1
|
Zhu L, Li Z, Sheng L, Zhang F, Ji W. Ginkgolide A attenuated apoptosis via inhibition of oxidative stress in mice with traumatic brain injury. Heliyon 2024; 10:e24759. [PMID: 38304806 PMCID: PMC10830544 DOI: 10.1016/j.heliyon.2024.e24759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 12/16/2023] [Accepted: 01/12/2024] [Indexed: 02/03/2024] Open
Abstract
Traumatic brain injury (TBI) is the main cause of death among young adults and the main cause of mortality and disability for all ages groups worldwide. Ginkgolides terpenoid compounds unique to Ginkgo biloba, which have protective effects on cardiovascular and cerebrovascular diseases. The aim of this study is to investigate whether ginkgolide A (GA) can improve TBI in mice and whether it can alleviate cell apoptosis in the brain of TBI mice by reducing oxidative stress. Mice received TBI and GA administration for 7 days. Neurological deficits were monitored and brain tissues were examined for molecular pathological markers. TBI mice had more severer neurobehavioral deficits compared with sham group, which could be improved by administration of GA. GA administration improveed Modified Neurological Severity Scale (mNSS) scores, Grid-Walking test and Rotarod test of TBI mice. The apoptosis increased in TBI mice, and reduced after GA treatment. The biomarkers of oxidative stress 8-OHdG and malondialdehyde (MDA) in the brain of TBI mice increased, while SOD reduced. These changes were reversed after GA administration. These outcomes showed that GA could raise neurobehavioral deficiency of TBI mice. GA treatment could attenuate apoptosis in TBI mice by reducing oxidative stress.
Collapse
Affiliation(s)
- Lei Zhu
- Department of Respiratory Disease, Children's Hospital of Soochow University, Suzhou, China
- Department of Intensive Care Unit, Xuzhou Children's Hospital, Xuzhou, China
| | - Zhengwei Li
- Department of Neurosurgery, Xuzhou Children's Hospital, Xuzhou, China
| | - Liping Sheng
- Department of Intensive Care Unit, Xuzhou Children's Hospital, Xuzhou, China
| | - Fengfei Zhang
- Department of Neurosurgery, Xuzhou Children's Hospital, Xuzhou, China
| | - Wei Ji
- Department of Respiratory Disease, Children's Hospital of Soochow University, Suzhou, China
| |
Collapse
|
2
|
Elder GA, Gama Sosa MA, De Gasperi R, Perez Garcia G, Perez GM, Abutarboush R, Kawoos U, Zhu CW, Janssen WGM, Stone JR, Hof PR, Cook DG, Ahlers ST. The Neurovascular Unit as a Locus of Injury in Low-Level Blast-Induced Neurotrauma. Int J Mol Sci 2024; 25:1150. [PMID: 38256223 PMCID: PMC10816929 DOI: 10.3390/ijms25021150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/11/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Blast-induced neurotrauma has received much attention over the past decade. Vascular injury occurs early following blast exposure. Indeed, in animal models that approximate human mild traumatic brain injury or subclinical blast exposure, vascular pathology can occur in the presence of a normal neuropil, suggesting that the vasculature is particularly vulnerable. Brain endothelial cells and their supporting glial and neuronal elements constitute a neurovascular unit (NVU). Blast injury disrupts gliovascular and neurovascular connections in addition to damaging endothelial cells, basal laminae, smooth muscle cells, and pericytes as well as causing extracellular matrix reorganization. Perivascular pathology becomes associated with phospho-tau accumulation and chronic perivascular inflammation. Disruption of the NVU should impact activity-dependent regulation of cerebral blood flow, blood-brain barrier permeability, and glymphatic flow. Here, we review work in an animal model of low-level blast injury that we have been studying for over a decade. We review work supporting the NVU as a locus of low-level blast injury. We integrate our findings with those from other laboratories studying similar models that collectively suggest that damage to astrocytes and other perivascular cells as well as chronic immune activation play a role in the persistent neurobehavioral changes that follow blast injury.
Collapse
Affiliation(s)
- Gregory A. Elder
- Neurology Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY 10468, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY 10029, USA;
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY 10029, USA; (M.A.G.S.); (R.D.G.)
- Mount Sinai Alzheimer’s Disease Research Center and the Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (C.W.Z.); (P.R.H.)
| | - Miguel A. Gama Sosa
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY 10029, USA; (M.A.G.S.); (R.D.G.)
- General Medical Research Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY 10468, USA
| | - Rita De Gasperi
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY 10029, USA; (M.A.G.S.); (R.D.G.)
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY 10468, USA;
| | - Georgina Perez Garcia
- Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY 10029, USA;
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY 10468, USA;
| | - Gissel M. Perez
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY 10468, USA;
| | - Rania Abutarboush
- Department of Neurotrauma, Operational and Undersea Medicine Directorate, Naval Medical ResearchCommand, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA; (R.A.); (U.K.); (S.T.A.)
- The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Usmah Kawoos
- Department of Neurotrauma, Operational and Undersea Medicine Directorate, Naval Medical ResearchCommand, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA; (R.A.); (U.K.); (S.T.A.)
- The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Carolyn W. Zhu
- Mount Sinai Alzheimer’s Disease Research Center and the Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (C.W.Z.); (P.R.H.)
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY 10468, USA;
- Department of Geriatrics and Palliative Care, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - William G. M. Janssen
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - James R. Stone
- Department of Radiology and Medical Imaging, University of Virginia, 480 Ray C Hunt Drive, Charlottesville, VA 22903, USA;
| | - Patrick R. Hof
- Mount Sinai Alzheimer’s Disease Research Center and the Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (C.W.Z.); (P.R.H.)
- Department of Geriatrics and Palliative Care, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - David G. Cook
- Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, 1660 S Columbian Way, Seattle, WA 98108, USA;
- Department of Medicine, University of Washington, 1959 NE Pacific St., Seattle, WA 98195, USA
| | - Stephen T. Ahlers
- Department of Neurotrauma, Operational and Undersea Medicine Directorate, Naval Medical ResearchCommand, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA; (R.A.); (U.K.); (S.T.A.)
| |
Collapse
|
3
|
Farkhondeh T, Zardast M, Rajabi S, Abdollahi-Karizno M, Roshanravan B, Havangi J, Aschner M, Samarghandian S. Neuroprotective Effects of Curcumin against Chronic Chlorpyrifos- Induced Oxidative Damage in Rat Brain Tissue. Curr Aging Sci 2024; 17:205-209. [PMID: 38347791 DOI: 10.2174/0118746098244014240119112706] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 09/02/2023] [Accepted: 09/08/2023] [Indexed: 09/10/2024]
Abstract
BACKGROUND Chlorpyrifos (CPF) is an organophosphate pesticide that inhibits acetylcholinesterase (AChE) activity. Investigations have also focused on its neurotoxicity, which is independent of AChE inhibition. Here, we evaluated the effect of CPF on oxidative indices in the brain tissue and explored the protective effect of curcumin (Cur) against its toxicity. METHODS Forty male Wistar rats were divided into five groups, each consisting of eight rats (n = 8) per group. Animals were administrated by oral gavage for 90 days with the following treatments: control (C), CPF, CPF + CUR 25 mg/kg, CPF + CUR50, and CPF + cur 100 received olive oil, CPF, CPF plus 25 mg/kg of CUR, CPF plus 50 mg/kg of CUR, and CPF plus 100 mg/kg of CUR, respectively. After anesthetization, animal brain tissues were obtained for assessment of oxidative stress indices. RESULTS The concentration of MDA significantly increased in the brains of the CPF group as compared to the control group (p < 0.01). Also, a significant decrease in MDA concentrations was observed in the brains of rats in the CPF + Cur 100 group compared to the CPF group (p < 0.05). A significant decrease was noted in the GSH concentration in the brains of the CPF group compared to the control group (p < 0.05). Treatment with Cur at 100 mg/kg exhibited a significant increase in GSH concentrations in the brains of the CPF-exposed group compared to the CPF group without Cur administration (p < 0.05). The concentration of NO exhibited a significant increase in the brains of the CPF group when compared to the control group (p < 0.05). Also, a significant decrease in NO concentration was observed in the brain tissue of the CPF + Cur 100 group compared to the CPF group (p < 0.05). CONCLUSION Our data establish that chronic exposure to CPF induced oxidative stress in brain tissue, which was reversed by CUR administration. Additional experimental and clinical investigations are needed to validate the efficacy of CUR as a potential antidote for CPF poisoning.
Collapse
Affiliation(s)
- Tahereh Farkhondeh
- Department of Toxicology and Pharmacology, School of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | - Mahmoud Zardast
- Department of Pathology, School of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Shahnaz Rajabi
- Department of Pathology, School of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | | | - Babak Roshanravan
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
| | - Jalal Havangi
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Saeed Samarghandian
- Healthy Ageing Research Centre, Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
4
|
Obinna UCHEWAO, Shallom EMECHETAS, Ogugua EGWUA, Joy EDEC, Augustine IBEGBUO. Neuromodulatory roles of PIPER GUINEENSE and honey against Lead-Induced neurotoxicity in social interactive behaviors and motor activities in rat models. AIMS Neurosci 2022; 9:460-478. [PMID: 36660078 PMCID: PMC9826751 DOI: 10.3934/neuroscience.2022026] [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: 07/25/2022] [Revised: 10/30/2022] [Accepted: 11/01/2022] [Indexed: 11/17/2022] Open
Abstract
Background Piper guineense and honey contain antioxidative, anti-inflammatory, and antimicrobial properties that can help restore neuronal and other cell damage. To investigate the neuromodulatory roles of p. guineense and honey against lead toxicity on the hippocampus and cerebellum, impairing social behaviors and motor activities. Methodology Thirty Wistar rats were separated into six groups of five rats each, marked with dye. Group A served as control; B was untreated lead; C was a medium dose of the extract (50 mg/kg) and honey (1000 mg/kg); D was a high dose of the extract (80 mg/kg) and honey (1500 mg/kg); E received extract (80 mg/kg), and F received honey (1500 mg/kg). All groups received 110 mg/kg of lead orally, except the control. Social interaction, antidepressant effects, and motor activities were studied using a sociability chamber (SC), Forced Swim Test (FST), and String methods. A blood sample was used to evaluate glutathione peroxidase (GPx) and glutathione oxide transaminase (GOT), while the lipid level was estimated using cerebellar homogenate. Neuronal damage, vacuolation, necrosis, cell degeneration, and alterations in both hippocampus and cerebellum marked untreated group, with decreased GPx and GOT activities followed by impaired motor activities, social behavior, memory, and motivation. Using SCT, group B spent significantly lesser time (47.60 ± 47.60) with stranger 1 compared to A (138.20 ± 34.05), while group C spent considerably more time with stranger 1 (86.80 ± 30.32) than group B at P ≥ 0.05. The treatment increased the enzyme level and restored histoarchitecture (Figures 1-12), improving motor activities, social behavior, memory, motivation, and social affiliation (Tables 3, 4, 2, and 6). The extract and honey may be helpful as neuromodulators in lead toxicity in a dose-dependent manner.
Collapse
Affiliation(s)
- UCHEWA O. Obinna
- * Correspondence: E-mail: ; Tel: +2348172628746 or +2347061644732
| | | | | | | | | |
Collapse
|
5
|
Gao Y, Wang C, Jiang D, An G, Jin F, Zhang J, Han G, Cui C, Jiang P. New insights into the interplay between autophagy and oxidative and endoplasmic reticulum stress in neuronal cell death and survival. Front Cell Dev Biol 2022; 10:994037. [PMID: 36187470 PMCID: PMC9524158 DOI: 10.3389/fcell.2022.994037] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 08/30/2022] [Indexed: 12/03/2022] Open
Abstract
Autophagy is a dynamic process that maintains the normal homeostasis of cells by digesting and degrading aging proteins and damaged organelles. The effect of autophagy on neural tissue is still a matter of debate. Some authors suggest that autophagy has a protective effect on nerve cells, whereas others suggest that autophagy also induces the death of nerve cells and aggravates nerve injury. In mammals, oxidative stress, autophagy and endoplasmic reticulum stress (ERS) constitute important defense mechanisms to help cells adapt to and survive the stress conditions caused by physiological and pathological stimuli. Under many pathophysiological conditions, oxidative stress, autophagy and ERS are integrated and amplified in cells to promote the progress of diseases. Over the past few decades, oxidative stress, autophagy and ERS and their interactions have been a hot topic in biomedical research. In this review, we summarize recent advances in understanding the interactions between oxidative stress, autophagy and ERS in neuronal cell death and survival.
Collapse
Affiliation(s)
- Yahao Gao
- Clinical Medical School, Jining Medical University, Jining, China
| | - Changshui Wang
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, China
| | - Di Jiang
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Gang An
- Clinical Medical School, Jining Medical University, Jining, China
| | - Feng Jin
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, China
| | - Junchen Zhang
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, China
| | - Guangkui Han
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, China
| | - Changmeng Cui
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, China
- *Correspondence: Changmeng Cui, ; Pei Jiang,
| | - Pei Jiang
- Department of Clinical Pharmacy, Jining First People’s Hospital, Jining Medical University, Jining, China
- *Correspondence: Changmeng Cui, ; Pei Jiang,
| |
Collapse
|
6
|
Yan C, Mao J, Yao C, Liu Y, Yan H, Jin W. Neuroprotective effects of mild hypothermia against traumatic brain injury by the involvement of the Nrf2/ARE pathway. Brain Behav 2022; 12:e2686. [PMID: 35803901 PMCID: PMC9392531 DOI: 10.1002/brb3.2686] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/01/2022] [Accepted: 06/14/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) is the leading cause of death and disability worldwide. Mild hypothermia (32-35°C) has been found to show neuroprotective effects against TBI. However, the specific mechanism is still elusive. In the current study, we explored the relationship between oxidative damage after TBI and treatment with mild hypothermia as well as the underlying molecular mechanisms. METHODS We used the closed cortex injury model to perform the brain injury and a temperature monitoring and control system to regulate the body temperature of mice after injury. Adult male C57BL/6 mice were adopted in this study and divided into four experimental groups. Tissue samples were harvested 24 h after injury. RESULTS First, our results showed that treatment with mild hypothermia significantly improved neurobehavioral dysfunction and alleviated brain edema after TBI. Moreover, treatment with mild hypothermia enhanced the activity of the antioxidant enzymes superoxide dismutase and glutathione peroxidase and reduced the accumulation of lipid peroxidation malondialdehyde. Importantly, the expression and activation of the nuclear factor erythroid 2-related factor 2-antioxidant response element (Nrf2-ARE) pathway were upregulated by mild hypothermia after TBI. Finally, treatment with hypothermia significantly decreased the cell apoptosis induced by TBI. CONCLUSION Our results showed that the protective effects of mild hypothermia after TBI may be achieved by the upregulation of the Nrf2-ARE pathway and revealed Nrf2 as a potentially important target to improve the prognosis of TBI.
Collapse
Affiliation(s)
- Chaolong Yan
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China.,Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.,Department of Neurosurgery, Zhongshan Hospital, The Affiliated Hospital of Fudan University, Shanghai, China
| | - Jiannan Mao
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Chenbei Yao
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yang Liu
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Huiying Yan
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Wei Jin
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China.,Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
7
|
Li H, Tofigh AM, Amirfakhraei A, Chen X, Tajik M, Xu D, Motevalli S. Modulation of astrocyte activity and improvement of oxidative stress through blockage of NO/NMDAR pathway improve posttraumatic stress disorder (PTSD)-like behavior induced by social isolation stress. Brain Behav 2022; 12:e2620. [PMID: 35605060 PMCID: PMC9304825 DOI: 10.1002/brb3.2620] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/21/2022] [Accepted: 04/27/2022] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND It has been well documented that social isolation stress (SIS) can induce posttraumatic stress disorder (PTSD)-like behavior in rodents, however, the underlying mechanism is remained misunderstood. In the current study, we aimed to elucidate the role of NO/NMDAR pathway in PTSD-like behavior through modulating of astrocyte activity and improvement of oxidative stress. METHODS Male NMRI mice were used to evaluate the memory function by using Morris water maze (MWM) and fear memory extinction by using freezing response. We used MK-801 (NMDAR-antagonist), L-NNA (NOS-inhibitor), NMDA (NMDAR-agonist), and L-arginine (NO-agent) to find a proper treatment. Also, immunohistochemistry, RT-PCR, and oxidative stress assays were used to evaluate the levels of astrocytes and oxidative stress. We used five mice in each experimental task. RESULTS Our results revealed that SIS could induce learning and memory dysfunction as well as impairment of fear memory extinction in MWM and freezing response tests, respectively. Also, we observed that combined treatment including blockage of NOS (by L-NNA, 0.5 mg/kg) and NMDAR (by MK-801, 0.001 mg/kg) at subeffective doses could result in improvement of both memory and fear memory. In addition, we observed that SIS significantly increases the GFAP expression and astrocyte activity, which results in significant imbalance in oxidative stress. Coadministration of MK-801 and L-NNA at subeffective doses not only decreases the expression of GFAP, but also regulates the oxidative stress imbalance CONCLUSION: Based on these results, it could be hypothesized that blockage of NO/NMDAR pathway might be a novel treatment for PTSD-like behavior in animals by inhibiting the astrocyte and regulating oxidative stress level.
Collapse
Affiliation(s)
- Hua Li
- Department of Neurology, 3201 Hospital, Xi'an Jiaotong University Health Science Center, Hanzhong, China
| | - Arash Mohammadi Tofigh
- Department of General Surgery, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azita Amirfakhraei
- Department of Psychology, Bandar Abbas Branch, Islamic Azad University, Bandar Abbas, Iran
| | - Xuan Chen
- Department of Neurosurgery, The 78th Group Army Hospital of Chinese PLA, Mudanjiang, China
| | - Michael Tajik
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Dongwei Xu
- Department of Neurosurgery, The 78th Group Army Hospital of Chinese PLA, Mudanjiang, China
| | - Saeid Motevalli
- Faculty of Social Sciences & Liberal Arts, Department of Psychology, UCSI University, Kuala Lumpur, Malaysia
| |
Collapse
|
8
|
Vázquez-Lorente H, Herrera-Quintana L, Molina-López J, Zapata-Soria M, Planells E. Need of nutritional assessment and monitoring in a population with acquired brain injury: an analytical cross-sectional study. Nutr Neurosci 2022; 26:525-534. [PMID: 35499860 DOI: 10.1080/1028415x.2022.2065815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Patients with acquired brain injury (ABI) may be at an increased risk of malnutrition due to the pathophysiology of their condition, which can affect their anthropometrical profile and therefore their quality of life. The present study analyzes the anthropometrical profile of these individuals, describes their dietary habits and nutrients intake and identifies the related risk factors influencing health status and quality of life. METHODS Twenty-three volunteers with ABI from the province of Granada (Spain) were recruited for this cross-sectional study. Nutritional assessment was quantitatively and qualitatively performed using a 72-h dietary record and a food frequency questionnaire, respectively. Body composition parameters were evaluated by bioelectrical impedance. RESULTS Low intakes of minerals related to bone health, extra virgin olive oil (EVOO) and fruits were reported. Moreover, women presented a poorer anthropometrical profile compared to men. With reference to age, a significant inverse correlation was observed with sarcopenic obesity, appendicular muscle mass index and Vitamin B6 intake [(r = -0.617, p < 0.01), (r = -0.475, p < 0.05) and (r = -0.498, p < 0.05), respectively]. Intake of EVOO was inversely correlated to body mass index (r = -0.767, p < 0.001). Lastly, a direct correlation was observed between the consumption of alcohol and sweets and snacks (r = 0.608, p < 0.01). CONCLUSION The main findings support the existence of poor dietary quality and anthropometrical profile. Interdisciplinary team assessment would be beneficial to enhance the quality of life and attenuate the development of comorbidities in subjects with ABI.
Collapse
Affiliation(s)
- H. Vázquez-Lorente
- Department of Physiology, School of Pharmacy, Institute of Nutrition and Food Technology “José Mataix”, University of Granada, Granada, Spain
| | - L. Herrera-Quintana
- Department of Physiology, School of Pharmacy, Institute of Nutrition and Food Technology “José Mataix”, University of Granada, Granada, Spain
| | - J. Molina-López
- Faculty of Education, Psychology and Sports Sciences, University of Huelva, Huelva, Spain
| | - M. Zapata-Soria
- Department of Physiology, School of Pharmacy, Institute of Nutrition and Food Technology “José Mataix”, University of Granada, Granada, Spain
- Brain Damage Association (AGREDACE), Granada, Spain
| | - E. Planells
- Department of Physiology, School of Pharmacy, Institute of Nutrition and Food Technology “José Mataix”, University of Granada, Granada, Spain
| |
Collapse
|
9
|
Fang J, Sheng R, Qin ZH. NADPH Oxidases in the Central Nervous System: Regional and Cellular Localization and the Possible Link to Brain Diseases. Antioxid Redox Signal 2021; 35:951-973. [PMID: 34293949 DOI: 10.1089/ars.2021.0040] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Significance: The significant role of reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (Nox) in signal transduction is mediated by the production of reactive oxygen species (ROS), especially in the central nervous system (CNS). The pathogenesis of some neurologic and psychiatric diseases is regulated by ROS, acting as a second messenger or pathogen. Recent Advances: In the CNS, the involvement of Nox-derived ROS has been implicated in the regulation of multiple signals, including cell survival/apoptosis, neuroinflammation, migration, differentiation, proliferation, and synaptic plasticity, as well as the integrity of the blood/brain barrier. In these processes, the intracellular signals mediated by the members of the Nox family vary among different tissues. The present review illuminates the regions and cellular, subcellular localization of Nox isoforms in the brain, the signal transduction, and the role of NOX enzymes in pathophysiology, respectively. Critical Issues: Different signal transduction cascades are coupled to ROS derived from various Nox homologues with varying degrees. Therefore, a critical issue worth noting is the varied role of the homologues of NOX enzymes in different signaling pathways and also they mediate different phenotypes in the diverse pathophysiological condition. This substantiates the effectiveness of selective Nox inhibitors in the CNS. Future Directions: Further investigation to elucidate the role of various homologues of NOX enzymes in acute and chronic brain diseases and signaling mechanisms, and the development of more specific NOX inhibitors for the treatment of CNS disease are urgently needed. Antioxid. Redox Signal. 35, 951-973.
Collapse
Affiliation(s)
- Jie Fang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, China
| | - Rui Sheng
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, China
| | - Zheng-Hong Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, China
| |
Collapse
|
10
|
Zhang Y, Cong P, Tong C, Jin H, Liu Y, Hou M. Melatonin pretreatment alleviates blast-induced oxidative stress in the hypothalamic-pituitary-gonadal axis by activating the Nrf2/HO-1 signaling pathway. Life Sci 2021; 280:119722. [PMID: 34153300 DOI: 10.1016/j.lfs.2021.119722] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 06/02/2021] [Accepted: 06/09/2021] [Indexed: 01/14/2023]
Abstract
Although melatonin has been demonstrated to exert a potent antioxidant effect, the ability of melatonin to alleviate blast-induced oxidative stress in the hypothalamic-pituitary-gonadal (HPG) axis remains unclear. This study aimed to elucidate the effects and underlying mechanism of melatonin pretreatment on the HPG axis disrupted by blast injury. Sixty C57BL/6 mice were randomly divided into control, blast, and blast + melatonin groups for behavioral experiments. The elevated maze experiment, open field experiment, and Morris Water Maze experiment were carried out on the 7th, 14th and 28th day after the blast injury. Fifty Sprague Dawley rats were randomly divided into control, blast, blast + melatonin, and blast + melatonin + luzindole groups for hormone assays and molecular and pathological experiments. Blood samples were used for HPG axis hormone detection and ELISA assays, and tissue samples were used to detect oxidative stress, inflammation, apoptosis, and stress-related protein levels. The results showed that melatonin pretreatment alleviated blast-induced behavioral abnormalities in mice and maintained the HPG axis hormone homeostasis in rats. Additionally, melatonin significantly reduced MDA5 expression and increased the expression of Nrf2/HO-1. Moreover, melatonin significantly inhibited NF-κB expression and upregulated IL-10 expression, and it reversed the blast-induced high expression of caspase-3 and Bax and the low expression of Bcl-2. Furthermore, luzindole counteracted melatonin inhibition of NF-κB and upregulated Nrf2/HO-1. Melatonin significantly alleviated blast-induced HPG axis hormone dyshomeostasis, behavioral abnormalities, oxidative stress, inflammation, and apoptosis, which may be achieved by upregulating the Nrf2/HO-1 signaling pathway. Our study suggested that melatonin pretreatment is a potential treatment for blast-induced HPG axis hormonal and behavioral abnormalities.
Collapse
Affiliation(s)
- Yin Zhang
- Graduate School, Dalian Medical University, Dalian, Liaoning Province 116044, China
| | - Peifang Cong
- Department of Emergency Medicine, The General Hospital of Northern Theater Command, Laboratory of Rescue Center of Severe Trauma PLA, Shenyang, Liaoning Province 116044, China
| | - Changci Tong
- Department of Emergency Medicine, The General Hospital of Northern Theater Command, Laboratory of Rescue Center of Severe Trauma PLA, Shenyang, Liaoning Province 116044, China
| | - Hongxu Jin
- Department of Emergency Medicine, The General Hospital of Northern Theater Command, Laboratory of Rescue Center of Severe Trauma PLA, Shenyang, Liaoning Province 116044, China
| | - Yunen Liu
- The Second Affiliated Hospital of Shenyang Medical College, The Veterans General Hospital of Liaoning Province, No. 20 Beijiu Road, Heping District, Shenyang 110001, China; Shenyang Medical College, No. 146, Huanghe North Street, Shenyang 110034, China.
| | - Mingxiao Hou
- Graduate School, Dalian Medical University, Dalian, Liaoning Province 116044, China; Department of Emergency Medicine, The General Hospital of Northern Theater Command, Laboratory of Rescue Center of Severe Trauma PLA, Shenyang, Liaoning Province 116044, China.
| |
Collapse
|
11
|
Huang TC, Luo L, Jiang SH, Chen C, He HY, Liang CF, Li WS, Wang H, Zhu L, Wang K, Guo Y. Targeting integrated stress response regulates microglial M1/M2 polarization and attenuates neuroinflammation following surgical brain injury in rat. Cell Signal 2021; 85:110048. [PMID: 34015470 DOI: 10.1016/j.cellsig.2021.110048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 05/01/2021] [Accepted: 05/16/2021] [Indexed: 12/24/2022]
Abstract
Integrated stress response (ISR) contributes to various neuropathological processes and acting as a therapy target in CNS injuries. However, the fundamental role of ISR in regulating microglial polarization remains largely unknown. Currently no proper pharmacological approaches to reverse microglia-driven neuroinflammation in surgical brain injury (SBI) have been reported. Here we found that inhibition of the crucial ISR effector, activating transcription factor 4 (ATF4), using the RNA interference suppressed the lipopolysaccharide (LPS)-stimulated microglial M1 polarization in vitro. Interestingly, counteracting ISR with a small-molecule ISR inhibitor (ISRIB) resulted in a significant microglial M1 towards M2 phenotype switching after LPS treatment. The potential underlying mechanisms may related to downregulate the intracellular NADPH oxidase 4 (NOX4) expression under the neuroinflammatory microenvironment. Notably, ISRIB ameliorated the infiltration of microglia and improved the neurobehavioral outcomes in the SBI rat model. Overall, our findings suggest that targeting ISR exerts a novel anti-inflammatory effect on microglia via regulating M1/M2 phenotype and may represent a potential therapeutic target to overcome neuroinflammation following SBI.
Collapse
Affiliation(s)
- Teng-Chao Huang
- Department of Neurosurgery, Third Affiliated Hospital of Sun Yat-sen University, Canton 510630, PR China; East China Institute of Digital Medical Engineering, Shangrao 334000, PR China
| | - Lun Luo
- Department of Neurosurgery, Third Affiliated Hospital of Sun Yat-sen University, Canton 510630, PR China
| | - Shi-Hai Jiang
- Department of Joint Replacement and Trauma Surgery, Third Affiliated Hospital of Sun Yat-sen University, Canton 510630, PR China; Institute for Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Germany
| | - Chuan Chen
- Department of Neurosurgery, Third Affiliated Hospital of Sun Yat-sen University, Canton 510630, PR China
| | - Hai-Yong He
- Department of Neurosurgery, Third Affiliated Hospital of Sun Yat-sen University, Canton 510630, PR China
| | - Chao-Feng Liang
- Department of Neurosurgery, Third Affiliated Hospital of Sun Yat-sen University, Canton 510630, PR China
| | - Wen-Sheng Li
- Department of Neurosurgery, Third Affiliated Hospital of Sun Yat-sen University, Canton 510630, PR China
| | - Hui Wang
- Department of Neurosurgery, Third Affiliated Hospital of Sun Yat-sen University, Canton 510630, PR China
| | - Lei Zhu
- Department of Burns, Plastic & Reconstructive Surgery, Third Affiliated Hospital of Sun Yat-sen University, Canton 510630, PR China
| | - Kun Wang
- Department of Joint Replacement and Trauma Surgery, Third Affiliated Hospital of Sun Yat-sen University, Canton 510630, PR China.
| | - Ying Guo
- Department of Neurosurgery, Third Affiliated Hospital of Sun Yat-sen University, Canton 510630, PR China.
| |
Collapse
|
12
|
Perez Garcia G, De Gasperi R, Gama Sosa MA, Perez GM, Otero-Pagan A, Pryor D, Abutarboush R, Kawoos U, Hof PR, Dickstein DL, Cook DG, Gandy S, Ahlers ST, Elder GA. Laterality and region-specific tau phosphorylation correlate with PTSD-related behavioral traits in rats exposed to repetitive low-level blast. Acta Neuropathol Commun 2021; 9:33. [PMID: 33648608 PMCID: PMC7923605 DOI: 10.1186/s40478-021-01128-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 02/07/2021] [Indexed: 12/14/2022] Open
Abstract
Military veterans who experience blast-related traumatic brain injuries often suffer from chronic cognitive and neurobehavioral syndromes. Reports of abnormal tau processing following blast injury have raised concerns that some cases may have a neurodegenerative basis. Rats exposed to repetitive low-level blast exhibit chronic neurobehavioral traits and accumulate tau phosphorylated at threonine 181 (Thr181). Using data previously reported in separate studies we tested the hypothesis that region-specific patterns of Thr181 phosphorylation correlate with behavioral measures also previously determined and reported in the same animals. Elevated p-tau Thr181 in anterior neocortical regions and right hippocampus correlated with anxiety as well as fear learning and novel object localization. There were no correlations with levels in amygdala or posterior neocortical regions. Particularly striking were asymmetrical effects on the right and left hippocampus. No systematic variation in head orientation toward the blast wave seems to explain the laterality. Levels did not correlate with behavioral measures of hyperarousal. Results were specific to Thr181 in that no correlations were observed for three other phospho-acceptor sites (threonine 231, serine 396, and serine 404). No consistent correlations were linked with total tau. These correlations are significant in suggesting that p-tau accumulation in anterior neocortical regions and the hippocampus may lead to disinhibited amygdala function without p-tau elevation in the amygdala itself. They also suggest an association linking blast injury with tauopathy, which has implications for understanding the relationship of chronic blast-related neurobehavioral syndromes in humans to neurodegenerative diseases.
Collapse
Affiliation(s)
- Georgina Perez Garcia
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY, 10468, USA
- Department of Neurology, Icahn School of Medicine At Mount Sinai, One Gustave Levy Place, New York, NY, 10029, USA
| | - Rita De Gasperi
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY, 10468, USA
- Department of Psychiatry, Icahn School of Medicine At Mount Sinai, One Gustave Levy Place, New York, NY, 10029, USA
| | - Miguel A Gama Sosa
- Department of Psychiatry, Icahn School of Medicine At Mount Sinai, One Gustave Levy Place, New York, NY, 10029, USA
- General Medical Research Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY, 10468, USA
| | - Gissel M Perez
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY, 10468, USA
| | - Alena Otero-Pagan
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY, 10468, USA
| | - Dylan Pryor
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY, 10468, USA
| | - Rania Abutarboush
- Department of Neurotrauma, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Usmah Kawoos
- Department of Neurotrauma, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Patrick R Hof
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine At Mount Sinai, One Gustave Levy Place, New York, NY, 10029, USA
- Department of Geriatrics and Palliative Care, Icahn School of Medicine At Mount Sinai, One Gustave Levy, New York, NY, 10029, USA
- Mount Sinai Alzheimer's Disease Research Center and Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine At Mount Sinai, One Gustave Levy Place, New York, NY, 10029, USA
| | - Dara L Dickstein
- Department of Pathology, Uniformed Services University of Health Sciences, 4301 Jones Bridge Rd., Bethesda, MD, 20814, USA
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of Health Sciences, 4301 Jones Bridge Rd., Bethesda, MD, 20814, USA
| | - David G Cook
- Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, 1660 S Columbian Way, Seattle, WA, 98108, USA
- Department of Medicine, University of Washington, 1959 NE Pacific St, Seattle, WA, 98195, USA
| | - Sam Gandy
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY, 10468, USA
- Department of Neurology, Icahn School of Medicine At Mount Sinai, One Gustave Levy Place, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine At Mount Sinai, One Gustave Levy Place, New York, NY, 10029, USA
- Mount Sinai Alzheimer's Disease Research Center and Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine At Mount Sinai, One Gustave Levy Place, New York, NY, 10029, USA
- Barbara and Maurice A. Deane Center for Wellness and Cognitive Health, and Mount Sinai NFL Neurological Care Center, Icahn School of Medicine At Mount Sinai, 5 East 98th Street, New York, NY, 10029, USA
| | - Stephen T Ahlers
- Department of Neurotrauma, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Gregory A Elder
- Department of Neurology, Icahn School of Medicine At Mount Sinai, One Gustave Levy Place, New York, NY, 10029, USA.
- Department of Psychiatry, Icahn School of Medicine At Mount Sinai, One Gustave Levy Place, New York, NY, 10029, USA.
- Mount Sinai Alzheimer's Disease Research Center and Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine At Mount Sinai, One Gustave Levy Place, New York, NY, 10029, USA.
- Neurology Service, James J. Peters Department of Veterans Affairs Medical Center, Neurology Service (3E16), 130 West Kingsbridge Road, Bronx, NY, 10468, USA.
| |
Collapse
|
13
|
Perez Garcia G, Perez GM, De Gasperi R, Gama Sosa MA, Otero-Pagan A, Pryor D, Abutarboush R, Kawoos U, Hof PR, Cook DG, Gandy S, Ahlers ST, Elder GA. Progressive Cognitive and Post-Traumatic Stress Disorder-Related Behavioral Traits in Rats Exposed to Repetitive Low-Level Blast. J Neurotrauma 2021; 38:2030-2045. [PMID: 33115338 DOI: 10.1089/neu.2020.7398] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Many military veterans who experienced blast-related traumatic brain injuries (TBI) in the conflicts in Iraq and Afghanistan currently have chronic cognitive and mental health problems including post-traumatic stress disorder (PTSD). Besides static symptoms, new symptoms may emerge or existing symptoms may worsen. TBI is also a risk factor for later development of neurodegenerative diseases. In rats exposed to repetitive low-level blast overpressure (BOP), robust and enduring cognitive and PTSD-related behavioral traits develop that are present for at least one year after blast exposure. Here we determined the time-course of the appearance of these traits by testing rats in the immediate post-blast period. Three cohorts of rats examined within the first eight weeks exhibited no behavioral phenotype or, in one cohort, features of anxiety. None showed the altered cued fear responses or impaired novel object recognition characteristic of the fully developed phenotype. Two cohorts retested 36 to 42 weeks after blast exposure exhibited the expanded behavioral phenotype including anxiety as well as altered cued fear learning and impaired novel object recognition. Combined with previous work, the chronic behavioral phenotype has been observed in six cohorts of blast-exposed rats studied at 3-4 months or longer after blast injury, and the three cohorts studied here document the progressive nature of the cognitive/behavioral phenotype. These studies suggest the existence of a latent, delayed emerging and progressive blast-induced cognitive and behavioral phenotype. The delayed onset has implications for the evolution of post-blast neurobehavioral syndromes in military veterans and its modeling in experimental animals.
Collapse
Affiliation(s)
- Georgina Perez Garcia
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA.,Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Gissel M Perez
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
| | - Rita De Gasperi
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Miguel A Gama Sosa
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,General Medical Research Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
| | - Alena Otero-Pagan
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
| | - Dylan Pryor
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
| | - Rania Abutarboush
- Department of Neurotrauma, Naval Medical Research Center, Silver Spring, Maryland, USA.,The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
| | - Usmah Kawoos
- Department of Neurotrauma, Naval Medical Research Center, Silver Spring, Maryland, USA.,The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
| | - Patrick R Hof
- Department of Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Department of Geriatrics and Palliative Care, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Department of Mount Sinai Alzheimer's Disease Research Center and Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - David G Cook
- Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, Washington, USA.,Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Sam Gandy
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA.,Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Barbara and Maurice A. Deane Center for Wellness and Cognitive Health, and the Mount Sinai NFL Neurological Care Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Stephen T Ahlers
- Department of Neurotrauma, Naval Medical Research Center, Silver Spring, Maryland, USA
| | - Gregory A Elder
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Department of Mount Sinai Alzheimer's Disease Research Center and Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Neurology Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
| |
Collapse
|
14
|
Ismael S, Ahmed HA, Adris T, Parveen K, Thakor P, Ishrat T. The NLRP3 inflammasome: a potential therapeutic target for traumatic brain injury. Neural Regen Res 2021; 16:49-57. [PMID: 32788447 PMCID: PMC7818859 DOI: 10.4103/1673-5374.286951] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Although the precise mechanisms contributing to secondary brain injury following traumatic brain injury are complex and obscure, a number of studies have demonstrated that inflammatory responses are an obvious and early feature in the pathogenesis of traumatic brain injury. Inflammasomes are multiprotein complexes that prompt the stimulation of caspase-1 and subsequently induce the maturation and secretion of proinflammatory cytokines, such as interleukin-1β and interleukin-18. These cytokines play a pivotal role in facilitating innate immune responses and inflammation. Among various inflammasome complexes, the NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome is the best characterized, a crucial role for NLRP3 has been demonstrated in various brain diseases, including traumatic brain injury. Several recent studies have revealed the contribution of NLRP3 inflammasome in identifying cellular damage and stimulating inflammatory responses to aseptic tissue injury after traumatic brain injury. Even more important, blocking or inhibiting the activation of the NLRP3 inflammasome may have substantial potential to salvage tissue damage during traumatic brain injury. In this review, we summarize recently described mechanisms that are involved in the activation and regulation of the NLRP3 inflammasome. Moreover, we review the recent investigations on the contribution of the NLRP3 inflammasome in the pathophysiology of TBI, and current advances and challenges in potential NLRP3-targeted therapies. A significant contribution of NLRP3 inflammasome activation to traumatic brain injury implies that therapeutic approaches focused on targeting specific inflammasome components could significantly improve the traumatic brain injury outcomes.
Collapse
Affiliation(s)
- Saifudeen Ismael
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Heba A Ahmed
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Tusita Adris
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | | | - Parth Thakor
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Tauheed Ishrat
- Department of Anatomy and Neurobiology; Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
15
|
Brain and blood biomarkers of tauopathy and neuronal injury in humans and rats with neurobehavioral syndromes following blast exposure. Mol Psychiatry 2021; 26:5940-5954. [PMID: 32094584 PMCID: PMC7484380 DOI: 10.1038/s41380-020-0674-z] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 12/31/2019] [Accepted: 01/30/2020] [Indexed: 12/14/2022]
Abstract
Traumatic brain injury (TBI) is a risk factor for the later development of neurodegenerative diseases that may have various underlying pathologies. Chronic traumatic encephalopathy (CTE) in particular is associated with repetitive mild TBI (mTBI) and is characterized pathologically by aggregation of hyperphosphorylated tau into neurofibrillary tangles (NFTs). CTE may be suspected when behavior, cognition, and/or memory deteriorate following repetitive mTBI. Exposure to blast overpressure from improvised explosive devices (IEDs) has been implicated as a potential antecedent for CTE amongst Iraq and Afghanistan Warfighters. In this study, we identified biomarker signatures in rats exposed to repetitive low-level blast that develop chronic anxiety-related traits and in human veterans exposed to IED blasts in theater with behavioral, cognitive, and/or memory complaints. Rats exposed to repetitive low-level blasts accumulated abnormal hyperphosphorylated tau in neuronal perikarya and perivascular astroglial processes. Using positron emission tomography (PET) and the [18F]AV1451 (flortaucipir) tau ligand, we found that five of 10 veterans exhibited excessive retention of [18F]AV1451 at the white/gray matter junction in frontal, parietal, and temporal brain regions, a typical localization of CTE tauopathy. We also observed elevated levels of neurofilament light (NfL) chain protein in the plasma of veterans displaying excess [18F]AV1451 retention. These findings suggest an association linking blast injury, tauopathy, and neuronal injury. Further study is required to determine whether clinical, neuroimaging, and/or fluid biomarker signatures can improve the diagnosis of long-term neuropsychiatric sequelae of mTBI.
Collapse
|
16
|
Desai A, Chen H, Kim HY. Multiple Mild Traumatic Brain Injuries Lead to Visual Dysfunction in a Mouse Model. J Neurotrauma 2019; 37:286-294. [PMID: 31530220 DOI: 10.1089/neu.2019.6602] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Visual dysfunction is a common occurrence after traumatic brain injury (TBI). We investigated in this study effects of single or multiple mild TBI on visual function in mice using a closed head injury model that permits unconstrained head movement after impact. Adult mice were briefly anesthetized with isoflurane and given one or three mild TBI with the closed head injury by mechanically engineered rotational acceleration (CHIMERA) device with an interinjury interval of 24 h. Mice were then tested in the Morris water maze, visual cliff, and open field tests from day 19 to day 32 and for visual evoked potential at 5 weeks after the last injury and euthanized. Mice with multiple TBI showed impaired performance in the visible platform water maze test and had increased errors in the visual cliff test. Further, there was a graded difference in visual evoked potential, with the single injury mice showing modest reduction in N1 amplitude whereas the multiple injuries produced significant reduction compared to sham and single injury groups. The optic tract of the injured mice showed increases in glial cell immunostaining. The increase in glial fibrillary acid protein immunostaining reached statistical significance for both injured groups whereas the ionized calcium binding adaptor molecule 1 immunostaining was only significantly increased in the optic tract of repeatedly injured mice. These results indicate that multiple injuries using CHIMERA may result in visual deficits, which can affect certain behavioral performances. The change in vision may be a useful marker when monitoring repeated TBI outcome and screening for protective agents from TBI.
Collapse
Affiliation(s)
- Abhishek Desai
- Laboratory of Molecular Signaling, NIAAA, NIH, Rockville, Maryland
| | - Huazhen Chen
- Laboratory of Molecular Signaling, NIAAA, NIH, Rockville, Maryland.,Center for Neuroscience and Regenerative Medicine at the Uniformed Services University of Health Sciences, Bethesda, Maryland
| | - Hee-Yong Kim
- Laboratory of Molecular Signaling, NIAAA, NIH, Rockville, Maryland
| |
Collapse
|
17
|
Gama Sosa MA, De Gasperi R, Perez Garcia GS, Perez GM, Searcy C, Vargas D, Spencer A, Janssen PL, Tschiffely AE, McCarron RM, Ache B, Manoharan R, Janssen WG, Tappan SJ, Hanson RW, Gandy S, Hof PR, Ahlers ST, Elder GA. Low-level blast exposure disrupts gliovascular and neurovascular connections and induces a chronic vascular pathology in rat brain. Acta Neuropathol Commun 2019; 7:6. [PMID: 30626447 PMCID: PMC6327415 DOI: 10.1186/s40478-018-0647-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 12/06/2018] [Indexed: 01/15/2023] Open
Abstract
Much concern exists over the role of blast-induced traumatic brain injury (TBI) in the chronic cognitive and mental health problems that develop in veterans and active duty military personnel. The brain vasculature is particularly sensitive to blast injury. The aim of this study was to characterize the evolving molecular and histologic alterations in the neurovascular unit induced by three repetitive low-energy blast exposures (3 × 74.5 kPa) in a rat model mimicking human mild TBI or subclinical blast exposure. High-resolution two-dimensional differential gel electrophoresis (2D-DIGE) and matrix-assisted laser desorption/ionization (MALDI) mass spectrometry of purified brain vascular fractions from blast-exposed animals 6 weeks post-exposure showed decreased levels of vascular-associated glial fibrillary acidic protein (GFAP) and several neuronal intermediate filament proteins (α-internexin and the low, middle, and high molecular weight neurofilament subunits). Loss of these proteins suggested that blast exposure disrupts gliovascular and neurovascular interactions. Electron microscopy confirmed blast-induced effects on perivascular astrocytes including swelling and degeneration of astrocytic endfeet in the brain cortical vasculature. Because the astrocyte is a major sensor of neuronal activity and regulator of cerebral blood flow, structural disruption of gliovascular integrity within the neurovascular unit should impair cerebral autoregulation. Disrupted neurovascular connections to pial and parenchymal blood vessels might also affect brain circulation. Blast exposures also induced structural and functional alterations in the arterial smooth muscle layer. Interestingly, by 8 months after blast exposure, GFAP and neuronal intermediate filament expression had recovered to control levels in isolated brain vascular fractions. However, despite this recovery, a widespread vascular pathology was still apparent at 10 months after blast exposure histologically and on micro-computed tomography scanning. Thus, low-level blast exposure disrupts gliovascular and neurovascular connections while inducing a chronic vascular pathology.
Collapse
|
18
|
Rama Rao KV, Iring S, Younger D, Kuriakose M, Skotak M, Alay E, Gupta RK, Chandra N. A Single Primary Blast-Induced Traumatic Brain Injury in a Rodent Model Causes Cell-Type Dependent Increase in Nicotinamide Adenine Dinucleotide Phosphate Oxidase Isoforms in Vulnerable Brain Regions. J Neurotrauma 2018; 35:2077-2090. [PMID: 29648986 PMCID: PMC6098412 DOI: 10.1089/neu.2017.5358] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Blast-induced traumatic brain injury (bTBI) is a leading cause of morbidity in soldiers on the battlefield and in training sites with long-term neurological and psychological pathologies. Previous studies from our laboratory demonstrated activation of oxidative stress pathways after blast injury, but their distribution among different brain regions and their impact on the pathogenesis of bTBI have not been explored. The present study examined the protein expression of two isoforms: nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 1 and 2 (NOX1, NOX2), corresponding superoxide production, a downstream event of NOX activation, and the extent of lipid peroxidation adducts of 4-hydroxynonenal (4HNE) to a range of proteins. Brain injury was evaluated 4 h after the shock-wave exposure, and immunofluorescence signal quantification was performed in different brain regions. Expression of NOX isoforms displayed a differential increase in various brain regions: in hippocampus and thalamus, there was the highest increase of NOX1, whereas in the frontal cortex, there was the highest increase of NOX2 expression. Cell-specific analysis of changes in NOX expression with respect to corresponding controls revealed that blast resulted in a higher increase of NOX1 and NOX 2 levels in neurons compared with astrocytes and microglia. Blast exposure also resulted in increased superoxide levels in different brain regions, and such changes were reflected in 4HNE protein adduct formation. Collectively, this study demonstrates that primary blast TBI induces upregulation of NADPH oxidase isoforms in different regions of the brain parenchyma and that neurons appear to be at higher risk for oxidative damage compared with other neural cells.
Collapse
Affiliation(s)
- Kakulavarapu V Rama Rao
- 1 Center for Injury Biomechanics, Materials, and Medicine, Department of Biomedical Engineering, New Jersey Institute of Technology , Newark, New Jersey
| | - Stephanie Iring
- 1 Center for Injury Biomechanics, Materials, and Medicine, Department of Biomedical Engineering, New Jersey Institute of Technology , Newark, New Jersey
| | - Daniel Younger
- 1 Center for Injury Biomechanics, Materials, and Medicine, Department of Biomedical Engineering, New Jersey Institute of Technology , Newark, New Jersey
| | - Matthew Kuriakose
- 1 Center for Injury Biomechanics, Materials, and Medicine, Department of Biomedical Engineering, New Jersey Institute of Technology , Newark, New Jersey
| | - Maciej Skotak
- 1 Center for Injury Biomechanics, Materials, and Medicine, Department of Biomedical Engineering, New Jersey Institute of Technology , Newark, New Jersey
| | - Eren Alay
- 1 Center for Injury Biomechanics, Materials, and Medicine, Department of Biomedical Engineering, New Jersey Institute of Technology , Newark, New Jersey
| | - Raj K Gupta
- 2 Department of Defense Blast Injury Research Program Coordinating Office, United States Army Medical Research and Materiel Command , Fort Detrick, Maryland
| | - Namas Chandra
- 1 Center for Injury Biomechanics, Materials, and Medicine, Department of Biomedical Engineering, New Jersey Institute of Technology , Newark, New Jersey
| |
Collapse
|
19
|
Ma MW, Wang J, Dhandapani KM, Wang R, Brann DW. NADPH oxidases in traumatic brain injury - Promising therapeutic targets? Redox Biol 2018; 16:285-293. [PMID: 29571125 PMCID: PMC5952873 DOI: 10.1016/j.redox.2018.03.005] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 03/09/2018] [Accepted: 03/10/2018] [Indexed: 12/22/2022] Open
Abstract
Traumatic brain injury (TBI) is a major cause of death and disability worldwide. Despite intense investigation, no neuroprotective agents for TBI have yet translated to the clinic. Recent efforts have focused on identifying potential therapeutic targets that underlie the secondary TBI pathology that evolves minutes to years following the initial injury. Oxidative stress is a key player in this complex cascade of secondary injury mechanisms and prominently contributes to neurodegeneration and neuroinflammation. NADPH oxidase (NOX) is a family of enzymes whose unique function is to produce reactive oxygen species (ROS). Human post-mortem and animal studies have identified elevated NOX2 and NOX4 levels in the injured brain, suggesting that these two NOXs are involved in the pathogenesis of TBI. In support of this, NOX2 and NOX4 deletion studies have collectively revealed that targeting NOX enzymes can reduce oxidative stress, attenuate neuroinflammation, promote neuronal survival, and improve functional outcomes following TBI. In addition, NOX inhibitor studies have confirmed these findings and demonstrated an extended critical window of efficacious TBI treatment. Finally, the translational potential, caveats, and future directions of the field are highlighted and discussed throughout the review.
Collapse
Affiliation(s)
- Merry W Ma
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Jing Wang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Krishnan M Dhandapani
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Ruimin Wang
- Department of Neurobiology, North China University of Science and Technology, Tangshan, Hebei, China
| | - Darrell W Brann
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
20
|
Byrd M, Dixon CE, Lucke-Wold B. Examining the Correlation between Acute Behavioral Manifestations of Concussion and the Underlying Pathophysiology of Chronic Traumatic Encephalopathy: A Pilot Study. JOURNAL OF NEUROLOGY AND PSYCHOLOGY 2018; 6. [PMID: 30079371 PMCID: PMC6075831 DOI: 10.13188/2332-3469.1000037] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Concussion in athletes can contribute to early neuropsychological changes that may be indicative of future neurodegenerative disease. One of the hallmark findings of chronic traumatic encephalopathy is anxiety and impulsive behavior that often develops early in the course of the disease. The behavioral dysfunction can be grouped into a broader category referred to as cognitive disruption. The current gold standard for diagnosing chronic neurodegeneration is post-mortem evaluation of tauopathy to identify neurofibrillary tau tangles in neurons. Few studies, however, have looked at clinical correlations between acute injury and chronic neurodegeneration in terms of behavior. This lack of focus towards translational study has limited advancements towards treatment. In this pilot investigation, the acute cognitive and emotional (anger, impulsivity, and anxiety) affects of concussion in a cohort of collegiate athletes (n = 30) are examined and compared to findings in the post-mortem pathologic features of chronic traumatic encephalopathy. Specifically, the role of the seroternergic system with alpha synuclein and tauopathy staining and the potential for early clinically relevant behavioral and pharmaceutical interventions was investigated. The purpose was to determine if athletes began demonstrating cognitive disruption present in post-mortem evaluation during the acute phase of injury. The acute data was collected via questionnaires within ten days of the athletes’ concussion diagnosis. Results demonstrated that 11 of 30 athletes (36%) scored in a diagnosable range of anxiety post-concussion, and athletes scored above the norm in state-anger (M = 22.9, SD = 9.99), indicating severe emotional disturbance. A limitation is that due to the long time frame from acute injury to the development of neurodegeneration individual athletes cannot be tracked in longevity thus limiting the findings to the realm of correlation. The findings from this pilot study warrant further investigation into the neuropsychological aspects for how to manage concussion and prevent degenerative disease.
Collapse
Affiliation(s)
- M Byrd
- Department of Exercise and Sports Psychology, West Virginia University, WV
| | - C E Dixon
- Department of Neurosurgery, University of Pittsburgh, PA
| | - B Lucke-Wold
- Department of Neurosurgery, West Virginia University, WV
| |
Collapse
|
21
|
Ma MW, Wang J, Dhandapani KM, Brann DW. Deletion of NADPH oxidase 4 reduces severity of traumatic brain injury. Free Radic Biol Med 2018; 117:66-75. [PMID: 29391196 DOI: 10.1016/j.freeradbiomed.2018.01.031] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 01/23/2018] [Accepted: 01/25/2018] [Indexed: 12/30/2022]
Abstract
Traumatic brain injury (TBI) contributes to over 30% of injury-related deaths and is a major cause of disability without effective clinical therapies. Oxidative stress contributes to neurodegeneration, neuroinflammation, and neuronal death to amplify the primary injury after TBI. NADPH oxidase (NOX) is a major source of reactive oxygen species following brain injury. Our current study addresses the functional role of the NOX4 isoform in the damaged cortex following TBI. Adult male C57BL/6 J and NOX4-/- mice received a controlled cortical impact and lesion size, NOX4 expression, oxidative stress, neurodegeneration, and cell death were assessed in the injured cerebral cortex. The results revealed that NOX4 mRNA and protein expression were significantly upregulated at 1-7 days post-TBI in the injured cerebral cortex. Expression of the oxidative stress markers, 8-OHdG, 4-HNE, and nitrotyrosine was upregulated at 2 and 4 days post-TBI in the WT injured cerebral cortex, and nitrotyrosine primarily colocalized with neurons. In the NOX4-/- mice, expression of these oxidative stress markers, 8-OHdG, 4-HNE, and nitrotyrosine were significantly attenuated at both timepoints. In addition, examination of NOX4-/- mice revealed a reduced number of apoptotic (TUNEL+) and degenerating (FJB+) cells in the perilesional cortex after TBI, as well as a smaller lesion size compared with the WT group. The results of this study implicate a functional role for NOX4 in TBI induced oxidative damage and neurodegeneration and raise the possibility that targeting NOX4 may have therapeutic efficacy in TBI.
Collapse
Affiliation(s)
- Merry W Ma
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA 30904, USA; Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Jing Wang
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA 30904, USA; Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Krishnan M Dhandapani
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA 30904, USA; Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Darrell W Brann
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA 30904, USA; Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| |
Collapse
|
22
|
Lucke-Wold BP, Logsdon AF, Nguyen L, Eltanahay A, Turner RC, Bonasso P, Knotts C, Moeck A, Maroon JC, Bailes JE, Rosen CL. Supplements, nutrition, and alternative therapies for the treatment of traumatic brain injury. Nutr Neurosci 2018; 21:79-91. [PMID: 27705610 PMCID: PMC5491366 DOI: 10.1080/1028415x.2016.1236174] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Studies using traditional treatment strategies for mild traumatic brain injury (TBI) have produced limited clinical success. Interest in treatment for mild TBI is at an all time high due to its association with the development of chronic traumatic encephalopathy and other neurodegenerative diseases, yet therapeutic options remain limited. Traditional pharmaceutical interventions have failed to transition to the clinic for the treatment of mild TBI. As such, many pre-clinical studies are now implementing non-pharmaceutical therapies for TBI. These studies have demonstrated promise, particularly those that modulate secondary injury cascades activated after injury. Because no TBI therapy has been discovered for mild injury, researchers now look to pharmaceutical supplementation in an attempt to foster success in human clinical trials. Non-traditional therapies, such as acupuncture and even music therapy are being considered to combat the neuropsychiatric symptoms of TBI. In this review, we highlight alternative approaches that have been studied in clinical and pre-clinical studies of TBI, and other related forms of neural injury. The purpose of this review is to stimulate further investigation into novel and innovative approaches that can be used to treat the mechanisms and symptoms of mild TBI.
Collapse
Affiliation(s)
- Brandon P. Lucke-Wold
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, USA
- Center for Neuroscience, West Virginia University School of Medicine, Morgantown, USA
| | - Aric F. Logsdon
- Center for Neuroscience, West Virginia University School of Medicine, Morgantown, USA
| | - Linda Nguyen
- Center for Neuroscience, West Virginia University School of Medicine, Morgantown, USA
| | - Ahmed Eltanahay
- Department of Neurosurgery, Oregon Health Sciences University, Portland, USA
| | - Ryan C. Turner
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, USA
| | - Patrick Bonasso
- Center for Neuroscience, West Virginia University School of Medicine, Morgantown, USA
| | - Chelsea Knotts
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, USA
| | - Adam Moeck
- Department of Surgery, Matigan Army Medical Center, Tacoma, WA, USA
| | - Joseph C. Maroon
- Department of Neurosurgery, University of Pittsburgh Medical Center, PA, USA
| | - Julian E. Bailes
- Department of Neurosurgery, Northshore Healthcare System, Evanston, IL, USA
| | - Charles L. Rosen
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, USA
| |
Collapse
|
23
|
Hylin MJ, Holden RC, Smith AC, Logsdon AF, Qaiser R, Lucke-Wold BP. Juvenile Traumatic Brain Injury Results in Cognitive Deficits Associated with Impaired Endoplasmic Reticulum Stress and Early Tauopathy. Dev Neurosci 2018; 40:175-188. [PMID: 29788004 PMCID: PMC6376969 DOI: 10.1159/000488343] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 03/12/2018] [Indexed: 02/05/2023] Open
Abstract
The leading cause of death in the juvenile population is trauma, and in particular neurotrauma. The juvenile brain response to neurotrauma is not completely understood. Endoplasmic reticulum (ER) stress has been shown to contribute to injury expansion and behavioral deficits in adult rodents and furthermore has been seen in adult postmortem human brains diagnosed with chronic traumatic encephalopathy. Whether endoplasmic reticulum stress is increased in juveniles with traumatic brain injury (TBI) is poorly delineated. We investigated this important topic using a juvenile rat controlled cortical impact (CCI) model. We proposed that ER stress would be significantly increased in juvenile rats following TBI and that this would correlate with behavioral deficits using a juvenile rat model. A juvenile rat (postnatal day 28) CCI model was used. Binding immunoglobulin protein (BiP) and C/EBP homologous protein (CHOP) were measured at 4 h in the ipsilateral pericontusion cortex. Hypoxia-inducible factor (HIF)-1α was measured at 48 h and tau kinase measured at 1 week and 30 days. At 4 h following injury, BiP and CHOP (markers of ER stress) were significantly elevated in rats exposed to TBI. We also found that HIF-1α was significantly upregulated 48 h following TBI showing delayed hypoxia. The early ER stress activation was additionally asso-ciated with the activation of a known tau kinase, glycogen synthase kinase-3β (GSK-3β), by 1 week. Tau oligomers measured by R23 were significantly increased by 30 days following TBI. The biochemical changes following TBI were associated with increased impulsive-like or anti-anxiety behavior measured with the elevated plus maze, deficits in short-term memory measured with novel object recognition, and deficits in spatial memory measured with the Morris water maze in juvenile rats exposed to TBI. These results show that ER stress was increased early in juvenile rats exposed to TBI, that these rats developed tau oligomers over the course of 30 days, and that they had significant short-term and spatial memory deficits following injury.
Collapse
Affiliation(s)
- Michael J. Hylin
- Neurotrauma and Rehabilitation Laboratory, Department of Psychology, Southern Illinois University, Carbondale, IL, USA
| | - Ryan C. Holden
- Neurotrauma and Rehabilitation Laboratory, Department of Psychology, Southern Illinois University, Carbondale, IL, USA
| | - Aidan C. Smith
- Neurotrauma and Rehabilitation Laboratory, Department of Psychology, Southern Illinois University, Carbondale, IL, USA
| | - Aric F. Logsdon
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Rabia Qaiser
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Brandon P. Lucke-Wold
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, USA
| |
Collapse
|
24
|
Dong XY, Feng Z. Wake-promoting effects of vagus nerve stimulation after traumatic brain injury: upregulation of orexin-A and orexin receptor type 1 expression in the prefrontal cortex. Neural Regen Res 2018; 13:244-251. [PMID: 29557373 PMCID: PMC5879895 DOI: 10.4103/1673-5374.226395] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Orexins, produced in the lateral hypothalamus, are important neuropeptides that participate in the sleep/wake cycle, and their expression coincides with the projection area of the vagus nerve in the brain. Vagus nerve stimulation has been shown to decrease the amounts of daytime sleep and rapid eye movement in epilepsy patients with traumatic brain injury. In the present study, we investigated whether vagus nerve stimulation promotes wakefulness and affects orexin expression. A rat model of traumatic brain injury was established using the free fall drop method. In the stimulated group, rats with traumatic brain injury received vagus nerve stimulation (frequency, 30 Hz; current, 1.0 mA; pulse width, 0.5 ms; total stimulation time, 15 minutes). In the antagonist group, rats with traumatic brain injury were intracerebroventricularly injected with the orexin receptor type 1 (OX1R) antagonist SB334867 and received vagus nerve stimulation. Changes in consciousness were observed after stimulation in each group. Enzyme-linked immunosorbent assay, western blot assay and immunohistochemistry were used to assess the levels of orexin-A and OX1R expression in the prefrontal cortex. In the stimulated group, consciousness was substantially improved, orexin-A protein expression gradually increased within 24 hours after injury and OX1R expression reached a peak at 12 hours, compared with rats subjected to traumatic brain injury only. In the antagonist group, the wake-promoting effect of vagus nerve stimulation was diminished, and orexin-A and OX1R expression were decreased, compared with that of the stimulated group. Taken together, our findings suggest that vagus nerve stimulation promotes the recovery of consciousness in comatose rats after traumatic brain injury. The upregulation of orexin-A and OX1R expression in the prefrontal cortex might be involved in the wake-promoting effects of vagus nerve stimulation.
Collapse
Affiliation(s)
- Xiao-Yang Dong
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Zhen Feng
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| |
Collapse
|
25
|
Lucke-Wold B, Seidel K, Udo R, Omalu B, Ornstein M, Nolan R, Rosen C, Ross J. Role of Tau Acetylation in Alzheimer's Disease and Chronic Traumatic Encephalopathy: The Way Forward for Successful Treatment. JOURNAL OF NEUROLOGY AND NEUROSURGERY 2017; 4. [PMID: 29276758 PMCID: PMC5738035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Progressive neurodegenerative diseases plague millions of individuals both in the United States and across the world. The current pathology of progressive neurodegenerative tauopathies, such as Alzheimer's disease (AD), Pick's disease, frontotemporal dementia (FTD), and progressive supranuclear palsy, primarily revolves around phosphorylation and hyperphosphorylation of the tau protein. However, more recent evidence suggests acetylation of tau protein at lysine 280 may be a critical step in molecular pathology of these neurodegenerative diseases prior to the tau hyperphosphorylation. Secondary injury cascades such as oxidative stress, endoplasmic reticulum stress, and neuroinflammation contribute to lasting damage within the brain and can be induced by a number of different risk factors. These injury cascades funnel into a common pathway of early tau acetylation, which may serve as the catalyst for progressive degeneration. The post translational modification of tau can result in production of toxic oligomers, contributing to reduced solubility as well as aggregation and formation of neurofibrillary tangles, the hallmark of AD pathology. Chronic Traumatic Encephalopathy (CTE), caused by repetitive brain trauma is also associated with a hyperphosphorylation of tau. We postulated acetylation of tau at lysine 280 in CTE disease could be present prior to the hyperphosphorylation and tested this hypothesis in CTE pathologic specimens. We also tested for ac-tau 280 in early stage Alzheimer's disease (Braak stage 1). Histopathological examination using the ac tau 280 antibody was performed in three Alzheimer's cases and three CTE patients. Presence of ac-tau 280 was confirmed in all cases at early sites of disease manifestation. These findings suggest that tau acetylation may precede tau phosphorylation and could be the first "triggering" event leading to neuronal loss. To the best of our knowledge, this is the first study to identify acetylation of the tau protein in CTE. Prevention of tau acetylation could possibly serve as a novel target for stopping neurodegeneration before it fully begins. In this study, we highlight what is known about tau acetylation and neurodegeneration.
Collapse
Affiliation(s)
- Brandon Lucke-Wold
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV
| | - Kay Seidel
- Dr. Senckenberg Chronomedical Institute, J. W. Goethe University, Frankfurt am Main, Germany
| | - Rub Udo
- Dr. Senckenberg Chronomedical Institute, J. W. Goethe University, Frankfurt am Main, Germany
| | - Bennet Omalu
- Department of Pathology, University of California Davis Medical Center, Davis, CA
| | | | - Richard Nolan
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV
| | - Charles Rosen
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV
| | - Joel Ross
- Cogwellin LLC 4 Industrial Way W, Eatontown NJ, USA
| |
Collapse
|
26
|
Kulbe JR, Hall ED. Chronic traumatic encephalopathy-integration of canonical traumatic brain injury secondary injury mechanisms with tau pathology. Prog Neurobiol 2017; 158:15-44. [PMID: 28851546 PMCID: PMC5671903 DOI: 10.1016/j.pneurobio.2017.08.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 08/09/2017] [Accepted: 08/17/2017] [Indexed: 12/14/2022]
Abstract
In recent years, a new neurodegenerative tauopathy labeled Chronic Traumatic Encephalopathy (CTE), has been identified that is believed to be primarily a sequela of repeated mild traumatic brain injury (TBI), often referred to as concussion, that occurs in athletes participating in contact sports (e.g. boxing, American football, Australian football, rugby, soccer, ice hockey) or in military combatants, especially after blast-induced injuries. Since the identification of CTE, and its neuropathological finding of deposits of hyperphosphorylated tau protein, mechanistic attention has been on lumping the disorder together with various other non-traumatic neurodegenerative tauopathies. Indeed, brains from suspected CTE cases that have come to autopsy have been confirmed to have deposits of hyperphosphorylated tau in locations that make its anatomical distribution distinct for other tauopathies. The fact that these individuals experienced repetitive TBI episodes during their athletic or military careers suggests that the secondary injury mechanisms that have been extensively characterized in acute TBI preclinical models, and in TBI patients, including glutamate excitotoxicity, intracellular calcium overload, mitochondrial dysfunction, free radical-induced oxidative damage and neuroinflammation, may contribute to the brain damage associated with CTE. Thus, the current review begins with an in depth analysis of what is known about the tau protein and its functions and dysfunctions followed by a discussion of the major TBI secondary injury mechanisms, and how the latter have been shown to contribute to tau pathology. The value of this review is that it might lead to improved neuroprotective strategies for either prophylactically attenuating the development of CTE or slowing its progression.
Collapse
Affiliation(s)
- Jacqueline R Kulbe
- Spinal Cord & Brain Injury Research Center, University of Kentucky College of Medicine, United States; Department of Neuroscience, University of Kentucky College of Medicine, United States
| | - Edward D Hall
- Spinal Cord & Brain Injury Research Center, University of Kentucky College of Medicine, United States; Department of Neuroscience, University of Kentucky College of Medicine, United States.
| |
Collapse
|
27
|
Lucke-Wold BP, Logsdon AF, Turner RC, Huber JD, Rosen CL. Endoplasmic Reticulum Stress Modulation as a Target for Ameliorating Effects of Blast Induced Traumatic Brain Injury. J Neurotrauma 2017; 34:S62-S70. [PMID: 28077004 PMCID: PMC5749601 DOI: 10.1089/neu.2016.4680] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Blast traumatic brain injury (bTBI) has been shown to contribute to progressive neurodegenerative disease. Recent evidence suggests that endoplasmic reticulum (ER) stress is a mechanistic link between acute neurotrauma and progressive tauopathy. We propose that ER stress contributes to extensive behavioral changes associated with a chronic traumatic encephalopathy (CTE)-like phenotype. Targeting ER stress is a promising option for the treatment of neurotrauma-related neurodegeneration, which warrants investigation. Utilizing our validated and clinically relevant Sprague-Dawley blast model, we investigated a time course of mechanistic changes that occur following bTBI (50 psi) including: ER stress activation, iron-mediated toxicity, and tauopathy via Western blot and immunohistochemistry. These changes were associated with behavioral alterations measured by the Elevated Plus Maze (EPM), Forced Swim Test (FST), and Morris Water Maze (MWM). Following characterization, salubrinal, an ER stress modulator, was given at a concentration of 1 mg/kg post-blast, and its mechanism of action was determined in vitro. bTBI significantly increased markers of injury in the cortex of the left hemisphere: p-PERK and p-eIF2α at 30 min, p-T205 tau at 6 h, and iron at 24 h. bTBI animals spent more time immobile on the FST at 72 h and more time in the open arm of the EPM at 7 days. Further, bTBI caused a significant learning disruption measured with MWM at 21 days post-blast, with persistent tau changes. Salubrinal successfully reduced ER stress markers in vivo and in vitro while significantly improving performance on the EPM. bTBI causes robust biochemical changes that contribute to neurodegeneration, but these changes may be targeted with ER stress modulators.
Collapse
Affiliation(s)
- Brandon P. Lucke-Wold
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Aric F. Logsdon
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, West Virginia
| | - Ryan C. Turner
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Jason D. Huber
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, West Virginia
| | - Charles L. Rosen
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, West Virginia
| |
Collapse
|
28
|
Defective methionine metabolism in the brain after repeated blast exposures might contribute to increased oxidative stress. Neurochem Int 2017; 112:234-238. [PMID: 28774719 DOI: 10.1016/j.neuint.2017.07.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 07/29/2017] [Indexed: 01/26/2023]
Abstract
Blast-induced traumatic brain injury (bTBI) is one of the major disabilities in Service Members returning from recent military operations. The neurobiological underpinnings of bTBI, which are associated with acute and chronic neuropathological and neurobehavioral deficits, are uncertain. Increased oxidative stress in the brain is reported to play a significant role promoting neuronal damage associated with both brain injury and neurodegenerative disorders. In this study, brains of rats exposed to repeated blasts in a shock tube underwent untargeted profiling of primary metabolism by automatic linear exchange/cold injection GC-TOF mass spectrometry and revealed acute and sub-acute disruptions in the metabolism of the essential amino acid methionine and associated antioxidants. Methionine sulfoxide, the oxidized metabolite of methionine, showed a sustained increase in the brain after blast exposure which was associated with a significant decrease in cysteine, the amino acid derived from methionine. Glutathione, the antioxidant synthesized from cysteine, also concomitantly decreased as did the antioxidant ascorbic acid. Reductions in ascorbic acid were accompanied by increased levels of its oxidized metabolite, dehydroascorbic acid and other metabolites such as threonic acid, isothreonic acid, glycolic acid and oxalic acid. Fluorometric analysis of the brains showed acute and sub-acute increase in total reactive oxygen species. In view of the fundamental importance of glutathione in the brain as an antioxidant, including its role in the reduction of dehydroascorbic acid to ascorbic acid, the disruptions in methionine metabolism elicited by blast exposure might prominently contribute to neuronal injury by promoting increased and sustained oxidative stress.
Collapse
|
29
|
NADPH Oxidase 2 Regulates NLRP3 Inflammasome Activation in the Brain after Traumatic Brain Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:6057609. [PMID: 28785377 PMCID: PMC5529650 DOI: 10.1155/2017/6057609] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 05/02/2017] [Accepted: 06/01/2017] [Indexed: 01/27/2023]
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability worldwide. After the initial primary mechanical injury, a complex secondary injury cascade involving oxidative stress and neuroinflammation follows, which may exacerbate the injury and complicate the healing process. NADPH oxidase 2 (NOX2) is a major contributor to oxidative stress in TBI pathology, and inhibition of NOX2 is neuroprotective. The NLRP3 inflammasome can become activated in response to oxidative stress, but little is known about the role of NOX2 in regulating NLRP3 inflammasome activation following TBI. In this study, we utilized NOX2 knockout mice to study the role of NOX2 in mediating NLRP3 inflammasome expression and activation following a controlled cortical impact. Expression of NLRP3 inflammasome components NLRP3 and apoptosis-associated speck-like protein containing a CARD (ASC), as well as its downstream products cleaved caspase-1 and interleukin-1β (IL-1β), was robustly increased in the injured cerebral cortex following TBI. Deletion of NOX2 attenuated the expression, assembly, and activity of the NLRP3 inflammasome via a mechanism that was associated with TXNIP, a sensor of oxidative stress. The results support the notion that NOX2-dependent inflammasome activation contributes to TBI pathology.
Collapse
|
30
|
Braun M, Vaibhav K, Saad NM, Fatima S, Vender JR, Baban B, Hoda MN, Dhandapani KM. White matter damage after traumatic brain injury: A role for damage associated molecular patterns. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2614-2626. [PMID: 28533056 DOI: 10.1016/j.bbadis.2017.05.020] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 05/09/2017] [Accepted: 05/16/2017] [Indexed: 12/12/2022]
Abstract
Traumatic brain injury (TBI) is a leading cause of mortality and long-term morbidity worldwide. Despite decades of pre-clinical investigation, therapeutic strategies focused on acute neuroprotection failed to improve TBI outcomes. This lack of translational success has necessitated a reassessment of the optimal targets for intervention, including a heightened focus on secondary injury mechanisms. Chronic immune activation correlates with progressive neurodegeneration for decades after TBI; however, significant challenges remain in functionally and mechanistically defining immune activation after TBI. In this review, we explore the burgeoning evidence implicating the acute release of damage associated molecular patterns (DAMPs), such as adenosine 5'-triphosphate (ATP), high mobility group box protein 1 (HMGB1), S100 proteins, and hyaluronic acid in the initiation of progressive neurological injury, including white matter loss after TBI. The role that pattern recognition receptors, including toll-like receptor and purinergic receptors, play in progressive neurological injury after TBI is detailed. Finally, we provide support for the notion that resident and infiltrating macrophages are critical cellular targets linking acute DAMP release with adaptive immune responses and chronic injury after TBI. The therapeutic potential of targeting DAMPs and barriers to clinical translational, in the context of TBI patient management, are discussed.
Collapse
Affiliation(s)
- Molly Braun
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Kumar Vaibhav
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, United States; Department of Medical Laboratory, Imaging & Radiologic Sciences, College of Allied Health Science, Augusta University, Augusta, GA, United States
| | - Nancy M Saad
- Department of Oral Biology, Dental College of Georgia, Augusta University, Augusta, GA, United States
| | - Sumbul Fatima
- Department of Medical Laboratory, Imaging & Radiologic Sciences, College of Allied Health Science, Augusta University, Augusta, GA, United States
| | - John R Vender
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Babak Baban
- Department of Oral Biology, Dental College of Georgia, Augusta University, Augusta, GA, United States; Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Md Nasrul Hoda
- Department of Medical Laboratory, Imaging & Radiologic Sciences, College of Allied Health Science, Augusta University, Augusta, GA, United States; Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Krishnan M Dhandapani
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, United States.
| |
Collapse
|
31
|
Logsdon AF, Lucke-Wold BP, Turner RC, Li X, Adkins CE, Mohammad AS, Huber JD, Rosen CL, Lockman PR. A mouse Model of Focal Vascular Injury Induces Astrocyte Reactivity, Tau Oligomers, and Aberrant Behavior. ARCHIVES OF NEUROSCIENCE 2017; 4. [PMID: 28758136 PMCID: PMC5529099 DOI: 10.5812/archneurosci.44254] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Neuropsychiatric symptom development has become more prevalent with 270,000 blast exposures occurring in the past 10 years in the United States. How blast injury leads to neuropsychiatric symptomology is currently unknown. Preclinical models of blast-induced traumatic brain injury have been used to demonstrate blood-brain barrier disruption, degenerative pathophysiology, and behavioral deficits. Vascular injury is a primary effect of neurotrauma that can trigger secondary injury cascades and neurodegeneration. Here we present data from a novel scaled and clinically relevant mouse blast model that was specifically developed to assess the outcome of vascular injury. We look at the biochemical effects and behavioral changes associated with blast injury in young-adult male BALB/c mice. We report that blast exposure causes focal vascular injury in the Somatosensory Barrel Field cortex, which leads to perivascular astrocyte reactivity, as well as acute aberrant behavior. Biochemical analysis revealed that mild blast exposure also invokes tauopathy, neuroinflammation, and oxidative stress. Overall, we propose our model to be used to evaluate focal blood-brain barrier disruption and to discover novel therapies for human neuropsychiatric symptoms.
Collapse
Affiliation(s)
- Aric F. Logsdon
- Department of Pharmaceutical Sciences, West Virginia University School of Medicine, Morgantown, WV 26506-9530, USA
| | - Brandon P. Lucke-Wold
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV 26506-9183, USA
| | - Ryan C. Turner
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV 26506-9183, USA
| | - Xinlan Li
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV 26506-9183, USA
| | - Chris E. Adkins
- Department of Pharmaceutical Sciences, West Virginia University School of Medicine, Morgantown, WV 26506-9530, USA
| | - Afroz S. Mohammad
- Department of Pharmaceutical Sciences, West Virginia University School of Medicine, Morgantown, WV 26506-9530, USA
| | - Jason D. Huber
- Department of Pharmaceutical Sciences, West Virginia University School of Medicine, Morgantown, WV 26506-9530, USA
| | - Charles L. Rosen
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV 26506-9183, USA
| | - Paul R. Lockman
- Department of Pharmaceutical Sciences, West Virginia University School of Medicine, Morgantown, WV 26506-9530, USA
- Corresponding author: Paul R. Lockman, Department of Pharmaceutical Sciences, West Virginia University School of Medicine, Morgantown, WV 26506-9530, USA,
| |
Collapse
|
32
|
Protection against Blast-Induced Traumatic Brain Injury by Increase in Brain Volume. BIOMED RESEARCH INTERNATIONAL 2017; 2017:2075463. [PMID: 28553646 PMCID: PMC5434276 DOI: 10.1155/2017/2075463] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 02/13/2017] [Accepted: 03/23/2017] [Indexed: 11/18/2022]
Abstract
Blast-induced traumatic brain injury (bTBI) is a leading cause of injuries in recent military conflicts and it is responsible for an increased number of civilian casualties by terrorist attacks. bTBI includes a variety of neuropathological changes depending on the intensity of blast overpressure (BOP) such as brain edema, neuronal degeneration, diffuse axonal damage, and vascular dysfunction with neurological manifestations of psychological and cognitive abnormalities. Internal jugular vein (IJV) compression is known to reduce intracranial compliance by causing an increase in brain volume and was shown to reduce brain damage during closed impact-induced TBI. We investigated whether IJV compression can attenuate signs of TBI in rats after exposure to BOP. Animals were exposed to three 110 ± 5 kPa BOPs separated by 30 min intervals. Exposure to BOP resulted in a significant decrease of neuronal nuclei (NeuN) together with upregulation of aquaporin-4 (AQP-4), 3-nitrotyrosine (3-NT), and endothelin 1 receptor A (ETRA) expression in frontal cortex and hippocampus one day following exposures. IJV compression attenuated this BOP-induced increase in 3-NT in cortex and ameliorated the upregulation of AQP-4 in hippocampus. These results suggest that elevated intracranial pressure and intracerebral volume have neuroprotective potential in blast-induced TBI.
Collapse
|
33
|
von Leden RE, Yauger YJ, Khayrullina G, Byrnes KR. Central Nervous System Injury and Nicotinamide Adenine Dinucleotide Phosphate Oxidase: Oxidative Stress and Therapeutic Targets. J Neurotrauma 2017; 34:755-764. [PMID: 27267366 PMCID: PMC5335782 DOI: 10.1089/neu.2016.4486] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Injury to the central nervous system (CNS) includes both traumatic brain and spinal cord injury (TBI and SCI, respectively). These injuries, which are heterogeneous and, therefore, difficult to treat, result in long-lasting functional, cognitive, and behavioral deficits. Severity of injury is determined by multiple factors, and is largely mediated by the activity of the CNS inflammatory system, including the primary CNS immune cells, microglia. The nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) family of enzymes is a primary source of reactive oxygen species (ROS), key inflammatory mediators after CNS injury. ROS play a central role in inflammation, contributing to cytokine translation and release, microglial polarization and activation, and clearance of damaged tissue. NOX has been suggested as a potential therapeutic target in CNS trauma, as inhibition of this enzyme family modulates inflammatory cell response and ROS production. The purpose of this review is to understand how the different NOX enzymes function and what role they play in the scope of CNS trauma.
Collapse
Affiliation(s)
| | - Young J. Yauger
- Neuroscience Program, Uniformed Services University, Bethesda, Maryland
| | - Guzal Khayrullina
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University, Bethesda, Maryland
| | - Kimberly R. Byrnes
- Neuroscience Program, Uniformed Services University, Bethesda, Maryland
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University, Bethesda, Maryland
| |
Collapse
|
34
|
Ma MW, Wang J, Zhang Q, Wang R, Dhandapani KM, Vadlamudi RK, Brann DW. NADPH oxidase in brain injury and neurodegenerative disorders. Mol Neurodegener 2017; 12:7. [PMID: 28095923 PMCID: PMC5240251 DOI: 10.1186/s13024-017-0150-7] [Citation(s) in RCA: 299] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 01/05/2017] [Indexed: 12/11/2022] Open
Abstract
Oxidative stress is a common denominator in the pathology of neurodegenerative disorders such as Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, amyotrophic lateral sclerosis, and multiple sclerosis, as well as in ischemic and traumatic brain injury. The brain is highly vulnerable to oxidative damage due to its high metabolic demand. However, therapies attempting to scavenge free radicals have shown little success. By shifting the focus to inhibit the generation of damaging free radicals, recent studies have identified NADPH oxidase as a major contributor to disease pathology. NADPH oxidase has the primary function to generate free radicals. In particular, there is growing evidence that the isoforms NOX1, NOX2, and NOX4 can be upregulated by a variety of neurodegenerative factors. The majority of recent studies have shown that genetic and pharmacological inhibition of NADPH oxidase enzymes are neuroprotective and able to reduce detrimental aspects of pathology following ischemic and traumatic brain injury, as well as in chronic neurodegenerative disorders. This review aims to summarize evidence supporting the role of NADPH oxidase in the pathology of these neurological disorders, explores pharmacological strategies of targeting this major oxidative stress pathway, and outlines obstacles that need to be overcome for successful translation of these therapies to the clinic.
Collapse
Affiliation(s)
- Merry W Ma
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, 1120 Fifteenth Street, Augusta, GA, 30912, USA
| | - Jing Wang
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, 1120 Fifteenth Street, Augusta, GA, 30912, USA
| | - Quanguang Zhang
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, 1120 Fifteenth Street, Augusta, GA, 30912, USA
| | - Ruimin Wang
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, 1120 Fifteenth Street, Augusta, GA, 30912, USA
| | - Krishnan M Dhandapani
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA.,Department of Neurosurgery, Medical College of Georgia, Augusta University, 1120 Fifteenth Street, Augusta, GA, 30912, USA
| | - Ratna K Vadlamudi
- Department of Obstetrics and Gynecology, University of Texas Health Science Center, 7703 Medical Drive, San Antonio, TX, 78229, USA
| | - Darrell W Brann
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA. .,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, 1120 Fifteenth Street, Augusta, GA, 30912, USA.
| |
Collapse
|
35
|
Lucke-Wold BP, Phillips M, Turner RC, Logsdon AF, Smith KE, Huber JD, Rosen CL, Regele JD. Elucidating the role of compression waves and impact duration for generating mild traumatic brain injury in rats. Brain Inj 2017; 31:98-105. [PMID: 27880054 PMCID: PMC5247354 DOI: 10.1080/02699052.2016.1218547] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND In total, 3.8 million concussions occur each year in the US leading to acute functional deficits, but the underlying histopathologic changes that occur are relatively unknown. In order to improve understanding of acute injury mechanisms, appropriately designed pre-clinical models must be utilized. METHODS The clinical relevance of compression wave injury models revolves around the ability to produce consistent histopathologic deficits. Mild traumatic brain injuries activate similar neuroinflammatory cascades, cell death markers and increases in amyloid precursor protein in both humans and rodents. Humans, however, infrequently succumb to mild traumatic brain injuries and, therefore, the intensity and magnitude of impacts must be inferred. Understanding compression wave properties and mechanical loading could help link the histopathologic deficits seen in rodents to what might be happening in human brains following concussions. RESULTS While the concept of linking duration and intensity of impact to subsequent histopathologic deficits makes sense, numerical modelling of compression waves has not been performed in this context. In this interdisciplinary work, numerical simulations were performed to study the creation of compression waves in an experimental model. CONCLUSION This work was conducted in conjunction with a repetitive compression wave injury paradigm in rats in order to better understand how the wave generation correlates with histopathologic deficits.
Collapse
Affiliation(s)
- Brandon P Lucke-Wold
- a Department of Neurosurgery
- b Center for Neuroscience, School of Medicine , West Virginia University , Morgantown , WV , USA
| | - Michael Phillips
- c Department of Aerospace Engineering , College of Engineering, Iowa State University , Ames , IA , USA
| | | | - Aric F Logsdon
- b Center for Neuroscience, School of Medicine , West Virginia University , Morgantown , WV , USA
- d Department of Pharmaceutical Sciences , School of Pharmacy, West Virginia University , Morgantown , WV , USA
| | - Kelly E Smith
- b Center for Neuroscience, School of Medicine , West Virginia University , Morgantown , WV , USA
- d Department of Pharmaceutical Sciences , School of Pharmacy, West Virginia University , Morgantown , WV , USA
| | - Jason D Huber
- d Department of Pharmaceutical Sciences , School of Pharmacy, West Virginia University , Morgantown , WV , USA
| | | | - Jonathan D Regele
- c Department of Aerospace Engineering , College of Engineering, Iowa State University , Ames , IA , USA
| |
Collapse
|
36
|
Logsdon AF, Lucke-Wold BP, Nguyen L, Matsumoto RR, Turner RC, Rosen CL, Huber JD. Salubrinal reduces oxidative stress, neuroinflammation and impulsive-like behavior in a rodent model of traumatic brain injury. Brain Res 2016; 1643:140-51. [PMID: 27131989 PMCID: PMC5578618 DOI: 10.1016/j.brainres.2016.04.063] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 04/07/2016] [Accepted: 04/26/2016] [Indexed: 02/05/2023]
Abstract
Traumatic brain injury (TBI) is the leading cause of trauma related morbidity in the developed world. TBI has been shown to trigger secondary injury cascades including endoplasmic reticulum (ER) stress, oxidative stress, and neuroinflammation. The link between secondary injury cascades and behavioral outcome following TBI is poorly understood warranting further investigation. Using our validated rodent blast TBI model, we examined the interaction of secondary injury cascades following single injury and how these interactions may contribute to impulsive-like behavior after a clinically relevant repetitive TBI paradigm. We targeted these secondary pathways acutely following single injury with the cellular stress modulator, salubrinal (SAL). We examined the neuroprotective effects of SAL administration on significantly reducing ER stress: janus-N-terminal kinase (JNK) phosphorylation and C/EBP homology protein (CHOP), oxidative stress: superoxide and carbonyls, and neuroinflammation: nuclear factor kappa beta (NFκB) activity, inducible nitric oxide synthase (iNOS) protein expression, and pro-inflammatory cytokines at 24h post-TBI. We then used the more clinically relevant repeat injury paradigm and observed elevated NFκB and iNOS activity. These injury cascades were associated with impulsive-like behavior measured on the elevated plus maze. SAL administration attenuated secondary iNOS activity at 72h following repetitive TBI, and most importantly prevented impulsive-like behavior. Overall, these results suggest a link between secondary injury cascades and impulsive-like behavior that can be modulated by SAL administration.
Collapse
Affiliation(s)
- Aric F Logsdon
- Department of Pharmaceutical Sciences, School of Pharmacy, Health Sciences Center, West Virginia University, One Medical Center Drive, Morgantown, WV, United States; Department of Neurosurgery, School of Medicine, West Virginia University, Morgantown, WV, United States; Centers for Neuroscience, School of Medicine, West Virginia University, Morgantown, WV, United States.
| | - Brandon P Lucke-Wold
- Department of Neurosurgery, School of Medicine, West Virginia University, Morgantown, WV, United States; Centers for Neuroscience, School of Medicine, West Virginia University, Morgantown, WV, United States.
| | - Linda Nguyen
- Department of Pharmaceutical Sciences, School of Pharmacy, Health Sciences Center, West Virginia University, One Medical Center Drive, Morgantown, WV, United States.
| | - Rae R Matsumoto
- Dean's Office, College of Pharmacy, Touro University California, Vallejo, CA, United States.
| | - Ryan C Turner
- Department of Neurosurgery, School of Medicine, West Virginia University, Morgantown, WV, United States; Centers for Neuroscience, School of Medicine, West Virginia University, Morgantown, WV, United States.
| | - Charles L Rosen
- Department of Neurosurgery, School of Medicine, West Virginia University, Morgantown, WV, United States; Centers for Neuroscience, School of Medicine, West Virginia University, Morgantown, WV, United States.
| | - Jason D Huber
- Department of Pharmaceutical Sciences, School of Pharmacy, Health Sciences Center, West Virginia University, One Medical Center Drive, Morgantown, WV, United States; Department of Neurosurgery, School of Medicine, West Virginia University, Morgantown, WV, United States; Centers for Neuroscience, School of Medicine, West Virginia University, Morgantown, WV, United States.
| |
Collapse
|
37
|
Turner RC, Lucke-Wold BP, Logsdon AF, Robson MJ, Lee JM, Bailes JE, Dashnaw ML, Huber JD, Petraglia AL, Rosen CL. Modeling Chronic Traumatic Encephalopathy: The Way Forward for Future Discovery. Front Neurol 2015; 6:223. [PMID: 26579067 PMCID: PMC4620695 DOI: 10.3389/fneur.2015.00223] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 10/09/2015] [Indexed: 02/05/2023] Open
Abstract
Despite the extensive media coverage associated with the diagnosis of chronic traumatic encephalopathy (CTE), our fundamental understanding of the disease pathophysiology remains in its infancy. Only recently have scientific laboratories and personnel begun to explore CTE pathophysiology through the use of preclinical models of neurotrauma. Some studies have shown the ability to recapitulate some aspects of CTE in rodent models, through the use of various neuropathological, biochemical, and/or behavioral assays. Many questions related to CTE development, however, remain unanswered. These include the role of impact severity, the time interval between impacts, the age at which impacts occur, and the total number of impacts sustained. Other important variables such as the location of impacts, character of impacts, and effect of environment/lifestyle and genetics also warrant further study. In this work, we attempt to address some of these questions by exploring work previously completed using single- and repetitive-injury paradigms. Despite some models producing some deficits similar to CTE symptoms, it is clear that further studies are required to understand the development of neuropathological and neurobehavioral features consistent with CTE-like features in rodents. Specifically, acute and chronic studies are needed that characterize the development of tau-based pathology.
Collapse
Affiliation(s)
- Ryan C. Turner
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Brandon P. Lucke-Wold
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Aric F. Logsdon
- Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, USA
| | - Matthew J. Robson
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - John M. Lee
- Department of Pathology and Laboratory Medicine, NorthShore University Health System, University of Chicago Pritzker School of Medicine, Evanston, IL, USA
| | - Julian E. Bailes
- Department of Neurosurgery, NorthShore University Health System, University of Chicago Pritzker School of Medicine, Evanston, IL, USA
| | - Matthew L. Dashnaw
- Department of Neurosurgery, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Jason D. Huber
- Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, USA
| | | | - Charles L. Rosen
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
| |
Collapse
|
38
|
Turner RC, Lucke-Wold BP, Logsdon AF, Robson MJ, Dashnaw ML, Huang JH, Smith KE, Huber JD, Rosen CL, Petraglia AL. The Quest to Model Chronic Traumatic Encephalopathy: A Multiple Model and Injury Paradigm Experience. Front Neurol 2015; 6:222. [PMID: 26539159 PMCID: PMC4611965 DOI: 10.3389/fneur.2015.00222] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 10/05/2015] [Indexed: 02/05/2023] Open
Abstract
Chronic neurodegeneration following a history of neurotrauma is frequently associated with neuropsychiatric and cognitive symptoms. In order to enhance understanding about the underlying pathophysiology linking neurotrauma to neurodegeneration, a multi-model preclinical approach must be established to account for the different injury paradigms and pathophysiologic mechanisms. We investigated the development of tau pathology and behavioral changes using a multi-model and multi-institutional approach, comparing the preclinical results to tauopathy patterns seen in post-mortem human samples from athletes diagnosed with chronic traumatic encephalopathy (CTE). We utilized a scaled and validated blast-induced traumatic brain injury model in rats and a modified pneumatic closed-head impact model in mice. Tau hyperphosphorylation was evaluated by western blot and immunohistochemistry. Elevated-plus maze and Morris water maze were employed to measure impulsive-like behavior and cognitive deficits respectively. Animals exposed to single blast (~50 PSI reflected peak overpressure) exhibited elevated AT8 immunoreactivity in the contralateral hippocampus at 1 month compared to controls (q = 3.96, p < 0.05). Animals exposed to repeat blast (six blasts over 2 weeks) had increased AT8 (q = 8.12, p < 0.001) and AT270 (q = 4.03, p < 0.05) in the contralateral hippocampus at 1 month post-injury compared to controls. In the modified controlled closed-head impact mouse model, no significant difference in AT8 was seen at 7 days, however a significant elevation was detected at 1 month following injury in the ipsilateral hippocampus compared to control (q = 4.34, p < 0.05). Elevated-plus maze data revealed that rats exposed to single blast (q = 3.53, p < 0.05) and repeat blast (q = 4.21, p < 0.05) spent more time in seconds exploring the open arms compared to controls. Morris water maze testing revealed a significant difference between groups in acquisition times on days 22-27. During the probe trial, single blast (t = 6.44, p < 0.05) and repeat blast (t = 8.00, p < 0.05) rats spent less time in seconds exploring where the platform had been located compared to controls. This study provides a multi-model example of replicating tau and behavioral changes in animals and provides a foundation for future investigation of CTE disease pathophysiology and therapeutic development.
Collapse
Affiliation(s)
- Ryan C. Turner
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Brandon P. Lucke-Wold
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Aric F. Logsdon
- Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, USA
| | - Matthew J. Robson
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Matthew L. Dashnaw
- Department of Neurosurgery, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Jason H. Huang
- Department of Neurosurgery, Baylor Scott and White Health System, Temple, TX, USA
| | - Kelly E. Smith
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, USA
| | - Jason D. Huber
- Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, USA
| | - Charles L. Rosen
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Anthony L. Petraglia
- Division of Neurosurgery, Rochester Regional Health, Rochester, NY, USA
- *Correspondence: Anthony L. Petraglia,
| |
Collapse
|