a controlled cortical impact mouse model of TBI. An extensive assessment of cognitive, motor and anxiety/depressive outcome measures were examined, and neuropathological and biochemical changes, over a 3-month period post-TBI. KEY RESULTS Lesion volume was significantly reduced at 3 months, which was preceded by a reduction in astrogliosis, microglia/macrophages and preservation of neurons in the injured brain at 2 weeks and/or 1 month post-TBI in mice receiving oral deferiprone. Deferiprone treatment showed significant improvements in neurological severity scores, locomotor/gait performance and cognitive function, and attenuated anxiety-like symptoms post-TBI. Deferiprone reduced iron levels, lipid peroxidation/oxidative stress and altered expression of neurotrophins in the injured brain over this period. CONCLUSION AND IMPLICATIONS Our findings support a detrimental role of iron in the injured brain and suggest that deferiprone (or similar iron chelators) may be promising therapeutic approaches to improve survival, functional outcomes and quality of life following TBI.
Collapse
Affiliation(s)
- Maria Daglas
- Synaptic Neurobiology Laboratory, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Australia
| | - Phan H Truong
- Synaptic Neurobiology Laboratory, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Australia
| | - Linh Q Miles
- Synaptic Neurobiology Laboratory, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Australia
| | - Sydney M A Juan
- Synaptic Neurobiology Laboratory, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Australia
| | - Shalini S Rao
- Synaptic Neurobiology Laboratory, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Australia
| | - Paul A Adlard
- Synaptic Neurobiology Laboratory, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
9
|
Vaghebin R, Khalili M, Amiresmaili S, Roghani M, Esmaeili Saber SS, Namdar H. Saphenous vein phlebotomy alleviates neuroinflammatory response and oxidative stress following traumatic brain injury. INTERDISCIPLINARY NEUROSURGERY 2022. [DOI: 10.1016/j.inat.2022.101626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022] Open
|
10
|
Smith AN, Shaughness M, Collier S, Hopkins D, Byrnes KR. Therapeutic targeting of microglia mediated oxidative stress after neurotrauma. Front Med (Lausanne) 2022; 9:1034692. [PMID: 36405593 PMCID: PMC9671221 DOI: 10.3389/fmed.2022.1034692] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/12/2022] [Indexed: 10/06/2023] Open
Abstract
Inflammation is a primary component of the central nervous system injury response. Traumatic brain and spinal cord injury are characterized by a pronounced microglial response to damage, including alterations in microglial morphology and increased production of reactive oxygen species (ROS). The acute activity of microglia may be beneficial to recovery, but continued inflammation and ROS production is deleterious to the health and function of other cells. Microglial nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX), mitochondria, and changes in iron levels are three of the most common sources of ROS. All three play a significant role in post-traumatic brain and spinal cord injury ROS production and the resultant oxidative stress. This review will evaluate the current state of therapeutics used to target these avenues of microglia-mediated oxidative stress after injury and suggest avenues for future research.
Collapse
Affiliation(s)
- Austin N. Smith
- Neuroscience Program, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Michael Shaughness
- Neuroscience Program, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Sean Collier
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Deanna Hopkins
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Kimberly R. Byrnes
- Neuroscience Program, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
11
|
Vinh To X, Mohamed AZ, Cumming P, Nasrallah FA. Subacute cytokine changes after a traumatic brain injury predict chronic brain microstructural alterations on advanced diffusion imaging in the male rat. Brain Behav Immun 2022; 102:137-150. [PMID: 35183698 DOI: 10.1016/j.bbi.2022.02.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 02/07/2022] [Accepted: 02/12/2022] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION The process of neuroinflammation occurring after traumatic brain injury (TBI) has received significant attention as a potential prognostic indicator and interventional target to improve patients' outcomes. Indeed, many of the secondary consequences of TBI have been attributed to neuroinflammation and peripheral inflammatory changes. However, inflammatory biomarkers in blood have not yet emerged as a clinical tool for diagnosis of TBI and predicting outcome. The controlled cortical impact model of TBI in the rodent gives reliable readouts of the dynamics of post-TBI neuroinflammation. We now extend this model to include a panel of plasma cytokine biomarkers measured at different time points post-injury, to test the hypothesis that these markers can predict brain microstructural outcome as quantified by advanced diffusion-weighted magnetic resonance imaging (MRI). METHODS Fourteen 8-10-week-old male rats were randomly assigned to sham surgery (n = 6) and TBI (n = 8) treatment with a single moderate-severe controlled cortical impact. We collected blood samples for cytokine analysis at days 1, 3, 7, and 60 post-surgery, and carried out standard structural and advanced diffusion-weighted MRI at day 60. We then utilized principal component regression to build an equation predicting different aspects of microstructural changes from the plasma inflammatory marker concentrations measured at different time points. RESULTS The TBI group had elevated plasma levels of IL-1β and several neuroprotective cytokines and chemokines (IL-7, CCL3, and GM-CSF) compared to the sham group from days 3 to 60 post-injury. The plasma marker panels obtained at day 7 were significantly associated with the outcome at day 60 of the trans-hemispheric cortical map transfer process that is a frequent finding in unilateral TBI models. DISCUSSION These results confirm and extend prior studies showing that day 7 post-injury is a critical temporal window for the reorganisation process following TBI. High plasma level of IL-1β and low plasma levels of the neuroprotective IL-7, CCL3, and GM-CSF of TBI animals at day 60 were associated with greater TBI pathology.
Collapse
Affiliation(s)
- Xuan Vinh To
- The Queensland Brain Institute, The University of Queensland, Queensland, Australia
| | - Abdalla Z Mohamed
- The Queensland Brain Institute, The University of Queensland, Queensland, Australia; Thompson Institute, University of the Sunshine Coast, Queensland, Australia
| | - Paul Cumming
- Department of Nuclear Medicine, Bern University Hospital, Bern, Switzerland; School of Psychology and Counselling, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Fatima A Nasrallah
- The Queensland Brain Institute, The University of Queensland, Queensland, Australia; The Centre for Advanced Imaging, The University of Queensland, Queensland, Australia.
| |
Collapse
|
12
|
Zhao H, Li Y, Zhang Y, He WY, Jin WN. Role of Immune and Inflammatory Mechanisms in Stroke: A Review of Current Advances. Neuroimmunomodulation 2022; 29:255-268. [PMID: 35640538 DOI: 10.1159/000524951] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 05/04/2022] [Indexed: 12/15/2022] Open
Abstract
Stroke accounts for a large proportion of morbidity and mortality burden in China. Moreover, there is a high prevalence of the leading risk factors for stroke, including hypertension and smoking. Understanding the underlying mechanisms and developing effective therapeutic interventions for patients with stroke is a key imperative. The pathophysiology of stroke involves a complex interplay between the immune and inflammatory mechanisms. Focal brain inflammation triggered by neuronal cell death and the release of factors such as damage-associated molecular patterns can further exacerbate neuronal injury; in addition, impairment of the blood-brain barrier, oxidative stress, microvascular dysfunction, and brain edema cause secondary brain injury. Immune cells, including microglia and other infiltrating inflammatory cells, play a key role in triggering focal and global brain inflammation. Anti-inflammatory therapies targeting the aforementioned mechanisms can alleviate primary and secondary brain injury in the aftermath of a stroke. Further experimental and clinical studies are required to explore the beneficial effects of anti-inflammatory drugs in stroke.
Collapse
Affiliation(s)
- Hui Zhao
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yan Li
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Ying Zhang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Wen-Yan He
- Center for Neuroinflammation, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wei-Na Jin
- Center for Neuroinflammation, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| |
Collapse
|
13
|
Post-injury ventricular enlargement associates with iron in choroid plexus but not with seizure susceptibility nor lesion atrophy-6-month MRI follow-up after experimental traumatic brain injury. Brain Struct Funct 2021; 227:145-158. [PMID: 34757444 PMCID: PMC8741668 DOI: 10.1007/s00429-021-02395-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 09/16/2021] [Indexed: 11/15/2022]
Abstract
Ventricular enlargement is one long-term consequence of a traumatic brain injury, and a risk factor for memory disorders and epilepsy. One underlying mechanisms of the chronic ventricular enlargement is disturbed cerebrospinal-fluid secretion or absorption by choroid plexus. We set out to characterize the different aspects of ventricular enlargement in lateral fluid percussion injury (FPI) rat model by magnetic resonance imaging (MRI) and discovered choroid plexus injury in rats that later developed hydrocephalus. We followed the brain pathology progression for 6 months and studied how the ventricular growth was associated with the choroid plexus injury, cortical lesion expansion, hemorrhagic load or blood perfusion deficits. We correlated MRI findings with the seizure susceptibility in pentylenetetrazol challenge and memory function in Morris water-maze. Choroid plexus injury was validated by ferric iron (Prussian blue) and cytoarchitecture (Nissl) stainings. We discovered choroid plexus injury that accumulates iron in 90% of FPI rats by MRI. The amount of the choroid plexus iron remained unaltered 1-, 3- and 6-month post-injury. During this time, the ventricles kept on growing bilaterally. Ventricular growth did not depend on the cortical lesion severity or the cortical hemorrhagic load suggesting a separate pathology. Instead, the results indicate choroidal injury as one driver of the post-traumatic hydrocephalus, since the higher the choroid plexus iron load the larger were the ventricles at 6 months. The ventricle size or the choroid plexus iron load did not associate with seizure susceptibility. Cortical hypoperfusion and memory deficits were worse in rats with greater ventricular growth.
Collapse
|
14
|
Traumatic brain injury augurs ill for prolonged deficits in the brain's structural and functional integrity following controlled cortical impact injury. Sci Rep 2021; 11:21559. [PMID: 34732737 PMCID: PMC8566513 DOI: 10.1038/s41598-021-00660-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 10/06/2021] [Indexed: 12/02/2022] Open
Abstract
Previous neuroimaging studies in rodents investigated effects of the controlled cortical impact (CCI) model of traumatic brain injury (TBI) within one-month post-TBI. This study extends this temporal window to monitor the structural–functional alterations from two hours to six months post-injury. Thirty-seven male Sprague–Dawley rats were randomly assigned to TBI and sham groups, which were scanned at two hours, 1, 3, 7, 14, 30, 60 days, and six months following CCI or sham surgery. Structural MRI, diffusion tensor imaging, and resting-state functional magnetic resonance imaging were acquired to assess the dynamic structural, microstructural, and functional connectivity alterations post-TBI. There was a progressive increase in lesion size associated with brain volume loss post-TBI. Furthermore, we observed reduced fractional anisotropy within 24 h and persisted to six months post-TBI, associated with acutely reduced axial diffusivity, and chronic increases in radial diffusivity post-TBI. Moreover, a time-dependent pattern of altered functional connectivity evolved over the six months’ follow-up post-TBI. This study extends the current understanding of the CCI model by confirming the long-term persistence of the altered microstructure and functional connectivity, which may hold a strong translational potential for understanding the long-term sequelae of TBI in humans.
Collapse
|
15
|
Rufus P, Moorthy RK, Joseph M, Rajshekhar V. Post Traumatic Hydrocephalus: Incidence, Pathophysiology and Outcomes. Neurol India 2021; 69:S420-S428. [PMID: 35102998 DOI: 10.4103/0028-3886.332264] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Background Post-traumatic hydrocephalus (PTH) is a sequel of traumatic brain injury (TBI) that is seen more often in patients undergoing decompressive craniectomy (DC). It is associated with prolonged hospital stay and unfavorable outcomes. Objective To study the incidence and risk factors for development of PTH in patients undergoing DC in our institution and to review the literature on PTH with respect to incidence, risk factors, pathophysiology, and outcomes of management. Methods Data from 95 patients (among 220 patients who underwent DC for TBI and fulfilled the inclusion criteria) over a 5-year period at Christian Medical College, Vellore were collected and analyzed to study the incidence and possible risk factors for development of PTH. A review of the literature on PTH was performed by searching PUBMED resources. Results Thirty (31.6%) out of 95 patients developed post-traumatic ventriculomegaly, of whom seven (7.3%) developed symptomatic PTH, necessitating placement of ventriculoperitoneal shunt (VPS). No risk factor for development of PTH could be identified. The reported incidence of PTH in the literature is from 0.07% to 29%, with patients undergoing DC having a higher incidence. Younger age, subarachnoid hemorrhage, severity of TBI, presence of subdural hygroma, and delayed cranioplasty after DC are the main risk factors reported in the literature. Conclusions PTH occurs in a significant proportion of patients with TBI and can lead to unfavorable outcomes. PTH has to be distinguished from asymptomatic ventriculomegaly as early as possible so that a CSF diversion procedure can be planned early during development of PTH.
Collapse
Affiliation(s)
- Phelix Rufus
- Department of Neurological Sciences, Christian Medical College, Vellore, Tamil Nadu, India
| | - Ranjith K Moorthy
- Department of Neurological Sciences, Christian Medical College, Vellore, Tamil Nadu, India
| | - Mathew Joseph
- Department of Neurological Sciences, Christian Medical College, Vellore, Tamil Nadu, India
| | - Vedantam Rajshekhar
- Department of Neurological Sciences, Christian Medical College, Vellore, Tamil Nadu, India
| |
Collapse
|
16
|
Fan D, Fang Q. Siderophores for medical applications: Imaging, sensors, and therapeutics. Int J Pharm 2021; 597:120306. [PMID: 33540031 DOI: 10.1016/j.ijpharm.2021.120306] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 01/07/2023]
Abstract
Siderophores are low-molecular-weight chelators produced by microorganisms to scavenge iron from the environment and deliver it to cells via specific receptors. Tremendous researches on the molecular basis of siderophore regulation, synthesis, secretion, and uptake have inspired their diverse applications in the medical field. Replacing iron with radionuclides in siderophores, such as the most prominent Ga-68 for positron emission tomography (PET), carves out ways for targeted imaging of infectious diseases and cancers. Additionally, the high affinity of siderophores for metal ions or microorganisms makes them a potent detecting moiety in sensors that can be used for diagnosis. As for therapeutics, the notable Trojan horse-inspired siderophore-antibiotic conjugates demonstrate enhanced toxicity against multi-drug resistant (MDR) pathogens. Besides, siderophores can tackle iron overload diseases and, when combined with moieties such as hydrogels and nanoparticles, a wide spectrum of iron-induced diseases and even cancers. In this review, we briefly outline the related mechanisms, before summarizing the siderophore-based applications in imaging, sensors, and therapeutics.
Collapse
Affiliation(s)
- Di Fan
- Laboratory of Theoretical and Computational Nanoscience, CAS Key Laboratory of Nanophotonic Materials and Devices, CAS Center for Excellence in Nanoscience, Beijing Key Laboratory of Ambient Particles Health Effects and Prevention Techniques, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, PR China
| | - Qiaojun Fang
- Laboratory of Theoretical and Computational Nanoscience, CAS Key Laboratory of Nanophotonic Materials and Devices, CAS Center for Excellence in Nanoscience, Beijing Key Laboratory of Ambient Particles Health Effects and Prevention Techniques, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, PR China; Sino-Danish Center for Education and Research, Beijing 101408, PR China.
| |
Collapse
|
17
|
Kosyakovsky J, Fine JM, Frey WH, Hanson LR. Mechanisms of Intranasal Deferoxamine in Neurodegenerative and Neurovascular Disease. Pharmaceuticals (Basel) 2021; 14:ph14020095. [PMID: 33513737 PMCID: PMC7911954 DOI: 10.3390/ph14020095] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 12/15/2022] Open
Abstract
Identifying disease-modifying therapies for neurological diseases remains one of the greatest gaps in modern medicine. Herein, we present the rationale for intranasal (IN) delivery of deferoxamine (DFO), a high-affinity iron chelator, as a treatment for neurodegenerative and neurovascular disease with a focus on its novel mechanisms. Brain iron dyshomeostasis with iron accumulation is a known feature of brain aging and is implicated in the pathogenesis of a number of neurological diseases. A substantial body of preclinical evidence and early clinical data has demonstrated that IN DFO and other iron chelators have strong disease-modifying impacts in Alzheimer’s disease (AD), Parkinson’s disease (PD), ischemic stroke, and intracranial hemorrhage (ICH). Acting by the disease-nonspecific pathway of iron chelation, DFO targets each of these complex diseases via multifactorial mechanisms. Accumulating lines of evidence suggest further mechanisms by which IN DFO may also be beneficial in cognitive aging, multiple sclerosis, traumatic brain injury, other neurodegenerative diseases, and vascular dementia. Considering its known safety profile, targeted delivery method, robust preclinical efficacy, multiple mechanisms, and potential applicability across many neurological diseases, the case for further development of IN DFO is considerable.
Collapse
Affiliation(s)
- Jacob Kosyakovsky
- School of Medicine, University of Virginia, 200 Jeanette Lancaster Way, Charlottesville, VA 22903, USA;
- HealthPartners Neuroscience Center, HealthPartners Institute, Saint Paul, MN 55130, USA; (W.H.F.II); (L.R.H.)
| | - Jared M. Fine
- HealthPartners Neuroscience Center, HealthPartners Institute, Saint Paul, MN 55130, USA; (W.H.F.II); (L.R.H.)
- Correspondence:
| | - William H. Frey
- HealthPartners Neuroscience Center, HealthPartners Institute, Saint Paul, MN 55130, USA; (W.H.F.II); (L.R.H.)
| | - Leah R. Hanson
- HealthPartners Neuroscience Center, HealthPartners Institute, Saint Paul, MN 55130, USA; (W.H.F.II); (L.R.H.)
| |
Collapse
|
18
|
Recombinant osteopontin provides protection for cerebral infarction by inhibiting the NLRP3 inflammasome in microglia. Brain Res 2020; 1751:147170. [PMID: 33157099 DOI: 10.1016/j.brainres.2020.147170] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/16/2020] [Accepted: 10/17/2020] [Indexed: 12/25/2022]
Abstract
Neuroinflammation is one of the most important secondary pathological events after cerebral infarction. Activation of NLRP3 inflammasome is a pivotal form of neuroinflammation. Osteopontin (OPN) is expressed during the subacute phase after cerebral infarction and has an important chemotactic effect on microglia. The aim of this study was to reveal the effect of recombinant OPN on brain injury after cerebral infarction and the regulation of NLRP3 inflammasome. We used the middle cerebral artery occlusion (MCAO) method-established focal cerebral ischemia model and LPS-induced inflammation model on neonate rat primary microglia. The effects of OPN on cerebral ischemic injury, neural function, microglia inflammation and NLRP3 inflammasome function were studied by immunofluorescence, staining, enzyme-linked immunosorbent assay and Western blot assay. We established MCAO cerebral ischemia and reperfusion injury model, and found that recombinant OPN reduced the volume of cerebral infarction and alleviated the ischemic injury degree of cerebral tissues, neurons, and neurological function. We found that OPN was also involved in the negative regulation of inflammasome and microglia activity in cerebral ischemic injury, and that OPN inhibited the activation of NLRP3 inflammasome and the function of microglia in a LPS-induced inflammatory model. Our findings show that recombinant OPN can reduce the ischemic infarct size and alleviate the cerebral ischemic injury of rats, which may be related to its efficient involvement in the inhibitory regulation of inflammasome and microglia inflammatory activation.
Collapse
|
19
|
Tang S, Gao P, Chen H, Zhou X, Ou Y, He Y. The Role of Iron, Its Metabolism and Ferroptosis in Traumatic Brain Injury. Front Cell Neurosci 2020; 14:590789. [PMID: 33100976 PMCID: PMC7545318 DOI: 10.3389/fncel.2020.590789] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 09/04/2020] [Indexed: 12/29/2022] Open
Abstract
Traumatic brain injury (TBI) is a structural and physiological disruption of brain function caused by external forces. It is a major cause of death and disability for patients worldwide. TBI includes both primary and secondary impairments. Iron overload and ferroptosis highly involved in the pathophysiological process of secondary brain injury. Ferroptosis is a form of regulatory cell death, as increased iron accumulation in the brain leads to lipid peroxidation, reactive oxygen species (ROS) production, mitochondrial dysfunction and neuroinflammatory responses, resulting in cellular and neuronal damage. For this reason, eliminating factors like iron deposition and inhibiting lipid peroxidation may be a promising therapy. Iron chelators can be used to eliminate excess iron and to alleviate some of the clinical manifestations of TBI. In this review we will focus on the mechanisms of iron and ferroptosis involving the manifestations of TBI, broaden our understanding of the use of iron chelators for TBI. Through this review, we were able to better find novel clinical therapeutic directions for further TBI study.
Collapse
Affiliation(s)
- Sicheng Tang
- Medical Clinic and Polyclinic IV, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
| | - Pan Gao
- Department of Translational Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Hanmin Chen
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangyue Zhou
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yibo Ou
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue He
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
20
|
Aravind A, Kosty J, Chandra N, Pfister BJ. Blast exposure predisposes the brain to increased neurological deficits in a model of blast plus blunt traumatic brain injury. Exp Neurol 2020; 332:113378. [PMID: 32553593 DOI: 10.1016/j.expneurol.2020.113378] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 05/20/2020] [Accepted: 06/08/2020] [Indexed: 10/24/2022]
Abstract
Soldiers are often exposed to more than one traumatic brain injury (TBI) over the course of their service. In recent years, more attention has been drawn to the increased risk of neurological deficits caused by the 'blast plus' polytrauma, which typically is a blast trauma combined with other forms of TBI. In this study, we investigated the behavioral and neuronal deficits resulting from a blast plus injury involving a mild-moderate blast followed by a mild blunt trauma using the fluid percussion injury model. We identified that the blast injury predisposed the brain to increased cognitive deficits, chronic ventricular enlargement, increased neurodegeneration at acute time points and chronic neuronal loss. Interestingly, a single blast and single blunt injury differed in their onset and manifestation of cognitive and regional neuronal loss. We also identified the presence of cleaved RIP1 from caspase 8 mediated apoptosis in the blunt injury while the blast injury did not activate immediate apoptosis but led to decreased hilar neuronal survival over time.
Collapse
Affiliation(s)
- Aswati Aravind
- Center for Injury Biomechanics, Materials and Medicine, Department of Biomedical Engineering, New Jersey Institute of Technology, 323 Dr M.L.K. Jr. Blvd, Newark, NJ 07102, USA
| | - Julianna Kosty
- Center for Injury Biomechanics, Materials and Medicine, Department of Biomedical Engineering, New Jersey Institute of Technology, 323 Dr M.L.K. Jr. Blvd, Newark, NJ 07102, USA
| | - Namas Chandra
- Center for Injury Biomechanics, Materials and Medicine, Department of Biomedical Engineering, New Jersey Institute of Technology, 323 Dr M.L.K. Jr. Blvd, Newark, NJ 07102, USA
| | - Bryan J Pfister
- Center for Injury Biomechanics, Materials and Medicine, Department of Biomedical Engineering, New Jersey Institute of Technology, 323 Dr M.L.K. Jr. Blvd, Newark, NJ 07102, USA.
| |
Collapse
|
21
|
Feng Z, Liu S, Chen Q, Tan Q, Xian J, Feng H, Chen Z, Li G. uPA alleviates kaolin-induced hydrocephalus by promoting the release and activation of hepatocyte growth factor in rats. Neurosci Lett 2020; 731:135011. [PMID: 32497735 DOI: 10.1016/j.neulet.2020.135011] [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: 02/19/2020] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 02/05/2023]
Abstract
Urokinase-type plasminogen activator (uPA) was demonstrated to alleviate kaolin-induced communicating hydrocephalus via inhibiting subarachnoid space fibrosis, but the exact mechanism remains elusive. Thus, this study was designed to investigate if hepatocyte growth factor (HGF), which plays a vital role in uPA-triggered inhibiting of fibrosis in multiple systems, is involved in this process in hydrocephalus. There were 2 parts in this study. First, hydrocephalus was induced in rats by basal cistern injection of kaolin. Then rats were treated with saline or uPA and brain tissue and CSF were collected for Western blot and enzyme-linked immuno sorbent assay (ELISA) four days later. Second, kaolin-induced hydrocephalus rats were treated with saline, uPA, uPA + PHA665752 (antagonist of HGF) or PHA665752. Some animals received MRI four weeks later and brains were used for immunofluorescence. The others were euthanized four days later for ELISA. Both levels of total and activated HGF in the CSF was increased after uPA injections, but related mRNA expression of HGF showed no statistical significance when compared with the control group. Further, the effects of uPA that alleviating ventricular enlargement, subarachnoid fibrosis and reactive astrocytosis were partially reversed by PHA665752. Moreover, PHA665752 partially abolished uPA-induced reduction of transforming growth factor- β1(TGF- β1) level in CSF. Our data suggest that uPA effectively inhibited subarachnoid fibrosis and restricted the development of communicating hydrocephalus in rats in part by promoting HGF release and activation, which may further regulate the TGF-β1 expression in CSF.
Collapse
Affiliation(s)
- Zhou Feng
- Department of Rehabilitation, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Shengyan Liu
- Chongqing Mental Health Center, Chongqing, 4001147, PR China
| | - Qianwei Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Qiang Tan
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Jishu Xian
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Zhi Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Gang Li
- Department of Cerebrovascular Disease, Affiliated Hospital of Zunyi Medical University, Zunyi Medical University, Guizhou, 563003, PR China.
| |
Collapse
|
22
|
Zhang L, Xiao H, Yu X, Deng Y. Minocycline attenuates neurological impairment and regulates iron metabolism in a rat model of traumatic brain injury. Arch Biochem Biophys 2020; 682:108302. [DOI: 10.1016/j.abb.2020.108302] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 02/08/2020] [Accepted: 02/08/2020] [Indexed: 02/08/2023]
|
23
|
Toro-Urrego N, Turner LF, Avila-Rodriguez MF. New Insights into Oxidative Damage and Iron Associated Impairment in Traumatic Brain Injury. Curr Pharm Des 2020; 25:4737-4746. [DOI: 10.2174/1381612825666191111153802] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 10/28/2019] [Indexed: 12/14/2022]
Abstract
:
Traumatic Brain Injury is considered one of the most prevalent causes of death around the world; more
than seventy millions of individuals sustain the condition per year. The consequences of traumatic brain injury on
brain tissue are complex and multifactorial, hence, the current palliative treatments are limited to improve patients’
quality of life. The subsequent hemorrhage caused by trauma and the ongoing oxidative process generated
by biochemical disturbances in the in the brain tissue may increase iron levels and reactive oxygen species. The
relationship between oxidative damage and the traumatic brain injury is well known, for that reason, diminishing
factors that potentiate the production of reactive oxygen species have a promissory therapeutic use. Iron chelators
are molecules capable of scavenging the oxidative damage from the brain tissue and are currently in use for ironoverload-
derived diseases.
:
Here, we show an updated overview of the underlying mechanisms of the oxidative damage after traumatic brain
injury. Later, we introduced the potential use of iron chelators as neuroprotective compounds for traumatic brain
injury, highlighting the action mechanisms of iron chelators and their current clinical applications.
Collapse
Affiliation(s)
- Nicolas Toro-Urrego
- Laboratorio de Citoarquitectura y Plasticidad Neuronal, Instituto de Investigaciones Cardiológicas, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Liliana F. Turner
- Grupo Modelos Experimentales para las Ciencias Zoohumanas - Departamento de Biología Facultad de Ciencias, Universidad del Tolima- Ibagué, Tolima, Colombia
| | - Marco F. Avila-Rodriguez
- Grupo Modelos Experimentales para las Ciencias Zoohumanas - Departamento de Ciencias Clínicas- Facultad de Ciencias de la Salud, Universidad del Tolima- Ibagué, Tolima, Colombia
| |
Collapse
|
24
|
Mohamed AZ, Corrigan F, Collins-Praino LE, Plummer SL, Soni N, Nasrallah FA. Evaluating spatiotemporal microstructural alterations following diffuse traumatic brain injury. Neuroimage Clin 2019; 25:102136. [PMID: 31865019 PMCID: PMC6931220 DOI: 10.1016/j.nicl.2019.102136] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 12/12/2019] [Accepted: 12/13/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Diffuse traumatic brain injury (TBI) is known to lead to microstructural changes within both white and grey matter detected in vivo with diffusion tensor imaging (DTI). Numerous studies have shown alterations in fractional anisotropy (FA) and mean diffusivity (MD) within prominent white matter tracts, but few have linked these to changes within the grey matter with confirmation via histological assessment. This is especially important as alterations in the grey matter may be predictive of long-term functional deficits. METHODS A total of 33 male Sprague Dawley rats underwent severe closed-head TBI. Eight animals underwent tensor-based morphometry (TBM) and DTI at baseline (pre-TBI), 24 hours (24 h), 7, 14, and 30 days post-TBI. Immunohistochemical analysis for the detection of ionised calcium-binding adaptor molecule 1 (IBA1) to assess microglia number and percentage of activated cells, β-amyloid precursor protein (APP) as a marker of axonal injury, and myelin basic protein (MBP) to investigate myelination was performed at each time-point. RESULTS DTI showed significant alterations in FA and RD in numerous white matter tracts including the corpus callosum, internal and external capsule, and optic tract and in the grey-matter in the cortex, thalamus, and hippocampus, with the most significant effects observed at 14 D post-TBI. TBM confirmed volumetric changes within the hippocampus and thalamus. Changes in DTI were in line with significant axonal injury noted at 24 h post-injury via immunohistochemical analysis of APP, with widespread microglial activation seen within prominent white matter tracts and the grey matter, which persisted to 30 D within the hippocampus and thalamus. Microstructural alterations in MBP+ve fibres were also noted within the hippocampus and thalamus, as well as the cortex. CONCLUSION This study confirms the widespread effects of diffuse TBI on white matter tracts which could be detected via DTI and extends these findings to key grey matter regions, with a comprehensive investigation of the whole brain. In particular, the hippocampus and thalamus appear to be vulnerable to ongoing pathology post-TBI, with DTI able to detect these alterations supporting the clinical utility in evaluating these regions post-TBI.
Collapse
Affiliation(s)
- Abdalla Z Mohamed
- Queensland Brain Institute, The University of Queensland, Building 79, Upland Road, Saint Lucia, Brisbane, QLD 4072, Australia
| | - Frances Corrigan
- Head Injury Laboratory, Division of Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Lyndsey E Collins-Praino
- Cognition, Aging and Neurodegenerative Disease Laboratory (CANDL), Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Stephanie L Plummer
- Translational Neuropathology Laboratory, Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Neha Soni
- Queensland Brain Institute, The University of Queensland, Building 79, Upland Road, Saint Lucia, Brisbane, QLD 4072, Australia
| | - Fatima A Nasrallah
- Queensland Brain Institute, The University of Queensland, Building 79, Upland Road, Saint Lucia, Brisbane, QLD 4072, Australia.
| |
Collapse
|
25
|
Wang K, Jing Y, Xu C, Zhao J, Gong Q, Chen S. HIF-1α and VEGF Are Involved in Deferoxamine-Ameliorated Traumatic Brain Injury. J Surg Res 2019; 246:419-426. [PMID: 31630885 DOI: 10.1016/j.jss.2019.09.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 08/01/2019] [Accepted: 09/13/2019] [Indexed: 12/29/2022]
Abstract
BACKGROUND Deferoxamine (DFX) has been reported to have neuroprotective effect. This study aimed to investigate the neuroprotective effect of DFX and its effect on hypoxia-inducible factor 1 alpha (HIF-1α) and vascular endothelial growth factor (VEGF) in rats after traumatic brain injury (TBI). MATERIALS AND METHODS Rats were randomly divided into sham operation, TBI + DFX, and TBI + vehicle groups. The rats in the TBI + DFX group were intraperitoneally injected with DFX 2 and 6 h after injury, thereafter once every 12 h. The rats in the TBI + vehicle group were intraperitoneally injected with saline at the same time points. At 6, 12, 24, and 48 h after TBI, 6 rats in each group were euthanized, and the brains were harvested. The expression of HIF-1α and VEGF in the pericontusional area was detected using real-time polymerase chain reaction and Western blot analysis. TBI-induced apoptosis was investigated using the TdT-mediated dUTP nick-end labeling (TUNEL) method. Three days after TBI, the density of microvessels was examined via immunohistochemical staining. RESULTS DFX treatment upregulated the expression of HIF-1α and VEGF after TBI. DFX treatment reduced apoptosis and improved the neurobehavioral score after TBI. The density of microvessels was higher in the TBI + DFX group than that in the TBI + vehicle group 3 d after TBI. CONCLUSIONS DFX can stimulate angiogenesis, inhibit apoptosis, and play a protective role after TBI. The protective effect of DFX may, at least in part, be through upregulating the expression of HIF-1α and its downstream target gene VEGF.
Collapse
Affiliation(s)
- Kai Wang
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China; Department of Neurosurgery, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, Huai'an, China
| | - Yao Jing
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Chen Xu
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jianwei Zhao
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Qiuyuan Gong
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Shiwen Chen
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| |
Collapse
|
26
|
Lower Iron Levels Predict Acute Hydrocephalus Following Aneurysmal Subarachnoid Hemorrhage. World Neurosurg 2019; 126:e907-e913. [PMID: 30872197 DOI: 10.1016/j.wneu.2019.03.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 02/28/2019] [Accepted: 03/01/2019] [Indexed: 11/22/2022]
Abstract
OBJECTIVE We tested the hypothesis that low serum iron levels are associated with acute hydrocephalus following aneurysmal subarachnoid hemorrhage (aSAH). METHODS Patients presenting with ruptured intracranial aneurysms were enrolled in the prospective observational study. Age, sex, history of diabetes, hypertension and hyperlipidemia, symptom onset, Fisher grade, Hunt-Hess grade, aneurysm location, hemoglobin, and serum iron were collected. Acute hydrocephalus was determined within 72 hours after subarachnoid hemorrhage. A propensity-score matching analysis was performed to correct imbalances in patient characteristics between hydrocephalus and non-hydrocephalus groups. RESULTS A total of 535 patients were included. Incidence of acute hydrocephalus was 20.0%. In multivariate logistic regression analysis, lower serum iron was considered as a risk factor of acute hydrocephalus, as well as delayed ischemic neurologic deficit and lower hemoglobin (P = 0.000). After propensity-score matching, lower serum iron was considered as an independent risk factor for acute hydrocephalus, whereas hemoglobin and delayed ischemic neurologic deficit were not. The matched hydrocephalus group had lower serum iron comparing with the matched non-hydrocephalus group (10.26 ± 5.33 mmol/L vs. 13.44 ± 5.18 mmol/L; P = 0.000). The optimal cut-off value for serum iron levels as a predictor for acute hydrocephalus in patients with aSAH was determined as 13.1 mmol/L in the receiver operating characteristic curve. Furthermore, lower serum iron levels (odds ratio 0.305; 95% confidence interval, 0.178-0.524; P = 0.000) and acute hydrocephalus (odds ratio 0.372; 95% confidence interval, 0.202-0.684; P = 0.001) were predictors of poor outcome, as well as higher Hunt-Hess grade and Fisher grade. CONCLUSIONS Lower serum iron levels after aSAH was a predictor of acute hydrocephalus and unfavorable outcome.
Collapse
|
27
|
Saletti PG, Ali I, Casillas-Espinosa PM, Semple BD, Lisgaras CP, Moshé SL, Galanopoulou AS. In search of antiepileptogenic treatments for post-traumatic epilepsy. Neurobiol Dis 2019; 123:86-99. [PMID: 29936231 PMCID: PMC6309524 DOI: 10.1016/j.nbd.2018.06.017] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 06/20/2018] [Indexed: 11/28/2022] Open
Abstract
Post-traumatic epilepsy (PTE) is diagnosed in 20% of individuals with acquired epilepsy, and can impact significantly the quality of life due to the seizures and other functional or cognitive and behavioral outcomes of the traumatic brain injury (TBI) and PTE. There is no available antiepileptogenic or disease modifying treatment for PTE. Animal models of TBI and PTE have been developed, offering useful insights on the value of inflammatory, neurodegenerative pathways, hemorrhages and iron accumulation, calcium channels and other target pathways that could be used for treatment development. Most of the existing preclinical studies test efficacy towards pathologies of functional recovery after TBI, while a few studies are emerging testing the effects towards induced or spontaneous seizures. Here we review the existing preclinical trials testing new candidate treatments for TBI sequelae and PTE, and discuss future directions for efforts aiming at developing antiepileptogenic and disease-modifying treatments.
Collapse
Affiliation(s)
- Patricia G Saletti
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Idrish Ali
- Department of Neuroscience, Central Clinical School, Monash University, The Alfred Hospital, Melbourne, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Melbourne, Australia
| | - Pablo M Casillas-Espinosa
- Department of Neuroscience, Central Clinical School, Monash University, The Alfred Hospital, Melbourne, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Melbourne, Australia
| | - Bridgette D Semple
- Department of Neuroscience, Central Clinical School, Monash University, The Alfred Hospital, Melbourne, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Melbourne, Australia
| | - Christos Panagiotis Lisgaras
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Solomon L Moshé
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx, NY, USA; Dominick P. Purpura Department of Neuroscience, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Einstein/Montefiore Epilepsy Center, Montefiore Medical Center, Bronx, NY, USA; Department of Pediatrics, Albert Einstein College of Medicine, Einstein/Montefiore Epilepsy Center, Montefiore Medical Center, Bronx, NY, USA
| | - Aristea S Galanopoulou
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx, NY, USA; Dominick P. Purpura Department of Neuroscience, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Einstein/Montefiore Epilepsy Center, Montefiore Medical Center, Bronx, NY, USA.
| |
Collapse
|
28
|
Wang H, Zhu X, Xiang H, Liao Z, Gao M, Luo Y, Wu P, Zhang Y, Ren M, Zhao H, Xu M. Effects of altitude changes on mild-to-moderate closed-head injury in rats following acute high-altitude exposure. Exp Ther Med 2019; 17:847-856. [PMID: 30651871 DOI: 10.3892/etm.2018.7020] [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: 05/28/2018] [Accepted: 10/12/2018] [Indexed: 11/05/2022] Open
Abstract
Mild-to-moderate closed-head injury (mmCHI) is an acute disease induced by high-altitudes. It is general practice to transfer patients to lower altitudes for treatment, but the pathophysiological changes at different altitudes following mmCHI remain unknown. The present study simulated acute high-altitude exposure (6,000 m above sea level) in rats to establish a model of mmCHI and recorded their vital signs. The rats were then randomly assigned into different altitude exposure groups (6,000, 4,500 and 3,000 m) and neurological severity score (NSS), body weight (BW), brain magnetic resonance imaging (MRI), brain water content (BWC) and the ratio of BW/BWC at 6, 12 and 24 h following mmCHI, and the glial fibrillary acidic protein levels were analysed in all groups. The results revealed that within the first 24 h following acute high-altitude exposure, mmCHI induced dehydration, brain oedema and neuronal damage. Brain injury in rats was significantly reversed following descent to 4,500 m compared with the results from 6,000 or 3,000 m. The results indicated that subjects should be transported as early as possible. Furthermore, avoiding large-span descent altitude was beneficial to reduce neurological impairment. The examination of brain-specific biomarkers and MRI may further be useful in determining the prognosis of high-altitude mmCHI. These results may provide guidance for rescuing high altitude injuries.
Collapse
Affiliation(s)
- Hao Wang
- Department of Neurosurgery, Daping Hospital, Third Military Medical University, Chongqing 400042, P.R. China
| | - Xiyan Zhu
- Chongqing Key Laboratory of Vehicle Crash/Bio-impact and Traffic Safety, Institute for Traffic Medicine, Third Military Medical University, Chongqing 400042, P.R. China
| | - Hongyi Xiang
- Chongqing Key Laboratory of Vehicle Crash/Bio-impact and Traffic Safety, Institute for Traffic Medicine, Third Military Medical University, Chongqing 400042, P.R. China
| | - Zhikang Liao
- Chongqing Key Laboratory of Vehicle Crash/Bio-impact and Traffic Safety, Institute for Traffic Medicine, Third Military Medical University, Chongqing 400042, P.R. China
| | - Mou Gao
- Affiliated Bayi Brain Hospital P.L.A Army General Hospital, Beijing 100038, P.R. China
| | - Yetao Luo
- School of Public Health and Management, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Pengfei Wu
- Department of Neurosurgery, Daping Hospital, Third Military Medical University, Chongqing 400042, P.R. China
| | - Yihua Zhang
- Department of Neurosurgery, Daping Hospital, Third Military Medical University, Chongqing 400042, P.R. China
| | - Mingliang Ren
- Department of Neurosurgery, Daping Hospital, Third Military Medical University, Chongqing 400042, P.R. China
| | - Hui Zhao
- Chongqing Key Laboratory of Vehicle Crash/Bio-impact and Traffic Safety, Institute for Traffic Medicine, Third Military Medical University, Chongqing 400042, P.R. China
| | - Minhui Xu
- Department of Neurosurgery, Daping Hospital, Third Military Medical University, Chongqing 400042, P.R. China
| |
Collapse
|
29
|
Daglas M, Adlard PA. The Involvement of Iron in Traumatic Brain Injury and Neurodegenerative Disease. Front Neurosci 2018; 12:981. [PMID: 30618597 PMCID: PMC6306469 DOI: 10.3389/fnins.2018.00981] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 12/07/2018] [Indexed: 12/18/2022] Open
Abstract
Traumatic brain injury (TBI) consists of acute and long-term pathophysiological sequelae that ultimately lead to cognitive and motor function deficits, with age being a critical risk factor for poorer prognosis. TBI has been recently linked to the development of neurodegenerative diseases later in life including Alzheimer’s disease, Parkinson’s disease, chronic traumatic encephalopathy, and multiple sclerosis. The accumulation of iron in the brain has been documented in a number of neurodegenerative diseases, and also in normal aging, and can contribute to neurotoxicity through a variety of mechanisms including the production of free radicals leading to oxidative stress, excitotoxicity and by promoting inflammatory reactions. A growing body of evidence similarly supports a deleterious role of iron in the pathogenesis of TBI. Iron deposition in the injured brain can occur via hemorrhage/microhemorrhages (heme-bound iron) or independently as labile iron (non-heme bound), which is considered to be more damaging to the brain. This review focusses on the role of iron in potentiating neurodegeneration in TBI, with insight into the intersection with neurodegenerative conditions. An important implication of this work is the potential for therapeutic approaches that target iron to attenuate the neuropathology/phenotype related to TBI and to also reduce the associated risk of developing neurodegenerative disease.
Collapse
Affiliation(s)
- Maria Daglas
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Paul A Adlard
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
30
|
Khalaf S, Ahmad AS, Chamara KR, Doré S. Unique Properties Associated with the Brain Penetrant Iron Chelator HBED Reveal Remarkable Beneficial Effects after Brain Trauma. J Neurotrauma 2018; 36:43-53. [PMID: 29743006 PMCID: PMC6306957 DOI: 10.1089/neu.2017.5617] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Iron is postulated to contribute to secondary injury after brain trauma through various pathways including oxidative stress and inflammation. Therefore, one goal is to limit iron toxicity by either directly limiting iron activity, or limiting the secondary cascade mediated by iron, therefore rescuing the brain from damage after trauma. The N,N'-Di(2-hydroxybenzyl)ethylenediamine-N,N'-diacetic acid monohydrochloride (HBED) is a unique iron chelator that has the ability to cross the intact blood-brain barrier; it has a higher affinity to iron, and it has a longer half-life than most commonly used chelators. A controlled-cortical impact model of traumatic brain injury (TBI) was induced in mice. Mice were subcutaneously injected with HBED immediately after TBI, then at 12 h after, followed by a twice-a-day regimen until an end-point of 3 days. Neurobehavioral tests were performed daily. Cortical injury volume, hemispheric enlargement, and hippocampal swelling were quantified. Perls' iron immunostaining along with markers of gliosis, oxidative stress, and aquaporin (AQP) 4 were also performed. Data revealed that HBED treatment significantly decreases motor deficits and improves recovery after TBI. It also reduces cortical injury volume by 36.6 ± 6.8% (p < 0.001), hippocampal swelling by 23.4 ± 3.8% (p < 0.05), and total hemispheric volume by 13.3 ± 2.7% (p < 0.01). These effects are related to a reduction in microgliosis and oxidiative stress markers in the impacted corpus callosum area by 39.8 ± 7.3%, and by 80.5 ± 0.8% (p < 0.05), respectively. AQP4 staining is also attenuated in the hippocampus of HBED-treated mice. Therefore, our results suggest that HBED should be considered as a therapeutic tool to facilitate the recovery process following brain trauma.
Collapse
Affiliation(s)
- Saher Khalaf
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, Florida
| | - Abdullah Shafique Ahmad
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, Florida
| | - K.V.D. Ranga Chamara
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, Florida
| | - Sylvain Doré
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, Florida
- Departments of Neurology, Psychiatry, Pharmaceutics, Psychology, and Neuroscience, University of Florida, Gainesville, Florida
| |
Collapse
|
31
|
Wang J, Zhang MY, Xu SQ, Cheng J, Yu ZJ, Hu XM. Down-regulation of telomerase reverse transcriptase-related anti-apoptotic function in a rat model of acrylamide induced neurobehavioral deficits. Biotech Histochem 2018; 93:512-518. [PMID: 29926741 DOI: 10.1080/10520295.2018.1471523] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
Although the precise mechanism is unknown, neuron apoptosis is believed to participate in neuropathy caused by acrylamide (ACR). Telomerase reverse transcriptase (TERT) exhibits an anti-apoptotic function, but its contribution to the pathogenesis of ACR neurotoxicity is unclear. We investigated adult male rats that were given 30, 40 and 50 mg/kg ACR three times/week for 4 weeks. We found that ACR treatment caused significant deficits in sensory/motor function as measured by gait score, landing foot spread distance, movement initiation test and tail immersion test. Histological examination showed that the cerebral cortex in all ACR treated animals exhibited fewer neurons and more condensed nuclei than normal cortex. A significant increase in apoptosis was found in the cerebral cortex of rat brains subjected to ACR treatment in a dose-dependent manner. The expression of TERT in the brain was significantly reduced by ACR treatment. The pro-apoptotic cleaved caspase-3 protein level was increased, while the anti-apoptotic Bcl-2 protein level was decreased by 30 - 50 mg/kg ACR. Our findings indicate that TERT and its downstream regulators of neuron apoptosis, including Bcl-2 and cleaved caspase-3, were involved in ACR neurotoxicity.
Collapse
Affiliation(s)
- J Wang
- a Department of Pharmacy , College of Medicine, Wuhan University of Science and Technology , Wuhan.,b Hubei Province Key Laboratory of Occupational Hazard Identification and Control , Wuhan University of Science and Technology , Wuhan
| | - M Y Zhang
- a Department of Pharmacy , College of Medicine, Wuhan University of Science and Technology , Wuhan.,b Hubei Province Key Laboratory of Occupational Hazard Identification and Control , Wuhan University of Science and Technology , Wuhan
| | - S Q Xu
- a Department of Pharmacy , College of Medicine, Wuhan University of Science and Technology , Wuhan.,b Hubei Province Key Laboratory of Occupational Hazard Identification and Control , Wuhan University of Science and Technology , Wuhan
| | - J Cheng
- a Department of Pharmacy , College of Medicine, Wuhan University of Science and Technology , Wuhan.,b Hubei Province Key Laboratory of Occupational Hazard Identification and Control , Wuhan University of Science and Technology , Wuhan
| | - Z J Yu
- a Department of Pharmacy , College of Medicine, Wuhan University of Science and Technology , Wuhan.,b Hubei Province Key Laboratory of Occupational Hazard Identification and Control , Wuhan University of Science and Technology , Wuhan
| | - X M Hu
- a Department of Pharmacy , College of Medicine, Wuhan University of Science and Technology , Wuhan.,b Hubei Province Key Laboratory of Occupational Hazard Identification and Control , Wuhan University of Science and Technology , Wuhan.,c College of Pharmacy , Shanghai University of Medicine & Health Sciences , Shanghai , China
| |
Collapse
|
32
|
Garton T, Hua Y, Xiang J, Xi G, Keep RF. Challenges for intraventricular hemorrhage research and emerging therapeutic targets. Expert Opin Ther Targets 2017; 21:1111-1122. [PMID: 29067856 PMCID: PMC6097191 DOI: 10.1080/14728222.2017.1397628] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Intraventricular hemorrhage (IVH) affects both premature infants and adults. In both demographics, it has high mortality and morbidity. There is no FDA approved therapy that improves neurological outcome in either population highlighting the need for additional focus on therapeutic targets and treatments emerging from preclinical studies. Areas covered: IVH induces both initial injury linked to the physical effects of the blood (mass effect) and secondary injury linked to the brain response to the hemorrhage. Preclinical studies have identified multiple secondary injury mechanisms following IVH, and particularly the role of blood components (e.g. hemoglobin, iron, thrombin). This review, with an emphasis on pre-clinical IVH research, highlights therapeutic targets and treatments that may be of use in prevention, acute care, or repair of damage. Expert opinion: An IVH is a potentially devastating event. Progress has been made in elucidating injury mechanisms, but this has still to translate to the clinic. Some pathways involved in injury also have beneficial effects (coagulation cascade/inflammation). A greater understanding of the downstream pathways involved in those pathways may allow therapeutic development. Iron chelation (deferoxamine) is in clinical trial for intracerebral hemorrhage and preclinical data suggest it may be a potential treatment for IVH.
Collapse
Affiliation(s)
- Thomas Garton
- a Department of Neurosurgery , University of Michigan , Ann Arbor , MI , USA
| | - Ya Hua
- a Department of Neurosurgery , University of Michigan , Ann Arbor , MI , USA
| | - Jianming Xiang
- a Department of Neurosurgery , University of Michigan , Ann Arbor , MI , USA
| | - Guohua Xi
- a Department of Neurosurgery , University of Michigan , Ann Arbor , MI , USA
| | - Richard F Keep
- a Department of Neurosurgery , University of Michigan , Ann Arbor , MI , USA
| |
Collapse
|
33
|
Xie Q, Bao X, Chen ZH, Xu Y, Keep RF, Muraszko KM, Xi G, Hua Y. Role of Protease-Activated Receptor-1 in Glioma Growth. ACTA NEUROCHIRURGICA. SUPPLEMENT 2017; 121:355-60. [PMID: 26463974 DOI: 10.1007/978-3-319-18497-5_61] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2023]
Abstract
Activation of a thrombin receptor, protease-activated receptor-1 (PAR-1), induces angiogenesis, cell proliferation, and invasion in tumors. The present study examined the effect of host PAR-1 gene deletion on glioma growth in a mouse model. F98 glioma cells were implanted into the right caudate of either wild type (WT) or PAR-1 knockout (KO) mice. Mice underwent magnetic resonance imaging (MRI) and the brains were used for measurements of brain water content and tumor mass. Levels of hypoxia-inducible factor-1 alpha (HIF-1α) and vascular endothelial growth factor (VEGF) were also measured by ELISA (enzyme-linked immunosorbent assay). We found brain water content in the ipsilateral hemisphere and the tumor mass were significantly lower in PAR-1 KO than WT mice at day 12 after implantation of F98 cells (p < 0.05). HIF-1α protein levels in the ipsilateral hemisphere were higher in the WT mice (373 ± 25 pg/g brain tissue vs 333 ± 35 pg/g brain tissue in PAR-1 KO mice, p < 0.05) 7 days after F98 cell implantation. VEGF protein levels were also higher in the ipsilateral hemisphere of WT mice (219 ± 21 vs 166 ± 22 pg/g brain tissue in PAR-1 KO mice, p < 0.01). In conclusion, PAR-1 has a role in glioma growth and could be a new therapeutic target for gliomas.
Collapse
Affiliation(s)
- Qing Xie
- Department of Neurosurgery, University of Michigan, 5018 BSRB, Ann Arbor, MI, 48109-2200, USA
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Xuhui Bao
- Department of Neurosurgery, University of Michigan, 5018 BSRB, Ann Arbor, MI, 48109-2200, USA
| | - Zhan Hong Chen
- Department of Neurosurgery, University of Michigan, 5018 BSRB, Ann Arbor, MI, 48109-2200, USA
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China
| | - Ying Xu
- Department of Neurosurgery, University of Michigan, 5018 BSRB, Ann Arbor, MI, 48109-2200, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, 5018 BSRB, Ann Arbor, MI, 48109-2200, USA
| | - Karin M Muraszko
- Department of Neurosurgery, University of Michigan, 5018 BSRB, Ann Arbor, MI, 48109-2200, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, 5018 BSRB, Ann Arbor, MI, 48109-2200, USA
| | - Ya Hua
- Department of Neurosurgery, University of Michigan, 5018 BSRB, Ann Arbor, MI, 48109-2200, USA.
| |
Collapse
|
34
|
Guo D, Li D, Li J, Li Y, Hu X, Guan F, Yang B. Topical application of the hematostatic agent Surgiflo® could attenuate brain injury in experimental TBI mice. Neurol Res 2017; 39:830-836. [PMID: 28566060 DOI: 10.1080/01616412.2017.1330815] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
OBJECT The pathologies resulting from traumatic brain injury (TBI) have been thoroughly studied, but rarely have the effects of bleeding and coagulation in the early stage of TBI been considered. In this study, we investigated the effects of topical Surgiflo® application on brain injury in experimental TBI mice using S100β, MAP-2 and mNSS scores. METHODS TBI was induced by modified weight drop injury in male C57BL/6 mice. The mice were then randomly divided into (i) the sham group, (ii) TBI mice applied with saline (vehicle), and (iii) TBI mice applied with Surgiflo® in the same volume. Modified neurological severity scores (mNSS) were measured on days 0 (before surgery), 1, 3, 7, and 28 to evaluate neurologic functional deficits. At day 28, the mice were sacrificed, and the forebrains were sliced. The effects of Surgiflo® on microtubule-associated protein 2 and serum S100β protein were examined by immunohistochemistry and electro-chemiluminescence immunoassay. RESULTS Serum S100β protein levels were significantly elevated at different time points (24 h, 3 days, 7 days) in the TBI groups (p < 0.01) compared to normal control groups. Surgiflo® induced a lower concentration of serum S100β protein levels at day 3 (p < 0.05) and day 7 (p < 0.05) compared to the TBI group applied with saline. H&E staining showed that Surgiflo® treatment led to a 45% decrease in cortical brain lesion volume and in subcortical white matter 28 days after TBI. Compared with the saline-treated group, the number of MAP2-positive cells was significantly increased in the perilesional area of the Surgiflo®-treated group. The Surgiflo®-treated group exhibited lower mNSS scores on days 7 and 28 than did the saline-treated group. DISCUSSION Surgiflo® treatment produced a significant decrease in serum S100β protein levels in TBI mouse models, which may lead to an improvement in the recovery of TBI models.
Collapse
Affiliation(s)
- Dewei Guo
- a Department of Neurosurgery , The First Affiliated Hospital of Zhengzhou University , Zhengzhou , P.R. China
| | - Dongpeng Li
- a Department of Neurosurgery , The First Affiliated Hospital of Zhengzhou University , Zhengzhou , P.R. China
| | - Jinghong Li
- b Department of Neurology , The First Affiliated Hospital of Zhengzhou University , Zhengzhou , P.R. China
| | - Yunfeng Li
- c Department of Neurological rehabilitation Laboratory , Xuchang Vocational Technical College , Xuchang , P.R. China
| | - Xiang Hu
- d Department of Neurological rehabilitation Laboratory , Shenzhen Beike Cell Engineering Institute , Shenzhen , P.R. China
| | - Fangxia Guan
- e Department of Bioengineering , Zhengzhou University , Zhengzhou , P.R. China
| | - Bo Yang
- a Department of Neurosurgery , The First Affiliated Hospital of Zhengzhou University , Zhengzhou , P.R. China
| |
Collapse
|
35
|
Hua C, Zhao G. Adult posthaemorrhagic hydrocephalus animal models. J Neurol Sci 2017; 379:39-43. [PMID: 28716276 DOI: 10.1016/j.jns.2017.05.038] [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: 08/23/2016] [Revised: 05/18/2017] [Accepted: 05/19/2017] [Indexed: 12/26/2022]
Abstract
Posthaemorrhagic hydrocephalus (PHH) is often associated with high morbidity and mortality and serves as an important clinical predictor of poor outcomes after intracranial haemorrhage (ICH). We are lack of effective medical intervention methods to improve functional outcomes in patients with PHH because little is still known about the mechanisms of PHH pathogenesis. Animal models play a key role in the study of PHH. Developed a suitable animal model that will help us to be better to find preventative strategies and improve the prognosis of patients with PHH. The purpose of this review is to summarize the body of knowledge gained from animal studies.
Collapse
Affiliation(s)
- Cong Hua
- Department of Neurosurgery of the First Clinical Hospital, Jilin University, Changchun, China.
| | - Gang Zhao
- Department of Neurosurgery of the First Clinical Hospital, Jilin University, Changchun, China.
| |
Collapse
|
36
|
Feng Z, Tan Q, Tang J, Li L, Tao Y, Chen Y, Yang Y, Luo C, Feng H, Zhu G, Chen Q, Chen Z. Intraventricular administration of urokinase as a novel therapeutic approach for communicating hydrocephalus. Transl Res 2017; 180:77-90.e2. [PMID: 27614013 DOI: 10.1016/j.trsl.2016.08.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 07/18/2016] [Accepted: 08/13/2016] [Indexed: 01/10/2023]
Abstract
Fibrosis of the subarachnoid space (SAS) after infection, inflammation, or hemorrhage can impair cerebrospinal fluid absorption and circulation, causing diffuse ventricular dilatation. In the present study, we tested the hypothesis that urokinase (also known as urokinase-type plasminogen activator [uPA]), a fibrinolytic agent, attenuates fibrosis and ventriculomegaly in a rat model of kaolin-induced communicating hydrocephalus and thus may have potential as a therapy for these conditions. Thirty microliters of sterile 25% kaolin suspension was injected into the basal cisterns of adult Sprague-Dawley rats to induce hydrocephalus, and 2 intraventricular injections of either uPA or vehicle (saline) were administered immediately and 3 days thereafter. Ventricular volumes were measured by magnetic resonance imaging (MRI) on days 3, 14, and 28 after kaolin injection. Fibrosis and reactive astrogliosis were evaluated on day 28 by immunofluorescence and Western blotting. Neurocognitive features were tested using the Morris water maze from days 23 to 28. MRI analysis demonstrated that kaolin administration successfully induced hydrocephalus in rats and that uPA treatment significantly attenuated ventricular enlargement. In addition, uPA inhibited the deposition of laminin and fibronectin, extracellular matrix molecules, in the SAS, attenuated gliosis, and improved learning and memory in kaolin-treated rats. Therefore, we concluded that uPA prevents the development of kaolin-induced communicating hydrocephalus by preventing the development of subarachnoid fibrosis and by eliciting improvements in neurocognition. The results of this study indicate that uPA may be a novel clinical therapy for communicating hydrocephalus.
Collapse
Affiliation(s)
- Zhou Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Qiang Tan
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Jun Tang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Lin Li
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Yihao Tao
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Yujie Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Yunfeng Yang
- Department of Neurosurgery, Sichuan Provincial Corps Hospital, Chinese People's Armed Police Forces, Leshan, People's Republic of China
| | - Chunxia Luo
- Department of Neurology, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Gang Zhu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Qianwei Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China.
| | - Zhi Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China.
| |
Collapse
|
37
|
Chen Q, Feng Z, Tan Q, Guo J, Tang J, Tan L, Feng H, Chen Z. Post-hemorrhagic hydrocephalus: Recent advances and new therapeutic insights. J Neurol Sci 2017; 375:220-230. [PMID: 28320134 DOI: 10.1016/j.jns.2017.01.072] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 01/25/2017] [Accepted: 01/30/2017] [Indexed: 02/07/2023]
Abstract
Post-hemorrhagic hydrocephalus (PHH), also referred to as progressive ventricular dilatation, is caused by disturbances in cerebrospinal fluid (CSF) flow or absorption following hemorrhage in the brain. As one of the most serious complications of neonatal/adult intraventricular hemorrhage (IVH), subarachnoid hemorrhage (SAH), and traumatic brain injury (TBI), PHH is associated with increased morbidity and disability of these events. Common sequelae of PHH include neurocognitive impairment, motor dysfunction, and growth impairment. Non-surgical measures to reduce increased intracranial pressure (ICP) in PHH have shown little success and most patients will ultimately require surgical management, such as external ventricular drainage and shunting which mostly by inserting a CSF drainage shunt. Unfortunately, shunt complications are common and the optimum time for intervention is unclear. To date, there remains no comprehensive strategy for PHH management and it becomes imperative that to explore new therapeutic targets and methods for PHH. Over past decades, increasing evidence have indicated that hemorrhage-derived blood and subsequent metabolic products may play a key role in the development of IVH-, SAH- and TBI-associated PHH. Several intervention strategies have recently been evaluated and cross-referenced. In this review, we summarized and discussed the common aspects of hydrocephalus following IVH, SAH and TBI, relevant experimental animal models, clinical translation of in vivo experiments, and potential preventive and therapeutic targets for PHH.
Collapse
Affiliation(s)
- Qianwei Chen
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University, Chongqing 400038, China
| | - Zhou Feng
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University, Chongqing 400038, China
| | - Qiang Tan
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University, Chongqing 400038, China
| | - Jing Guo
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University, Chongqing 400038, China; Department of Neurosurgery, The 211st Hospital of PLA, Harbin 150086, China
| | - Jun Tang
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University, Chongqing 400038, China
| | - Liang Tan
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University, Chongqing 400038, China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University, Chongqing 400038, China.
| | - Zhi Chen
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
38
|
Progress of Research on Diffuse Axonal Injury after Traumatic Brain Injury. Neural Plast 2016; 2016:9746313. [PMID: 28078144 PMCID: PMC5204088 DOI: 10.1155/2016/9746313] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 10/25/2016] [Accepted: 11/15/2016] [Indexed: 12/02/2022] Open
Abstract
The current work reviews the concept, pathological mechanism, and process of diagnosing of DAI. The pathological mechanism underlying DAI is complicated, including axonal breakage caused by axonal retraction balls, discontinued protein transport along the axonal axis, calcium influx, and calpain-mediated hydrolysis of structural protein, degradation of axonal cytoskeleton network, the changes of transport proteins such as amyloid precursor protein, and changes of glia cells. Based on the above pathological mechanism, the diagnosis of DAI is usually made using methods such as CT, traditional and new MRI, biochemical markers, and neuropsychological assessment. This review provides a basis in literature for further investigation and discusses the pathological mechanism. It may also facilitate improvement of the accuracy of diagnosis for DAI, which may come to play a critical role in breaking through the bottleneck of the clinical treatment of DAI and improving the survival and quality of life of patients through clear understanding of pathological mechanisms and accurate diagnosis.
Collapse
|
39
|
Garton T, Keep RF, Hua Y, Xi G. Brain iron overload following intracranial haemorrhage. Stroke Vasc Neurol 2016; 1:172-184. [PMID: 28959481 PMCID: PMC5435218 DOI: 10.1136/svn-2016-000042] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 11/01/2016] [Accepted: 11/02/2016] [Indexed: 12/15/2022] Open
Abstract
Intracranial haemorrhages, including intracerebral haemorrhage (ICH), intraventricular haemorrhage (IVH) and subarachnoid haemorrhage (SAH), are leading causes of morbidity and mortality worldwide. In addition, haemorrhage contributes to tissue damage in traumatic brain injury (TBI). To date, efforts to treat the long-term consequences of cerebral haemorrhage have been unsatisfactory. Incident rates and mortality have not showed significant improvement in recent years. In terms of secondary damage following haemorrhage, it is becoming increasingly apparent that blood components are of integral importance, with haemoglobin-derived iron playing a major role. However, the damage caused by iron is complex and varied, and therefore, increased investigation into the mechanisms by which iron causes brain injury is required. As ICH, IVH, SAH and TBI are related, this review will discuss the role of iron in each, so that similarities in injury pathologies can be more easily identified. It summarises important components of normal brain iron homeostasis and analyses the existing evidence on iron-related brain injury mechanisms. It further discusses treatment options of particular promise.
Collapse
Affiliation(s)
- Thomas Garton
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Ya Hua
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
40
|
Guo D, Wilkinson DA, Thompson BG, Pandey AS, Keep RF, Xi G, Hua Y. MRI Characterization in the Acute Phase of Experimental Subarachnoid Hemorrhage. Transl Stroke Res 2016; 8:234-243. [PMID: 27896625 DOI: 10.1007/s12975-016-0511-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 11/06/2016] [Accepted: 11/08/2016] [Indexed: 12/12/2022]
Abstract
A number of mechanisms have been proposed for the early brain injury after subarachnoid hemorrhage (SAH). In this study, we investigated the radiographic characteristics and influence of gender on early brain injury after experimental SAH. SAH was induced by endovascular perforation in male and female rats. Magnetic resonance imaging was performed in a 7.0-T Varian MR scanner at 24 h after SAH. The occurrence and size of T2 lesions, ventricular dilation, and white matter injury (WMI) were determined on T2-weighted images (T2WI). The effects of SAH on heme oxygenase-1 and fibrin/fibrinogen were examined by Western blotting and immunohistochemistry. SAH severity was assessed using a MRI grading system, and neurological function was evaluated according to a modified Garcia's scoring system. T2 hyperintensity areas and enlarged ventricles were observed in T2WI coronal sections 24 h after SAH. The overall incidence of T2 lesions, WMI, and hydrocephalus was 54, 20, and 63%, respectively. Female rats had a higher incidence of T2 hyperintensity lesions and hydrocephalus, as well as larger T2 lesion volumes and higher average ventricular volume. SAH rats graded at 3-4 (our previously validated MRI grading scale) had larger T2 lesion volumes, more hydrocephalus, and worse neurological function compared with those graded at 0-2. In conclusion, T2 lesion, WMI, and hydrocephalus were the most prevalent MRI characteristics 24 h after experimental SAH. The T2 lesion area matched with fibrinogen/fibrin positive staining in the acute phase of SAH. SAH induced more severe brain injury in females compared to males in the acute phase of SAH.
Collapse
Affiliation(s)
- Dewei Guo
- Department of Neurosurgery, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA.,Department of Neurosurgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - D Andrew Wilkinson
- Department of Neurosurgery, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - B Gregory Thompson
- Department of Neurosurgery, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Aditya S Pandey
- Department of Neurosurgery, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Ya Hua
- Department of Neurosurgery, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA.
| |
Collapse
|
41
|
Dang G, Yang Y, Wu G, Hua Y, Keep RF, Xi G. Early Erythrolysis in the Hematoma After Experimental Intracerebral Hemorrhage. Transl Stroke Res 2016; 8:174-182. [PMID: 27783383 DOI: 10.1007/s12975-016-0505-3] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 10/11/2016] [Accepted: 10/13/2016] [Indexed: 12/14/2022]
Abstract
Erythrolysis occurs in the clot after intracerebral hemorrhage (ICH), and the release of hemoglobin causes brain injury, but it is unclear when such lysis occurs. The present study examined early erythrolysis in rats. ICH rats had an intracaudate injection of 100 μl autologous blood, and sham rats had a needle insertion. All rats had T2 and T2* magnetic response imaging (MRI) scanning, and brains were used for histology and CD163 (a hemoglobin scavenger receptor) and DARPP-32 (a neuronal marker) immunohistochemistry. There was marked heterogeneity within the hematoma on T2* MRI, with a hyperintense or isointense core and a hypointense periphery. Hematoxylin and eosin staining in the same animals showed significant erythrolysis in the core with the formation of erythrocyte ghosts. The degree of erythrolysis correlated with the severity of perihematomal neuronal loss. Perihematomal CD163 was increased by day 1 after ICH and may be involved in clearing hemoglobin caused by early hemolysis. Furthermore, ICH resulted in more severe erythrolysis, neuronal loss, and perihematomal CD163 upregulation in spontaneously hypertensive rats compared to Wistar-Kyoto rats. In conclusion, T2*MRI-detectable early erythrolysis occurred in the clot after ICH and activated CD163. Hypertension is associated with enhanced erythrolysis in the hematoma.
Collapse
Affiliation(s)
- Ge Dang
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA.,Department of Neurology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yuefan Yang
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA.,Department of Neurosurgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Gang Wu
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Ya Hua
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA. .,R5018 BSRB, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA.
| |
Collapse
|
42
|
LeBlanc RH, Chen R, Selim MH, Hanafy KA. Heme oxygenase-1-mediated neuroprotection in subarachnoid hemorrhage via intracerebroventricular deferoxamine. J Neuroinflammation 2016; 13:244. [PMID: 27618864 PMCID: PMC5020472 DOI: 10.1186/s12974-016-0709-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 09/02/2016] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Subarachnoid hemorrhage (SAH) is a devastating disease that affects over 30,000 Americans per year. Previous animal studies have explored the therapeutic effects of deferoxamine (DFX) via its iron-chelating properties after SAH, but none have assessed the necessity of microglial/macrophage heme oxygenase-1 (HO-1 or Hmox1) in DFX neuroprotection, nor has the efficacy of an intracerebroventricular (ICV) administration route been fully examined. We explored the therapeutic efficacy of systemic and ICV DFX in a SAH mouse model and its effect on microglial/macrophage HO-1. METHODS Wild-type (WT) mice were split into the following treatment groups: SAH sham + vehicle, SAH + vehicle, SAH + intraperitoneal (IP) DFX, and SAH + ICV DFX. For each experimental group, neuronal damage, cognitive outcome, vasospasm, cerebral and hematogenous myeloid cell populations, cerebral IL-6 concentration, and mitochondrial superoxide anion production were measured. HO-1 co-localization to microglia was measured using confocal images. Trans-wells with WT or HO-1(-/-) microglia and hippocampal neurons were treated with vehicle, red blood cells (RBCs), or RBCs with DFX; neuronal damage, TNF-α concentration, and microglial HO-1 expression were measured. HO-1 conditional knockouts were used to study myeloid, neuronal, and astrocyte HO-1 involvement in DFX-induced neuroprotection and cognitive recovery. RESULTS DFX treatment after SAH decreased cortical damage and improved cognitive outcome after SAH yet had no effect on vasospasm; ICV DFX was most neuroprotective. ICV DFX treatment after SAH decreased cerebral IL-6 concentration and trended towards decreased mitochondrial superoxide anion production. ICV DFX treatment after SAH effected an increase in HO-1 co-localization to microglia. DFX treatment of WT microglia with RBCs in the trans-wells showed decreased neuronal damage; this effect was abolished in HO-1(-/-) microglia. ICV DFX after SAH decreased neuronal damage and improved cognition in Hmox1 (fl/fl) control and Nes (Cre) :Hmox1 (fl/fl) mice, but not LyzM (Cre) :Hmox1 (fl/fl) mice. CONCLUSIONS DFX neuroprotection is independent of vasospasm. ICV DFX treatment provides superior neuroprotection in a mouse model of SAH. Mechanisms of DFX neuroprotection after SAH may involve microglial/macrophage HO-1 expression. Monitoring patient HO-1 expression during DFX treatment for hemorrhagic stroke may help clinicians identify patients that are more likely to respond to treatment.
Collapse
Affiliation(s)
- Robert H LeBlanc
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, 3 Blackfan Circle, Boston, MA, 02140, USA
| | - Ruiya Chen
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, 3 Blackfan Circle, Boston, MA, 02140, USA
| | - Magdy H Selim
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, 3 Blackfan Circle, Boston, MA, 02140, USA
| | - Khalid A Hanafy
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, 3 Blackfan Circle, Boston, MA, 02140, USA. .,Division of Neurointensive Care Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 3 Blackfan Circle, Boston, MA, 02140, USA.
| |
Collapse
|
43
|
Early expression of serum neutrophil gelatinase-associated lipocalin (NGAL) is associated with neurological severity immediately after traumatic brain injury. J Neurol Sci 2016; 368:392-8. [DOI: 10.1016/j.jns.2016.07.060] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Revised: 06/28/2016] [Accepted: 07/25/2016] [Indexed: 11/19/2022]
|
44
|
An C, Jiang X, Pu H, Hong D, Zhang W, Hu X, Gao Y. Severity-Dependent Long-Term Spatial Learning-Memory Impairment in a Mouse Model of Traumatic Brain Injury. Transl Stroke Res 2016; 7:512-520. [DOI: 10.1007/s12975-016-0483-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 06/28/2016] [Accepted: 07/11/2016] [Indexed: 01/04/2023]
|
45
|
Intraventricular Hemorrhage: the Role of Blood Components in Secondary Injury and Hydrocephalus. Transl Stroke Res 2016; 7:447-451. [DOI: 10.1007/s12975-016-0480-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 06/22/2016] [Indexed: 01/24/2023]
|
46
|
Shishido H, Toyota Y, Hua Y, Keep RF, Xi G. Role of lipocalin 2 in intraventricular haemoglobin-induced brain injury. Stroke Vasc Neurol 2016; 1:37-43. [PMID: 28959462 PMCID: PMC5435192 DOI: 10.1136/svn-2016-000009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 04/12/2016] [Accepted: 04/14/2016] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Our recent studies have shown that blood components, including haemoglobin and iron, contribute to hydrocephalus development and brain injury after intraventricular haemorrhage (IVH). The current study investigated the role of lipocalin 2 (LCN2), a protein involved in iron handling, in the ventricular dilation and neuroinflammation caused by brain injury in a mouse model of IVH. DESIGN Female wild-type (WT) C57BL/6 mice and LCN2-deficient (LCN2-/-) mice had an intraventricular injection of haemoglobin, and control mice received an equivalent amount of saline. MRI was performed presurgery and postsurgery to measure ventricular volume and the brains were used for either immunohistochemistry or western blot. RESULTS Ventricular dilation was observed in WT mice at 24 h after haemoglobin (25 mg/mL, 20 µL) injection (12.5±2.4 vs 8.6±1.5 mm3 in the control, p<0.01). Western blotting showed that LCN2 was significantly upregulated in the periventricular area (p<0.01). LCN2 was mainly expressed in astrocytes, whereas the LCN2 receptor was detected in astrocytes, microglia/macrophages and neurons. Haemoglobin-induced ventricle dilation and glia activation were less in LCN2-/- mice (p<0.01). Injection of high-dose haemoglobin (50 mg/mL) resulted in lower mortality in LCN2-/- mice (27% vs 86% in WT; p<0.05). CONCLUSIONS Intraventricular haemoglobin caused LCN2 upregulation and ventricular dilation. Haemoglobin resulted in lower mortality and less ventricular dilation in LCN2-/- mice. These results suggest that LCN2 has a role in haemoglobin-induced brain injury and may be a therapeutic target for IVH.
Collapse
Affiliation(s)
- Hajime Shishido
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Yasunori Toyota
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Ya Hua
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
47
|
Xu H. New concept of the pathogenesis and therapeutic orientation of acquired communicating hydrocephalus. Neurol Sci 2016; 37:1387-91. [DOI: 10.1007/s10072-016-2589-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 04/19/2016] [Indexed: 12/12/2022]
|
48
|
Cheng T, Wang W, Li Q, Han X, Xing J, Qi C, Lan X, Wan J, Potts A, Guan F, Wang J. Cerebroprotection of flavanol (-)-epicatechin after traumatic brain injury via Nrf2-dependent and -independent pathways. Free Radic Biol Med 2016; 92:15-28. [PMID: 26724590 PMCID: PMC4769660 DOI: 10.1016/j.freeradbiomed.2015.12.027] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 12/09/2015] [Accepted: 12/23/2015] [Indexed: 12/13/2022]
Abstract
Traumatic brain injury (TBI), which leads to disability, dysfunction, and even death, is a prominent health problem worldwide with no effective treatment. A brain-permeable flavonoid named (-)-epicatechin (EC) modulates redox/oxidative stress and has been shown to be beneficial for vascular and cognitive function in humans and for ischemic and hemorrhagic stroke in rodents. Here we examined whether EC is able to protect the brain against TBI-induced brain injury in mice and if so, whether it exerts neuroprotection by modulating the NF-E2-related factor (Nrf2) pathway. We used the controlled cortical impact model to mimic TBI. EC was administered orally at 3h after TBI and then every 24h for either 3 or 7 days. We evaluated lesion volume, brain edema, white matter injury, neurologic deficits, cognitive performance and emotion-like behaviors, neutrophil infiltration, reactive oxygen species (ROS), and a variety of injury-related protein markers. Nrf2 knockout mice were used to determine the role of the Nrf2 signaling pathway after EC treatment. In wild-type mice, EC significantly reduced lesion volume, edema, and cell death and improved neurologic function on days 3 and 28; cognitive performance and depression-like behaviors were also improved with EC administration. In addition, EC reduced white matter injury, heme oxygenase-1 expression, and ferric iron deposition after TBI. These changes were accompanied by attenuation of neutrophil infiltration and oxidative insults, reduced activity of matrix metalloproteinase 9, decreased Keap 1 expression, increased Nrf2 nuclear accumulation, and increased expression of superoxide dismutase 1 and quinone 1. However, EC did not significantly reduce lesion volume or improve neurologic deficits in Nrf2 knockout mice after TBI. Our results show that EC protects the TBI brain by activating the Nrf2 pathway, inhibiting heme oxygenase-1 protein expression, and reducing iron deposition. The latter two effects could represent an Nrf2-independent mechanism in this model of TBI.
Collapse
Affiliation(s)
- Tian Cheng
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, PR China; Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA
| | - Wenzhu Wang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA
| | - Qian Li
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA
| | - Xiaoning Han
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA
| | - Jing Xing
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA
| | - Cunfang Qi
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA
| | - Xi Lan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA
| | - Jieru Wan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA
| | - Alexa Potts
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA
| | - Fangxia Guan
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, PR China; School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450000, PR China.
| | - Jian Wang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, PR China; Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
49
|
Del Bigio MR, Di Curzio DL. Nonsurgical therapy for hydrocephalus: a comprehensive and critical review. Fluids Barriers CNS 2016; 13:3. [PMID: 26846184 PMCID: PMC4743412 DOI: 10.1186/s12987-016-0025-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 01/15/2016] [Indexed: 12/13/2022] Open
Abstract
Pharmacological interventions have been tested experimentally and clinically to prevent hydrocephalus and avoid the need for shunting beginning in the 1950s. Clinical trials of varied quality have not demonstrated lasting and convincing protective effects through manipulation of cerebrospinal fluid production, diuresis, blood clot fibrinolysis, or manipulation of fibrosis in the subarachnoid compartment, although there remains some promise in the latter areas. Acetazolamide bolus seems to be useful for predicting shunt response in adults with hydrocephalus. Neuroprotection in the situation of established hydrocephalus has been tested experimentally beginning more recently. Therapies designed to modify blood flow or pulsation, reduce inflammation, reduce oxidative damage, or protect neurons are so far of limited success; more experimental work is needed in these areas. As has been recommended for preclinical studies in stroke and brain trauma, stringent conditions should be met for preclinical studies in hydrocephalus.
Collapse
Affiliation(s)
- Marc R Del Bigio
- Department of Pathology, University of Manitoba; Children's Hospital Research Institute of Manitoba, Diagnostic Services Manitoba, 401 Brodie Centre, 715 McDermot Avenue, Winnipeg, MB, R3E 3P5, Canada.
| | - Domenico L Di Curzio
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Canada.
| |
Collapse
|
50
|
Lekic T, Krafft PR, Klebe D, Flores J, Rolland WB, Tang J, Zhang JH. PAR-1, -4, and the mTOR Pathway Following Germinal Matrix Hemorrhage. ACTA NEUROCHIRURGICA. SUPPLEMENT 2016; 121:213-6. [PMID: 26463951 DOI: 10.1007/978-3-319-18497-5_38] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Germinal matrix hemorrhage (GMH) is the most common cause of neurological complications of prematurity and has lasting implications. PAR-1 and PAR-4 receptors are involved with upstream signaling pathways following brain hemorrhage in adult models of stroke, of which the mammalian target of rapamycin (mTOR) is a potential downstream mediator. Therefore, we hypothesized a role for PAR-1, -4/ mTOR signaling following GMH brain injury. Postnatal day 7 Sprague-Dawley rats were subjected to GMH through stereotactic infusion of collagenase into the right ganglionic eminence. Rodents were euthanized at 72 h (short term), or 4 weeks (long term). Short-term mTOR expression was evaluated by Western blot in the context of PAR-1 (SCH-79797) and PAR-4 (P4pal10) inhibition. Pups in the long-term group were administered the selective mTOR inhibitor (rapamycin) with neurobehavioral and brain pathological examinations performed at 4 weeks. Pharmacological PAR-1, -4 antagonism normalized the increased mTOR expression following GMH. Early inhibition of mTOR by rapamycin improved long-term outcomes in rats. Mammalian-TOR signaling plays an important role in brain injury following neonatal GMH, possibly involving upstream PAR-1, -4 mechanisms.
Collapse
Affiliation(s)
- Tim Lekic
- Division of Physiology and Pharmacology, School of Medicine, Loma Linda, CA, USA
| | - Paul R Krafft
- Division of Physiology and Pharmacology, School of Medicine, Loma Linda, CA, USA
| | - Damon Klebe
- Division of Physiology and Pharmacology, School of Medicine, Loma Linda, CA, USA
| | - Jerry Flores
- Division of Physiology and Pharmacology, School of Medicine, Loma Linda, CA, USA
| | - William B Rolland
- Division of Physiology and Pharmacology, School of Medicine, Loma Linda, CA, USA
| | - Jiping Tang
- Division of Physiology and Pharmacology, School of Medicine, Loma Linda, CA, USA
| | - John H Zhang
- Division of Physiology and Pharmacology, School of Medicine, Loma Linda, CA, USA. .,Department of Neurosurgery, School of Medicine, Loma Linda, CA, USA. .,Department of Physiology and Pharmacology, Loma Linda University School of Medicine, 11041 Campus Street, Risley Hall Rm 219, Loma Linda, CA, 92354, USA.
| |
Collapse
|