1
|
Bayiroglu AF, Acar G, Gulbahce-Mutlu E, Baltaci SB, Mogulkoc R, Baltaci AK. Dietary zinc status is associated with ZnT3 (SLC30A3), IL-6 gene expressions and spinal cord tissue damage in spinal cord tissue in a cuprizone-induced rat Multiple Sclerosis model. J Trace Elem Med Biol 2024; 86:127540. [PMID: 39383661 DOI: 10.1016/j.jtemb.2024.127540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/26/2024] [Accepted: 09/29/2024] [Indexed: 10/11/2024]
Abstract
The aim of this study was to investigate the effects of dietary zinc status on spinal cord tissue damage and ZnT3, IL-6 gene expressions in a cuprizone-induced rat Multiple Sclerosis (MS) model. The study was carried out on 46 adult male rats of the genus Wistar. The animals used in the study were divided into 5 groups (G) (Control 6, other groups 10). G1, Control. G2, Sham-MS: Carboxy-methyl-cellulose (KMS) solution in which Cuprizon was dissolved was given to rats by gavage daily for 8 weeks at the rate of 1 % of daily feed consumption. MS was formed by giving 1 % of the daily feed consumption cuprizon in KMS solution by gavage to the animals in G3, 4 and 5 for 8 weeks. G4 was fed with a zinc deficient (50 µg/kg zinc) diet. G5 was given intraperitoneal (ip) zinc sulfate (5 mg/kg/day) supplementation. MS formation in animals was determined by Rotarod tests and Myelin Basic Protein (MBP) gene expression analysis. ZNT3 and IL-6 gene expression levels in spinal cord tissue samples of animals by Real-Time-PCR method; MDA and GSH levels were determined by ELISA method. The highest spinal cord MDA and IL-6 levels were obtained in G3 and G4 (P<0.05). Zinc supplementation in G5 prevented the increase in the mentioned parameters and turned them into control values (P<0.05). The spinal cord GSH and ZnT3 levels of G3 and G4 were lower than all other groups (P<0.05). Zinc supplementation prevented suppression in the same parameters in G5 and reached the control values (P<0.05). The findings of the current study suggest that zinc supplementation in addition to treatment for MS may be beneficial in reducing the severity of the disease.
Collapse
Affiliation(s)
- Aysenur Feyza Bayiroglu
- Bandirma Onyedi Eylul University, Medical Faculty, Department of Physiology, Bandırma, Turkey
| | - Gozde Acar
- Selcuk University, Medical Faculty, Department of Physiology, Konya, Turkey
| | - Elif Gulbahce-Mutlu
- KTO Karatay University, Medical Faculty, Department of Medical Biology, Konya, Turkey
| | - Saltuk Bugra Baltaci
- İstanbul Medipol University, Medical Faculty, Department of Physiology, İstanbul, Turkey
| | - Rasim Mogulkoc
- Selcuk University, Medical Faculty, Department of Physiology, Konya, Turkey
| | | |
Collapse
|
2
|
Jahromi HM, Rafati A, Karbalay-Doust S, Keshavarz S, Naseh M. The combination treatment of hypothermia and intranasal insulin ameliorates the structural and functional changes in a rat model of traumatic brain injury. Brain Struct Funct 2024; 229:947-957. [PMID: 38498064 DOI: 10.1007/s00429-024-02769-5] [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: 04/19/2023] [Accepted: 01/26/2024] [Indexed: 03/19/2024]
Abstract
The present study aimed to investigate the combination effects of hypothermia (HT) and intranasal insulin (INS) on structural changes of the hippocampus and cognitive impairments in the traumatic brain injury (TBI) rat model. The rats were divided randomly into the following five groups (n = 10): Sham, TBI, TBI with HT treatment for 3 h (TBI + HT), TBI with INS (ten microliters of insulin) treatment daily for 7 days (TBI + INS), and TBI with combining HT and INS (TBI + HT + INS). At the end of the 7th day, the open field and the Morris water maze tests were done for evaluation of anxiety-like behavior and memory performance. Then, after sacrificing, the brain was removed for stereological study. TBI led to an increase in the total volume of hippocampal subfields CA1 and DG and a decrease in the total number of neurons and non-neuronal cells in both sub-regions, which was associated with anxiety-like behavior and memory impairment. Although, the combination of HT and INS prevented the increased hippocampal volume and cell loss and improved behavioral performances in the TBI group. Our study suggests that the combined treatment of HT and INS could prevent increased hippocampal volume and cell loss in CA1 and DG sub-regions and consequently improve anxiety-like behaviors and memory impairment following TBI.
Collapse
Affiliation(s)
- Hadi Moatamed Jahromi
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Physiology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Rafati
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Physiology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saied Karbalay-Doust
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Anatomy Department, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Somaye Keshavarz
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Physiology, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Maryam Naseh
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
3
|
Zinc in the Brain: Friend or Foe? Int J Mol Sci 2020; 21:ijms21238941. [PMID: 33255662 PMCID: PMC7728061 DOI: 10.3390/ijms21238941] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/22/2020] [Accepted: 11/24/2020] [Indexed: 12/12/2022] Open
Abstract
Zinc is a trace metal ion in the central nervous system that plays important biological roles, such as in catalysis, structure, and regulation. It contributes to antioxidant function and the proper functioning of the immune system. In view of these characteristics of zinc, it plays an important role in neurophysiology, which leads to cell growth and cell proliferation. However, after brain disease, excessively released and accumulated zinc ions cause neurotoxic damage to postsynaptic neurons. On the other hand, zinc deficiency induces degeneration and cognitive decline disorders, such as increased neuronal death and decreased learning and memory. Given the importance of balance in this context, zinc is a biological component that plays an important physiological role in the central nervous system, but a pathophysiological role in major neurological disorders. In this review, we focus on the multiple roles of zinc in the brain.
Collapse
|
4
|
Effects of Transient Receptor Potential Cation 5 (TRPC5) Inhibitor, NU6027, on Hippocampal Neuronal Death after Traumatic Brain Injury. Int J Mol Sci 2020; 21:ijms21218256. [PMID: 33158109 PMCID: PMC7662546 DOI: 10.3390/ijms21218256] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 10/30/2020] [Accepted: 11/03/2020] [Indexed: 12/20/2022] Open
Abstract
Traumatic brain injury (TBI) can cause physical, cognitive, social, and behavioral changes that can lead to permanent disability or death. After primary brain injury, translocated free zinc can accumulate in neurons and lead to secondary events such as oxidative stress, inflammation, edema, swelling, and cognitive impairment. Under pathological conditions, such as ischemia and TBI, excessive zinc release, and accumulation occurs in neurons. Based on previous research, it hypothesized that calcium as well as zinc would be influx into the TRPC5 channel. Therefore, we hypothesized that the suppression of TRPC5 would prevent neuronal cell death by reducing the influx of zinc and calcium. To test our hypothesis, we used a TBI animal model. After the TBI, we immediately injected NU6027 (1 mg/kg, intraperitoneal), TRPC5 inhibitor, and then sacrificed animals 24 h later. We conducted Fluoro-Jade B (FJB) staining to confirm the presence of degenerating neurons in the hippocampal cornus ammonis 3 (CA3). After the TBI, the degenerating neuronal cell count was decreased in the NU6027-treated group compared with the vehicle-treated group. Our findings suggest that the suppression of TRPC5 can open a new therapeutic window for a reduction of the neuronal death that may occur after TBI.
Collapse
|
5
|
Baraibar AM, Hernández-Guijo JM. Micromolar concentrations of Zn 2+ depress cellular excitability through a blockade of calcium current in rat adrenal slices. Toxicology 2020; 444:152543. [PMID: 32858065 DOI: 10.1016/j.tox.2020.152543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/08/2020] [Accepted: 07/25/2020] [Indexed: 11/30/2022]
Abstract
The present work, using chromaffin cells in rat adrenal slices (RCCs), aims to describe what type of ionic current alterations induced by zinc underlies their effects reported on synaptic transmission. Thus, Zn2+ blocked calcium channels of RCCs in a time- and concentration-dependent manner with an IC50 of 391 μM. This blockade was partially reversed upon washout and was greater at more depolarizing holding potentials (i.e. 32 ± 5% at -110 mV, and 43 ± 6% at -50 mV, after 5 min perfusion). In ω-toxins-sensitive calcium channels (N-, P- and Q-types), Zn2+caused a lower blockade of ICa, 33.3%, than in ω-toxins-resistant ones (L-type, 55.3%; and R-type, 90%). This compound inhibited calcium current at all test potentials and shows a shift of the I-V curve to more depolarized values of about 10 mV. The sodium current was not blocked by acute application of high Zn2+concentrations. Voltage-dependent potassium current was marginally affected by high Zn2+ concentrations showing no concentration-dependence. Nevertheless, calcium- and voltage-dependent potassium current was drastically depressed in a dose-dependent manner, with an IC50 of 453 μM. This blockade was related to the prevention of Ca2+ influx through voltage-dependent calcium channels coupled to BK channels. Under current-clamp conditions, RCCs exhibit a resting potential of -50.7 mV, firing spontaneous APs (1-2 spikes/s) generated by the opening of Na+ and Ca2+-channels, and terminated by the activation of voltage and Ca2+-activated K+-channels (BK). We found that the blockade of these ionic currents by Zn2+ led to a drastic alteration of cellular excitability with a depolarization of the membrane potential, the slowdown and broadening of the APs and the severe reduction of the after hyperpolarization (AHP) which led to a decrease in the APs firing frequency. Taken together, these results point to a neurotoxic action evoked by zinc that is associated with changes to cellular excitability by blocking the ionic currents responsible for both the neurotransmitter release and the action potentials firing.
Collapse
Affiliation(s)
- Andrés M Baraibar
- Department of Neuroscience, University of Minnesota, 4-260 Wallin Medical Biosciences Building, 2101 6th Street SE, Minneapolis, MN, 55455, USA
| | - Jesús M Hernández-Guijo
- Department of Pharmacology and Therapeutic, Univ. Autónoma de Madrid, Av. Arzobispo Morcillo 4, 28029, Madrid, Spain; Instituto Teófilo Hernando, Facultad de Medicina, Univ. Autónoma de Madrid, Av. Arzobispo Morcillo 4, 28029, Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS, Hospital Ramón y Cajal, Ctra. de Colmenar Viejo, Km. 9,100, 28029, Madrid, Spain.
| |
Collapse
|
6
|
Transient Receptor Potential Melastatin 2 (TRPM2) Inhibition by Antioxidant, N-Acetyl-l-Cysteine, Reduces Global Cerebral Ischemia-Induced Neuronal Death. Int J Mol Sci 2020; 21:ijms21176026. [PMID: 32825703 PMCID: PMC7504640 DOI: 10.3390/ijms21176026] [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: 07/09/2020] [Revised: 08/18/2020] [Accepted: 08/20/2020] [Indexed: 12/14/2022] Open
Abstract
A variety of pathogenic mechanisms, such as cytoplasmic calcium/zinc influx, reactive oxygen species production, and ionic imbalance, have been suggested to play a role in cerebral ischemia induced neurodegeneration. During the ischemic state that occurs after stroke or heart attack, it is observed that vesicular zinc can be released into the synaptic cleft, and then translocated into the cytoplasm via various cation channels. Transient receptor potential melastatin 2 (TRPM2) is highly distributed in the central nervous system and has high sensitivity to oxidative damage. Several previous studies have shown that TRPM2 channel activation contributes to neuroinflammation and neurodegeneration cascades. Therefore, we examined whether anti-oxidant treatment, such as with N-acetyl-l-cysteine (NAC), provides neuroprotection via regulation of TRPM2, following global cerebral ischemia (GCI). Experimental animals were then immediately injected with NAC (150 mg/kg/day) for 3 and 7 days, before sacrifice. We demonstrated that NAC administration reduced activation of GCI-induced neuronal death cascades, such as lipid peroxidation, microglia and astroglia activation, free zinc accumulation, and TRPM2 over-activation. Therefore, modulation of the TRPM2 channel can be a potential therapeutic target to prevent ischemia-induced neuronal death.
Collapse
|
7
|
Choi BY, Lee SH, Choi HC, Lee SK, Yoon HS, Park JB, Chung WS, Suh SW. Alcohol dependence treating agent, acamprosate, prevents traumatic brain injury-induced neuron death through vesicular zinc depletion. Transl Res 2019; 207:1-18. [PMID: 30731068 DOI: 10.1016/j.trsl.2019.01.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 01/11/2019] [Accepted: 01/14/2019] [Indexed: 12/29/2022]
Abstract
Acamprosate, also known as N-acetyl homotaurine, is an N-methyl-d-aspartate receptor antagonist that is used for treating alcohol dependence. Although the exact mechanism of acamprosate has not been clearly established, it appears to work by promoting a balance between the excitatory and inhibitory neurotransmitters, glutamate, and gamma-aminobutyric acid, respectively. Several studies have demonstrated that acamprosate provides neuroprotection against ischemia-induced brain injury. However, no studies have been performed evaluating the effect of acamprosate on traumatic brain injury (TBI). In the present study, we sought to evaluate the therapeutic potential of acamprosate to protect against neuronal death following TBI. Rats were given oral acamprosate (200 mg/kg/d for 2weeks) and then subjected to a controlled cortical impact injury localized over the parietal cortex. Histologic analysis was performed at 3hours, 24hours, and 7days after TBI. We found that acamprosate treatment reduced the concentration of vesicular glutamate and zinc in the hippocampus. Consequently, this reduced vesicular glutamate and zinc level resulted in a reduction of reactive oxygen species production after TBI. When evaluated 24hours after TBI, acamprosate administration reduced the number of degenerating neurons, zinc accumulation, blood-brain barrier disruption, neutrophil infiltration, and dendritic loss. Acamprosate also reduced glial activation and neuronal loss at 7days after TBI. In addition, acamprosate rescued TBI-induced neurologic and cognitive dysfunction. The present study demonstrates that acamprosate attenuates TBI-induced brain damage by depletion of vesicular glutamate and zinc levels. Therefore, this study suggests that acamprosate may have high therapeutic potential for prevention of TBI-induced neuronal death.
Collapse
Affiliation(s)
- Bo Young Choi
- Department of Physiology, Hallym University, College of Medicine, Chuncheon, Korea
| | - Song Hee Lee
- Department of Physiology, Hallym University, College of Medicine, Chuncheon, Korea
| | - Hui Chul Choi
- Department of Neurology, Hallym University, College of Medicine, Chuncheon, Korea
| | - Sang-Kyu Lee
- Department of Psychiatry, Hallym University, College of Medicine, Chuncheon, Korea
| | | | - Jae Bong Park
- Department of Biochemistry, Hallym University, College of Medicine, Chuncheon, Korea
| | - Won Suk Chung
- Department of Biological Science, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Sang Won Suh
- Department of Physiology, Hallym University, College of Medicine, Chuncheon, Korea.
| |
Collapse
|
8
|
Portbury SD, Sedjahtera A, Perrones K, Sgambelloni C, Zhang M, Crack PJ, Finkelstein DI, Adlard PA. Metal chaperones: a novel therapeutic strategy for brain injury? Brain Inj 2018; 33:305-312. [PMID: 30507321 DOI: 10.1080/02699052.2018.1552988] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE This study sought to assess the potential efficacy of a novel class of metal chaperone on the outcomes in an animal model of a controlled cortical impact. This work was predicated on previous observations that this class of compound has exhibited neuroprotective potential in other models of aging and neurodegeneration. RESEARCH DESIGN The study employed a controlled cortical impact traumatic brain injury in three month old mice with subsequent behavioral and cellular assessments to determine therapeutic efficacy. METHODS Cognitive (Y-maze) and motor assessments (Rotarod and Open Field) were employed to determine behavioral end points. Histological-based methods were utilized to assess neuronal integrity, astrocytosis, and lesion volume. OUTCOMES We demonstrate here that acute post-injury treatment with PBT2 (Prana Biotechnology) is sufficient to maintain neuronal integrity (evidenced by decreased lesion area and increased numbers of neurons; decreased astrocytosis was also present) and to normalize performance in cognitive testing (Y-maze). These effects occurred within days and were maintained for the entire duration of the study (26 days post-injury). These data support the further interrogation of the utility of metal chaperones for the treatment and/or prevention of the neuroanatomical, biochemical, and behavioral deficits that occur following brain injuries of different etiologies.
Collapse
Affiliation(s)
- S D Portbury
- a The Florey Institute of Neuroscience and Mental Health, Dementia Theme , Parkville, Victoria , Australia
| | - A Sedjahtera
- a The Florey Institute of Neuroscience and Mental Health, Dementia Theme , Parkville, Victoria , Australia
| | - K Perrones
- a The Florey Institute of Neuroscience and Mental Health, Dementia Theme , Parkville, Victoria , Australia
| | - C Sgambelloni
- a The Florey Institute of Neuroscience and Mental Health, Dementia Theme , Parkville, Victoria , Australia
| | - M Zhang
- b Department of Pharmacology and Therapeutics , The University of Melbourne , Parkville, Victoria , Australia
| | - P J Crack
- b Department of Pharmacology and Therapeutics , The University of Melbourne , Parkville, Victoria , Australia
| | - D I Finkelstein
- a The Florey Institute of Neuroscience and Mental Health, Dementia Theme , Parkville, Victoria , Australia
| | - P A Adlard
- a The Florey Institute of Neuroscience and Mental Health, Dementia Theme , Parkville, Victoria , Australia.,c The University of Melbourne , Melbourne , Australia
| |
Collapse
|
9
|
Administration of Protocatechuic Acid Reduces Traumatic Brain Injury-Induced Neuronal Death. Int J Mol Sci 2017; 18:ijms18122510. [PMID: 29168791 PMCID: PMC5751113 DOI: 10.3390/ijms18122510] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 11/17/2017] [Accepted: 11/21/2017] [Indexed: 01/27/2023] Open
Abstract
Protocatechuic acid (PCA) was first purified from green tea and has shown numerous biological activities, including anti-apoptotic, anti-inflammatory, and anti-atherosclerotic effects. The effect of PCA on traumatic brain injury (TBI)-induced neuronal death has not previously been evaluated. TBI is defined as damage to the brain resulting from external mechanical force, such as rapid acceleration or deceleration, impact, blast waves, or penetration by a projectile. TBI causes neuronal death in the hippocampus and cerebral cortex. The present study aimed to evaluate the therapeutic potential of PCA on TBI-induced neuronal death. Here, TBI was induced by a controlled cortical impact model using rats. PCA (30 mg/kg) was injected into the intraperitoneal (ip) space immediately after TBI. Neuronal death was evaluated with Fluoro Jade-B (FJB) staining at 24 h after TBI. Oxidative injury was detected by 4-hydroxy-2-nonenal (4HNE), glutathione (GSH) concentration was analyzed by glutathione adduct with N-ethylmaleimide (GS-NEM) staining at 24 h after TBI, and microglial activation in the hippocampus was detected by CD11b immunohistochemistry at one week after TBI. We found that the proportion of degenerating neurons, oxidative injury, GSH depletion, and microglia activation in the hippocampus and cortex were all reduced by PCA treatment following TBI. Therefore, our study suggests that PCA may have therapeutic potential in preventing TBI-induced neuronal death.
Collapse
|
10
|
Galgano M, Toshkezi G, Qiu X, Russell T, Chin L, Zhao LR. Traumatic Brain Injury: Current Treatment Strategies and Future Endeavors. Cell Transplant 2017; 26:1118-1130. [PMID: 28933211 PMCID: PMC5657730 DOI: 10.1177/0963689717714102] [Citation(s) in RCA: 319] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 10/16/2016] [Accepted: 10/18/2016] [Indexed: 01/04/2023] Open
Abstract
Traumatic brain injury (TBI) presents in various forms ranging from mild alterations of consciousness to an unrelenting comatose state and death. In the most severe form of TBI, the entirety of the brain is affected by a diffuse type of injury and swelling. Treatment modalities vary extensively based on the severity of the injury and range from daily cognitive therapy sessions to radical surgery such as bilateral decompressive craniectomies. Guidelines have been set forth regarding the optimal management of TBI, but they must be taken in context of the situation and cannot be used in every individual circumstance. In this review article, we have summarized the current status of treatment for TBI in both clinical practice and basic research. We have put forth a brief overview of the various subtypes of traumatic injuries, optimal medical management, and both the noninvasive and invasive monitoring modalities, in addition to the surgical interventions necessary in particular instances. We have overviewed the main achievements in searching for therapeutic strategies of TBI in basic science. We have also discussed the future direction for developing TBI treatment from an experimental perspective.
Collapse
Affiliation(s)
- Michael Galgano
- Department of Neurosurgery, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Gentian Toshkezi
- Department of Neurosurgery, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Xuecheng Qiu
- Department of Neurosurgery, SUNY Upstate Medical University, Syracuse, NY, USA
- VA Health Care Upstate New York, Syracuse VA Medical Center, Syracuse, NY, USA
| | - Thomas Russell
- Department of Neurosurgery, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Lawrence Chin
- Department of Neurosurgery, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Li-Ru Zhao
- Department of Neurosurgery, SUNY Upstate Medical University, Syracuse, NY, USA
- VA Health Care Upstate New York, Syracuse VA Medical Center, Syracuse, NY, USA
| |
Collapse
|
11
|
Nyanzu M, Siaw-Debrah F, Ni H, Xu Z, Wang H, Lin X, Zhuge Q, Huang L. Improving on Laboratory Traumatic Brain Injury Models to Achieve Better Results. Int J Med Sci 2017; 14:494-505. [PMID: 28539826 PMCID: PMC5441042 DOI: 10.7150/ijms.18075] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 01/31/2017] [Indexed: 11/30/2022] Open
Abstract
Experimental modeling of traumatic brain injury (TBI) in animals has identified several potential means and interventions that might have beneficial applications for treating traumatic brain injury clinically. Several of these interventions have been applied and tried with humans that are at different phases of testing (completed, prematurely terminated and others in progress). The promising results achieved in the laboratory with animal models have not been replicated with human trails as expected. This review will highlight some insights and significance attained via laboratory animal modeling of TBI as well as factors that require incorporation into the experimental studies that could help in translating results from laboratory to the bedside. Major progress has been made due to laboratory studies; in explaining the mechanisms as well as pathophysiological features of brain damage after TBI. Attempts to intervene in the cascade of events occurring after TBI all rely heavily on the knowledge from basic laboratory investigations. In looking to discover treatment, this review will endeavor to sight and state some central discrepancies between laboratory models and clinical scenarios.
Collapse
Affiliation(s)
- Mark Nyanzu
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325000, China.,Department of Neurosurgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Felix Siaw-Debrah
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325000, China.,Department of Neurosurgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Haoqi Ni
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325000, China.,Department of Neurosurgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Zhu Xu
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325000, China.,Department of Neurosurgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Hua Wang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325000, China.,Department of Neurosurgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Xiao Lin
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325000, China.,Department of Neurosurgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Qichuan Zhuge
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325000, China.,Department of Neurosurgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Lijie Huang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325000, China.,Department of Neurosurgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| |
Collapse
|
12
|
Shijo K, Sutton RL, Ghavim SS, Harris NG, Bartnik-Olson BL. Metabolic fate of glucose in rats with traumatic brain injury and pyruvate or glucose treatments: A NMR spectroscopy study. Neurochem Int 2016; 102:66-78. [PMID: 27919624 DOI: 10.1016/j.neuint.2016.11.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 11/30/2016] [Accepted: 11/30/2016] [Indexed: 12/27/2022]
Abstract
Administration of sodium pyruvate (SP; 9.08 μmol/kg, i.p.), ethyl pyruvate (EP; 0.34 μmol/kg, i.p.) or glucose (GLC; 11.1 μmol/kg, i.p.) to rats after unilateral controlled cortical impact (CCI) injury has been reported to reduce neuronal loss and improve cerebral metabolism. In the present study these doses of each fuel or 8% saline (SAL; 5.47 nmoles/kg) were administered immediately and at 1, 3, 6 and 23 h post-CCI. At 24 h all CCI groups and non-treated Sham injury controls were infused with [1,2 13C] glucose for 68 min 13C nuclear magnetic resonance (NMR) spectra were obtained from cortex + hippocampus tissues from left (injured) and right (contralateral) hemispheres. All three fuels increased lactate labeling to a similar degree in the injured hemisphere. The amount of lactate labeled via the pentose phosphate and pyruvate recycling (PPP + PR) pathway increased in CCI-SAL and was not improved by SP, EP, and GLC treatments. Oxidative metabolism, as assessed by glutamate labeling, was reduced in CCI-SAL animals. The greatest improvement in oxidative metabolism was observed in animals treated with SP and fewer improvements after EP or GLC treatments. Compared to SAL, all three fuels restored glutamate and glutamine labeling via pyruvate carboxylase (PC), suggesting improved astrocyte metabolism following fuel treatment. Only SP treatments restored the amount of [4 13C] glutamate labeled by the PPP + PR pathway to sham levels. Milder injury effects in the contralateral hemisphere appear normalized by either SP or EP treatments, as increases in the total pool of 13C lactate and labeling of lactate in glycolysis, or decreases in the ratio of PC/PDH labeling of glutamine, were found only for CCI-SAL and CCI-GLC groups compared to Sham. The doses of SP, EP and GLC examined in this study all enhanced lactate labeling and restored astrocyte-specific PC activity but differentially affected neuronal metabolism after CCI injury. The restoration of astrocyte metabolism by all three fuel treatments may partially underlie their abilities to improve cerebral glucose utilization and to reduce neuronal loss following CCI injury.
Collapse
Affiliation(s)
- Katsunori Shijo
- Brain Injury Research Center, Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, Box 956901, CA, USA.
| | - Richard L Sutton
- Brain Injury Research Center, Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, Box 956901, CA, USA.
| | - Sima S Ghavim
- Brain Injury Research Center, Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, Box 956901, CA, USA.
| | - Neil G Harris
- Brain Injury Research Center, Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, Box 956901, CA, USA.
| | | |
Collapse
|
13
|
Choi BY, Kim IY, Kim JH, Lee BE, Lee SH, Kho AR, Sohn M, Suh SW. Zinc plus cyclo-(His-Pro) promotes hippocampal neurogenesis in rats. Neuroscience 2016; 339:634-643. [DOI: 10.1016/j.neuroscience.2016.10.035] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 09/23/2016] [Accepted: 10/12/2016] [Indexed: 10/20/2022]
|
14
|
Choi BY, Kim IY, Kim JH, Kho AR, Lee SH, Lee BE, Sohn M, Koh JY, Suh SW. Zinc transporter 3 (ZnT3) gene deletion reduces spinal cord white matter damage and motor deficits in a murine MOG-induced multiple sclerosis model. Neurobiol Dis 2016; 94:205-12. [DOI: 10.1016/j.nbd.2016.06.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 05/20/2016] [Accepted: 06/27/2016] [Indexed: 12/25/2022] Open
|
15
|
Decreased cysteine uptake by EAAC1 gene deletion exacerbates neuronal oxidative stress and neuronal death after traumatic brain injury. Amino Acids 2016; 48:1619-29. [DOI: 10.1007/s00726-016-2221-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 03/21/2016] [Indexed: 12/22/2022]
|
16
|
Choi BY, Lee BE, Kim JH, Kim HJ, Sohn M, Song HK, Chung TN, Suh SW. Colchicine induced intraneuronal free zinc accumulation and dentate granule cell degeneration. Metallomics 2015; 6:1513-20. [PMID: 24874779 DOI: 10.1039/c4mt00067f] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Colchicine has been discovered to inhibit many inflammatory processes such as gout, familial Mediterranean fever, pericarditis and Behcet disease. Other than these beneficial anti-inflammatory effects, colchicine blocks microtubule-assisted axonal transport, which results in the selective loss of dentate granule cells of the hippocampus. The mechanism of the colchicine-induced dentate granule cell death and depletion of mossy fiber terminals still remains unclear. In the present study, we hypothesized that colchicine-induced dentate granule cell death may be caused by accumulation of labile intracellular zinc. 10 μg kg(-1) of colchicine was injected into the adult rat hippocampus and then brain sections were evaluated at 1 day or 1 week later. Neuronal cell death was evaluated by H&E staining or Fluoro-Jade B. Zinc accumulation and vesicular zinc were detected by N-(6-methoxy-8-quinolyl)-para-toluene sulfonamide (TSQ) staining. To test whether an extracellular zinc chelator can prevent this process, CaEDTA was injected into the hippocampus over a 5 min period with colchicine. To test whether other microtubule toxins also produce similar effects as colchicine, vincristine was injected into the hippocampus. The present study found that colchicine injection induced intracellular zinc accumulation in the dentate granule cells and depleted vesicular zinc from mossy fiber terminals. Injection of a zinc chelator, CaEDTA, did not block the zinc accumulation and neuronal death. Vincristine also produced intracellular zinc accumulation and neuronal death. These results suggest that colchicine-induced dentate granule cell death is caused by blocking axonal zinc flow and accumulation of intracellular labile zinc.
Collapse
Affiliation(s)
- Bo Young Choi
- Department of Physiology, College of Medicine, Hallym University, 1-Okcheon Dong, Chuncheon, Korea 200-702.
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Titus DJ, Furones C, Atkins CM, Dietrich WD. Emergence of cognitive deficits after mild traumatic brain injury due to hyperthermia. Exp Neurol 2014; 263:254-62. [PMID: 25447938 DOI: 10.1016/j.expneurol.2014.10.020] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 10/16/2014] [Accepted: 10/22/2014] [Indexed: 02/04/2023]
Abstract
Mild elevations in core temperature can occur in individuals involved in strenuous activities that are risky for potentially sustaining a mild traumatic brain injury (mTBI) or concussion. Recently, we have discovered that mild elevations in brain temperature can significantly aggravate the histopathological consequences of mTBI. However, whether this exacerbation of brain pathology translates into behavioral deficits is unknown. Therefore, we investigated the behavioral consequences of elevating brain temperature to mildly hyperthermic levels prior to mTBI. Adult male Sprague Dawley rats underwent mild fluid-percussion brain injury or sham surgery while normothermic (37 °C) or hyperthermic (39 °C) and were allowed to recover for 7 days. Animals were then assessed for cognition using the water maze and cue and contextual fear conditioning. We found that mTBI alone at normothermia had no effect on long-term cognitive measures whereas mTBI animals that were hyperthermic for 15 min prior to and for 4h after brain injury were significantly impaired on long-term retention for both the water maze and fear conditioning. In contrast, hyperthermic mTBI animals cooled within 15 min to normothermia demonstrated no significant long-term cognitive deficits. Mild TBI irrespective of temperature manipulations resulted in significant short-term working memory deficits. Cortical atrophy and contusions were detected in all mTBI treatment groups and contusion volume was significantly less in hyperthermic mTBI animals that were cooled as compared to hyperthermic mTBI animals that remained hyperthermic. These results indicate that brain temperature is an important variable for mTBI outcome and that mildly elevated temperatures at the time of injury result in persistent cognitive deficits. Importantly, cooling to normothermia after mTBI prevents the development of long-term cognitive deficits caused by hyperthermia. Reducing temperature to normothermic levels soon after mTBI represents a rational approach to potentially mitigate the long-term consequences of mTBI.
Collapse
Affiliation(s)
- David J Titus
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Concepcion Furones
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Coleen M Atkins
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - W Dalton Dietrich
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
18
|
Choi BY, Kim JH, Kim HJ, Lee BE, Kim IY, Sohn M, Suh SW. Zinc chelation reduces traumatic brain injury-induced neurogenesis in the subgranular zone of the hippocampal dentate gyrus. J Trace Elem Med Biol 2014; 28:474-81. [PMID: 25200616 DOI: 10.1016/j.jtemb.2014.07.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Numerous studies have demonstrated that traumatic brain injury (TBI) increases hippocampal neurogenesis in the rodent brain. However, the mechanisms underlying increased neurogenesis after TBI remain unknown. Continuous neurogenesis occurs in the subgranular zone (SGZ) of the hippocampal dentate gyrus (DG) in the adult brain. The mechanism that maintains active neurogenesis in the hippocampal area is not known. A high level of vesicular zinc is localized in the presynaptic terminals of the SGZ (mossy fiber). The mossy fiber of dentate granular cells contains high levels of chelatable zinc in their terminal vesicles, which can be released into the extracellular space during neuronal activity. Previously, our lab presented findings indicating that a possible correlation may exist between synaptic zinc localization and high rates of neurogenesis in this area after hypoglycemia or epilepsy. Using a weight drop animal model to mimic human TBI, we tested our hypothesis that zinc plays a key role in modulating hippocampal neurogenesis after TBI. Thus, we injected a zinc chelator, clioquinol (CQ, 30mg/kg), into the intraperitoneal space to reduce brain zinc availability twice per day for 1 week. Neuronal death was evaluated with Fluoro Jade-B and NeuN staining to determine whether CQ has neuroprotective effects after TBI. The number of degenerating neurons (FJB (+)) and live neurons (NeuN (+)) was similar in vehicle and in CQ-treated rats at 1 week after TBI. Neurogenesis was evaluated using BrdU, Ki67 and doublecortin (DCX) immunostaining 1 week after TBI. The number of BrdU, Ki67 and DCX positive cell was increased after TBI. However, the number of BrdU, Ki67 and DCX positive cells was significantly decreased by CQ treatment. The present study shows that zinc chelation did not prevent neurodegeneration but did reduce TBI-induced progenitor cell proliferation and neurogenesis. Therefore, this study suggests that zinc has an essential role for modulating hippocampal neurogenesis after TBI.
Collapse
Affiliation(s)
- Bo Young Choi
- Department of Physiology, Hallym University, College of Medicine, Chuncheon, Republic of Korea
| | - Jin Hee Kim
- Department of Physiology, Hallym University, College of Medicine, Chuncheon, Republic of Korea
| | - Hyun Jung Kim
- Department of Physiology, Hallym University, College of Medicine, Chuncheon, Republic of Korea
| | - Bo Eun Lee
- Department of Physiology, Hallym University, College of Medicine, Chuncheon, Republic of Korea
| | - In Yeol Kim
- Department of Physiology, Hallym University, College of Medicine, Chuncheon, Republic of Korea
| | - Min Sohn
- Inha University, Department of Nursing, Incheon, Republic of Korea
| | - Sang Won Suh
- Department of Physiology, Hallym University, College of Medicine, Chuncheon, Republic of Korea.
| |
Collapse
|
19
|
Wang HC, Sun CF, Chen H, Chen MS, Shen G, Ma YB, Wang BD. Where are we in the modelling of traumatic brain injury? Models complicated by secondary brain insults. Brain Inj 2014; 28:1491-503. [PMID: 25111457 DOI: 10.3109/02699052.2014.943288] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Hong-Cai Wang
- Department of Neurosurgery, Li Hui Li Hospital of Medical Centre of Ningbo
NingboPR China
| | - Cheng-Feng Sun
- Department of Neurosurgery, Li Hui Li Hospital of Medical Centre of Ningbo
NingboPR China
| | - Hai Chen
- Department of Neurosurgery, Li Hui Li Hospital of Medical Centre of Ningbo
NingboPR China
| | - Mao-Song Chen
- Department of Neurosurgery, Li Hui Li Hospital of Medical Centre of Ningbo
NingboPR China
| | - Gang Shen
- Department of Neurosurgery, Li Hui Li Hospital of Medical Centre of Ningbo
NingboPR China
| | - Yan-Bin Ma
- Department of Neurosurgery, NO.3 People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine
ShanghaiPR China
| | - Bo-Ding Wang
- Department of Neurosurgery, Li Hui Li Hospital of Medical Centre of Ningbo
NingboPR China
| |
Collapse
|
20
|
Subacute zinc administration and L-NAME caused an increase of NO, zinc, lipoperoxidation, and caspase-3 during a cerebral hypoxia-ischemia process in the rat. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:240560. [PMID: 23997853 PMCID: PMC3749594 DOI: 10.1155/2013/240560] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 07/04/2013] [Indexed: 11/17/2022]
Abstract
Zinc or L-NAME administration has been shown to be protector agents, decreasing oxidative stress and cell death. However, the treatment with zinc and L-NAME by intraperitoneal injection has not been studied. The aim of our work was to study the effect of zinc and L-NAME administration on nitrosative stress and cell death. Male Wistar rats were treated with ZnCl2 (2.5 mg/kg each 24 h, for 4 days) and N-ω-nitro-L-arginine-methyl ester (L-NAME, 10 mg/kg) on the day 5 (1 hour before a common carotid-artery occlusion (CCAO)). The temporoparietal cortex and hippocampus were dissected, and zinc, nitrites, and lipoperoxidation were assayed at different times. Cell death was assayed by histopathology using hematoxylin-eosin staining and caspase-3 active by immunostaining. The subacute administration of zinc before CCAO decreases the levels of zinc, nitrites, lipoperoxidation, and cell death in the late phase of the ischemia. L-NAME administration in the rats treated with zinc showed an increase of zinc levels in the early phase and increase of zinc, nitrites, and lipoperoxidation levels, cell death by necrosis, and the apoptosis in the late phase. These results suggest that the use of these two therapeutic strategies increased the injury caused by the CCAO, unlike the alone administration of zinc.
Collapse
|
21
|
Kim JH, Jang BG, Choi BY, Kwon LM, Sohn M, Song HK, Suh SW. Zinc chelation reduces hippocampal neurogenesis after pilocarpine-induced seizure. PLoS One 2012; 7:e48543. [PMID: 23119054 PMCID: PMC3485345 DOI: 10.1371/journal.pone.0048543] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 09/26/2012] [Indexed: 12/03/2022] Open
Abstract
Several studies have shown that epileptic seizures increase hippocampal neurogenesis in the adult. However, the mechanism underlying increased neurogenesis after seizures remains largely unknown. Neurogenesis occurs in the subgranular zone (SGZ) of the hippocampus in the adult brain, although an understanding of why it actively occurs in this region has remained elusive. A high level of vesicular zinc is localized in the presynaptic terminals of the SGZ. Previously, we demonstrated that a possible correlation may exist between synaptic zinc localization and high rates of neurogenesis in this area after hypoglycemia. Using a lithium-pilocarpine model, we tested our hypothesis that zinc plays a key role in modulating hippocampal neurogenesis after seizure. Then, we injected the zinc chelator, clioquinol (CQ, 30 mg/kg), into the intraperitoneal space to reduce brain zinc availability. Neuronal death was detected with Fluoro Jade-B and NeuN staining to determine whether CQ has neuroprotective effects after seizure. The total number of degenerating and live neurons was similar in vehicle and in CQ treated rats at 1 week after seizure. Neurogenesis was evaluated using BrdU, Ki67 and doublecortin (DCX) immunostaining 1 week after seizure. The number of BrdU, Ki67 and DCX positive cell was increased after seizure. However, the number of BrdU, Ki67 and DCX positive cells was significantly decreased by CQ treatment. Intracellular zinc chelator, N,N,N0,N-Tetrakis (2-pyridylmethyl) ethylenediamine (TPEN), also reduced seizure-induced neurogenesis in the hippocampus. The present study shows that zinc chelation does not prevent neurodegeneration but does reduce seizure-induced progenitor cell proliferation and neurogenesis. Therefore, this study suggests that zinc has an essential role for modulating hippocampal neurogenesis after seizure.
Collapse
Affiliation(s)
- Jin Hee Kim
- Department of Physiology, Hallym University, College of Medicine, Chuncheon, Korea
| | - Bong Geom Jang
- Department of Physiology, Hallym University, College of Medicine, Chuncheon, Korea
| | - Bo Young Choi
- Department of Physiology, Hallym University, College of Medicine, Chuncheon, Korea
| | - Lyo Min Kwon
- Department of Physiology, Hallym University, College of Medicine, Chuncheon, Korea
| | - Min Sohn
- Inha University, Department of Nursing, Incheon, Korea
| | - Hong Ki Song
- Department of Neurology, College of Medicine, Hallym University, Chunchon, Korea
- * E-mail: (HKS); (SWS)
| | - Sang Won Suh
- Department of Physiology, Hallym University, College of Medicine, Chuncheon, Korea
- * E-mail: (HKS); (SWS)
| |
Collapse
|
22
|
Choi BY, Jang BG, Kim JH, Lee BE, Sohn M, Song HK, Suh SW. Prevention of traumatic brain injury-induced neuronal death by inhibition of NADPH oxidase activation. Brain Res 2012; 1481:49-58. [PMID: 22975130 DOI: 10.1016/j.brainres.2012.08.032] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Revised: 08/13/2012] [Accepted: 08/16/2012] [Indexed: 11/30/2022]
Abstract
The present study aimed to evaluate the therapeutic potential of apocynin, an NADPH oxidase assembly inhibitor, on traumatic brain injury. Rat traumatic brain injury (TBI) was performed using a weight drop model. Apocynin (100mg/kg) was injected into the intraperitoneal space 15 min before TBI. Reactive oxygen species (ROS) in the hippocampal CA3 pyramidal neurons were detected by dihydroethidium (dHEt) at 3h after TBI. Oxidative injury was detected by 4-hydroxy-2-nonenal (4HNE) at 6h after TBI. Blood-brain barrier disruption was detected by IgG extravasation and neuronal death was evaluated with Fluoro Jade-B staining 24h after TBI. Microglia activation was detected by CD11b immunohistochemistry in the hippocampus at 1 week after TBI. ROS production was inhibited by apocynin administration in the hippocampal CA3 pyramidal neurons. This pre-treatment with apocynin decreased the blood-brain barrier disruption, the number of degenerating neurons in the hippocampal CA3 region and microglial activation after TBI. The present study indicates that apocynin pre-treatment prevents TBI-induced ROS production, thus decreasing BBB disruption, neuronal death and microglial activation. Therefore, the present study suggests that inhibition of NADPH oxidase by apocynin may have a high therapeutic potential to reduce traumatic brain injury-induced neuronal death.
Collapse
Affiliation(s)
- Bo Yong Choi
- Department of Physiology, Hallym University, College of Medicine, Chuncheon, South Korea
| | | | | | | | | | | | | |
Collapse
|
23
|
Won SJ, Choi BY, Yoo BH, Sohn M, Ying W, Swanson RA, Suh SW. Prevention of traumatic brain injury-induced neuron death by intranasal delivery of nicotinamide adenine dinucleotide. J Neurotrauma 2012; 29:1401-9. [PMID: 22352983 DOI: 10.1089/neu.2011.2228] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
Traumatic brain injury (TBI) is one of the most devastating injuries experienced by military personnel, as well as the general population, and can result in acute and chronic complications such as cognitive impairments. Since there are currently no effective tools for the treatment of TBI, it is of great importance to determine the mechanisms of neuronal death that characterize this insult. Several studies have indicated that TBI-induced neuronal death arises in part due to excessive activation of poly(ADP-ribose) polymerase-1 (PARP-1), which results in nicotinamide adenine dinucleotide (NAD⁺) depletion and subsequent energy failure. In this study, we investigated whether intranasal administration of NAD⁺ could reduce neuronal death after TBI. Rats were subjected to a weight-drop TBI model that induces cortical and hippocampal neuronal death. The intranasal administration of NAD⁺ (20 mg/kg) immediately after TBI protected neurons in CA1, CA3, and dentate gyrus of the hippocampus, but not in the cortex. In addition, delayed microglial activation normally seen after TBI was reduced by NAD⁺ treatment at 7 days after insult. Neuronal superoxide production and PARP-1 accumulation after TBI were not inhibited by NAD⁺ treatment, indicating that reactive oxygen species (ROS) production and PARP-1 activation are events that occur upstream of NAD⁺ depletion. This study suggests that intranasal delivery of NAD⁺ represents a novel, inexpensive, and non-toxic intervention for preventing TBI-induced neuronal death.
Collapse
Affiliation(s)
- Seok Joon Won
- Department of Neurology, University of California-San Francisco, San Francisco, California, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Udayabanu M, Kumaran D, Katyal A. Free chelatable zinc modulates the cholinergic function during hypobaric hypoxia-induced neuronal damage: an in vivo study. Neuroscience 2012; 202:434-45. [DOI: 10.1016/j.neuroscience.2011.11.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Revised: 11/08/2011] [Accepted: 11/08/2011] [Indexed: 10/15/2022]
|
25
|
Meeusen JW, Tomasiewicz H, Nowakowski A, Petering DH. TSQ (6-methoxy-8-p-toluenesulfonamido-quinoline), a common fluorescent sensor for cellular zinc, images zinc proteins. Inorg Chem 2011; 50:7563-73. [PMID: 21774459 DOI: 10.1021/ic200478q] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Zn(2+) is a necessary cofactor for thousands of mammalian proteins. Research has suggested that transient fluxes of cellular Zn(2+) are also involved in processes such as apoptosis. Observations of Zn(2+) trafficking have been collected using Zn(2+) responsive fluorescent dyes. A commonly used Zn(2+) fluorophore is 6-methoxy-8-p-toluenesulfonamido-quinoline (TSQ). The chemical species responsible for TSQ's observed fluorescence in resting or activated cells have not been characterized. Parallel fluorescence microscopy and spectrofluorometry of LLC-PK(1) cells incubated with TSQ demonstrated punctate staining that concentrated around the nucleus and was characterized by an emission maximum near 470 nm. Addition of cell permeable Zn-pyrithione resulted in greatly increased, diffuse fluorescence that shifted the emission peak to 490 nm, indicative of the formation of Zn(TSQ)(2). TPEN (N,N,N'N'-tetrakis(-)[2-pyridylmethyl]-ethylenediamine), a cell permeant Zn(2+) chelator, largely quenched TSQ fluorescence returning the residual fluorescence to the 470 nm emission maximum. Gel filtration chromatography of cell supernatant from LLC-PK(1) cells treated with TSQ revealed that TSQ fluorescence (470 nm emission) eluted with the proteome fractions. Similarly, addition of TSQ to proteome prior to chromatography resulted in 470 nm fluorescence emission that was not observed in smaller molecular weight fractions. It is hypothesized that Zn-TSQ fluorescence, blue-shifted from the 490 nm emission maximum of Zn(TSQ)(2), results from ternary complex, TSQ-Zn-protein formation. As an example, Zn-carbonic anhydrase formed a ternary adduct with TSQ characterized by a fluorescence emission maximum of 470 nm and a dissociation constant of 1.55 × 10(-7) M. Quantification of TSQ-Zn-proteome fluorescence indicated that approximately 8% of cellular Zn(2+) was imaged by TSQ. These results were generalized to other cell types and model Zn-proteins.
Collapse
Affiliation(s)
- Jeffrey W Meeusen
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53201, USA
| | | | | | | |
Collapse
|
26
|
Fukushima M, Lee SM, Moro N, Hovda DA, Sutton RL. Metabolic and histologic effects of sodium pyruvate treatment in the rat after cortical contusion injury. J Neurotrauma 2010; 26:1095-110. [PMID: 19594384 DOI: 10.1089/neu.2008.0771] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
This study determined the effects of intraperitoneal sodium pyruvate (SP) treatment on the levels of circulating fuels and on cerebral microdialysis levels of glucose (MD(glc)), lactate (MD(lac)), and pyruvate (MD(pyr)), and the effects of SP treatment on neuropathology after left cortical contusion injury (CCI) in rats. SP injection (1000 mg/kg) 5 min after sham injury (Sham-SP) or CCI (CCI-SP) significantly increased arterial pyruvate (p < 0.005) and lactate (p < 0.001) compared to that of saline-treated rats with CCI (CCI-Sal). Serum glucose also increased significantly in CCI-SP compared to that in CCI-Sal rats (p < 0.05), but not in Sham-SP rats. MD(pyr) was not altered after CCI-Sal, whereas MD(lac) levels within the cerebral cortex significantly increased bilaterally (p < 0.05) and those for MD(glc) decreased bilaterally (p < 0.05). MD(pyr) levels increased significantly in both Sham-SP and CCI-SP rats (p < 0.05 vs. CCI-Sal) and were higher in left/injured cortex of the CCI-SP group (p < 0.05 vs. sham-SP). In CCI-SP rats the contralateral MD(lac) decreased below CCI-Sal levels (p < 0.05) and the ipsilateral MD(glc) levels exceeded those of CCI-Sal rats (p < 0.05). Rats with a single low (500 mg/kg) or high dose (1000 mg/kg) SP treatment had fewer damaged cortical cells 6 h post-CCI than did saline-treated rats (p < 0.05), but three hourly injections of SP (1000 mg/kg) were needed to significantly reduce contusion volume 2 weeks after CCI. Thus, a single intraperitoneal SP treatment increases circulating levels of three potential brain fuels, attenuates a CCI-induced reduction in extracellular glucose while increasing extracellular levels of pyruvate, but not lactate, and can attenuate cortical cell damage occurring within 6 h of injury. Enduring (2 week) neuronal protection was achieved only with multiple SP treatments within the first 2 h post-CCI, perhaps reflecting the need for additional fuel throughout the acute period of increased metabolic demands induced by CCI.
Collapse
Affiliation(s)
- Masamichi Fukushima
- Brain Injury Research Center, David Geffen School of Medicine at UCLA, Los Angeles, California 90095-7039, USA
| | | | | | | | | |
Collapse
|
27
|
Clioquinol inhibits zinc-triggered caspase activation in the hippocampal CA1 region of a global ischemic gerbil model. PLoS One 2010; 5:e11888. [PMID: 20686690 PMCID: PMC2912365 DOI: 10.1371/journal.pone.0011888] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2010] [Accepted: 07/09/2010] [Indexed: 11/29/2022] Open
Abstract
Background Excessive release of chelatable zinc from excitatory synaptic vesicles is involved in the pathogenesis of selective neuronal cell death following transient forebrain ischemia. The present study was designed to examine the neuroprotective effect of a membrane-permeable zinc chelator, clioquinol (CQ), in the CA1 region of the gerbil hippocampus after transient global ischemia. Methodology/Principal Findings The common carotid arteries were occluded bilaterally, and CQ (10 mg/kg, i.p.) was injected into gerbils once a day. The zinc chelating effect of CQ was examined with TSQ fluorescence and autometallography. Neuronal death, the expression levels of caspases and apoptosis inducing factor (AIF) were evaluated using TUNEL, in situ hybridization and Western blotting, respectively. We were able to show for the first time that CQ treatment attenuates the ischemia-induced zinc accumulation in the CA1 pyramidal neurons, accompanied by less neuronal loss in the CA1 field of the hippocampus after ischemia. Furthermore, the expression levels of caspase-3, -9, and AIF were significantly decreased in the hippocampus of CQ-treated gerbils. Conclusions/Significance The present study indicates that the neuroprotective effect of CQ is related to downregulation of zinc-triggered caspase activation in the hippocampal CA1 region of gerbils with global ischemia.
Collapse
|
28
|
Chemical blocking of zinc ions in CNS increases neuronal damage following traumatic brain injury (TBI) in mice. PLoS One 2010; 5:e10131. [PMID: 20396380 PMCID: PMC2852423 DOI: 10.1371/journal.pone.0010131] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2009] [Accepted: 03/12/2010] [Indexed: 12/02/2022] Open
Abstract
Background Traumatic brain injury (TBI) is one of the leading causes of disability and death among young people. Although much is already known about secondary brain damage the full range of brain tissue responses to TBI remains to be elucidated. A population of neurons located in cerebral areas associated with higher cognitive functions harbours a vesicular zinc pool co-localized with glutamate. This zinc enriched pool of synaptic vesicles has been hypothesized to take part in the injurious signalling cascade that follows pathological conditions such as seizures, ischemia and traumatic brain injury. Pathological release of excess zinc ions from pre-synaptic vesicles has been suggested to mediate cell damage/death to postsynaptic neurons. Methodology/Principal Findings In order to substantiate the influence of vesicular zinc ions on TBI, we designed a study in which damage and zinc movements were analysed in several different ways. Twenty-four hours after TBI ZnT3-KO mice (mice without vesicular zinc) were compared to littermate Wild Type (WT) mice (mice with vesicular zinc) with regard to histopathology. Furthermore, in order to evaluate a possible neuro-protective dimension of chemical blocking of vesicular zinc, we treated lesioned mice with either DEDTC or selenite. Our study revealed that chemical blocking of vesicular zinc ions, either by chelation with DEDTC or accumulation in zinc-selenium nanocrystals, worsened the effects on the aftermath of TBI in the WT mice by increasing the number of necrotic and apoptotic cells within the first 24 hours after TBI, when compared to those of chemically untreated WT mice. Conclusion/Significance ZnT3-KO mice revealed more damage after TBI compared to WT controls. Following treatment with DEDTC or selenium an increase in the number of both dead and apoptotic cells were seen in the controls within the first 24 hours after TBI while the degree of damage in the ZnT3-KO mice remained largely unchanged. Further analyses revealed that the damage development in the two mouse strains was almost identical after either zinc chelation or zinc complexion therapy.
Collapse
|
29
|
Chan KWY, Chow AM, Chan KC, Yang J, Wu EX. Magnetic resonance spectroscopy of the brain under mild hypothermia indicates changes in neuroprotection-related metabolites. Neurosci Lett 2010; 475:150-5. [PMID: 20362032 DOI: 10.1016/j.neulet.2010.03.066] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2010] [Revised: 03/24/2010] [Accepted: 03/25/2010] [Indexed: 11/25/2022]
Abstract
Brain hypothermia has demonstrated pronounced neuroprotective effect in patients with cardiac arrest, ischemia and acute liver failure. However, its underlying neuroprotective mechanisms remain to be elucidated in order to improve therapeutic outcomes. Single voxel proton magnetic resonance spectroscopy ((1)H-MRS) was performed using a 7 Tesla MRI scanner on normal Sprague-Dawley rats (N=8) in the same voxel under normothermia (36.5 degrees C) and 30min mild hypothermia (33.5 degrees C). Levels of various brain proton metabolites were compared. The level of lactate (Lac) and myo-inositol (mI) increased in the cortex during hypothermia. In the thalamus, taurine (Tau), a cryogen in brain, increased and choline (Cho) decreased. These metabolic alterations indicated the onset of a number of neuroprotective processes that include attenuation of energy metabolism, excitotoxic pathways, brain osmolytes and thermoregulation, thus protecting neuronal cells from damage. These experimental findings demonstrated that (1)H-MRS can be applied to investigate the changes of specific metabolites and corresponding neuroprotection mechanisms in vivo noninvasively, and ultimately improve our basic understanding of hypothermia and ability to optimize its therapeutic efficacy.
Collapse
Affiliation(s)
- Kannie W Y Chan
- Laboratory of Biomedical Imaging and Signal Processing, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | | | | | | | | |
Collapse
|
30
|
Abstract
Hypothermia reduces neuronal damage after cerebral ischemia and traumatic brain injury, while hyperthermia exacerbates damage from these insults. Previously we have shown that temperature-dependent modulation of excitotoxic neuronal death is mediated in part by temperature-dependent changes in the synaptic release/translocation of Zn(2+). In this study, we hypothesize that brain temperature also affects hypoglycemia-induced neuronal death by modulation of vesicular Zn(2+) release from presynaptic terminals. To test our hypothesis, we used a rat model of insulin-induced hypoglycemia. Here we found that hypoglycemia-induced neuronal injury was significantly affected by brain temperature, that is, hypothermia inhibited while hyperthermia aggravated neuronal death. To investigate the mechanism of temperature-dependent neuronal death after hypoglycemia, we measured zinc release/translocation, reactive oxygen species (ROS) production, and microglia activation. Here we found that hypoglycemia-induced Zn(2+) release/translocation, ROS production, and microglia activation were inhibited by hypothermia but aggravated by hyperthermia. Even when the insult was accompanied by hyperthermic conditions, zinc chelation inhibited ROS production and microglia activation. Zinc chelation during hyperthermia reduced neuronal death, superoxide production, and microglia activation, which was comparable to the protective effects of hypothermia. We conclude that neuronal death after hypoglycemia is temperature-dependent and is mediated by increased Zn(2+) release, superoxide production, and microglia activation.
Collapse
|
31
|
Shibuta S, Varathan S, Kamibayashi T, Mashimo T. Small temperature variations alter edaravone-induced neuroprotection of cortical cultures exposed to prolonged hypoxic episodes. Br J Anaesth 2010; 104:52-8. [PMID: 19923134 DOI: 10.1093/bja/aep320] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Edaravone, a free radical scavenger, has been shown to be neuroprotective in vivo and in vitro. However, the impact of small temperature variations on its neuroprotective actions remains unknown. METHODS We examined the degree of neuroprotection conferred by various concentrations of edaravone on cortical cultures exposed to prolonged hypoxia (24 h) under three conditions: mild hypothermia (32 degrees C), normothermia (37 degrees C), and mild hyperthermia (39 degrees C). The survival of cortical neurones from E16 Wistar rats (SR) was evaluated using photomicrographs taken before and after exposure to hypoxia. RESULTS The mean survival of neurones exposed to hypoxia at normothermia was 14.7 (sem 1.8)%. The addition of 50 microM edaravone significantly improved the mean survival to 40.5 (4.7)%. This improvement was noted at higher doses of edaravone (5 microM < or =) but not at lower doses (< or =500 nM). With mild hypothermia and prolonged hypoxia without edaravone, neuroprotection was significantly improved with a mean survival of 63.0 (5.2)%. This neuroprotective effect was not enhanced with the addition of edaravone, even at the highest dose. Hypoxia-induced neurotoxicity was aggravated by mild hyperthermia as reflected by a mean survival of 9.1 (2.1)%. However, higher concentrations of edaravone inhibited the deleterious effect of mild hyperthermia, thereby demonstrating a significant neuroprotective effect. The survival of neurones subjected to both hyperthermia and edaravone was the same as that of neurones exposed to normothermia and edaravone. CONCLUSIONS Temperature is a potential factor in determining whether edaravone confers a neuroprotective effect when applied during prolonged hypoxic insults.
Collapse
Affiliation(s)
- S Shibuta
- Department of Anaesthesiology and Intensive Care Medicine, Graduate School of Medicine D7, Osaka University, 2-2, Yamadaoka, Suita 565-0871, Japan.
| | | | | | | |
Collapse
|
32
|
Suh SW, Yoo BH, Won SJ. Role of zinc in hypoglycemia-induced neuron death. FUTURE NEUROLOGY 2009. [DOI: 10.2217/fnl.09.51] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The mechanism of hypoglycemia-induced neuronal death has been extensively studied since a Swedish scientist, Roland Auer, developed a rat model of insulin-induced hypoglycemia one quarter of a century ago. Currently, mounting data derived from these studies are making it possible to propose an ever more complete mechanism of hypoglycemia-induced neuron death. Although salient differences between the two insults do exist, the pattern and progression of neuron death after acute/severe hypoglycemia have been observed to share many similarities with ischemia-induced neuron death. One important consideration is that the divalent cation zinc is a common neurotoxic factor in both conditions. For the last decade, our laboratory has offered evidence for the hypothesis that synaptic vesicular zinc release and subsequent translocation into the postsynaptic neuron are key upstream events in both neurological injuries of ischemia and hypoglycemia. The release of vesicular zinc from presynaptic terminals depends on nitric oxide production, which is initiated by glucose reperfusion after hypoglycemia. Postsynaptic intracellular influx of zinc after hypoglycemia increases reactive oxygen species production and PARP-1 activation, which eventually leads to neuronal death. Thus, hypoglycemia-induced neuronal death is not simply caused by a lack of neuronal glucose availability but rather by sequential zinc-mediated events that impinge upon the process of neuronal death. This paper focuses on the role of zinc in hypoglycemia-induced neuron death; first, the temporal events of zinc release and translocation after severe hypoglycemia; second, the role of zinc on reactive oxygen species production; and third, the role of zinc on poly(ADP-ribose) polymerase 1 activation. Here, we also speculate on possible intervention strategies to prevent hypoglycemia-induced neuronal death.
Collapse
Affiliation(s)
- Sang Won Suh
- Department of Neurology, University of California San Francisco & Veterans Affairs Medical Center, 4150 Clement Street, San Francisco, CA 94121, USA and Department of Physiology, College of Medicine, Hallym University, Chun Cheon, Korea 200–702
| | - Byung Hoon Yoo
- Department of Neurology, University of California San Francisco & Veterans Affairs Medical Center, 4150 Clement Street, San Francisco, CA 94121, USA and Departments of Anesthesiology, Sanggye Paik Hospital, Inje University, School of Medicine, Seoul, Korea
| | - Seok Joon Won
- Department of Neurology, University of California San Francisco & Veterans Affairs Medical Center, 4150 Clement Street, San Francisco, CA 94121, USA
| |
Collapse
|
33
|
Abstract
In the adult brain, neurogenesis occurs in the subgranular zone of the dentate gyrus (DG), where high levels of vesicular zinc are localized in the presynaptic terminals. To determine whether zinc has a role in modulating hippocampal neurogenesis under normal or pathologic conditions, we manipulated the level of vesicular zinc experimentally. To reduce hippocampal vesicular zinc, rats were either fed a zinc-deficient diet or treated with a zinc chelator, clioquinol (CQ). The number of progenitor cells and immature neurons was decreased significantly in the DG after 6 weeks of dietary zinc deprivation. Conversely, the number of progenitor cells and immature neurons was restored after a 2-week reversal to a normal zinc-containing diet. Similarly, a 1-week treatment with the zinc chelator, CQ, reduced the number of progenitor cells. The results of our previous study showed that hypoglycemia increased hippocampal neurogenesis. This study shows that zinc chelation reduced hypoglycemia-induced progenitor cell proliferation and neurogenesis. Finally, the role of vesicular zinc on neurogenesis was further assessed in zinc transporter 3 (ZnT3) gene deleted mice. Zinc transporter 3 knockout (KO) mice had significantly fewer proliferating progenitor cells and immature neurons after hypoglycemia. Our data provide converging evidence in support of the essential role zinc has in modulating hippocampal neurogenesis.
Collapse
|
34
|
Zhu L, Wang HD, Yu XG, Jin W, Qiao L, Lu TJ, Hu ZL, Zhou J. Erythropoietin prevents zinc accumulation and neuronal death after traumatic brain injury in rat hippocampus: in vitro and in vivo studies. Brain Res 2009; 1289:96-105. [PMID: 19615349 DOI: 10.1016/j.brainres.2009.07.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2009] [Revised: 07/02/2009] [Accepted: 07/04/2009] [Indexed: 02/07/2023]
Abstract
Erythropoietin (Epo) has been gaining great interest for its potential neuroprotective effect in various neurological insults. However, the molecular mechanism underlying how Epo exerts the function is not clear. Recent studies have indicated that Zn(2+) may have a key role in selective cell death in excitotoxicity after injury. In the present study, we studied the effect of recombinant human Epo (rhEpo) in zinc-induced neurotoxicity both in vitro and in vivo. Exposure of cultured hippocampal neurons to 200 muM ZnC1(2) for 20 min resulted in remarkable neuronal injury, revealed by assessing neuronal morphology. By measuring mitochondrial function using MTT assay, we found that application of rhEpo (0.1 U/ml) 24 h before zinc exposure resulted in a significant increase of neuronal survival (0.6007+/-0.2280 Epo group vs 0.2333+/-0.1249 in control group; n=4, p<0.01). Furthermore, we demonstrated that administration of rhEpo (5,000 IU/kg, intraperitoneal) 30 min after traumatic brain injury (TBI) in rats dramatically protected neuronal death indicated by ZP4 staining, a new zinc-specific fluorescent sensor which has been widely used to indicate neuronal damage after excitotoxic injury (n=5/group, p<0.05). Neuronal damage was also assessed by Fluoro-Jade B (FJB) staining, a highly specific fluorescent marker for the degenerating neurons. Consistent with ZP4 staining, we found the beneficial effects of rhEpo on neuronal survival in hippocampus after TBI (n=5/group, p<0.05). Our results suggest that rhEpo can significantly reduce the pathological Zn(2+) accumulation in rat hippocampus after TBI as well as zinc-induced cell death in cultured cells, which may potentially contribute to its neuronal protection after excitotoxic brain damage.
Collapse
Affiliation(s)
- Lin Zhu
- Department of Neurosurgery, Jinling Hospital, Nanjing, Jiangsu Province, China
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Kim J, Kim TY, Hwang JJ, Lee JY, Shin JH, Gwag BJ, Koh JY. Accumulation of labile zinc in neurons and astrocytes in the spinal cords of G93A SOD-1 transgenic mice. Neurobiol Dis 2009; 34:221-9. [DOI: 10.1016/j.nbd.2009.01.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2008] [Revised: 01/07/2009] [Accepted: 01/09/2009] [Indexed: 10/21/2022] Open
|
36
|
Pedersen MØ, Larsen A, Stoltenberg M, Penkowa M. Cell death in the injured brain: roles of metallothioneins. ACTA ACUST UNITED AC 2008; 44:1-27. [PMID: 19348909 DOI: 10.1016/j.proghi.2008.10.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2008] [Accepted: 10/02/2008] [Indexed: 10/21/2022]
Abstract
In traumatic brain injury (TBI), the primary, irreversible damage associated with the moment of impact consists of cells dying from necrosis. This contributes to fuelling a chronic central nervous system (CNS) inflammation with increased formation of proinflammatory cytokines, enzymes and reactive oxygen species (ROS). ROS promote oxidative stress, which leads to neurodegeneration and ultimately results in programmed cell death (secondary injury). Since this delayed, secondary tissue loss occurs days to months following the primary injury it provides a therapeutic window where potential neuroprotective treatment could alleviate ongoing neurodegeneration, cell death and neurological impairment following TBI. Various neuroprotective drug candidates have been described, tested and proven effective in pre-clinical studies, including glutamate receptor antagonists, calcium-channel blockers, and caspase inhibitors. However, most of the scientific efforts have failed in translating the experimental results into clinical trials. Despite intensive research, effective neuroprotective therapies are lacking in the clinic, and TBI continues to be a major cause of morbidity and mortality. This paper provides an overview of the TBI pathophysiology leading to cell death and neurological impairment. We also discuss endogenously expressed neuroprotectants and drug candidates, which at this stage may still hold the potential for treating brain injured patients.
Collapse
Affiliation(s)
- Mie Ø Pedersen
- Section of Neuroprotection, Department of Neuroscience and Pharmacology, Faculty of Health Sciences, The Panum Institute, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen, Denmark.
| | | | | | | |
Collapse
|
37
|
Suh SW. Detection of zinc translocation into apical dendrite of CA1 pyramidal neuron after electrical stimulation. J Neurosci Methods 2008; 177:1-13. [PMID: 18929598 DOI: 10.1016/j.jneumeth.2008.09.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2008] [Revised: 09/08/2008] [Accepted: 09/09/2008] [Indexed: 11/19/2022]
Abstract
Translocation of the endogenous cation zinc from presynaptic terminals to postsynaptic neurons after brain insult has been implicated as a potential neurotoxic event. Several studies have previously demonstrated that a brief electrical stimulation is sufficient to induce the translocation of zinc from presynaptic vesicles into the cytoplasm (soma) of postsynaptic neurons. In the present work I have extended those findings in three ways: (i) providing evidence that zinc translocation occurs into apical dendrites, (ii) presenting data that there is an apparent translocation into apical dendrites when only a zinc-containing synaptic input is stimulated, and (iii) presenting data that there is no zinc translocation into apical dendrite of ZnT3 KO mice following electrical stimulation. Hippocampal slices were preloaded with the "trappable" zinc fluorescent probe, Newport Green. After washout, a single apical dendrite in the stratum radiatum of hippocampal CA1 area was selected and focused on. Burst stimulation (100Hz, 500microA, 0.2ms, monopolar) was delivered to either the adjacent Schaffer-collateral inputs (zinc-containing) or to the adjacent temporo-ammonic inputs (zinc-free) to the CA1 dendrites. Stimulation of the Schaffer collaterals increased the dendritic fluorescence, which was blocked by TTX, low-Ca medium, or the extracellular zinc chelator, CaEDTA. Stimulation of the temporo-ammonic pathway caused no significant rise in the fluorescence. Genetic depletion of vesicular zinc by ZnT3 KO showed no stimulation-induced apical dendrite zinc rise. The present study provides evidence that synaptically released zinc translocates into postsynaptic neurons through the apical dendrites of CA1 pyramidal neurons during physiological synaptic activity.
Collapse
Affiliation(s)
- Sang Won Suh
- Department of Neurology, University of California, San Francisco and Veterans Affairs Medical Center, San Francisco, CA 94121, USA.
| |
Collapse
|
38
|
Abstract
Traumatic brain injury is a leading cause of morbidity and death in both industrialized and developing countries. To date, there is no targeted pharmacological treatment that effectively limits the progression of secondary injury. The delayed progression of deterioration of grey and white matter gives hope that a meaningful intervention can be applied in a realistic timeframe following initial trauma. In this review we discuss new insights into the subcellular mechanisms of secondary injury that have highlighted numerous potential targets for intervention.
Collapse
Affiliation(s)
- Eugene Park
- Cara Phelan Centre for Trauma Research, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ont
| | | | | |
Collapse
|
39
|
Taylor AN, Rahman SU, Sanders NC, Tio DL, Prolo P, Sutton RL. Injury Severity Differentially Affects Short- and Long-Term Neuroendocrine Outcomes of Traumatic Brain Injury. J Neurotrauma 2008; 25:311-23. [DOI: 10.1089/neu.2007.0486] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Anna N. Taylor
- Department of Neurobiology, Brain Research Institute and Brain Injury Research Center, David Geffen School of Medicine at UCLA, Los Angeles, California; and West Los Angeles Healthcare Center, Veterans Administration, Greater Los Angeles Healthcare System, Los Angeles, California
| | - Shayan U. Rahman
- Division of Neurosurgery, Department of Surgery, and Brain Injury Research Center, David Geffen School of Medicine at UCLA, Los Angeles, California
| | | | - Delia L. Tio
- Department of Neurobiology, Brain Research Institute and Brain Injury Research Center, David Geffen School of Medicine at UCLA, Los Angeles, California; and West Los Angeles Healthcare Center, Veterans Administration, Greater Los Angeles Healthcare System, Los Angeles, California
| | - Paolo Prolo
- Division of Oral Biology, UCLA School of Dentistry, Los Angeles, California
| | - Richard L. Sutton
- Division of Neurosurgery, Department of Surgery, and Brain Injury Research Center, David Geffen School of Medicine at UCLA, Los Angeles, California
| |
Collapse
|
40
|
Doering P, Danscher G, Larsen A, Bruhn M, Søndergaard C, Stoltenberg M. Changes in the vesicular zinc pattern following traumatic brain injury. Neuroscience 2007; 150:93-103. [DOI: 10.1016/j.neuroscience.2007.09.066] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2007] [Revised: 08/13/2007] [Accepted: 09/11/2007] [Indexed: 02/05/2023]
|
41
|
Suh SW, Aoyama K, Alano CC, Anderson CM, Hamby AM, Swanson RA. Zinc inhibits astrocyte glutamate uptake by activation of poly(ADP-ribose) polymerase-1. Mol Med 2007; 13:344-9. [PMID: 17728843 PMCID: PMC1952665 DOI: 10.2119/2007-00043.suh] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2007] [Accepted: 06/05/2007] [Indexed: 12/21/2022] Open
Abstract
Several processes by which astrocytes protect neurons during ischemia are now well established. However, less is known about how neurons themselves may influence these processes. Neurons release zinc (Zn2+) from presynaptic terminals during ischemia, seizure, head trauma, and hypoglycemia, and modulate postsynaptic neuronal function. Peak extracellular zinc may reach concentrations as high as 400 microM. Excessive levels of free, ionic zinc can initiate DNA damage and the subsequent activation of poly(ADP-ribose) polymerase 1 (PARP-1), which in turn lead to NAD+ and ATP depletion when DNA damage is extensive. In this study, cultured cortical astrocytes were used to explore the effects of zinc on astrocyte glutamate uptake, an energy-dependent process that is critical for neuron survival. Astrocytes incubated with 100 or 400 microM of zinc for 30 min showed significant decreases in ATP levels and glutamate uptake capacity. These changes were prevented by the PARP inhibitors benzamide or DPQ (3,4-dihydro-5-[4-(1-piperidinyl)butoxyl]-1(2H)-isoquinolinone) or PARP-1 gene deletion (PARP-1 KO). These findings suggest that release of Zn2+ from neurons during brain insults could induce PARP-1 activation in astrocytes, leading to impaired glutamate uptake and exacerbation of neuronal injury.
Collapse
Affiliation(s)
- Sang Won Suh
- Department of Neurology, University of California, San Francisco, California, USA.
| | | | | | | | | | | |
Collapse
|
42
|
Atkins CM, Oliva AA, Alonso OF, Chen S, Bramlett HM, Hu BR, Dietrich WD. Hypothermia treatment potentiates ERK1/2 activation after traumatic brain injury. Eur J Neurosci 2007; 26:810-9. [PMID: 17666079 DOI: 10.1111/j.1460-9568.2007.05720.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Traumatic brain injury (TBI) results in significant hippocampal pathology and hippocampal-dependent memory loss, both of which are alleviated by hypothermia treatment. To elucidate the molecular mechanisms regulated by hypothermia after TBI, rats underwent moderate parasagittal fluid-percussion brain injury. Brain temperature was maintained at normothermic or hypothermic temperatures for 30 min prior and up to 4 h after TBI. The ipsilateral hippocampus was assayed with Western blotting. We found that hypothermia potentiated extracellular signal-regulated kinase 1/2 (ERK1/2) activation and its downstream effectors, p90 ribosomal S6 kinase (p90RSK) and the transcription factor cAMP response element-binding protein. Phosphorylation of another p90RSK substrate, Bad, also increased with hypothermia after TBI. ERK1/2 regulates mRNA translation through phosphorylation of mitogen-activated protein kinase-interacting kinase 1 (Mnk1) and the translation factor eukaryotic initiation factor 4E (eIF4E). Hypothermia also potentiated the phosphorylation of both Mnk1 and eIF4E. Augmentation of ERK1/2 activation and its downstream signalling components may be one molecular mechanism that hypothermia treatment elicits to improve functional outcome after TBI.
Collapse
Affiliation(s)
- Coleen M Atkins
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Hellmich HL, Eidson KA, Capra BA, Garcia JM, Boone DR, Hawkins BE, Uchida T, Dewitt DS, Prough DS. Injured Fluoro-Jade-positive hippocampal neurons contain high levels of zinc after traumatic brain injury. Brain Res 2006; 1127:119-26. [PMID: 17109824 PMCID: PMC2896019 DOI: 10.1016/j.brainres.2006.09.094] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2006] [Revised: 09/12/2006] [Accepted: 09/13/2006] [Indexed: 10/23/2022]
Abstract
Hippocampal damage contributes to cognitive dysfunction after traumatic brain injury (TBI). We previously showed that Fluoro-Jade, a fluorescent stain that labels injured, degenerating brain neurons, quantifies the extent of hippocampal injury after experimental fluid percussion TBI in rats. Coincidentally, we observed that injured neurons in the rat hippocampus also stained with Newport Green, a fluorescent dye specific for free ionic zinc. Here, we show that, regardless of injury severity or therapeutic intervention, the post-TBI population of injured neurons in rat hippocampal subfields CA1, CA3 and dentate gyrus is indistinguishable, both in numbers and anatomical distribution, from the population of neurons containing high levels of zinc. Treatment with lamotrigine, which inhibits presynaptic release of glutamate and presumably zinc that is co-localized with glutamate, reduced numbers of Fluoro-Jade-positive and Newport Green-positive neurons equally as did treatment with nicardipine, which blocks voltage-gated calcium channels through which zinc enters neurons. To confirm using molecular techniques that Fluoro-Jade and Newport Green-positive neurons are equivalent populations, we isolated total RNA from 25 Fluoro-Jade-positive and 25 Newport Green-positive pyramidal neurons obtained by laser capture microdissection (LCM) from the CA3 subfield, linearly amplified the mRNA and used quantitative ribonuclease protection analysis to demonstrate similar expression of mRNA for selected TBI-induced genes. Our data suggest that therapeutic interventions aimed at reducing neurotoxic zinc levels after TBI may reduce hippocampal neuronal injury.
Collapse
Affiliation(s)
- Helen L Hellmich
- Department of Anesthesiology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555-0830, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|