101
|
Abstract
Traumatic brain injury (TBI) is a serious global public health problem. Survivors of TBI often suffer from long-term disability, which puts a heavy burden on society and families. Unfortunately, up to now, there is no efficacious treatment for TBI patients in clinical practice. As a reducing gas, hydrogen has been shown to be neuroprotective in multiple cerebral disease models; however, its efficacy in TBI remains controversial. In this review, we will focus on the results of hydrogen in experimental TBI, elaborate the potential mechanisms, and put forward for future researches based on our current understanding and views.
Collapse
Affiliation(s)
- Hong-Wei Hu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Zhi-Guo Chen
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Jian-Gang Liu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
102
|
Sheng J, Li T, Zhuang D, Cai S, Yang J, Ding F, Chen X, Tian F, Huang M, Li L, Li K, Chen W. The Monocyte-to-Lymphocyte Ratio at Hospital Admission Is a Novel Predictor for Acute Traumatic Intraparenchymal Hemorrhage Expansion after Cerebral Contusion. Mediators Inflamm 2020; 2020:5483981. [PMID: 33456370 PMCID: PMC7785383 DOI: 10.1155/2020/5483981] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 11/27/2020] [Accepted: 12/12/2020] [Indexed: 02/05/2023] Open
Abstract
PURPOSE To explore the potential of monocyte-to-lymphocyte ratio (MLR) at hospital admission for predicting acute traumatic intraparenchymal hematoma (tICH) expansion in patients with cerebral contusion. Patients and Methods. This multicenter, observational study included patients with available at-hospital admission (baseline) and follow-up computed tomography for volumetric analysis (retrospective development cohort: 1146 patients; prospective validation cohort: 207 patients). Semiautomated software assessed tICH expansion (defined as ≥33% or 5 mL absolute growth). MLR was acquired from routine blood tests upon admission. We constructed two predictive models: basic combined model of clinical and imaging variables and MLR combined model of both MLR and other variables in the basic model. Receiver operating characteristic (ROC) analysis and decision curve analysis (DCA) were used to estimate the performance of MLR for predicting acute tICH expansion. RESULTS MLR was significantly larger in patients with acute tICH expansion compared to those without acute tICH expansion (mean [SD], 1.08 [1.05] vs. 0.59 [0.37], P < 0.001). A nonlinear positive relationship between MLR and the incidence of acute tICH expansion was observed. Multivariate logistic regression indicated MLR as an independent risk factor for acute tICH expansion (odds ratio (OR), 5.88; 95% confidence interval (CI), 4.02-8.61). The power of the multivariate model for predicting acute tICH expansion was substantially improved with the inclusion of MLR (AUC 0.86 vs. AUC 0.74, P < 0.001), as was also observed in an external validation cohort (AUC 0.83 vs. AUC 0.71, P < 0.001). The net benefit of MLR model was higher between threshold probabilities of 20-100% in DCA. For clinical application, a nomogram derived from the multivariate model with MLR was introduced. In addition, MLR was positively associated with 6-month unfavorable outcome. CONCLUSION MLR is a novel predictor for traumatic parenchymatous hematoma expansion. A nomogram derived from the MLR model may provide an easy-to-use tool for predicting acute tICH expansion and promoting the individualized treatment of patients with hemorrhagic cerebral contusion. MLR is associated with long-term outcome after cerebral contusion.
Collapse
Affiliation(s)
- Jiangtao Sheng
- Department of Microbiology and Immunology & Key Immunopathology Laboratory of Guangdong Province, Shantou University Medical College, Shantou, Guangdong, China
| | - Tian Li
- Department of Microbiology and Immunology & Key Immunopathology Laboratory of Guangdong Province, Shantou University Medical College, Shantou, Guangdong, China
| | - Dongzhou Zhuang
- Department of Neurosurgery, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Shirong Cai
- Department of Neurosurgery, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Jinhua Yang
- Department of Neurosurgery, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Faxiu Ding
- Department of Neurosurgery, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Xiaoxuan Chen
- Department of Microbiology and Immunology & Key Immunopathology Laboratory of Guangdong Province, Shantou University Medical College, Shantou, Guangdong, China
| | - Fei Tian
- Department of Neurosurgery, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Mindong Huang
- Department of Neurosurgery, Affiliated Jieyang Hospital of Sun Yat-sen University, Jieyang, Guangdong, China
| | - Lianjie Li
- Department of Neurosurgery, Fuzhou General Hospital of Xiamen University Medical College, Fuzhou, Fujian, China
| | - Kangsheng Li
- Department of Microbiology and Immunology & Key Immunopathology Laboratory of Guangdong Province, Shantou University Medical College, Shantou, Guangdong, China
| | - Weiqiang Chen
- Department of Neurosurgery, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
103
|
Zhang Y, Wang Z, Peng J, Gerner ST, Yin S, Jiang Y. Gut microbiota-brain interaction: An emerging immunotherapy for traumatic brain injury. Exp Neurol 2020; 337:113585. [PMID: 33370556 DOI: 10.1016/j.expneurol.2020.113585] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 12/14/2020] [Accepted: 12/20/2020] [Indexed: 02/06/2023]
Abstract
Individuals suffering from traumatic brain injury (TBI) often experience the activation of the immune system, resulting in declines in cognitive and neurological function after brain injury. Despite decades of efforts, approaches for clinically effective treatment are sparse. Evidence on the association between current therapeutic strategies and clinical outcomes after TBI is limited to poorly understood mechanisms. For decades, an increasing number of studies suggest that the gut-brain axis (GBA), a bidirectional communication system between the central nervous system (CNS) and the gastrointestinal tract, plays a critical role in systemic immune response following neurological diseases. In this review, we detail current knowledge of the immune pathologies of GBA after TBI. These processes may provide a new therapeutic target and rehabilitation strategy developed and used in clinical treatment of TBI patients.
Collapse
Affiliation(s)
- Yuxuan Zhang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Zhaoyang Wang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Jianhua Peng
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Sichuan Clinical Research Center for Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Stefan T Gerner
- Department of Neurology, University Hospital Erlangen-Nuremberg, Erlangen 91054, Germany
| | - Shigang Yin
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.
| | - Yong Jiang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Sichuan Clinical Research Center for Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
104
|
Nieves MD, Furmanski O, Doughty ML. Sensorimotor dysfunction in a mild mouse model of cortical contusion injury without significant neuronal loss is associated with increases in inflammatory proteins with innate but not adaptive immune functions. J Neurosci Res 2020; 99:1533-1549. [PMID: 33269491 DOI: 10.1002/jnr.24766] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/16/2020] [Accepted: 11/01/2020] [Indexed: 12/12/2022]
Abstract
Traumatic brain injury is a leading cause of mortality and morbidity in the United States. Acute trauma to the brain triggers chronic secondary injury mechanisms that contribute to long-term neurological impairment. We have developed a single, unilateral contusion injury model of sensorimotor dysfunction in adult mice. By targeting a topographically defined neurological circuit with a mild impact, we are able to track sustained behavioral deficits in sensorimotor function in the absence of tissue cavitation or neuronal loss in the contused cortex of these mice. Stereological histopathology and multiplex enzyme-linked immunosorbent assay proteomic screening confirm contusion resulted in chronic gliosis and the robust expression of innate immune cytokines and monocyte attractant chemokines IL-1β, IL-5, IL-6, TNFα, CXCL1, CXCL2, CXCL10, CCL2, and CCL3 in the contused cortex. In contrast, the expression of neuroinflammatory proteins with adaptive immune functions was not significantly modulated by injury. Our data support widespread activation of innate but not adaptive immune responses, confirming an association between sensorimotor dysfunction with innate immune activation in the absence of tissue or neuronal loss in our mice.
Collapse
Affiliation(s)
- Michael D Nieves
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Department of Anatomy, Physiology & Genetics, F.E. Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Graduate Program in Neuroscience, F.E. Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Orion Furmanski
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Department of Anatomy, Physiology & Genetics, F.E. Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Martin L Doughty
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Department of Anatomy, Physiology & Genetics, F.E. Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Graduate Program in Neuroscience, F.E. Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| |
Collapse
|
105
|
Chen T, Zheng M, Li Y, Liu S, He L. The role of CCR5 in the protective effect of Esculin on lipopolysaccharide-induced depressive symptom in mice. J Affect Disord 2020; 277:755-764. [PMID: 33065814 DOI: 10.1016/j.jad.2020.08.065] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 07/18/2020] [Accepted: 08/26/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND The purpose of this study was to evaluate whether Esculin could improve the depressive symptom induced by LPS in mice and explore the role of CCR5 in its potential mechanism. METHODS Mice were stimulated with LPS to establish depression model and treated with Esculin. The emotional alteration was assessed via behavior tests. The ELISA assay and western blot analysis were applied to detect the expressions of inflammatory cytokines and correlative proteins. RESULTS As a result, Esculin played a protective role in LPS-induced depressive dysfunction, which was possible through the reduction of M1 microglia, and elevation of M2 microglia by inhibiting TLR4/NF-κB signaling pathway regulated by CCR5. Besides, Esculin led to up-regulation of the CREB/BDNF neuroprotective pathway, and suppression of inflammatory cytokines both in the central and peripheral system. BV2 cells were stimulated with LPS to further elucidate the accordant mechanism in vitro. Molecular docking results suggested that Esc bound to CCR5 at amino acid residues TYR187 and THR105 through hydrogen-bonding. LIMITATIONS Transgenic animals might be useful for the further investigation. CONCLUSIONS From the overall results, we concluded that Esculin might exert a beneficial effect on LPS-induced depression in mice and represent an effective treatment for depression.
Collapse
Affiliation(s)
- Tong Chen
- Department of Pharmacology, China Pharmaceutical University, Longmian Avenue, Nanjing 211198, China.
| | - Menglin Zheng
- Department of Pharmacology, China Pharmaceutical University, Longmian Avenue, Nanjing 211198, China
| | - Yixuan Li
- Department of Pharmacology, China Pharmaceutical University, Longmian Avenue, Nanjing 211198, China
| | - Shengnan Liu
- Department of Pharmacology, China Pharmaceutical University, Longmian Avenue, Nanjing 211198, China
| | - Ling He
- Department of Pharmacology, China Pharmaceutical University, Longmian Avenue, Nanjing 211198, China.
| |
Collapse
|
106
|
Antagonism of Protease-Activated Receptor 4 Protects Against Traumatic Brain Injury by Suppressing Neuroinflammation via Inhibition of Tab2/NF-κB Signaling. Neurosci Bull 2020; 37:242-254. [PMID: 33111257 DOI: 10.1007/s12264-020-00601-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 07/19/2020] [Indexed: 12/23/2022] Open
Abstract
Traumatic brain injury (TBI) triggers the activation of the endogenous coagulation mechanism, and a large amount of thrombin is released to curb uncontrollable bleeding through thrombin receptors, also known as protease-activated receptors (PARs). However, thrombin is one of the most critical factors in secondary brain injury. Thus, the PARs may be effective targets against hemorrhagic brain injury. Since the PAR1 antagonist has an increased bleeding risk in clinical practice, PAR4 blockade has been suggested as a more promising treatment. Here, we explored the expression pattern of PAR4 in the brain of mice after TBI, and explored the effect and possible mechanism of BMS-986120 (BMS), a novel selective and reversible PAR4 antagonist on secondary brain injury. Treatment with BMS protected against TBI in mice. mRNA-seq analysis, Western blot, and qRT-PCR verification in vitro showed that BMS significantly inhibited thrombin-induced inflammation in astrocytes, and suggested that the Tab2/ERK/NF-κB signaling pathway plays a key role in this process. Our findings provide reliable evidence that blocking PAR4 is a safe and effective intervention for TBI, and suggest that BMS has a potential clinical application in the management of TBI.
Collapse
|
107
|
Low Molecular Weight Dextran Sulfate (ILB ®) Administration Restores Brain Energy Metabolism Following Severe Traumatic Brain Injury in the Rat. Antioxidants (Basel) 2020; 9:antiox9090850. [PMID: 32927770 PMCID: PMC7555574 DOI: 10.3390/antiox9090850] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/06/2020] [Accepted: 09/08/2020] [Indexed: 02/07/2023] Open
Abstract
Traumatic brain injury (TBI) is the leading cause of death and disability in people less than 40 years of age in Western countries. Currently, there are no satisfying pharmacological treatments for TBI patients. In this study, we subjected rats to severe TBI (sTBI), testing the effects of a single subcutaneous administration, 30 min post-impact, of a new low molecular weight dextran sulfate, named ILB®, at three different dose levels (1, 5, and 15 mg/kg body weight). A group of control sham-operated animals and one of untreated sTBI rats were used for comparison (each group n = 12). On day 2 or 7 post-sTBI animals were sacrificed and the simultaneous HPLC analysis of energy metabolites, N-acetylaspartate (NAA), oxidized and reduced nicotinic coenzymes, water-soluble antioxidants, and biomarkers of oxidative/nitrosative stress was carried out on deproteinized cerebral homogenates. Compared to untreated sTBI rats, ILB® improved energy metabolism by increasing ATP, ATP/ adenosine diphosphate ratio (ATP/ADP ratio), and triphosphate nucleosides, dose-dependently increased NAA concentrations, protected nicotinic coenzyme levels and their oxidized over reduced ratios, prevented depletion of ascorbate and reduced glutathione (GSH), and decreased oxidative (malondialdehyde formation) and nitrosative stress (nitrite + nitrate production). Although needing further experiments, these data provide the first evidence that a single post-injury injection of a new low molecular weight dextran sulfate (ILB®) has beneficial effects on sTBI metabolic damages. Due to the absence of adverse effects in humans, ILB® represents a promising therapeutic agent for the treatment of sTBI patients.
Collapse
|
108
|
Korotkov A, Puhakka N, Gupta SD, Vuokila N, Broekaart DWM, Anink JJ, Heiskanen M, Karttunen J, van Scheppingen J, Huitinga I, Mills JD, van Vliet EA, Pitkänen A, Aronica E. Increased expression of miR142 and miR155 in glial and immune cells after traumatic brain injury may contribute to neuroinflammation via astrocyte activation. Brain Pathol 2020; 30:897-912. [PMID: 32460356 PMCID: PMC7540383 DOI: 10.1111/bpa.12865] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 02/17/2020] [Accepted: 05/15/2020] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) is associated with the pathological activation of immune-competent cells in the brain, such as astrocytes, microglia and infiltrating immune blood cells, resulting in chronic inflammation and gliosis. This may contribute to the secondary injury after TBI, thus understanding of these processes is crucial for the development of effective treatments of post-traumatic pathologies. MicroRNAs (miRNAs, miRs) are small noncoding RNAs, functioning as posttranscriptional regulators of gene expression. The increased expression of inflammation-associated microRNAs miR155 and miR142 has been reported after TBI in rats. However, expression of these miRNAs in the human brain post-TBI is not studied and their functions are not well understood. Moreover, circulating miR155 and miR142 are candidate biomarkers. Therefore, we characterized miR142 and miR155 expression in the perilesional cortex and plasma of rats that underwent lateral fluid-percussion injury, a model for TBI and in the human perilesional cortex post-TBI. We demonstrated higher miR155 and miR142 expression in the perilesional cortex of rats 2 weeks post-TBI. In plasma, miR155 was associated with proteins and miR142 with extracellular vesicles, however their expression did not change. In the human perilesional cortex miR155 was most prominently expressed by activated astrocytes, whereas miR142 was expressed predominantly by microglia, macrophages and lymphocytes. Pro-inflammatory medium from macrophage-like cells stimulated miR155 expression in astrocytes and overexpression of miR142 in these cells further potentiated a pro-inflammatory state of activated astrocytes. We conclude that miR155 and miR142 promote brain inflammation via astrocyte activation and may be involved in the secondary brain injury after TBI.
Collapse
Affiliation(s)
- Anatoly Korotkov
- Department of (Neuro)Pathology, Amsterdam NeuroscienceAmsterdam UMC, University of AmsterdamMeibergdreef 9Amsterdam1105 AZthe Netherlands
| | - Noora Puhakka
- Department of Neurology, A. I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFI‐70211Finland
| | - Shalini Das Gupta
- Department of Neurology, A. I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFI‐70211Finland
| | - Niina Vuokila
- Department of Neurology, A. I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFI‐70211Finland
| | - Diede W. M. Broekaart
- Department of (Neuro)Pathology, Amsterdam NeuroscienceAmsterdam UMC, University of AmsterdamMeibergdreef 9Amsterdam1105 AZthe Netherlands
| | - Jasper J. Anink
- Department of (Neuro)Pathology, Amsterdam NeuroscienceAmsterdam UMC, University of AmsterdamMeibergdreef 9Amsterdam1105 AZthe Netherlands
| | - Mette Heiskanen
- Department of Neurology, A. I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFI‐70211Finland
| | - Jenni Karttunen
- Department of Neurology, A. I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFI‐70211Finland
| | - Jackelien van Scheppingen
- Department of (Neuro)Pathology, Amsterdam NeuroscienceAmsterdam UMC, University of AmsterdamMeibergdreef 9Amsterdam1105 AZthe Netherlands
- Department of NeuroimmunologyNetherlands Institute for NeuroscienceMeibergdreef 47Amsterdam1105 BAthe Netherlands
| | - Inge Huitinga
- Department of NeuroimmunologyNetherlands Institute for NeuroscienceMeibergdreef 47Amsterdam1105 BAthe Netherlands
| | - James D. Mills
- Department of (Neuro)Pathology, Amsterdam NeuroscienceAmsterdam UMC, University of AmsterdamMeibergdreef 9Amsterdam1105 AZthe Netherlands
| | - Erwin A. van Vliet
- Department of (Neuro)Pathology, Amsterdam NeuroscienceAmsterdam UMC, University of AmsterdamMeibergdreef 9Amsterdam1105 AZthe Netherlands
- Swammerdam Institute for Life Sciences, Center for NeuroscienceUniversity of AmsterdamScience Park 904Amsterdam1090 GEthe Netherlands
| | - Asla Pitkänen
- Department of Neurology, A. I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFI‐70211Finland
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Amsterdam NeuroscienceAmsterdam UMC, University of AmsterdamMeibergdreef 9Amsterdam1105 AZthe Netherlands
- Stichting Epilepsie Instellingen Nederland (SEIN)Heemstedethe Netherlands
| |
Collapse
|
109
|
Green TRF, Ortiz JB, Wonnacott S, Williams RJ, Rowe RK. The Bidirectional Relationship Between Sleep and Inflammation Links Traumatic Brain Injury and Alzheimer's Disease. Front Neurosci 2020; 14:894. [PMID: 32982677 PMCID: PMC7479838 DOI: 10.3389/fnins.2020.00894] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 07/31/2020] [Indexed: 12/18/2022] Open
Abstract
Traumatic brain injury (TBI) and Alzheimer's disease (AD) are diseases during which the fine-tuned autoregulation of the brain is lost. Despite the stark contrast in their causal mechanisms, both TBI and AD are conditions which elicit a neuroinflammatory response that is coupled with physical, cognitive, and affective symptoms. One commonly reported symptom in both TBI and AD patients is disturbed sleep. Sleep is regulated by circadian and homeostatic processes such that pathological inflammation may disrupt the chemical signaling required to maintain a healthy sleep profile. In this way, immune system activation can influence sleep physiology. Conversely, sleep disturbances can exacerbate symptoms or increase the risk of inflammatory/neurodegenerative diseases. Both TBI and AD are worsened by a chronic pro-inflammatory microenvironment which exacerbates symptoms and worsens clinical outcome. Herein, a positive feedback loop of chronic inflammation and sleep disturbances is initiated. In this review, the bidirectional relationship between sleep disturbances and inflammation is discussed, where chronic inflammation associated with TBI and AD can lead to sleep disturbances and exacerbated neuropathology. The role of microglia and cytokines in sleep disturbances associated with these diseases is highlighted. The proposed sleep and inflammation-mediated link between TBI and AD presents an opportunity for a multifaceted approach to clinical intervention.
Collapse
Affiliation(s)
- Tabitha R. F. Green
- BARROW Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ, United States
- Department of Child Health, University of Arizona College of Medicine – Phoenix, Phoenix, AZ, United States
| | - J. Bryce Ortiz
- BARROW Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ, United States
- Department of Child Health, University of Arizona College of Medicine – Phoenix, Phoenix, AZ, United States
| | - Sue Wonnacott
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Robert J. Williams
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Rachel K. Rowe
- BARROW Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ, United States
- Department of Child Health, University of Arizona College of Medicine – Phoenix, Phoenix, AZ, United States
- Phoenix Veteran Affairs Health Care System, Phoenix, AZ, United States
| |
Collapse
|
110
|
Pterostilbene Attenuates Cocultured BV-2 Microglial Inflammation-Mediated SH-SY5Y Neuronal Oxidative Injury via SIRT-1 Signalling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3986348. [PMID: 32831997 PMCID: PMC7426790 DOI: 10.1155/2020/3986348] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 07/02/2020] [Indexed: 12/17/2022]
Abstract
Microglial inflammation plays an important part in the progression of multiple neurological diseases, including neurodegenerative diseases, stroke, depression, and traumatic encephalopathy. Here, we aimed to explore the role of pterostilbene (PTE) in the microglial inflammatory response and subsequent damage of cocultured neural cells and partially explain the underlying mechanisms. In the coculture system of lipopolysaccharide-activated BV-2 microglia and SH-SY5Y neuroblastoma, PTE (only given to BV-2) exhibited protection on SH-SY5Y cells, evidenced by improved SH-SY5Y morphology and viability and LDH release. It also attenuated SH-SY5Y apoptosis and oxidative stress, evidenced by TUNEL and DCFH-DA staining, as well as MDA, SOD, and GSH levels. Moreover, PTE upregulated SIRT-1 expression and suppressed acetylation of NF-κB p65 subunit in BV-2 microglia, thus decreasing the inflammatory factors, including TNF-α and IL-6. Furthermore, the effects above were reversed by SIRT-1 inhibitor EX527. These results suggest that PTE reduces the microglia-mediated inflammatory response and alleviates subsequent neuronal apoptosis and oxidative injury via increasing SIRT-1 expression and inhibiting the NF-κB signalling pathway.
Collapse
|
111
|
Eastman CL, D'Ambrosio R, Ganesh T. Modulating neuroinflammation and oxidative stress to prevent epilepsy and improve outcomes after traumatic brain injury. Neuropharmacology 2020; 172:107907. [PMID: 31837825 PMCID: PMC7274911 DOI: 10.1016/j.neuropharm.2019.107907] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 11/26/2019] [Accepted: 12/05/2019] [Indexed: 12/14/2022]
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability in young adults worldwide. TBI survival is associated with persistent neuropsychiatric and neurological impairments, including posttraumatic epilepsy (PTE). To date, no pharmaceutical treatment has been found to prevent PTE or ameliorate neurological/neuropsychiatric deficits after TBI. Brain trauma results in immediate mechanical damage to brain cells and blood vessels that may never be fully restored given the limited regenerative capacity of brain tissue. This primary insult unleashes cascades of events, prominently including neuroinflammation and massive oxidative stress that evolve over time, expanding the brain injury, but also clearing cellular debris and establishing homeostasis in the region of damage. Accumulating evidence suggests that oxidative stress and neuroinflammatory sequelae of TBI contribute to posttraumatic epileptogenesis. This review will focus on possible roles of reactive oxygen species (ROS), their interactions with neuroinflammation in posttraumatic epileptogenesis, and emerging therapeutic strategies after TBI. We propose that inhibitors of the professional ROS-generating enzymes, the NADPH oxygenases and myeloperoxidase alone, or combined with selective inhibition of cyclooxygenase mediated signaling may have promise for the treatment or prevention of PTE and other sequelae of TBI. This article is part of the special issue entitled 'New Epilepsy Therapies for the 21st Century - From Antiseizure Drugs to Prevention, Modification and Cure of Epilepsy'.
Collapse
Affiliation(s)
- Clifford L Eastman
- Department of Neurological Surgery, 325 Ninth Ave., Seattle, WA, 98104, USA.
| | - Raimondo D'Ambrosio
- Department of Neurological Surgery, 325 Ninth Ave., Seattle, WA, 98104, USA; Regional Epilepsy Center, University of Washington, 325 Ninth Ave., Seattle, WA, 98104, USA
| | - Thota Ganesh
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Rd, Atlanta, GA, 30322, Georgia.
| |
Collapse
|
112
|
Guedes VA, Devoto C, Leete J, Sass D, Acott JD, Mithani S, Gill JM. Extracellular Vesicle Proteins and MicroRNAs as Biomarkers for Traumatic Brain Injury. Front Neurol 2020; 11:663. [PMID: 32765398 PMCID: PMC7378746 DOI: 10.3389/fneur.2020.00663] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 06/02/2020] [Indexed: 12/12/2022] Open
Abstract
Traumatic brain injury (TBI) is a heterogeneous condition, associated with diverse etiologies, clinical presentations and degrees of severity, and may result in chronic neurobehavioral sequelae. The field of TBI biomarkers is rapidly evolving to address the many facets of TBI pathology and improve its clinical management. Recent years have witnessed a marked increase in the number of publications and interest in the role of extracellular vesicles (EVs), which include exosomes, cell signaling, immune responses, and as biomarkers in a number of pathologies. Exosomes have a well-defined lipid bilayer with surface markers that reflect the cell of origin and an aqueous core that contains a variety of biological material including proteins (e.g., cytokines and growth factors) and nucleic acids (e.g., microRNAs). The presence of proteins associated with neurodegenerative changes such as amyloid-β, α-synuclein and phosphorylated tau in exosomes suggests a role in the initiation and propagation of neurological diseases. However, mechanisms of cell communication involving exosomes in the brain and their role in TBI pathology are poorly understood. Exosomes are promising TBI biomarkers as they can cross the blood-brain barrier and can be isolated from peripheral fluids, including serum, saliva, sweat, and urine. Exosomal content is protected from enzymatic degradation by exosome membranes and reflects the internal environment of their cell of origin, offering insights into tissue-specific pathological processes. Challenges in the clinical use of exosomal cargo as biomarkers include difficulty in isolating pure exosomes, variable yields of the isolation processes, quantification of vesicles, and lack of specificity of exosomal markers. Moreover, there is no consensus regarding nomenclature and characteristics of EV subtypes. In this review, we discuss current technical limitations and challenges of using exosomes and other EVs as blood-based biomarkers, highlighting their potential as diagnostic and prognostic tools in TBI.
Collapse
Affiliation(s)
- Vivian A Guedes
- National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, United States
| | - Christina Devoto
- National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, United States
| | - Jacqueline Leete
- National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, United States
| | - Delia Sass
- National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, United States
| | - Jedidiah D Acott
- National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, United States
| | - Sara Mithani
- National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, United States
| | - Jessica M Gill
- National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
113
|
Meier TB, Nitta ME, Teague TK, Nelson LD, McCrea MA, Savitz J. Prospective study of the effects of sport-related concussion on serum kynurenine pathway metabolites. Brain Behav Immun 2020; 87:715-724. [PMID: 32147388 PMCID: PMC7316609 DOI: 10.1016/j.bbi.2020.03.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 02/18/2020] [Accepted: 03/02/2020] [Indexed: 02/07/2023] Open
Abstract
Reports of neurodegenerative and psychiatric disease in former athletes have increased public concern about the acute and chronic effects of sport-related concussions (SRC). The biological factors underlying individual differences in the psychiatric sequalae of SRC and their role in potential long-term negative outcomes have not been determined. One understudied biological consequence of the known inflammatory response to concussion is the activation of a key immunoregulatory pathway, the kynurenine pathway (KP). Activation of the KP produces several neuroactive metabolites that have been associated with psychiatric and neurodegenerative diseases. We tested the hypothesis that SRC results in an elevation of serum KP metabolites with neurotoxic properties (quinolinic acid [QuinA], 3-hydroxykynurenine [3HK]) together with a reduction in the neuroprotective metabolite kynurenic acid (KynA), and that these metabolites would predict post-concussion psychological symptoms. Additionally, because brain injury is thought to prime the immune system, a secondary goal was to test the hypothesis that athletes with acute SRC and a history of prior SRC would have elevated neurotoxic relative to neuroprotective KP metabolites compared to athletes that were concussed for the first time. High school and collegiate football players (N = 1136) were enrolled at a preseason baseline visit that included clinical testing and blood specimen collection. Athletes that suffered a SRC (N = 59) completed follow-up visits within 6-hours (early-acute), at 24-48 h (late-acute) and at 8, 15, and 45 days post-injury. Uninjured contact sport (CC; N = 54) and non-contact sport athletes completed similar visits and served as controls (NCC; N = 30). SRC athletes had significantly elevated psychological symptoms, assessed using the Brief Symptom Inventory-18 (BSI), acutely following injury relative to both control groups. There was a group-by-visit interaction on the ratio of KynA to 3HK in serum, a neuroprotective index, with elevated KynA/3HK in athletes with SRC at the early-acute visit relative to later visits. Importantly, athletes with greater elevation in this neuroprotective index at the early-acute visit reported fewer depressive symptoms at the late-acute visit. Finally, SRC athletes with prior concussion had significantly lower serum KynA/QuinA at all visits compared to SRC athletes with no prior concussion, an effect driven by elevated QuinA in SRC athletes with prior concussion. These results suggest that early-acute activation of the KynA branch of the KP may protect against the development of depressive symptoms following concussion. Furthermore, they highlight the potential of serum QuinA as a biomarker for repetitive head injury and provide insight into possible mechanisms linking prior concussion with subsequent injury.
Collapse
Affiliation(s)
- Timothy B. Meier
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI,Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI,Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI,Corresponding Author: Timothy B. Meier, PhD, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, Phone: 414-955-7310, Fax: 414-955-0115,
| | - Morgan E. Nitta
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI,Department of Psychology, Marquette University, Milwaukee, WI
| | - T. Kent Teague
- Department of Surgery, University of Oklahoma School of Community Medicine, Tulsa, OK,Department of Psychiatry, University of Oklahoma School of Community Medicine, Tulsa, OK,Department of Pharmaceutical Sciences, University of Oklahoma College of Pharmacy, Tulsa, OK.,Department of Biochemistry and Microbiology, Oklahoma State University Center for Health Sciences, Tulsa, OK
| | - Lindsay D. Nelson
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI,Department of Neurology, Medical College of Wisconsin, Milwaukee, WI
| | - Michael A. McCrea
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI,Department of Neurology, Medical College of Wisconsin, Milwaukee, WI
| | - Jonathan Savitz
- Laureate Institute for Brain Research, Tulsa, OK,Oxley College of Health Sciences, The University of Tulsa, Tulsa OK
| |
Collapse
|
114
|
Paudel YN, Angelopoulou E, Piperi C, Othman I, Shaikh MF. HMGB1-Mediated Neuroinflammatory Responses in Brain Injuries: Potential Mechanisms and Therapeutic Opportunities. Int J Mol Sci 2020; 21:ijms21134609. [PMID: 32610502 PMCID: PMC7370155 DOI: 10.3390/ijms21134609] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/17/2020] [Accepted: 06/23/2020] [Indexed: 12/14/2022] Open
Abstract
Brain injuries are devastating conditions, representing a global cause of mortality and morbidity, with no effective treatment to date. Increased evidence supports the role of neuroinflammation in driving several forms of brain injuries. High mobility group box 1 (HMGB1) protein is a pro-inflammatory-like cytokine with an initiator role in neuroinflammation that has been implicated in Traumatic brain injury (TBI) as well as in early brain injury (EBI) after subarachnoid hemorrhage (SAH). Herein, we discuss the implication of HMGB1-induced neuroinflammatory responses in these brain injuries, mediated through binding to the receptor for advanced glycation end products (RAGE), toll-like receptor4 (TLR4) and other inflammatory mediators. Moreover, we provide evidence on the biomarker potential of HMGB1 and the significance of its nucleocytoplasmic translocation during brain injuries along with the promising neuroprotective effects observed upon HMGB1 inhibition/neutralization in TBI and EBI induced by SAH. Overall, this review addresses the current advances on neuroinflammation driven by HMGB1 in brain injuries indicating a future treatment opportunity that may overcome current therapeutic gaps.
Collapse
Affiliation(s)
- Yam Nath Paudel
- Neuropharmacology Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor 47500, Malaysia;
- Correspondence: (Y.N.P.); (C.P.); (M.F.S.); Tel.: +6-01-8396-0285 (Y.N.P.); +30-210-746-2610 (C.P.); +60-3-5514-6000 (ext. 44483) or +60-3-5514-4483 (M.F.S.); Fax: +30-210-746-2703 (C.P.); +601-4283-2410 (M.F.S.)
| | - Efthalia Angelopoulou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
- Correspondence: (Y.N.P.); (C.P.); (M.F.S.); Tel.: +6-01-8396-0285 (Y.N.P.); +30-210-746-2610 (C.P.); +60-3-5514-6000 (ext. 44483) or +60-3-5514-4483 (M.F.S.); Fax: +30-210-746-2703 (C.P.); +601-4283-2410 (M.F.S.)
| | - Iekhsan Othman
- Neuropharmacology Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor 47500, Malaysia;
| | - Mohd. Farooq Shaikh
- Neuropharmacology Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor 47500, Malaysia;
- Correspondence: (Y.N.P.); (C.P.); (M.F.S.); Tel.: +6-01-8396-0285 (Y.N.P.); +30-210-746-2610 (C.P.); +60-3-5514-6000 (ext. 44483) or +60-3-5514-4483 (M.F.S.); Fax: +30-210-746-2703 (C.P.); +601-4283-2410 (M.F.S.)
| |
Collapse
|
115
|
Mukherjee S, Arisi GM, Mims K, Hollingsworth G, O'Neil K, Shapiro LA. Neuroinflammatory mechanisms of post-traumatic epilepsy. J Neuroinflammation 2020; 17:193. [PMID: 32552898 PMCID: PMC7301453 DOI: 10.1186/s12974-020-01854-w] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 05/25/2020] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) occurs in as many as 64-74 million people worldwide each year and often results in one or more post-traumatic syndromes, including depression, cognitive, emotional, and behavioral deficits. TBI can also increase seizure susceptibility, as well as increase the incidence of epilepsy, a phenomenon known as post-traumatic epilepsy (PTE). Injury type and severity appear to partially predict PTE susceptibility. However, a complete mechanistic understanding of risk factors for PTE is incomplete. MAIN BODY From the earliest days of modern neuroscience, to the present day, accumulating evidence supports a significant role for neuroinflammation in the post-traumatic epileptogenic progression. Notably, substantial evidence indicates a role for astrocytes, microglia, chemokines, and cytokines in PTE progression. Although each of these mechanistic components is discussed in separate sections, it is highly likely that it is the totality of cellular and neuroinflammatory interactions that ultimately contribute to the epileptogenic progression following TBI. CONCLUSION This comprehensive review focuses on the neuroinflammatory milieu and explores putative mechanisms involved in the epileptogenic progression from TBI to increased seizure-susceptibility and the development of PTE.
Collapse
Affiliation(s)
- Sanjib Mukherjee
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX, USA
| | - Gabriel M Arisi
- Department of Physiology, Federal University of Sao Paulo - Escola Paulista de Medicina, Sao Paulo, Brazil.
| | - Kaley Mims
- Texas A&M University, College Station, TX, USA
| | | | | | - Lee A Shapiro
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX, USA.
| |
Collapse
|
116
|
Zhu Z, Chuckowree JA, Musgrove R, Dickson TC, Blizzard CA. The pathologic outcomes and efficacy of epothilone treatment following traumatic brain injury is determined by age. Neurobiol Aging 2020; 93:85-96. [PMID: 32480164 DOI: 10.1016/j.neurobiolaging.2020.03.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/27/2020] [Accepted: 03/28/2020] [Indexed: 02/05/2023]
Abstract
Traumatic brain injury (TBI) can affect individuals at any age, with the potential of causing lasting neurologic consequences. The lack of effective therapeutic solutions and recommendations for patients that acquire a TBI can be attributed, at least in part, to an inability to confidently predict long-term outcomes following TBI, and how the response of the brain differs across the life span. The purpose of this study was to determine how age specifically affects TBI outcomes in a preclinical model. Male Thy1-YFPH mice, that express yellow fluorescent protein in the cytosol of a subset of Layer V pyramidal neurons in the neocortex, were subjected to a lateral fluid percussion injury over the right parietal cortex at distinct time points throughout the life span (1.5, 3, and 12 months of age). We found that the degree of neuronal injury, astrogliosis, and microglial activation differed depending on the age of the animal when the injury occurred. Furthermore, age affected the initial injury response and how it resolved over time. Using the microtubule stabilizing agent Epothilone D, to potentially protect against these pathologic outcomes, we found that the neuronal response was different depending on age. This study clearly shows that age must be taken into account in neurologic studies and preclinical trials involving TBI, and that future therapeutic interventions must be tailored to age.
Collapse
Affiliation(s)
- Zhendan Zhu
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Jyoti A Chuckowree
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Ruth Musgrove
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Tracey C Dickson
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Catherine A Blizzard
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia.
| |
Collapse
|
117
|
Van Eldik LJ, Sawaki L, Bowen K, Laskowitz DT, Noveck RJ, Hauser B, Jordan L, Spears TG, Wu H, Watt K, Raja S, Roy SM, Watterson DM, Guptill JT. First-in-Human Studies of MW01-6-189WH, a Brain-Penetrant, Antineuroinflammatory Small-Molecule Drug Candidate: Phase 1 Safety, Tolerability, Pharmacokinetic, and Pharmacodynamic Studies in Healthy Adult Volunteers. Clin Pharmacol Drug Dev 2020; 10:131-143. [PMID: 32255549 PMCID: PMC7541708 DOI: 10.1002/cpdd.795] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 02/18/2020] [Indexed: 11/18/2022]
Abstract
MW01‐6‐189WH (MW189) is a novel central nervous system–penetrant small‐molecule drug candidate that selectively attenuates stressor‐induced proinflammatory cytokine overproduction and is efficacious in intracerebral hemorrhage and traumatic brain injury animal models. We report first‐in‐human, randomized, double‐blind, placebo‐controlled phase 1 studies to evaluate the safety, tolerability, and pharmacokinetics (PK) of single and multiple ascending intravenous doses of MW189 in healthy adult volunteers. MW189 was safe and well tolerated in single and multiple doses up to 0.25 mg/kg, with no clinically significant concerns. The most common drug‐related treatment‐emergent adverse event was infusion‐site reactions, likely related to drug solution acidity. No clinically concerning changes were seen in vital signs, electrocardiograms, physical or neurological examinations, or safety laboratory results. PK analysis showed dose‐proportional increases in plasma concentrations of MW189 after single or multiple doses, with approximately linear kinetics and no significant drug accumulation. Steady state was achieved by dose 3 for all dosing cohorts. A pilot pharmacodynamic study administering low‐dose endotoxin to induce a systemic inflammatory response was done to evaluate the effects of a single intravenous dose of MW189 on plasma cytokine levels. MW189 treatment resulted in lower levels of the proinflammatory cytokine TNF‐α and higher levels of the anti‐inflammatory cytokine IL‐10 compared with placebo treatment. The outcomes are consistent with the pharmacological mechanism of MW189. Overall, the safety profile, PK properties, and pharmacodynamic effect support further development of MW189 for patients with acute brain injury.
Collapse
Affiliation(s)
- Linda J. Van Eldik
- Sanders‐Brown Center on AgingUniversity of KentuckyLexingtonKentuckyUSA
- Department of NeuroscienceUniversity of KentuckyLexingtonKentuckyUSA
- Spinal Cord and Brain Injury Research CenterUniversity of KentuckyLexingtonKentuckyUSA
| | - Lumy Sawaki
- Department of Physical Medicine & RehabilitationUniversity of KentuckyLexingtonKentuckyUSA
| | - Karen Bowen
- Bluegrass Research Consultants, Inc.VersaillesKentuckyUSA
| | - Daniel T. Laskowitz
- Department of NeurologyDuke UniversityDurhamNorth CarolinaUSA
- Duke Clinical Research InstituteDurhamNorth CarolinaUSA
| | | | - Byron Hauser
- Duke Early Phase Clinical Research UnitDurhamNorth CarolinaUSA
| | - Lynn Jordan
- Duke Early Phase Clinical Research UnitDurhamNorth CarolinaUSA
| | | | - Huali Wu
- Duke Clinical Research InstituteDurhamNorth CarolinaUSA
| | - Kevin Watt
- Duke Clinical Research InstituteDurhamNorth CarolinaUSA
| | - Shruti Raja
- Department of NeurologyDuke UniversityDurhamNorth CarolinaUSA
- Duke Early Phase Clinical Research UnitDurhamNorth CarolinaUSA
| | | | | | - Jeffrey T. Guptill
- Department of NeurologyDuke UniversityDurhamNorth CarolinaUSA
- Duke Clinical Research InstituteDurhamNorth CarolinaUSA
- Duke Early Phase Clinical Research UnitDurhamNorth CarolinaUSA
| |
Collapse
|
118
|
Hellewell SC, Nguyen VPB, Jayasena RN, Welton T, Grieve SM. Characteristic patterns of white matter tract injury in sport-related concussion: An image based meta-analysis. Neuroimage Clin 2020; 26:102253. [PMID: 32278315 PMCID: PMC7152675 DOI: 10.1016/j.nicl.2020.102253] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/21/2020] [Accepted: 03/20/2020] [Indexed: 12/16/2022]
Abstract
Sports-related concussion (SRC) is sustained by millions of people per year, yet the spatiotemporal patterns of white matter (WM) injury remain poorly understood. Several SRC studies have implemented the standardised approach Tract-Based Spatial Statistics (TBSS). The aim of this image-based meta-analysis was to identify consensus patterns of SRC-related WM injury across TBSS studies. We included studies comparing the diffusion MRI measurement fractional anisotropy (FA) in SRC or subconcussive injury vs. controls using TBSS, as FA is the most frequently examined diffusion tensor imaging metric. Authors of eligible studies were contacted to request unthresholded statistical map outputs from TBSS, and image-based meta-analyses were performed using Seed-Based d-Mapping. Eight studies contributed to our meta-analyses, comprising 174 SRC or subconcussive injury participants and 160 controls. Our primary meta-analysis (n = 8), encompassing subjects with acute SRC (n = 2), chronic SRC (n = 4) and subconcussive injuries (n = 2) revealed dominant bilateral increased FA in the superior longitudinal fasciculus (SLF) and internal capsule. Subconcussive injury was associated with small clusters of increased and decreased FA in the arcuate fasciculus compared to control. In acute SRC, we found diffuse foci of raised FA at WM/grey matter border-zone associated with the bilateral SLF and right inferior fronto-occipital fasciculus. In contrast, chronic SRC had a pattern of deep WM alteration, asymmetrically located in the right optic radiations and arcuate fasciculus. Our findings represent the most powerful analysis of TBSS data in SRC, supporting the use of this approach to analyse diffusion data. TBSS is sensitive to WM abnormalities resulting from SRC or subconcussive injury over a range of temporal and clinical scenarios. Our data show spatially concordant patterns of WM injury unique to subconcussive, acute and chronic phases, highlighting the future utility of diffusion MRI for concussion diagnosis.
Collapse
Affiliation(s)
- Sarah C Hellewell
- Sydney Translational Imaging Laboratory, Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Vy P B Nguyen
- Sydney Translational Imaging Laboratory, Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Ruchira N Jayasena
- Sydney Translational Imaging Laboratory, Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Thomas Welton
- Sydney Translational Imaging Laboratory, Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Stuart M Grieve
- Sydney Translational Imaging Laboratory, Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2006, Australia; Department of Radiology, Royal Prince Alfred Hospital, Camperdown, Sydney, NSW, Australia.
| |
Collapse
|
119
|
Cordaro M, Cuzzocrea S, Crupi R. An Update of Palmitoylethanolamide and Luteolin Effects in Preclinical and Clinical Studies of Neuroinflammatory Events. Antioxidants (Basel) 2020; 9:antiox9030216. [PMID: 32150935 PMCID: PMC7139331 DOI: 10.3390/antiox9030216] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 02/06/2023] Open
Abstract
The inflammation process represents of a dynamic series of phenomena that manifest themselves with an intense vascular reaction. Neuroinflammation is a reply from the central nervous system (CNS) and the peripheral nervous system (PNS) to a changed homeostasis. There are two cell systems that mediate this process: the glia of the CNS and the lymphocites, monocytes, and macrophages of the hematopoietic system. In both the peripheral and central nervous systems, neuroinflammation plays an important role in the pathogenesis of neurodegenerative diseases, such as Parkinson’s and Alzheimer’s diseases, and in neuropsychiatric illnesses, such as depression and autism spectrum disorders. The resolution of neuroinflammation is a process that allows for inflamed tissues to return to homeostasis. In this process the important players are represented by lipid mediators. Among the naturally occurring lipid signaling molecules, a prominent role is played by the N-acylethanolamines, namely N-arachidonoylethanolamine and its congener N-palmitoylethanolamine, which is also named palmitoylethanolamide or PEA. PEA possesses a powerful neuroprotective and anti-inflammatory power but has no antioxidant effects per se. For this reason, its co-ultramicronization with the flavonoid luteolin is more efficacious than either molecule alone. Inhibiting or modulating the enzymatic breakdown of PEA represents a complementary therapeutic approach to treating neuroinflammation. The aim of this review is to discuss the role of ultramicronized PEA and co-ultramicronized PEA with luteolin in several neurological diseases using preclinical and clinical approaches.
Collapse
Affiliation(s)
- Marika Cordaro
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Via Consolare Valeria 1, 98100 Messina, Italy;
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Via F. Stagno D’Alcontres 31, 98166 Messina, Italy;
- Department of Pharmacology and Physiology, Saint Louis University, St. Louis, MO 63103, USA
- Correspondence: ; Tel.: +390-906-765-208
| | - Rosalia Crupi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Via F. Stagno D’Alcontres 31, 98166 Messina, Italy;
| |
Collapse
|
120
|
Collins JM, Woodhouse A, Bye N, Vickers JC, King AE, Ziebell JM. Pathological Links between Traumatic Brain Injury and Dementia: Australian Pre-Clinical Research. J Neurotrauma 2020; 37:782-791. [PMID: 32046575 DOI: 10.1089/neu.2019.6906] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) can cause persistent cognitive changes and ongoing neurodegeneration in the brain. Accumulating epidemiological and pathological evidence implicates TBI in the development of Alzheimer's disease, the most common cause of dementia. Further, the TBI-induced form of dementia, called chronic traumatic encephalopathy, shares many pathological hallmarks present in multiple different diseases which cause dementia. The inflammatory and neuritic responses to TBI and dementia overlap, indicating that they may share common pathological mechanisms and that TBI may ultimately cause a pathological cascade culminating in the development of dementia. This review explores Australian pre-clinical research investigating the pathological links between TBI and dementia.
Collapse
Affiliation(s)
- Jessica M Collins
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Adele Woodhouse
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Nicole Bye
- School of Pharmacy, and College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - James C Vickers
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia.,School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Anna E King
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Jenna M Ziebell
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
121
|
Chen Y, Qin C, Huang J, Tang X, Liu C, Huang K, Xu J, Guo G, Tong A, Zhou L. The role of astrocytes in oxidative stress of central nervous system: A mixed blessing. Cell Prolif 2020; 53:e12781. [PMID: 32035016 PMCID: PMC7106951 DOI: 10.1111/cpr.12781] [Citation(s) in RCA: 150] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 12/17/2019] [Accepted: 01/20/2020] [Indexed: 02/05/2023] Open
Abstract
Central nervous system (CNS) maintains a high level of metabolism, which leads to the generation of large amounts of free radicals, and it is also one of the most vulnerable organs to oxidative stress. Emerging evidences have shown that, as the key homeostatic cells in CNS, astrocytes are deeply involved in multiple aspects of CNS function including oxidative stress regulation. Besides, the redox level in CNS can in turn affect astrocytes in morphology and function. The complex and multiple roles of astrocytes indicate that their correct performance is crucial for the normal functioning of the CNS, and its dysfunction may result in the occurrence and progression of various neurological disorders. To date, the influence of astrocytes in CNS oxidative stress is rarely reviewed. Therefore, in this review we sum up the roles of astrocytes in redox regulation and the corresponding mechanisms under both normal and different pathological conditions.
Collapse
Affiliation(s)
- Yaxing Chen
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, China
| | - Chen Qin
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, China
| | - Jianhan Huang
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Tang
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, China
| | - Chang Liu
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, China
| | - Keru Huang
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, China
| | - Jianguo Xu
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, China
| | - Gang Guo
- State Key Laboratory of Biotherapy, West China Medical School, Sichuan University, Chengdu, China
| | - Aiping Tong
- State Key Laboratory of Biotherapy, West China Medical School, Sichuan University, Chengdu, China
| | - Liangxue Zhou
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
122
|
Liu R, Wang J, Liang S, Zhang G, Yang X. Role of NKCC1 and KCC2 in Epilepsy: From Expression to Function. Front Neurol 2020; 10:1407. [PMID: 32010056 PMCID: PMC6978738 DOI: 10.3389/fneur.2019.01407] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 12/23/2019] [Indexed: 01/21/2023] Open
Abstract
As a main inhibitory neurotransmitter in the central nervous system, γ-aminobutyric acid (GABA) activates chloride-permeable GABAa receptors (GABAa Rs) and induces chloride ion (Cl−) flow, which relies on the intracellular chloride concentration ([Cl−]i) of the postsynaptic neuron. The Na-K-2Cl cotransporter isoform 1 (NKCC1) and the K-Cl cotransporter isoform 2 (KCC2) are two main cation-chloride cotransporters (CCCs) that have been implicated in human epilepsy. NKCC1 and KCC2 reset [Cl−]i by accumulating and extruding Cl−, respectively. Previous studies have shown that the profile of NKCC1 and KCC2 in neonatal neurons may reappear in mature neurons under some pathophysiological conditions, such as epilepsy. Although increasing studies focusing on the expression of NKCC1 and KCC2 have suggested that impaired chloride plasticity may be closely related to epilepsy, additional neuroelectrophysiological research aimed at studying the functions of NKCC1 and KCC2 are needed to understand the exact mechanism by which they induce epileptogenesis. In this review, we aim to briefly summarize the current researches surrounding the expression and function of NKCC1 and KCC2 in epileptogenesis and its implications on the treatment of epilepsy. We will also explore the potential for NKCC1 and KCC2 to be therapeutic targets for the development of novel antiepileptic drugs.
Collapse
Affiliation(s)
- Ru Liu
- Neuroelectrophysiological Laboratory, Xuanwu Hospital, Capital Medical University, Beijing, China.,Center of Epilepsy, Center for Brain Disorders Research, Capital Medical University, Beijing, China.,Center of Epilepsy, Beijing Institute of Brain Disorders, Beijing, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Junling Wang
- Neuroelectrophysiological Laboratory, Xuanwu Hospital, Capital Medical University, Beijing, China.,Center of Epilepsy, Center for Brain Disorders Research, Capital Medical University, Beijing, China.,Center of Epilepsy, Beijing Institute of Brain Disorders, Beijing, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Shuli Liang
- Department of Functional Neurosurgery, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Guojun Zhang
- Department of Functional Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiaofeng Yang
- Neuroelectrophysiological Laboratory, Xuanwu Hospital, Capital Medical University, Beijing, China.,Center of Epilepsy, Center for Brain Disorders Research, Capital Medical University, Beijing, China.,Center of Epilepsy, Beijing Institute of Brain Disorders, Beijing, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| |
Collapse
|
123
|
Rusiecki J, Levin LI, Wang L, Byrne C, Krishnamurthy J, Chen L, Galdzicki Z, French LM. Blast traumatic brain injury and serum inflammatory cytokines: a repeated measures case-control study among U.S. military service members. J Neuroinflammation 2020; 17:20. [PMID: 31931830 PMCID: PMC6958571 DOI: 10.1186/s12974-019-1624-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 10/28/2019] [Indexed: 12/11/2022] Open
Abstract
Background There is a paucity of human data on exposure to blast traumatic brain injury (bTBI) and the corresponding systemic cytokine immune response at later time points (i.e., months, years) post-injury. Methods We conducted a repeated measures, case-control study, examining associations of serum levels of pro- and anti-inflammatory cytokines, measured both pre- and post-deployment with having mild and moderate/severe bTBI. Utilizing serum from the Department of Defense Serum Repository cytokines were measured via an ELISA-based array for 15 cytokines. We compared pre- vs. post-levels among mild cases, moderate/severe cases, and controls and carried out case-control comparisons, using paired t- tests and generalized linear models. Results The average time between bTBI and post-deployment/bTBI serum among cases was 315.8 days. From pre- to post-deployment/bTBI, levels of interleukin 8 (IL-8) were decreased among both mild cases (μ = − 83.43 pg/ml; s.e. = 21.66) and moderate/severe cases (μ = − 107.67 pg/ml; s.e. = 28.74 pg/ml), while levels increased among controls (μ = 32.86 pg/ml; s.e. = 30.29). The same pattern occurred for matrix metallopeptidase 3 (MMP3), with levels decreasing for moderate/severe cases (μ = − 3369.24 pg/ml; s.e. = 1701.68) and increasing for controls (μ = 1859.60 pg/ml; s.e. = 1737.51) from pre- to post-deployment/bTBI. Evidence was also suggestive of case-control differences, from pre- to post-deployment/bTBI for interleukin 1 alpha (IL-1α), interleukin 4 (IL-4), and interleukin 6 (IL-6) among moderate/severe cases. Conclusion The findings of this longitudinal study indicate that in the chronic phase of bTBI, levels of IL-8 and MMP3 may be substantially lower than pre-injury. These results need confirmation in other studies, potentially those that account for treatment differences, which was not possible in our study.
Collapse
Affiliation(s)
- Jennifer Rusiecki
- Department of Preventive Medicine and Biostatistics, F. Edward Hébert School of Medicine, Uniformed Services University, Bethesda, MD, USA.
| | - Lynn I Levin
- Statistics and Epidemiology Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Li Wang
- Department of Preventive Medicine and Biostatistics, F. Edward Hébert School of Medicine, Uniformed Services University, Bethesda, MD, USA
| | - Celia Byrne
- Department of Preventive Medicine and Biostatistics, F. Edward Hébert School of Medicine, Uniformed Services University, Bethesda, MD, USA
| | - Jayasree Krishnamurthy
- Department of Pediatrics, F. Edward Hébert School of Medicine, Uniformed Services University, Bethesda, MD, USA
| | - Ligong Chen
- Department of Preventive Medicine and Biostatistics, F. Edward Hébert School of Medicine, Uniformed Services University, Bethesda, MD, USA
| | - Zygmunt Galdzicki
- Department of Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University, Bethesda, MD, USA
| | - Louis M French
- National Intrepid Center of Excellence, Walter Reed National Military Medical Center, Bethesda, MD, USA
| |
Collapse
|
124
|
Fesharaki-Zadeh A, Miyauchi JT, St. Laurent-Arriot K, Tsirka SE, Bergold PJ. Increased Behavioral Deficits and Inflammation in a Mouse Model of Co-Morbid Traumatic Brain Injury and Post-Traumatic Stress Disorder. ASN Neuro 2020; 12:1759091420979567. [PMID: 33342261 PMCID: PMC7755938 DOI: 10.1177/1759091420979567] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/03/2020] [Accepted: 11/08/2020] [Indexed: 01/21/2023] Open
Abstract
Comorbid post-traumatic stress disorder with traumatic brain injury (TBI) produce more severe affective and cognitive deficits than PTSD or TBI alone. Both PTSD and TBI produce long-lasting neuroinflammation, which may be a key underlying mechanism of the deficits observed in co-morbid TBI/PTSD. We developed a model of co-morbid TBI/PTSD by combining the closed head (CHI) model of TBI with the chronic variable stress (CVS) model of PTSD and examined multiple behavioral and neuroinflammatory outcomes. Male C57/Bl6 mice received sham treatment, CHI, CVS, CHI then CVS (CHI → CVS) or CVS then CHI (CVS → CHI). The CVS → CHI group had deficits in Barnes maze or active place avoidance not seen in the other groups. The CVS → CHI, CVS and CHI → CVS groups displayed increased basal anxiety level, based on performance on elevated plus maze. The CVS → CHI had impaired performance on Barnes Maze, and Active Place Avoidance. These performance deficits were strongly correlated with increased hippocampal Iba-1 level an indication of activated MP/MG. These data suggest that greater cognitive deficits in the CVS → CHI group were due to increased inflammation. The increased deficits and neuroinflammation in the CVS → CHI group suggest that the order by which a subject experiences TBI and PTSD is a major determinant of the outcome of brain injury in co-morbid TBI/PTSD.
Collapse
Affiliation(s)
- Arman Fesharaki-Zadeh
- Department of Psychiatry, State University of New York, Downstate Medical Center, Brooklyn, New York
| | - Jeremy T. Miyauchi
- Department of Physiology, State University of New York, Downstate Medical Center, Brooklyn, New York
| | - Karrah St. Laurent-Arriot
- Department of Pharmacology, State University of New York, Downstate Medical Center, Brooklyn, New York
| | - Stella E. Tsirka
- Department of Physiology, State University of New York, Downstate Medical Center, Brooklyn, New York
| | - Peter J. Bergold
- Department of Pharmacology, State University of New York, Downstate Medical Center, Brooklyn, New York
- Department of Neurology, State University of New York, Downstate Medical Center, Brooklyn, New York
- Department of Pharmacological Sciences, Stony Brook Medicine, Stony Brook University, Stony Brook, New York
| |
Collapse
|
125
|
Fei M, Wang H, Zhou M, Deng C, Zhang L, Han Y. Podoplanin influences the inflammatory phenotypes and mobility of microglia in traumatic brain injury. Biochem Biophys Res Commun 2019; 523:361-367. [PMID: 31866008 DOI: 10.1016/j.bbrc.2019.12.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 12/01/2019] [Indexed: 01/15/2023]
Abstract
Traumatic brain injury (TBI) represents a major cause of death and disability worldwide. Exacerbated neuroinflammation following TBI causes secondary injury. Podoplanin (PDPN) is a small transmembrane mucin-like glycoprotein that promotes the inflammatory response in different tissues and cells. However, the contribution of PDPN to neuroinflammation and microglial activation is unknown. Here, we found that PDPN was correlated with microglial activation after TBI in mice. Meanwhile, PDPN expression could be induced by trauma-related stimuli, such as lipopolysaccharide (LPS), ATP, H2O2 and hemoglobin (Hb), in primary microglia. Furthermore, with Hb treatment in vitro, knockdown of PDPN could decrease the proportion of M1-like microglia and increase the proportion of M2-like microglia via reduced secretion of IL-1β and TNF-α and increased secretion of IL-10 and TGF-β compared to the control microglia. Immunofluorescence also showed that CD86-positive microglia were decreased and CD206-positive microglia were elevated in the PDPN-KD group. Additionally, PDPN knockdown impaired microglial mobility and phagocytosis and decreased the expression of matrix metalloproteinases (mainly MMP2 and MMP9). In summary, PDPN plays an important role in microglia-mediated inflammation and may serve as a potential target for TBI treatment.
Collapse
Affiliation(s)
- Maoxing Fei
- Department of Neurosurgery, Jinling Hospital, Nanjing Medical University, Nanjing, 210002, PR China
| | - Handong Wang
- Department of Neurosurgery, Jinling Hospital, Nanjing Medical University, Nanjing, 210002, PR China; Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, PR China.
| | - Mengliang Zhou
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, PR China.
| | - Chulei Deng
- Department of Neurosurgery, Jinling Hospital, Southern Medical University, Guangzhou, 510000, PR China
| | - Li Zhang
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, PR China
| | - Yanling Han
- Department of Neurosurgery, Jinling Hospital, Nanjing, 210002, PR China
| |
Collapse
|
126
|
Gano A, Mondello JE, Doremus-Fitzwater TL, Deak T. Rapid alterations in neuroimmune gene expression after acute ethanol: Timecourse, sex differences and sensitivity to cranial surgery. J Neuroimmunol 2019; 337:577083. [PMID: 31675629 PMCID: PMC6866658 DOI: 10.1016/j.jneuroim.2019.577083] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 10/04/2019] [Accepted: 10/05/2019] [Indexed: 01/08/2023]
Abstract
Prior work has established that that an acute ethanol challenge mimicking high intensity alcohol consumption increased IL-6 and suppressed IL-1β and TNFα mRNA in intoxication, with the opposite pattern seen in withdrawal. These experiments utilized Sprague-Dawley rats to further extend these results across time course (from 45 min to 6 h after ethanol), sex, and central versus peripheral expression. Furthermore, these data show that cannulation surgery may selectively modify the central neuroimmune response to ethanol. These findings highlight a unique plasticity of IL-6 that is specific to central structures and responsive to alterations by environmental factors.
Collapse
Affiliation(s)
- Anny Gano
- Developmental Exposure Alcohol Research Center (DEARC), Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States of America
| | - Jamie E Mondello
- Developmental Exposure Alcohol Research Center (DEARC), Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States of America
| | - Tamara L Doremus-Fitzwater
- Department of Psychology, Williams Hall, Ithaca College, 953 Danby Road, Ithaca, NY 14850, United States of America
| | - Terrence Deak
- Developmental Exposure Alcohol Research Center (DEARC), Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States of America.
| |
Collapse
|
127
|
Targeting high-mobility group box protein 1 (HMGB1) in pediatric traumatic brain injury: Chronic neuroinflammatory, behavioral, and epileptogenic consequences. Exp Neurol 2019; 320:112979. [DOI: 10.1016/j.expneurol.2019.112979] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 05/29/2019] [Accepted: 06/18/2019] [Indexed: 11/18/2022]
|
128
|
Lee HF, Lin JS, Chang CF. Acute Kahweol Treatment Attenuates Traumatic Brain Injury Neuroinflammation and Functional Deficits. Nutrients 2019; 11:nu11102301. [PMID: 31569604 PMCID: PMC6835740 DOI: 10.3390/nu11102301] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 09/19/2019] [Accepted: 09/24/2019] [Indexed: 12/11/2022] Open
Abstract
Traumatic brain injury (TBI) affects millions worldwide with devastating long-term effects on health and cognition. Emerging data suggest that targeting the immune response may offer promising strategies to alleviate TBI outcomes; kahweol, an anti-inflammatory diterpene that remains in unfiltered coffee, has been shown to be beneficial in neuronal recovery. Here, we examined whether kahweol could alleviate brain trauma-induced injury in a mouse model of TBI and its underlying mechanisms. TBI was induced by controlled cortical impact (CCI) and various doses of kahweol were intraperitoneally administered following injury. Contusion volume, brain edema, neurobehavioral deficits, and protein expression and activity were evaluated in both short-term and long-term recovery. We found that kahweol treatments significantly reduced secondary brain injury and improved neurobehavioral outcomes in TBI mice. These changes were accompanied by the attenuation of proinflammatory cytokine secretion, decreased microglia/macrophage activation, and reduction of neutrophil and leukocyte infiltration. In addition, continuous kahweol treatment further improved short-term TBI outcomes compared to single-dosage. Collectively, our data showed that kahweol protects against TBI by reducing immune responses and may serve as a potential therapeutic intervention for TBI patients.
Collapse
Affiliation(s)
- Hung-Fu Lee
- Department of Neurosurgery, Cheng Hsin General Hospital, Taipei 11220, Taiwan.
| | - Jhih Syuan Lin
- Department of Neurosurgery, Cheng Hsin General Hospital, Taipei 11220, Taiwan.
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan.
| | - Che-Feng Chang
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan.
| |
Collapse
|
129
|
Ma Y, Liu T, Fu J, Fu S, Hu C, Sun B, Fan X, Zhu J. Lactobacillus acidophilus Exerts Neuroprotective Effects in Mice with Traumatic Brain Injury. J Nutr 2019; 149:1543-1552. [PMID: 31174208 DOI: 10.1093/jn/nxz105] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/11/2019] [Accepted: 04/26/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) causes dysbiosis and intestinal barrier disruption, which further exacerbate brain damage via an inflammatory pathway. Gut microbiota remodeling by Lactobacillus acidophilus (LA) is a potential intervention. OBJECTIVE The aim of this study was to investigate the neuroprotective effects of LA in TBI and elucidated underlying mechanisms. METHODS C57BL/6 male mice (aged 8-9 wk) were subjected to weight-drop impact and gavaged with saline (TBI + vehicle) or LA (1 × 1010 CFU) (TBI + LA) on the day of injury and each day after for 1, 3, or 7 d. The sham + vehicle mice underwent craniotomy without brain injury and were gavaged with saline. Sensorimotor functions were determined pre-TBI and 1, 3, and 7 d postinjury. Indexes of neuroinflammation, peripheral inflammation, and intestinal barrier function were measured on days 3 and 7. Microbiota composition was measured 3 d postinjury. The data were mainly analyzed by 2-factor ANOVA. RESULTS Compared with sham + vehicle mice, the TBI + vehicle mice exhibited impairments in the neurological severity score (+692%, day 3; +600%, day 7) and rotarod test (-58%, day 3; -45%, day 7) (P < 0.05), which were rescued by LA. The numbers of microglia (total and activated) and astrocytes and concentrations of TNF-α and IL1-β in the perilesional cortex were elevated in the TBI + vehicle mice on day 3 or 7 compared with sham + vehicle mice (P < 0.05) and were normalized by LA. Compared with sham + vehicle mice, the TBI + vehicle mice exhibited increased serum concentrations of endotoxin and TNF-α, and intestinal barrier permeability (D-lactate) on days 3 and 7 (P < 0.05), and these changes were alleviated by LA. Three days postinjury, the microbiota composition was disrupted in the TBI + vehicle mice compared with sham + vehicle mice (P < 0.05), which was restored by LA. CONCLUSION Our results demonstrate that LA exerts neuroprotective effects that may be associated with gut microbiota remodeling in TBI mice.
Collapse
Affiliation(s)
- Yuanyuan Ma
- Department of Basic Nursing, School of Nursing, Third Military Medical University (Army Medical University), Chongqing, China.,Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Tianyao Liu
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jingjing Fu
- Department of Basic Nursing, School of Nursing, Third Military Medical University (Army Medical University), Chongqing, China
| | - Shaoli Fu
- Department of Basic Nursing, School of Nursing, Third Military Medical University (Army Medical University), Chongqing, China
| | - Chen Hu
- Department of Basic Nursing, School of Nursing, Third Military Medical University (Army Medical University), Chongqing, China
| | - Bo Sun
- Department of Basic Nursing, School of Nursing, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xiaotang Fan
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jingci Zhu
- Department of Basic Nursing, School of Nursing, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
130
|
De Bellis A, Bellastella G, Maiorino MI, Costantino A, Cirillo P, Longo M, Pernice V, Bellastella A, Esposito K. The role of autoimmunity in pituitary dysfunction due to traumatic brain injury. Pituitary 2019; 22:236-248. [PMID: 30847776 DOI: 10.1007/s11102-019-00953-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE Traumatic brain injury (TBI) is one of the most common causes of mortality and long-term disability and it is associated with an increased prevalence of neuroendocrine dysfunctions. Post-traumatic hypopituitarism (PTHP) results in major physical, psychological and social consequences leading to impaired quality of life. PTHP can occur at any time after traumatic event, evolving through various ways and degrees of deficit, requiring appropriate screening for early detection and treatment. Although the PTHP pathophysiology remains to be elucitated, on the basis of proposed hypotheses it seems to be the result of combined pathological processes, with a possible role played by hypothalamic-pituitary autoimmunity (HPA). This review is aimed at focusing on this possible role in the development of PTHP and its potential clinical consequences, on the basis of the data so far appeared in the literature and of some results of personal studies on this issue. METHODS Scrutinizing the data so far appeared in literature on this topic, we have found only few studies evaluating the autoimmune pattern in affected patients, searching in particular for antipituitary and antihypothalamus autoantibodies (APA and AHA, respectively) by simple indirect immunofluorescence. RESULTS The presence of APA and/or AHA at high titers was associated with an increased risk of onset/persistence of PTHP. CONCLUSIONS HPA seems to contribute to TBI-induced pituitary damage and related PTHP. However, further prospective studies in a larger cohort of patients are needed to define etiopathogenic and diagnostic role of APA/AHA in development of post-traumatic hypothalamic/pituitary dysfunctions after a TBI.
Collapse
Affiliation(s)
- Annamaria De Bellis
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy.
| | - Giuseppe Bellastella
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Maria Ida Maiorino
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Angela Costantino
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Paolo Cirillo
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Miriam Longo
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Vlenia Pernice
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Antonio Bellastella
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Katherine Esposito
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
131
|
Neuroinflammation, the thread connecting neurological disease : Cluster: "Neuroinflammatory mechanisms in neurodegenerative disorders". Acta Neuropathol 2019; 137:689-691. [PMID: 30968314 DOI: 10.1007/s00401-019-02009-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 04/05/2019] [Indexed: 02/02/2023]
|
132
|
von Leden RE, Parker KN, Bates AA, Noble-Haeusslein LJ, Donovan MH. The emerging role of neutrophils as modifiers of recovery after traumatic injury to the developing brain. Exp Neurol 2019; 317:144-154. [PMID: 30876905 DOI: 10.1016/j.expneurol.2019.03.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 03/03/2019] [Accepted: 03/08/2019] [Indexed: 12/16/2022]
Abstract
The innate immune response plays a critical role in traumatic brain injury (TBI), contributing to ongoing pathogenesis and worsening long-term outcomes. Here we focus on neutrophils, one of the "first responders" to TBI. These leukocytes are recruited to the injured brain where they release a host of toxic molecules including free radicals, proteases, and pro-inflammatory cytokines, all of which promote secondary tissue damage. There is mounting evidence that the developing brain is more vulnerable to injury that the adult brain. This vulnerability to greater damage from TBI is, in part, attributed to relatively low antioxidant reserves coupled with an early robust immune response. The latter is reflected in enhanced sensitivity to cytokines and a prolonged recruitment of neutrophils into both cortical and subcortical regions. This review considers the contribution of neutrophils to early secondary pathogenesis in the injured developing brain and raises the distinct possibility that these leukocytes, which exhibit phenotypic plasticity, may also be poised to support wound healing. We provide a basic review of the development, life cycle, and granular contents of neutrophils and evaluate their potential as therapeutic targets for early neuroprotection and functional recovery after injury at early age. While neutrophils have been broadly studied in neurotrauma, we are only beginning to appreciate their diverse roles in the developing brain and the extent to which their acute manipulation may result in enduring neurological recovery when TBI is superimposed upon brain development.
Collapse
Affiliation(s)
- Ramona E von Leden
- Department of Neurology, Dell Medical School, The University of Texas at Austin, 1701 Trinity St., Austin, TX 78712, USA.
| | - Kaila N Parker
- Department of Psychology, Behavioral Neuroscience, The University of Texas at Austin, 108 E. Dean Keeton St., Austin, TX 78712, USA.
| | - Adrian A Bates
- Institute for Neuroscience, The University of Texas at Austin, 100 E. 24(th) St., Austin, TX 78712, USA.
| | - Linda J Noble-Haeusslein
- Department of Neurology, Dell Medical School, The University of Texas at Austin, 1701 Trinity St., Austin, TX 78712, USA; Department of Psychology, Behavioral Neuroscience, The University of Texas at Austin, 108 E. Dean Keeton St., Austin, TX 78712, USA; Institute for Neuroscience, The University of Texas at Austin, 100 E. 24(th) St., Austin, TX 78712, USA.
| | - Michael H Donovan
- Department of Neurology, Dell Medical School, The University of Texas at Austin, 1701 Trinity St., Austin, TX 78712, USA.
| |
Collapse
|
133
|
Jiang JP, Niu XG, Dai C, Ma K, Xu HY, Cheng SX, Zhang ZW, Duan F, Zhu X, Wang YT, Chen XY, Zhang S. Neurological functional evaluation based on accurate motions in big animals with traumatic brain injury. Neural Regen Res 2019; 14:991-996. [PMID: 30762010 PMCID: PMC6404497 DOI: 10.4103/1673-5374.250578] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
An accurate and effective neurological evaluation is indispensable in the treatment and rehabilitation of traumatic brain injury. However, most of the existing evaluation methods in basic research and clinical practice are not objective or intuitive for assessing the neurological function of big animals, and are also difficult to use to qualify the extent of damage and recovery. In the present study, we established a big animal model of traumatic brain injury by impacting the cortical motor region of beagles. At 2 weeks after successful modeling, we detected neurological deficiencies in the animal model using a series of techniques, including three-dimensional motion capture, electromyogram and ground reaction force. These novel technologies may play an increasingly important role in the field of traumatic brain injury diagnosis and rehabilitation in the future. The experimental protocol was approved by the Animal Care and Use Committee of Logistics University of People’s Armed Police Force (approval No. 2017-0006.2).
Collapse
Affiliation(s)
- Ji-Peng Jiang
- Center for Neurology and Neurosurgery, Medical Center of People's Armed Policed Force, Key Laboratory of Neurotrauma Repair of Tianjin, Tianjin, China
| | - Xue-Gang Niu
- Department of Neurosurgery, Tianjin Fourth Central Hospital, Tianjin, China
| | - Chen Dai
- Center for Neurology and Neurosurgery, Medical Center of People's Armed Policed Force, Key Laboratory of Neurotrauma Repair of Tianjin, Tianjin, China
| | - Ke Ma
- Center for Neurology and Neurosurgery, Medical Center of People's Armed Policed Force, Key Laboratory of Neurotrauma Repair of Tianjin, Tianjin, China
| | - Hui-You Xu
- Center for Neurology and Neurosurgery, Medical Center of People's Armed Policed Force, Key Laboratory of Neurotrauma Repair of Tianjin, Tianjin, China
| | - Shi-Xiang Cheng
- Center for Neurology and Neurosurgery, Medical Center of People's Armed Policed Force, Key Laboratory of Neurotrauma Repair of Tianjin, Tianjin, China
| | - Zhi-Wen Zhang
- Department of Automation, College of Computer and Control Engineering, Nankai University, Tianjin, China
| | - Feng Duan
- Department of Automation, College of Computer and Control Engineering, Nankai University, Tianjin, China
| | - Xu Zhu
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Yu-Ting Wang
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Xu-Yi Chen
- Center for Neurology and Neurosurgery, Medical Center of People's Armed Policed Force, Key Laboratory of Neurotrauma Repair of Tianjin, Tianjin, China
| | - Sai Zhang
- Center for Neurology and Neurosurgery, Medical Center of People's Armed Policed Force, Key Laboratory of Neurotrauma Repair of Tianjin, Tianjin, China
| |
Collapse
|