1
|
Henry RJ, Barrett JP, Vaida M, Khan NZ, Makarevich O, Ritzel RM, Faden AI, Stoica BA. Interaction of high-fat diet and brain trauma alters adipose tissue macrophages and brain microglia associated with exacerbated cognitive dysfunction. J Neuroinflammation 2024; 21:113. [PMID: 38685031 PMCID: PMC11058055 DOI: 10.1186/s12974-024-03107-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 04/22/2024] [Indexed: 05/02/2024] Open
Abstract
Obesity increases the morbidity and mortality of traumatic brain injury (TBI). Detailed analyses of transcriptomic changes in the brain and adipose tissue were performed to elucidate the interactive effects between high-fat diet-induced obesity (DIO) and TBI. Adult male mice were fed a high-fat diet (HFD) for 12 weeks prior to experimental TBI and continuing after injury. High-throughput transcriptomic analysis using Nanostring panels of the total visceral adipose tissue (VAT) and cellular components in the brain, followed by unsupervised clustering, principal component analysis, and IPA pathway analysis were used to determine shifts in gene expression patterns and molecular pathway activity. Cellular populations in the cortex and hippocampus, as well as in VAT, during the chronic phase after combined TBI-HFD showed amplification of central and peripheral microglia/macrophage responses, including superadditive changes in selected gene expression signatures and pathways. Furthermore, combined TBI and HFD caused additive dysfunction in Y-Maze, Novel Object Recognition (NOR), and Morris water maze (MWM) cognitive function tests. These novel data suggest that HFD-induced obesity and TBI can independently prime and support the development of altered states in brain microglia and VAT, including the disease-associated microglia/macrophage (DAM) phenotype observed in neurodegenerative disorders. The interaction between HFD and TBI promotes a shift toward chronic reactive microglia/macrophage transcriptomic signatures and associated pro-inflammatory disease-altered states that may, in part, underlie the exacerbation of cognitive deficits. Thus, targeting of HFD-induced reactive cellular phenotypes, including in peripheral adipose tissue immune cell populations, may serve to reduce microglial maladaptive states after TBI, attenuating post-traumatic neurodegeneration and neurological dysfunction.
Collapse
Affiliation(s)
- Rebecca J Henry
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA.
- Department of Anatomy and Neuroscience, School of Medicine, University College Cork, Cork, Ireland.
| | - James P Barrett
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Maria Vaida
- Harrisburg University of Science and Technology, 326 Market St, Harrisburg, PA, USA
| | - Niaz Z Khan
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Oleg Makarevich
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Rodney M Ritzel
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alan I Faden
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Bogdan A Stoica
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
- VA Maryland Health Care System, Baltimore VA Medical Center, Baltimore, MD, 21201, USA
| |
Collapse
|
2
|
Vincent JC, Garnett CN, Watson JB, Higgins EK, Macheda T, Sanders L, Roberts KN, Shahidehpour RK, Blalock EM, Quan N, Bachstetter AD. IL-1R1 signaling in TBI: assessing chronic impacts and neuroinflammatory dynamics in a mouse model of mild closed-head injury. J Neuroinflammation 2023; 20:248. [PMID: 37884959 PMCID: PMC10601112 DOI: 10.1186/s12974-023-02934-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 10/17/2023] [Indexed: 10/28/2023] Open
Abstract
Neuroinflammation contributes to secondary injury cascades following traumatic brain injury (TBI), with alternating waves of inflammation and resolution. Interleukin-1 (IL-1), a critical neuroinflammatory mediator originating from brain endothelial cells, microglia, astrocytes, and peripheral immune cells, is acutely overexpressed after TBI, propagating secondary injury and tissue damage. IL-1 affects blood-brain barrier permeability, immune cell activation, and neural plasticity. Despite the complexity of cytokine signaling post-TBI, we hypothesize that IL-1 signaling specifically regulates neuroinflammatory response components. Using a closed-head injury (CHI) TBI model, we investigated IL-1's role in the neuroinflammatory cascade with a new global knock-out (gKO) mouse model of the IL-1 receptor (IL-1R1), which efficiently eliminates all IL-1 signaling. We found that IL-1R1 gKO attenuated behavioral impairments 14 weeks post-injury and reduced reactive microglia and astrocyte staining in the neocortex, corpus callosum, and hippocampus. We then examined whether IL-1R1 loss altered acute neuroinflammatory dynamics, measuring gene expression changes in the neocortex at 3, 9, 24, and 72 h post-CHI using the NanoString Neuroinflammatory panel. Of 757 analyzed genes, IL-1R1 signaling showed temporal specificity in neuroinflammatory gene regulation, with major effects at 9 h post-CHI. IL-1R1 signaling specifically affected astrocyte-related genes, selectively upregulating chemokines like Ccl2, Ccl3, and Ccl4, while having limited impact on cytokine regulation, such as Tnfα. This study provides further insight into IL-1R1 function in amplifying the neuroinflammatory cascade following CHI in mice and demonstrates that suppression of IL-1R1 signaling offers long-term protective effects on brain health.
Collapse
Affiliation(s)
- Jonathan C Vincent
- Department of Neuroscience, University of Kentucky, 741 S. Limestone St., Lexington, KY, 40536, USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
- MD/PhD Program, University of Kentucky, Lexington, KY, USA
| | - Colleen N Garnett
- Department of Neuroscience, University of Kentucky, 741 S. Limestone St., Lexington, KY, 40536, USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - James B Watson
- Department of Neuroscience, University of Kentucky, 741 S. Limestone St., Lexington, KY, 40536, USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, USA
| | - Emma K Higgins
- Department of Neuroscience, University of Kentucky, 741 S. Limestone St., Lexington, KY, 40536, USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, USA
| | - Teresa Macheda
- Department of Neuroscience, University of Kentucky, 741 S. Limestone St., Lexington, KY, 40536, USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, USA
| | - Lydia Sanders
- Department of Neuroscience, University of Kentucky, 741 S. Limestone St., Lexington, KY, 40536, USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, USA
| | - Kelly N Roberts
- Department of Neuroscience, University of Kentucky, 741 S. Limestone St., Lexington, KY, 40536, USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, USA
| | - Ryan K Shahidehpour
- Department of Neuroscience, University of Kentucky, 741 S. Limestone St., Lexington, KY, 40536, USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Eric M Blalock
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Ning Quan
- Department of Biomedical Science, Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Adam D Bachstetter
- Department of Neuroscience, University of Kentucky, 741 S. Limestone St., Lexington, KY, 40536, USA.
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, USA.
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
3
|
Yanckello LM, Chang YH, Sun M, Chlipala G, Green SJ, Lei Z, Ericsson AC, Xing X, Hammond TC, Bachstetter AD, Lin AL. Inulin supplementation prior to mild traumatic brain injury mitigates gut dysbiosis, and brain vascular and white matter deficits in mice. FRONTIERS IN MICROBIOMES 2022; 1:986951. [PMID: 36756543 PMCID: PMC9903356 DOI: 10.3389/frmbi.2022.986951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Introduction Mild traumatic brain injury (mTBI) has been shown to negatively alter bacterial diversity and composition within the gut, known as dysbiosis, in rodents and humans. These changes cause secondary consequences systemically through decreased bacterial metabolites such as short chain fatty acids (SCFAs) which play a role in inflammation and metabolism. The goal of the study was to identify if giving prebiotic inulin prior to closed head injury (CHI) could mitigate gut dysbiosis, increase SCFAs, and improve recovery outcomes, including protecting cerebral blood flow (CBF) and white matter integrity (WMI) in young mice. Methods We fed mice at 2 months of age with either inulin or control diet (with cellulose as fiber source) for two months before the CHI and continued till the end of the study. We analyzed gut microbiome composition and diversity, determined SCFAs levels, and measured CBF and WMI using MRI. We compared the results with Naïve and Sham-injury mice at 24 hours, 1.5 months, and 3-4 months post-injury. Results We found that both CHI and Sham mice had time-dependent changes in gut composition and diversity after surgery. Inulin significantly reduced the abundance of pathobiont bacteria, such as E. coli, Desulfovibrio spp and Pseudomonas aeruginosa, in Sham and CHI mice compared to mice fed with control diet. On the other hand, inulin increased SCFAs-producing bacteria, such as Bifidobacterium spp and Lactobacillus spp, increased levels of SCFAs, including butyrate and propionate, and significantly altered beta diversity as early as 24 hours post-injury, which lasted up to 3-4 months post-injury. The mitigation of dysbiosis is associated with protection of WMI in fimbria, internal and external capsule, and CBF in the right hippocampus of CHI mice, suggesting protection of memory and cognitive functions. Discussion The results indicate that giving inulin prior to CHI could promote recovery outcome through gut microbiome modulation. As inulin, microbiome analysis, and MRI are readily to be used in humans, the findings from the study may pave a way for a cost-effective, accessible intervention for those at risk of sustaining a head injury, such as military personnel or athletes in contact sports.
Collapse
Affiliation(s)
- Lucille M. Yanckello
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, United States
| | - Ya-Hsuan Chang
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, United States
| | - McKenna Sun
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States
| | - George Chlipala
- Research Informatics Core, University of Illinois Chicago, Chicago, IL, United States
| | - Stefan J. Green
- Genomics and Microbiome Core Facility, Rush University, Chicago, IL, United States
| | - Zhentian Lei
- Metabolomics Center, University of Missouri, Columbia, MO, United States
- Department of Biochemistry, University of Missouri, Columbia, MO, United States
| | - Aaron C. Ericsson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| | - Xin Xing
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States
- Department of Computer Science, University of Kentucky, Lexington, KY, United States
| | - Tyler C. Hammond
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| | - Adam D. Bachstetter
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
- Spinal Cord and Brain Injury Research Center, University of Kentucky, KY, United States
| | - Ai-Ling Lin
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, United States
- Department of Radiology, University of Missouri, Columbia, MO, United States
- Institute for Data Science &Informatics, University of Missouri, Columbia, MO, United States
- Department of Biological Sciences, University of Missouri, Columbia, MO, United States
| |
Collapse
|
4
|
Macheda T, Roberts K, Bachstetter AD. Electromagnetic Controlled Closed-Head Model of Mild Traumatic Brain Injury in Mice. J Vis Exp 2022:10.3791/64556. [PMID: 36279529 PMCID: PMC10550048 DOI: 10.3791/64556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Highly reproducible animal models of traumatic brain injury (TBI), with well-defined pathologies, are needed for testing therapeutic interventions and understanding the mechanisms of how a TBI alters brain function. The availability of multiple animal models of TBI is necessary to model the different aspects and severities of TBI seen in people. This manuscript describes the use of a midline closed head injury (CHI) to develop a mouse model of mild TBI. The model is considered mild because it does not produce structural brain lesions based on neuroimaging or gross neuronal loss. However, a single impact creates enough pathology that cognitive impairment is measurable at least 1 month after injury. A step-by-step protocol to induce a CHI in mice using a stereotaxically guided electromagnetic impactor is defined in the paper. The benefits of the mild midline CHI model include the reproducibility of the injury-induced changes with low mortality. The model has been temporally characterized up to 1 year after the injury for neuroimaging, neurochemical, neuropathological, and behavioral changes. The model is complementary to open skull models of controlled cortical impact using the same impactor device. Thus, labs can model both mild diffuse TBI and focal moderate-to-severe TBI with the same impactor.
Collapse
Affiliation(s)
- Teresa Macheda
- Spinal Cord & Brain Injury Research Center, University of Kentucky
| | - Kelly Roberts
- Spinal Cord & Brain Injury Research Center, University of Kentucky
| | - Adam D Bachstetter
- Spinal Cord & Brain Injury Research Center, University of Kentucky; Department of Neuroscience, University of Kentucky; Sanders-Brown Center on Aging, University of Kentucky;
| |
Collapse
|
5
|
Zajac DJ, Shaw BC, Braun DJ, Green SJ, Morganti JM, Estus S. Exogenous Short Chain Fatty Acid Effects in APP/PS1 Mice. Front Neurosci 2022; 16:873549. [PMID: 35860296 PMCID: PMC9289923 DOI: 10.3389/fnins.2022.873549] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
Elucidating the impact of the gut microbiome on Alzheimer's Disease (AD) is an area of intense interest. Short chain fatty acids (SCFAs) are major microbiota metabolites that have been implicated as a mediator of gut microbiome effects in the brain. Here, we tested the effects of SCFA-treated water vs. saline-treated water on APPswe/PSEN1dE9 mice maintained under standard laboratory conditions. Mice were treated with SCFAs from five months of age until ten months of age, when they were evaluated for microbiome profile, impaired spatial memory as evaluated with the radial arm water maze, astrocyte activation as measured by Gfap expression and amyloid burden as assessed by histochemistry and MSD ELISA. We report that SCFA treatment increased alpha-diversity and impacted the gut microbiome profile by increasing, in part, the relative abundance of several bacteria that typically produce SCFAs. However, SCFA treatment did not significantly affect behavior. Similarly, SCFAs did not affect cortical or hippocampal astrocyte activation observed in the APP/PS1 mice. Lastly, although robust levels of soluble and insoluble amyloid were present in the APP/PS1 mice, SCFA treatment had no effect on these indices. Overall, our findings are that SCFA treatment modifies the microbiome in a fashion that may increase further SCFA production. However, SCFA treatment did not alter behavior, astrocyte activation, nor amyloid neuropathology in APP/PS1 mice maintained with a conventional microbiome.
Collapse
Affiliation(s)
- Diana J. Zajac
- Department of Physiology and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States
| | - Benjamin C. Shaw
- Department of Physiology and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States
| | - David J. Braun
- Department of Neuroscience and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States
| | - Stefan J. Green
- Genome Research Core, Research Resources Center, University of Illinois at Chicago, Chicago, IL, United States
| | - Joshua M. Morganti
- Department of Neuroscience and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States
| | - Steven Estus
- Department of Physiology and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
6
|
Therapeutic treatment with the anti-inflammatory drug candidate MW151 may partially reduce memory impairment and normalizes hippocampal metabolic markers in a mouse model of comorbid amyloid and vascular pathology. PLoS One 2022; 17:e0262474. [PMID: 35081152 PMCID: PMC8791470 DOI: 10.1371/journal.pone.0262474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 12/24/2021] [Indexed: 12/03/2022] Open
Abstract
Alzheimer’s disease (AD) is the leading cause of dementia in the elderly, but therapeutic options are lacking. Despite long being able to effectively treat the ill-effects of pathology present in various rodent models of AD, translation of these strategies to the clinic has so far been disappointing. One potential contributor to this situation is the fact that the vast majority of AD patients have other dementia-contributing comorbid pathologies, the most common of which are vascular in nature. This situation is modeled relatively infrequently in basic AD research, and almost never in preclinical studies. As part of our efforts to develop small molecule, anti-inflammatory therapeutics for neurological injury and disease, we have recently been exploring potentially promising treatments in preclinical multi-morbidity contexts. In the present study, we generated a mouse model of mixed amyloid and hyperhomocysteinemia (HHcy) pathology in which to test the efficacy of one of our anti-inflammatory compounds, MW151. HHcy can cause cerebrovascular damage and is an independent risk factor for both AD dementia and vascular contributions to cognitive impairment and dementia. We found that MW151 was able to partially rescue hippocampal-dependent spatial memory and learning deficits in this comorbidity context, and further, that the benefit is associated with a normalization of hippocampal metabolites detectable via magnetic resonance spectroscopy. These findings provide evidence that MW151 in particular, and potentially anti-inflammatory treatment more generally, may be beneficial in AD patients with comorbid vascular pathology.
Collapse
|
7
|
Yanckello LM, Fanelli B, McCulloch S, Xing X, Sun M, Hammond TC, Colwell R, Gu Z, Ericsson AC, Chang YH, Bachstetter AD, Lin AL. Inulin Supplementation Mitigates Gut Dysbiosis and Brain Impairment Induced by Mild Traumatic Brain Injury during Chronic Phase. JOURNAL OF CELLULAR IMMUNOLOGY 2022; 4:50-64. [PMID: 35611116 PMCID: PMC9126115 DOI: 10.33696/immunology.4.132] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Mild traumatic brain injury (mTBI) has been shown to acutely alter the gut microbiome diversity and composition, known as dysbiosis, which can further exacerbate metabolic and vascular changes in the brain in both humans and rodents. However, it remains unknown how mTBI affects the gut microbiome in the chronic phase recovery (past one week post injury). It is also unknown if injury recovery can be improved by mitigating dysbiosis. The goal of the study is to fill the knowledge gap. First, we aim to understand how mTBI alters the gut microbiome through the chronic period of recovery (3 months post injury). In addition, as the gut microbiome can be modulated by diet, we also investigated if prebiotic inulin, a fermentable fiber that promotes growth of beneficial bacteria and metabolites, would mitigate dysbiosis, improve systemic metabolism, and protect brain structural and vascular integrity when administered after 3 months post closed head injury (CHI). We found that CHI given to male mice at 4 months of age induced gut dysbiosis which peaked at 1.5 months post injury, reduced cerebral blood flow (CBF) and altered brain white matter integrity. Interestingly, we also found that Sham mice had transient dysbiosis, which peaked 24 hours after injury and then normalized. After 8 weeks of inulin feeding, CHI mice had increased abundance of beneficial/anti-inflammatory bacteria, reduced abundance of pathogenic bacteria, enriched levels of short-chain fatty acids, and restored CBF in both hippocampi and left thalamus, compared to the CHI-control fed and Sham groups. Using machine learning, we further identified top bacterial species that separate Sham and CHI mice with and without the diet. Our results indicate that there is an injury- and time-dependent dysbiosis between CHI and Sham mice; inulin is effective to mitigate dysbiosis and improve brain injury recovery in the CHI mice. As there are currently no effective treatments for mTBI, the study may have profound implications for developing therapeutics or preventive interventions in the future.
Collapse
Affiliation(s)
- Lucille M. Yanckello
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, United States of America
| | - Brian Fanelli
- CosmosID Inc., Rockville, MD, United States of America
| | | | - Xin Xing
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America
- Department of Computer Science, University of Kentucky, Lexington, KY, United States of America
| | - McKenna Sun
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America
| | - Tyler C. Hammond
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America
| | - Rita Colwell
- CosmosID Inc., Rockville, MD, United States of America
| | - Zezong Gu
- Department of Pathology & Anatomical Sciences, University of Missouri, Columbia, MO, United States of America
- Harry S. Truman Memorial Veteran Hospital, Columbia, MO, United States of America
| | - Aaron C. Ericsson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States of America
| | - Ya-Hsuan Chang
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, United States of America
| | - Adam D. Bachstetter
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America
- Spinal Cord and Brain Injury Research Center, University of Kentucky, KY, United States of America
| | - Ai-Ling Lin
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, United States of America
- Department of Radiology, University of Missouri, Columbia, MO, United States of America
- Institute for Data Science &Informatics, University of Missouri, Columbia, MO United States of America
| |
Collapse
|
8
|
Moss LD, Sode D, Patel R, Lui A, Hudson C, Patel NA, Bickford PC. Intranasal delivery of exosomes from human adipose derived stem cells at forty-eight hours post injury reduces motor and cognitive impairments following traumatic brain injury. Neurochem Int 2021; 150:105173. [PMID: 34453976 PMCID: PMC8511339 DOI: 10.1016/j.neuint.2021.105173] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/18/2021] [Accepted: 08/24/2021] [Indexed: 12/14/2022]
Abstract
The neuroprotective role of human adipose-derived stems cells (hASCs) has raised great interest in regenerative medicine due to their ability to modulate their surrounding environment. Our group has demonstrated that exosomes derived from hASC (hASCexo) are a cell-free regenerative approach to long term recovery following traumatic brain injury (TBI). Previously, we demonstrated the efficacy of exosome treatment with intravenous delivery at 3 h post TBI in rats. Here, we show efficacy of exosomes through intranasal delivery at 48 h post TBI in mice lengthening the therapeutic window of treatment and therefore increasing possible translation to clinical studies. Our findings demonstrate significant recovery of motor impairment assessed by an elevated body swing test in mice treated with exosomes containing MALAT1 compared to both TBI mice without exosomes and exosomes depleted of MALAT1. Significant cognitive improvement was seen in the reversal trial of 8 arm radial arm water maze in mice treated with exosomes containing MALAT1. Furthermore, cortical damage was significantly reduced in mice treated with exosomes containing MALAT1 as well as decreased MHCII+ staining of microglial cells. Mice without exosomes or treated with exosomes depleted of MALAT1 did not show similar recovery. Results demonstrate both inflammation related genes and NRTK3 (TrkC) are target genes modulated by hASC exosomes and further that MALAT1 in hASC exosomes regulates expression of full length TrkC thereby activating the MAPK pathway and promoting recovery. Exosomes are a promising therapeutic approach following TBI with a therapeutic window of at least 48 h and contain long noncoding RNA's, specifically MALAT1 that play a vital role in the mechanism of action.
Collapse
Affiliation(s)
- Lauren D Moss
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Derek Sode
- Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Rekha Patel
- James A. Haley Veterans Hospital, Research Service, Tampa, FL, USA
| | - Ashley Lui
- Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Charles Hudson
- James A. Haley Veterans Hospital, Research Service, Tampa, FL, USA
| | - Niketa A Patel
- James A. Haley Veterans Hospital, Research Service, Tampa, FL, USA; Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
| | - Paula C Bickford
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA; James A. Haley Veterans Hospital, Research Service, Tampa, FL, USA.
| |
Collapse
|
9
|
Serpa RO, Ferguson L, Larson C, Bailard J, Cooke S, Greco T, Prins ML. Pathophysiology of Pediatric Traumatic Brain Injury. Front Neurol 2021; 12:696510. [PMID: 34335452 PMCID: PMC8319243 DOI: 10.3389/fneur.2021.696510] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 06/21/2021] [Indexed: 11/23/2022] Open
Abstract
The national incidence of traumatic brain injury (TBI) exceeds that of any other disease in the pediatric population. In the United States the Centers for Disease Control and Prevention (CDC) reports 697,347 annual TBIs in children ages 0–19 that result in emergency room visits, hospitalization or deaths. There is a bimodal distribution within the pediatric TBI population, with peaks in both toddlers and adolescents. Preclinical TBI research provides evidence for age differences in acute pathophysiology that likely contribute to long-term outcome differences between age groups. This review will examine the timecourse of acute pathophysiological processes during cerebral maturation, including calcium accumulation, glucose metabolism and cerebral blood flow. Consequences of pediatric TBI are complicated by the ongoing maturational changes allowing for substantial plasticity and windows of vulnerabilities. This review will also examine the timecourse of later outcomes after mild, repeat mild and more severe TBI to establish developmental windows of susceptibility and altered maturational trajectories. Research progress for pediatric TBI is critically important to reveal age-associated mechanisms and to determine knowledge gaps for future studies.
Collapse
Affiliation(s)
- Rebecka O Serpa
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| | - Lindsay Ferguson
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| | - Cooper Larson
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| | - Julie Bailard
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| | - Samantha Cooke
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| | - Tiffany Greco
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| | - Mayumi L Prins
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
10
|
Verboon LN, Patel HC, Greenhalgh AD. The Immune System's Role in the Consequences of Mild Traumatic Brain Injury (Concussion). Front Immunol 2021; 12:620698. [PMID: 33679762 PMCID: PMC7928307 DOI: 10.3389/fimmu.2021.620698] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 01/25/2021] [Indexed: 12/14/2022] Open
Abstract
Mild traumatic brain injury (mild TBI), often referred to as concussion, is the most common form of TBI and affects millions of people each year. A history of mild TBI increases the risk of developing emotional and neurocognitive disorders later in life that can impact on day to day living. These include anxiety and depression, as well as neurodegenerative conditions such as chronic traumatic encephalopathy (CTE) and Alzheimer's disease (AD). Actions of brain resident or peripherally recruited immune cells are proposed to be key regulators across these diseases and mood disorders. Here, we will assess the impact of mild TBI on brain and patient health, and evaluate the recent evidence for immune cell involvement in its pathogenesis.
Collapse
Affiliation(s)
- Laura N. Verboon
- Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, The University of Manchester, Manchester, United Kingdom
| | - Hiren C. Patel
- Division of Cardiovascular Sciences, Salford Royal National Health Service Foundation Trust, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
- Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance National Health Service Group, University of Manchester, Manchester, United Kingdom
| | - Andrew D. Greenhalgh
- Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, The University of Manchester, Manchester, United Kingdom
- Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance National Health Service Group, University of Manchester, Manchester, United Kingdom
- Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
11
|
Traumatic Brain Injury Causes Chronic Cortical Inflammation and Neuronal Dysfunction Mediated by Microglia. J Neurosci 2021; 41:1597-1616. [PMID: 33452227 DOI: 10.1523/jneurosci.2469-20.2020] [Citation(s) in RCA: 233] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/03/2020] [Accepted: 12/14/2020] [Indexed: 01/02/2023] Open
Abstract
Traumatic brain injury (TBI) can lead to significant neuropsychiatric problems and neurodegenerative pathologies, which develop and persist years after injury. Neuroinflammatory processes evolve over this same period. Therefore, we aimed to determine the contribution of microglia to neuropathology at acute [1 d postinjury (dpi)], subacute (7 dpi), and chronic (30 dpi) time points. Microglia were depleted with PLX5622, a CSF1R antagonist, before midline fluid percussion injury (FPI) in male mice and cortical neuropathology/inflammation was assessed using a neuropathology mRNA panel. Gene expression associated with inflammation and neuropathology were robustly increased acutely after injury (1 dpi) and the majority of this expression was microglia independent. At 7 and 30 dpi, however, microglial depletion reversed TBI-related expression of genes associated with inflammation, interferon signaling, and neuropathology. Myriad suppressed genes at subacute and chronic endpoints were attributed to neurons. To understand the relationship between microglia, neurons, and other glia, single-cell RNA sequencing was completed 7 dpi, a critical time point in the evolution from acute to chronic pathogenesis. Cortical microglia exhibited distinct TBI-associated clustering with increased type-1 interferon and neurodegenerative/damage-related genes. In cortical neurons, genes associated with dopamine signaling, long-term potentiation, calcium signaling, and synaptogenesis were suppressed. Microglial depletion reversed the majority of these neuronal alterations. Furthermore, there was reduced cortical dendritic complexity 7 dpi, reduced neuronal connectively 30 dpi, and cognitive impairment 30 dpi. All of these TBI-associated functional and behavioral impairments were prevented by microglial depletion. Collectively, these studies indicate that microglia promote persistent neuropathology and long-term functional impairments in neuronal homeostasis after TBI.SIGNIFICANCE STATEMENT Millions of traumatic brain injuries (TBIs) occur in the United States alone each year. Survivors face elevated rates of cognitive and psychiatric complications long after the inciting injury. Recent studies of human brain injury link chronic neuroinflammation to adverse neurologic outcomes, suggesting that evolving inflammatory processes may be an opportunity for intervention. Here, we eliminate microglia to compare the effects of diffuse TBI on neurons in the presence and absence of microglia and microglia-mediated inflammation. In the absence of microglia, neurons do not undergo TBI-induced changes in gene transcription or structure. Microglial elimination prevented TBI-induced cognitive changes 30 d postinjury (dpi). Therefore, microglia have a critical role in disrupting neuronal homeostasis after TBI, particularly at subacute and chronic timepoints.
Collapse
|
12
|
Quinpirole-Mediated Regulation of Dopamine D2 Receptors Inhibits Glial Cell-Induced Neuroinflammation in Cortex and Striatum after Brain Injury. Biomedicines 2021; 9:biomedicines9010047. [PMID: 33430188 PMCID: PMC7825629 DOI: 10.3390/biomedicines9010047] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/29/2020] [Accepted: 01/04/2021] [Indexed: 12/13/2022] Open
Abstract
Brain injury is a significant risk factor for chronic gliosis and neurodegenerative diseases. Currently, no treatment is available for neuroinflammation caused by the action of glial cells following brain injury. In this study, we investigated the quinpirole-mediated activation of dopamine D2 receptors (D2R) in a mouse model of traumatic brain injury (TBI). We also investigated the neuroprotective effects of quinpirole (a D2R agonist) against glial cell-induced neuroinflammation secondary to TBI in adult mice. After the brain injury, we injected quinpirole into the TBI mice at a dose of 1 mg/kg daily intraperitoneally for 7 days. Our results showed suppression of D2R expression and deregulation of downstream signaling molecules in ipsilateral cortex and striatum after TBI on day 7. Quinpirole administration regulated D2R expression and significantly reduced glial cell-induced neuroinflammation via the D2R/Akt/glycogen synthase kinase 3 beta (GSK3-β) signaling pathway after TBI. Quinpirole treatment concomitantly attenuated increase in glial cells, neuronal apoptosis, synaptic dysfunction, and regulated proteins associated with the blood–brain barrier, together with the recovery of lesion volume in the TBI mouse model. Additionally, our in vitro results confirmed that quinpirole reversed the microglial condition media complex-mediated deleterious effects and regulated D2R levels in HT22 cells. This study showed that quinpirole administration after TBI reduced secondary brain injury-induced glial cell activation and neuroinflammation via regulation of the D2R/Akt/GSK3-β signaling pathways. Our study suggests that quinpirole may be a safe therapeutic agent against TBI-induced neurodegeneration.
Collapse
|
13
|
Wang WX, Prajapati P, Vekaria HJ, Spry M, Cloud AL, Sullivan PG, Springer JE. Temporal changes in inflammatory mitochondria-enriched microRNAs following traumatic brain injury and effects of miR-146a nanoparticle delivery. Neural Regen Res 2021; 16:514-522. [PMID: 32985480 PMCID: PMC7996041 DOI: 10.4103/1673-5374.293149] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNA molecules that regulate post-transcriptional gene expression and contribute to all aspects of cellular function. We previously reported that the activities of several mitochondria-enriched miRNAs regulating inflammation (i.e., miR-142-3p, miR-142-5p, and miR-146a) are altered in the hippocampus at 3-12 hours following a severe traumatic brain injury. In the present study, we investigated the temporal expression profile of these inflammatory miRNAs in mitochondria and cytosol fractions at more chronic post-injury times following severe controlled cortical impact injury in rats. In addition, several inflammatory genes were analyzed in the cytosol fractions. The analysis showed that while elevated levels were observed in cytoplasm, the mitochondria-enriched miRNAs, miR-142-3p and miR-142-5p continued to be significantly reduced in mitochondria from injured hippocampi for at least 3 days and returned to near normal levels at 7 days post-injury. Although not statistically significant, miR-146a also remained at reduced levels for up to 3 days following controlled cortical impact injury, and recovered by 7 days. In contrast, miRNAs that are not enriched in mitochondria, including miR-124a, miR-150, miR-19b, miR-155, and miR-223 were either increased or demonstrated no change in their levels in mitochondrial fractions for 7 days. The one exception was that miR-223 levels were reduced in mitochondria at 1 day following injury. No major alterations were observed in sham operated animals. This temporal pattern was unique to mitochondria-enriched miRNAs and correlated with injury-induced changes in mitochondrial bioenergetics as well as expression levels of several inflammatory markers. These observations suggested a potential compartmental re-distribution of the mitochondria-enriched inflammatory miRNAs and may reflect an intracellular mechanism by which specific miRNAs regulate injury-induced inflammatory signaling. To test this, we utilized a novel peptide-based nanoparticle strategy for in vitro and in vivo delivery of a miR-146a mimic as a potential therapeutic strategy for targeting nuclear factor-kappaB inflammatory modulators in the injured brain. Nanoparticle delivery of miR-146a to BV-2 or SH-SY5Y cells significantly reduced expression of TNF receptor-associated factor 6 (TRAF6) and interleukin-1 receptor-associated kinase 1 (IRAK1), two important modulators of the nuclear factor-kappaB (NF-κB) pro-inflammatory pathway. Moreover, injections of miR-146a containing nanoparticles into the brain immediately following controlled cortical impact injury significantly reduced hippocampal TNF receptor-associated factor 6 and interleukin-1 receptor-associated kinase 1 levels. Taken together, our studies demonstrate the subcellular alteration of inflammatory miRNAs after traumatic brain injury and establish proof of principle that nanoparticle delivery of miR-146a has therapeutic potential for modulating pro-inflammatory effectors in the injured brain. All of the studies performed were approved by the University of Kentucky Institutional Animal Care and Usage Committee (IACUC protocol # 2014-1300) on August 17, 2017.
Collapse
Affiliation(s)
- Wang-Xia Wang
- Sanders Brown Center on Aging; Spinal Cord and Brain Injury Research Center; Department of Pathology & Laboratory Medicine, University of Kentucky, Lexington, KY, USA
| | - Paresh Prajapati
- Spinal Cord and Brain Injury Research Center; Department of Neuroscience, University of Kentucky, Lexington, KY, USA
| | - Hemendra J Vekaria
- Spinal Cord and Brain Injury Research Center; Department of Neuroscience, University of Kentucky, Lexington, KY, USA
| | - Malinda Spry
- Spinal Cord and Brain Injury Research Center; Department of Neuroscience, University of Kentucky, Lexington, KY, USA
| | - Amber L Cloud
- Spinal Cord and Brain Injury Research Center; Department of Neuroscience, University of Kentucky, Lexington, KY, USA
| | - Patrick G Sullivan
- Spinal Cord and Brain Injury Research Center; Department of Neuroscience, University of Kentucky, Lexington, KY, USA
| | - Joe E Springer
- Spinal Cord and Brain Injury Research Center; Department of Neuroscience, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
14
|
Fakhoury M, Shakkour Z, Kobeissy F, Lawand N. Depression following traumatic brain injury: a comprehensive overview. Rev Neurosci 2020; 32:289-303. [PMID: 33661587 DOI: 10.1515/revneuro-2020-0037] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 10/21/2020] [Indexed: 12/19/2022]
Abstract
Traumatic brain injury (TBI) represents a major health concern affecting the neuropsychological health; TBI is accompanied by drastic long-term adverse complications that can influence many aspects of the life of affected individuals. A substantial number of studies have shown that mood disorders, particularly depression, are the most frequent complications encountered in individuals with TBI. Post-traumatic depression (P-TD) is present in approximately 30% of individuals with TBI, with the majority of individuals experiencing symptoms of depression during the first year following head injury. To date, the mechanisms of P-TD are far from being fully understood, and effective treatments that completely halt this condition are still lacking. The aim of this review is to outline the current state of knowledge on the prevalence and risk factors of P-TD, to discuss the accompanying brain changes at the anatomical, molecular and functional levels, and to discuss current approaches used for the treatment of P-TD.
Collapse
Affiliation(s)
- Marc Fakhoury
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Zaynab Shakkour
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Nada Lawand
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,Department of Neurology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
15
|
Macheda T, Roberts KN, Morganti JM, Braun DJ, Bachstetter AD. Optimization and validation of a modified radial-arm water maze protocol using a murine model of mild closed head traumatic brain injury. PLoS One 2020; 15:e0232862. [PMID: 32810143 PMCID: PMC7433858 DOI: 10.1371/journal.pone.0232862] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 06/27/2020] [Indexed: 11/25/2022] Open
Abstract
Cognitive impairments can be a significant problem after a traumatic brain injury (TBI), which affects millions worldwide each year. There is a need for establish reproducible cognitive assays in rodents to better understand disease mechanisms and to develop therapeutic interventions towards treating TBI-induced impairments. Our goal was to validate and standardize the radial arm water maze (RAWM) test as an assay to screen for cognitive impairments caused by TBI. RAWM is a visuo-spatial learning test, originally designed for use with rats, and later adapted for mice. The present study investigates whether test procedures, such us the presence of extra-maze cues influences learning and memory performance. C57BL/6 mice were tested in an 8-arm RAWM using a four-day protocol. We demonstrated that two days of training, exposing the mice to extra-maze cues and a visible platform, influenced learning and memory performance. Mice that did not receive training performed poorer compared to mice trained. To further validate our RAWM protocol, we used scopolamine. We, also, demonstrated that a single mild closed head injury (CHI) caused deficits in this task at two weeks post-CHI. Our data supported the use of 7 trials per day and a spaced training protocol as key factor to unmask memory impairment following CHI. Here, we provide a detailed standard operating procedure for RAWM test, which can be applied to a variety of mouse models including neurodegenerative diseases and pathology, as well as when pharmacological approaches are used.
Collapse
Affiliation(s)
- Teresa Macheda
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States of America
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America
| | - Kelly N. Roberts
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States of America
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America
| | - Josh M. Morganti
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States of America
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America
| | - David J. Braun
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America
| | - Adam D. Bachstetter
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States of America
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America
- * E-mail:
| |
Collapse
|
16
|
Gao J, Chen N, Li N, Xu F, Wang W, Lei Y, Shi J, Gong Q. Neuroprotective Effects of Trilobatin, a Novel Naturally Occurring Sirt3 Agonist from Lithocarpus polystachyus Rehd., Mitigate Cerebral Ischemia/Reperfusion Injury: Involvement of TLR4/NF-κB and Nrf2/Keap-1 Signaling. Antioxid Redox Signal 2020; 33:117-143. [PMID: 32212827 DOI: 10.1089/ars.2019.7825] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Aims: Neuroinflammation and oxidative stress are deemed the prime causes of brain injury after cerebral ischemia/reperfusion (I/R). Since the silent mating-type information regulation 2 homologue 3 (Sirt3) pathway plays an imperative role in protecting against neuroinflammation and oxidative stress, it has been verified as a target to treat ischemia stroke. Therefore, we attempted to seek novel Sirt3 agonist and explore its underlying mechanism for stroke treatment both in vivo and in vitro. Results: Trilobatin (TLB) not only dramatically suppressed neuroinflammation and oxidative stress injury after middle cerebral artery occlusion in rats, but also effectively mitigated oxygen and glucose deprivation/reoxygenation injury in primary cultured astrocytes. These beneficial effects, along with the reduced proinflammatory cytokines via suppressing Toll-like receptor 4 (TLR4) signaling pathway, lessened oxidative injury via activating nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathways, in keeping with the findings in vivo. Intriguingly, the TLB-mediated neuroprotection on cerebral I/R injury was modulated by reciprocity between TLR4-mediated neuroinflammatory responses and Nrf2 antioxidant responses as evidenced by molecular docking and silencing TLR4 and Nrf2, respectively. Most importantly, TLB not only directly bonded to Sirt3 but also increased Sirt3 expression and activity, indicating that Sirt3 might be a promising therapeutic target of TLB. Innovation: TLB is a naturally occurring Sirt3 agonist with potent neuroprotective effects via regulation of TLR4/nuclear factor-kappa B and Nrf2/Kelch-like ECH-associated protein 1 (Keap-1) signaling pathways both in vivo and in vitro. Conclusion: Our findings indicate that TLB protects against cerebral I/R-induced neuroinflammation and oxidative injury through the regulation of neuroinflammatory and oxidative responses via TLR4, Nrf2, and Sirt3, suggesting that TLB might be a promising Sirt3 agonist against ischemic stroke.
Collapse
Affiliation(s)
- Jianmei Gao
- Department of Clinical Pharmacotherapeutics, School of Pharmacy, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Nana Chen
- Department of Clinical Pharmacotherapeutics, School of Pharmacy, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Na Li
- Department of Clinical Pharmacotherapeutics, School of Pharmacy, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Fan Xu
- Department of Clinical Pharmacotherapeutics, School of Pharmacy, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Wei Wang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Yaying Lei
- Department of Clinical Pharmacotherapeutics, School of Pharmacy, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Jingshan Shi
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Qihai Gong
- Department of Clinical Pharmacotherapeutics, School of Pharmacy, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China
| |
Collapse
|
17
|
Witcher KG, Dziabis JE, Bray CE, Gordillo AJ, Kumar JE, Eiferman DS, Godbout JP, Kokiko-Cochran ON. Comparison between midline and lateral fluid percussion injury in mice reveals prolonged but divergent cortical neuroinflammation. Brain Res 2020; 1746:146987. [PMID: 32592739 DOI: 10.1016/j.brainres.2020.146987] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 05/25/2020] [Accepted: 06/13/2020] [Indexed: 01/19/2023]
Abstract
Animal models are critical for determining the mechanisms mediating traumatic brain injury-induced (TBI) neuropathology. Fluid percussion injury (FPI) is a widely used model of brain injury typically applied either midline or parasagittally (lateral). Midline FPI induces a diffuse TBI, while lateral FPI induces both focal cortical injury (ipsilateral hemisphere) and diffuse injury (contralateral hemisphere). Nonetheless, discrete differences in neuroinflammation and neuropathology between these two versions of FPI remain unclear. The purpose of this study was to compare acute (4-72 h) and subacute (7 days) neuroinflammatory responses between midline and lateral FPI. Midline FPI resulted in longer righting reflex times than lateral FPI. At acute time points, the inflammatory responses to the two different injuries were similar. For instance, there was evidence of monocytes and cytokine mRNA expression in the brain with both injuries acutely. Midline FPI had the highest proportion of brain monocytes and highest IL-1β/TNFα mRNA expression 24 h later. NanoString nCounter analysis 7 days post-injury revealed robust and prolonged expression of inflammatory-related genes in the cortex after midline FPI compared to lateral FPI; however, Iba-1 cortical immunoreactivity was increased with lateral FPI. Thus, midline and lateral FPI caused similar cortical neuroinflammatory responses acutely and mRNA expression of inflammatory genes was detectable in the brain 7 days later. The primary divergence was that inflammatory gene expression was greater and more diverse subacutely after midline FPI. These results provide novel insight to variations between midline and lateral FPI, which may recapitulate unique temporal pathogenesis.
Collapse
Affiliation(s)
- Kristina G Witcher
- Department of Neuroscience, The Ohio State University, 333 W 10(th) Ave, Columbus, OH 43210, USA; Institute for Behavioral Medicine Research, The Ohio State University, 460 Medical Center Dr, Columbus, OH 43210, USA
| | - Julia E Dziabis
- Department of Neuroscience, The Ohio State University, 333 W 10(th) Ave, Columbus, OH 43210, USA; Institute for Behavioral Medicine Research, The Ohio State University, 460 Medical Center Dr, Columbus, OH 43210, USA
| | - Chelsea E Bray
- Department of Neuroscience, The Ohio State University, 333 W 10(th) Ave, Columbus, OH 43210, USA; Institute for Behavioral Medicine Research, The Ohio State University, 460 Medical Center Dr, Columbus, OH 43210, USA
| | - Alan J Gordillo
- Department of Neuroscience, The Ohio State University, 333 W 10(th) Ave, Columbus, OH 43210, USA; Institute for Behavioral Medicine Research, The Ohio State University, 460 Medical Center Dr, Columbus, OH 43210, USA
| | - Julia E Kumar
- Department of Neuroscience, The Ohio State University, 333 W 10(th) Ave, Columbus, OH 43210, USA; Institute for Behavioral Medicine Research, The Ohio State University, 460 Medical Center Dr, Columbus, OH 43210, USA
| | - Daniel S Eiferman
- Department of Surgery, The Ohio State University, 395 W 12(th) Ave, Columbus, OH 43210, USA
| | - Jonathan P Godbout
- Department of Neuroscience, The Ohio State University, 333 W 10(th) Ave, Columbus, OH 43210, USA; Center for Brain and Spinal Cord Repair, The Ohio State University, 460 W 12(th) Ave, Columbus, OH 43210, USA; Institute for Behavioral Medicine Research, The Ohio State University, 460 Medical Center Dr, Columbus, OH 43210, USA
| | - Olga N Kokiko-Cochran
- Department of Neuroscience, The Ohio State University, 333 W 10(th) Ave, Columbus, OH 43210, USA; Center for Brain and Spinal Cord Repair, The Ohio State University, 460 W 12(th) Ave, Columbus, OH 43210, USA; Institute for Behavioral Medicine Research, The Ohio State University, 460 Medical Center Dr, Columbus, OH 43210, USA.
| |
Collapse
|
18
|
Bachstetter AD, Morganti JM, Bodnar CN, Webster SJ, Higgins EK, Roberts KN, Snider H, Meier SE, Nation GK, Goulding DS, Hamm M, Powell DK, Vandsburger M, Van Eldik LJ, Abisambra JF. The effects of mild closed head injuries on tauopathy and cognitive deficits in rodents: Primary results in wild type and rTg4510 mice, and a systematic review. Exp Neurol 2020; 326:113180. [PMID: 31930992 PMCID: PMC7373372 DOI: 10.1016/j.expneurol.2020.113180] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 12/02/2019] [Accepted: 01/09/2020] [Indexed: 12/17/2022]
Abstract
In humans, the majority of sustained traumatic brain injuries (TBIs) are classified as 'mild' and most often a result of a closed head injury (CHI). The effects of a non-penetrating CHI are not benign and may lead to chronic pathology and behavioral dysfunction, which could be worsened by repeated head injury. Clinical-neuropathological correlation studies provide evidence that conversion of tau into abnormally phosphorylated proteotoxic intermediates (p-tau) could be part of the pathophysiology triggered by a single TBI and enhanced by repeated TBIs. However, the link between p-tau and CHI in rodents remains controversial. To address this question experimentally, we induced a single CHI or two CHIs to WT or rTg4510 mice. We found that 2× CHI increased tau phosphorylation in WT mice and rTg4510 mice. Behavioral characterization in WT mice found chronic deficits in the radial arm water maze in 2× CHI mice that had partially resolved in the 1× CHI mice. Moreover, using Manganese-Enhanced Magnetic Resonance Imaging with R1 mapping - a novel functional neuroimaging technique - we found greater deficits in the rTg4510 mice following 2× CHI compared to 1× CHI. To integrate our findings with prior work in the field, we conducted a systematic review of rodent mild repetitive CHI studies. Following Prisma guidelines, we identified 25 original peer-reviewed papers. Results from our experiments, as well as our systematic review, provide compelling evidence that tau phosphorylation is modified by experimental mild TBI studies; however, changes in p-tau levels are not universally reported. Together, our results provide evidence that repetitive TBIs can result in worse and more persistent neurological deficits compared to a single TBI, but the direct link between the worsened outcome and elevated p-tau could not be established.
Collapse
Affiliation(s)
- Adam D Bachstetter
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States of America; Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America.
| | - Josh M Morganti
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States of America; Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America
| | - Colleen N Bodnar
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States of America; Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America
| | - Scott J Webster
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America
| | - Emma K Higgins
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States of America; Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America
| | - Kelly N Roberts
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States of America; Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America
| | - Henry Snider
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States of America; Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America
| | - Shelby E Meier
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America; Department of Physiology, University of Kentucky, Lexington, KY, United States of America
| | - Grant K Nation
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America; Department of Physiology, University of Kentucky, Lexington, KY, United States of America
| | - Danielle S Goulding
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America
| | - Matthew Hamm
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, United States of America
| | - David K Powell
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America; Department of Biomedical Engineering, University of Kentucky, Lexington, KY, United States of America
| | - Moriel Vandsburger
- Department of Bioengineering, University of California Berkeley, Berkeley, CA, United States of America
| | - Linda J Van Eldik
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States of America; Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America
| | - Jose F Abisambra
- Department of Physiology, University of Kentucky, Lexington, KY, United States of America; Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, United States of America.
| |
Collapse
|
19
|
Bodnar CN, Roberts KN, Higgins EK, Bachstetter AD. A Systematic Review of Closed Head Injury Models of Mild Traumatic Brain Injury in Mice and Rats. J Neurotrauma 2019; 36:1683-1706. [PMID: 30661454 PMCID: PMC6555186 DOI: 10.1089/neu.2018.6127] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Mild TBI (mTBI) is a significant health concern. Animal models of mTBI are essential for understanding mechanisms, and pathological outcomes, as well as to test therapeutic interventions. A variety of closed head models of mTBI that incorporate different aspects (i.e., biomechanics) of the mTBI have been reported. The aim of the current review was to compile a comprehensive list of the closed head mTBI rodent models, along with the common data elements, and outcomes, with the goal to summarize the current state of the field. Publications were identified from a search of PubMed and Web of Science and screened for eligibility following PRISMA guidelines. Articles were included that were closed head injuries in which the authors classified the injury as mild in rats or mice. Injury model and animal-specific common data elements, as well as behavioral and histological outcomes, were collected and compiled from a total of 402 articles. Our results outline the wide variety of methods used to model mTBI. We also discovered that female rodents and both young and aged animals are under-represented in experimental mTBI studies. Our findings will aid in providing context comparing the injury models and provide a starting point for the selection of the most appropriate model of mTBI to address a specific hypothesis. We believe this review will be a useful starting place for determining what has been done and what knowledge is missing in the field to reduce the burden of mTBI.
Collapse
Affiliation(s)
- Colleen N. Bodnar
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky
| | - Kelly N. Roberts
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky
| | - Emma K. Higgins
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky
| | - Adam D. Bachstetter
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
20
|
Morganti-Kossmann MC, Semple BD, Hellewell SC, Bye N, Ziebell JM. The complexity of neuroinflammation consequent to traumatic brain injury: from research evidence to potential treatments. Acta Neuropathol 2019; 137:731-755. [PMID: 30535946 DOI: 10.1007/s00401-018-1944-6] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 11/19/2018] [Accepted: 12/01/2018] [Indexed: 12/18/2022]
Abstract
This review recounts the definitions and research evidence supporting the multifaceted roles of neuroinflammation in the injured brain following trauma. We summarise the literature fluctuating from the protective and detrimental properties that cytokines, leukocytes and glial cells play in the acute and chronic stages of TBI, including the intrinsic factors that influence cytokine responses and microglial functions relative to genetics, sex, and age. We elaborate on the pros and cons that cytokines, chemokines, and microglia play in brain repair, specifically neurogenesis, and how such conflicting roles may be harnessed therapeutically to sustain the survival of new neurons. With a brief review of the clinical and experimental findings demonstrating early and chronic inflammation impacts on outcomes, we focus on the clinical conditions that may be amplified by neuroinflammation, ranging from acute seizures to chronic epilepsy, neuroendocrine dysfunction, dementia, depression, post-traumatic stress disorder and chronic traumatic encephalopathy. Finally, we provide an overview of the therapeutic agents that have been tested to reduce inflammation-driven secondary pathological cascades and speculate the future promise of alternative drugs.
Collapse
Affiliation(s)
- Maria Cristina Morganti-Kossmann
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, VIC, Australia.
- Australian New Zealand Intensive Care Research Centre, Melbourne, VIC, Australia.
| | - Bridgette D Semple
- Department of Neuroscience, Central Clinical School, The Alfred Hospital, Monash University, Melbourne, VIC, Australia
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, Australia
| | - Sarah C Hellewell
- Sydney Translational Imaging Laboratory, Charles Perkins Centre, Heart Research Institute, University of Sydney, Sydney, NSW, Australia
| | - Nicole Bye
- Department of Pharmacy, College of Health and Medicine, University of Tasmania, Sandy Bay, TAS, Australia
| | - Jenna M Ziebell
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia
| |
Collapse
|
21
|
Flygt J, Ruscher K, Norberg A, Mir A, Gram H, Clausen F, Marklund N. Neutralization of Interleukin-1β following Diffuse Traumatic Brain Injury in the Mouse Attenuates the Loss of Mature Oligodendrocytes. J Neurotrauma 2018; 35:2837-2849. [PMID: 29690837 PMCID: PMC6247990 DOI: 10.1089/neu.2018.5660] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Traumatic brain injury (TBI) commonly results in injury to the components of the white matter tracts, causing post-injury cognitive deficits. The myelin-producing oligodendrocytes (OLs) are vulnerable to TBI, although may potentially be replaced by proliferating oligodendrocyte progenitor cells (OPCs). The cytokine interleukin-1β (IL-1β) is a key mediator of the complex inflammatory response, and when neutralized in experimental TBI, behavioral outcome was improved. To evaluate the role of IL-1β on oligodendrocyte cell death and OPC proliferation, 116 adult male mice subjected to sham injury or the central fluid percussion injury (cFPI) model of traumatic axonal injury, were analyzed at two, seven, and 14 days post-injury. At 30 min post-injury, mice were randomly administered an IL-1β neutralizing or a control antibody. OPC proliferation (5-ethynyl 2'- deoxyuridine (EdU)/Olig2 co-labeling) and mature oligodendrocyte cell loss was evaluated in injured white matter tracts. Microglia/macrophages immunohistochemistry and ramification using Sholl analysis were also evaluated. Neutralizing IL-1β resulted in attenuated cell death, indicated by cleaved caspase-3 expression, and attenuated loss of mature OLs from two to seven days post-injury in brain-injured animals. IL-1β neutralization also attenuated the early, two day post-injury increase of microglia/macrophage immunoreactivity and altered their ramification. The proliferation of OPCs in brain-injured animals was not altered, however. Our data suggest that IL-1β is involved in the TBI-induced loss of OLs and early microglia/macrophage activation, although not the OPC proliferation. Attenuated oligodendrocyte cell loss may contribute to the improved behavioral outcome observed by IL-1β neutralization in this mouse model of diffuse TBI.
Collapse
Affiliation(s)
- Johanna Flygt
- 1 Department of Neuroscience, Section of Neurosurgery, Uppsala University , Uppsala, Sweden
| | - Karsten Ruscher
- 2 Novartis Institutes of Biomedical Research , Basel, Switzerland
| | - Amanda Norberg
- 1 Department of Neuroscience, Section of Neurosurgery, Uppsala University , Uppsala, Sweden
| | - Anis Mir
- 3 Lund University, Skane University Hospital , Department of Clinical Sciences Lund, Neurosurgery, Lund, Sweden
| | - Hermann Gram
- 3 Lund University, Skane University Hospital , Department of Clinical Sciences Lund, Neurosurgery, Lund, Sweden
| | - Fredrik Clausen
- 1 Department of Neuroscience, Section of Neurosurgery, Uppsala University , Uppsala, Sweden
| | - Niklas Marklund
- 1 Department of Neuroscience, Section of Neurosurgery, Uppsala University , Uppsala, Sweden .,3 Lund University, Skane University Hospital , Department of Clinical Sciences Lund, Neurosurgery, Lund, Sweden
| |
Collapse
|
22
|
Lyons DN, Vekaria H, Macheda T, Bakshi V, Powell DK, Gold BT, Lin AL, Sullivan PG, Bachstetter AD. A Mild Traumatic Brain Injury in Mice Produces Lasting Deficits in Brain Metabolism. J Neurotrauma 2018; 35:2435-2447. [PMID: 29808778 PMCID: PMC6196750 DOI: 10.1089/neu.2018.5663] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Metabolic uncoupling has been well-characterized during the first minutes-to-days after a traumatic brain injury (TBI), yet mitochondrial bioenergetics during the weeks-to-months after a brain injury is poorly defined, particularly after a mild TBI. We hypothesized that a closed head injury (CHI) would be associated with deficits in mitochondrial bioenergetics at one month after the injury. A significant decrease in state-III (adenosine triphosphate production) and state-V (complex-I) driven mitochondrial respiration was found at one month post-injury in adult C57Bl/6J mice. Isolation of synaptic mitochondria demonstrated that the deficit in state-III and state-V was primarily neuronal. Injured mice had a temporally consistent deficit in memory recall at one month post-injury. Using proton magnetic resonance spectroscopy (1H MRS) at 7-Tesla, we found significant decreases in phosphocreatine, N-Acetylaspartic acid, and total choline. We also found regional variations in cerebral blood flow, including both hypo- and hyperperfusion, as measured by a pseudocontinuous arterial spin labeling MR sequence. Our results highlight a chronic deficit in mitochondrial bioenergetics associated with a CHI that may lead toward a novel approach for neurorestoration after a mild TBI. MRS provides a potential biomarker for assessing the efficacy of candidate treatments targeted at improving mitochondrial bioenergetics.
Collapse
Affiliation(s)
- Danielle N Lyons
- 1 Spinal Cord & Brain Injury Research Center, University of Kentucky , Lexington Kentucky.,2 Department of Neuroscience, University of Kentucky , Lexington Kentucky
| | - Hemendra Vekaria
- 1 Spinal Cord & Brain Injury Research Center, University of Kentucky , Lexington Kentucky.,2 Department of Neuroscience, University of Kentucky , Lexington Kentucky
| | - Teresa Macheda
- 1 Spinal Cord & Brain Injury Research Center, University of Kentucky , Lexington Kentucky.,2 Department of Neuroscience, University of Kentucky , Lexington Kentucky
| | - Vikas Bakshi
- 4 Sanders-Brown Center on Aging, University of Kentucky , Lexington Kentucky.,5 Department of Pharmacology and Nutritional Sciences, University of Kentucky , Lexington Kentucky
| | - David K Powell
- 2 Department of Neuroscience, University of Kentucky , Lexington Kentucky.,3 Department of Biomedical Engineering, University of Kentucky , Lexington Kentucky
| | - Brian T Gold
- 2 Department of Neuroscience, University of Kentucky , Lexington Kentucky
| | - Ai-Ling Lin
- 4 Sanders-Brown Center on Aging, University of Kentucky , Lexington Kentucky.,5 Department of Pharmacology and Nutritional Sciences, University of Kentucky , Lexington Kentucky
| | - Patrick G Sullivan
- 1 Spinal Cord & Brain Injury Research Center, University of Kentucky , Lexington Kentucky.,2 Department of Neuroscience, University of Kentucky , Lexington Kentucky
| | - Adam D Bachstetter
- 1 Spinal Cord & Brain Injury Research Center, University of Kentucky , Lexington Kentucky.,2 Department of Neuroscience, University of Kentucky , Lexington Kentucky
| |
Collapse
|
23
|
Späni CB, Braun DJ, Van Eldik LJ. Sex-related responses after traumatic brain injury: Considerations for preclinical modeling. Front Neuroendocrinol 2018; 50:52-66. [PMID: 29753798 PMCID: PMC6139061 DOI: 10.1016/j.yfrne.2018.03.006] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/26/2018] [Accepted: 03/29/2018] [Indexed: 12/18/2022]
Abstract
Traumatic brain injury (TBI) has historically been viewed as a primarily male problem, since men are more likely to experience a TBI because of more frequent participation in activities that increase risk of head injuries. This male bias is also reflected in preclinical research where mostly male animals have been used in basic and translational science. However, with an aging population in which TBI incidence is increasingly sex-independent due to falls, and increasing female participation in high-risk activities, the attention to potential sex differences in TBI responses and outcomes will become more important. These considerations are especially relevant in designing preclinical animal models of TBI that are more predictive of human responses and outcomes. This review characterizes sex differences following TBI with a special emphasis on the contribution of the female sex hormones, progesterone and estrogen, to these differences. This information is potentially important in developing and customizing TBI treatments.
Collapse
Affiliation(s)
- Claudia B Späni
- Sanders-Brown Center on Aging, University of Kentucky, 101 Sanders-Brown Bldg., 800 S. Limestone Street, Lexington, KY 40536, USA.
| | - David J Braun
- Sanders-Brown Center on Aging, University of Kentucky, 101 Sanders-Brown Bldg., 800 S. Limestone Street, Lexington, KY 40536, USA.
| | - Linda J Van Eldik
- Sanders-Brown Center on Aging, University of Kentucky, 101 Sanders-Brown Bldg., 800 S. Limestone Street, Lexington, KY 40536, USA; Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky, B481, BBSRB, 741 S. Limestone Street, Lexington, KY 40536, USA; Department of Neuroscience, College of Medicine, University of Kentucky, UK Medical Center MN 150, Lexington, KY 40536, USA.
| |
Collapse
|
24
|
Sports-Related Concussion Results in Differential Expression of Nuclear Factor-κB Pathway Genes in Peripheral Blood During the Acute and Subacute Periods. J Head Trauma Rehabil 2018; 31:269-76. [PMID: 26479397 DOI: 10.1097/htr.0000000000000191] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To determine changes in global gene expression in peripheral leukocytes in the acute and subacute periods following a sports-related concussion in athletes. SETTING Samples were collected at 2 universities in Rochester, New York. PARTICIPANTS Fifteen contact sport athletes who experienced a sports-related concussion, and 16 nonconcussed teammates served as controls. DESIGN Blood samples were collected at the start of the season (baseline), within 6 hours of injury (acute), and at 7 days (subacute) postinjury. Differential gene expression was measured using the GeneChip 3' in vitro transcription Expression kit and Affymetrix microarrays, and genes with fold difference of 2 or more were identified using Partek. MAIN MEASURES Whole genome differential gene expression, and cognitive and balance measures to asses for clinical symptoms pre- and postinjury. RESULTS In the concussed athletes, we observed 67 downregulated and 4 upregulated genes in the acute period and 63 downregulated and 2 upregulated genes in the subacute period compared with baseline. Of these, there were 28 genes from both time points involved in the inflammatory response. No significant differences in gene expression were detected in the control group. CONCLUSIONS Our findings suggest that recovery from sports-related concussion relates to modulation of inflammation through cytokine and chemokine gene pathways, which can contribute to future development of personalized therapeutic agents.
Collapse
|
25
|
Schiavone S, Trabace L. Small Molecules: Therapeutic Application in Neuropsychiatric and Neurodegenerative Disorders. Molecules 2018; 23:molecules23020411. [PMID: 29438357 PMCID: PMC6017408 DOI: 10.3390/molecules23020411] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 02/05/2018] [Accepted: 02/07/2018] [Indexed: 12/13/2022] Open
Abstract
In recent years, an increasing number of studies have been published, focusing on the potential therapeutic use of small catalytic agents with strong biological properties. So far, most of these works have only regarded specific clinical fields, such as oncology, infectivology and general pathology, in particular with respect to the treatment of significant inflammatory processes. However, interesting data on possible therapeutic applications of small molecules for the treatment of neuropsychiatric and neurodegenerative illnesses are emerging, especially with respect to the possibility to modulate the cellular redox state. Indeed, a crucial role of redox dysregulation in the pathogenesis of these disorders has been widely demonstrated by both pre-clinical and clinical studies, being the reduction of the total amount of free radicals a promising novel therapeutic approach for these diseases. In this review, we focused our interest on studies published during the last ten years reporting therapeutic potential of small molecules for the treatment of neuropsychiatric and neurodegenerative disorders, also based on the biological efficiency of these compounds in detecting intracellular disturbances induced by increased production of reactive oxygen species.
Collapse
Affiliation(s)
- Stefania Schiavone
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli, 20, 71122 Foggia, Italy.
| | - Luigia Trabace
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli, 20, 71122 Foggia, Italy.
| |
Collapse
|
26
|
Bodnar CN, Morganti JM, Bachstetter AD. Depression following a traumatic brain injury: uncovering cytokine dysregulation as a pathogenic mechanism. Neural Regen Res 2018; 13:1693-1704. [PMID: 30136679 PMCID: PMC6128046 DOI: 10.4103/1673-5374.238604] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
A substantial number of individuals have long-lasting adverse effects from a traumatic brain injury (TBI). Depression is one of these long-term complications that influences many aspects of life. Depression can limit the ability to return to work, and even worsen cognitive function and contribute to dementia. The mechanistic cause for the increased depression risk associated with a TBI remains to be defined. As TBI results in chronic neuroinflammation, and priming of glia to a secondary challenge, the inflammatory theory of depression provides a promising framework for investigating the cause of depression following a TBI. Increases in cytokines similar to those seen in depression in the general population are also increased following a TBI. Biomarker levels of cytokines peak within hours-to-days after the injury, yet pro-inflammatory cytokines may still be elevated above physiological levels months-to-years following TBI, which is the time frame in which post-TBI depression can persist. As tumor necrosis factor α and interleukin 1 can signal directly at the neuronal synapse, pathophysiological levels of these cytokines can detrimentally alter neuronal synaptic physiology. The purpose of this review is to outline the current evidence for the inflammatory hypothesis of depression specifically as it relates to depression following a TBI. Moreover, we will illustrate the potential synaptic mechanisms by which tumor necrosis factor α and interleukin 1 could contribute to depression. The association of inflammation with the development of depression is compelling; however, in the context of post-TBI depression, the role of inflammation is understudied. This review attempts to highlight the need to understand and treat the psychological complications of a TBI, potentially by neuroimmune modulation, as the neuropsychiatric disabilities can have a great impact on the rehabilitation from the injury, and overall quality of life.
Collapse
Affiliation(s)
- Colleen N Bodnar
- Spinal Cord & Brain Injury Research Center, University of Kentucky; Department of Neuroscience, University of Kentucky, Lexington, KY, USA
| | - Josh M Morganti
- Department of Neuroscience, University of Kentucky; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Adam D Bachstetter
- Spinal Cord & Brain Injury Research Center, University of Kentucky; Department of Neuroscience, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
27
|
Inhibition of the integrated stress response reverses cognitive deficits after traumatic brain injury. Proc Natl Acad Sci U S A 2017; 114:E6420-E6426. [PMID: 28696288 DOI: 10.1073/pnas.1707661114] [Citation(s) in RCA: 159] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of long-term neurological disability, yet the mechanisms underlying the chronic cognitive deficits associated with TBI remain unknown. Consequently, there are no effective treatments for patients suffering from the long-lasting symptoms of TBI. Here, we show that TBI persistently activates the integrated stress response (ISR), a universal intracellular signaling pathway that responds to a variety of cellular conditions and regulates protein translation via phosphorylation of the translation initiation factor eIF2α. Treatment with ISRIB, a potent drug-like small-molecule inhibitor of the ISR, reversed the hippocampal-dependent cognitive deficits induced by TBI in two different injury mouse models-focal contusion and diffuse concussive injury. Surprisingly, ISRIB corrected TBI-induced memory deficits when administered weeks after the initial injury and maintained cognitive improvement after treatment was terminated. At the physiological level, TBI suppressed long-term potentiation in the hippocampus, which was fully restored with ISRIB treatment. Our results indicate that ISR inhibition at time points late after injury can reverse memory deficits associated with TBI. As such, pharmacological inhibition of the ISR emerges as a promising avenue to combat head trauma-induced chronic cognitive deficits.
Collapse
|
28
|
Rehman SU, Ahmad A, Yoon GH, Khan M, Abid MN, Kim MO. Inhibition of c-Jun N-Terminal Kinase Protects Against Brain Damage and Improves Learning and Memory After Traumatic Brain Injury in Adult Mice. Cereb Cortex 2017; 28:2854-2872. [DOI: 10.1093/cercor/bhx164] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 06/12/2017] [Indexed: 12/26/2022] Open
Affiliation(s)
- Shafiq Ur Rehman
- Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Ashfaq Ahmad
- Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Gwang-Ho Yoon
- Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Mehtab Khan
- Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Muhammad Noman Abid
- Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Myeong Ok Kim
- Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| |
Collapse
|
29
|
Zhou Z, Bachstetter AD, Späni CB, Roy SM, Watterson DM, Van Eldik LJ. Retention of normal glia function by an isoform-selective protein kinase inhibitor drug candidate that modulates cytokine production and cognitive outcomes. J Neuroinflammation 2017; 14:75. [PMID: 28381303 PMCID: PMC5382362 DOI: 10.1186/s12974-017-0845-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 03/20/2017] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Brain p38α mitogen-activated protein kinase (MAPK), a potential therapeutic target for cognitive dysfunction based on the neuroinflammation-synaptic dysfunction cycle of pathophysiology progression, offers an innovative pharmacological strategy via inhibiting the same activated target in both glia and neurons, thereby enhancing the possibility for efficacy. The highly selective, brain-penetrant p38αMAPK inhibitor MW150 attenuates cognitive dysfunction in two distinct Alzheimer's disease (AD)-relevant models and avoids the problems encountered with previous mixed-kinase inhibitor drug candidates. Therefore, it is essential that the glial effects of this CNS-active kinase inhibitor be addressed in order to anticipate future use in clinical investigations. METHODS We explored the effects of MW150 on glial biology in the AD-relevant APP/PS1 knock-in (KI) mouse model where we previously showed efficacy in suppression of hippocampal-dependent associative and spatial memory deficits. MW150 (2.5 mg/kg/day) was administered daily to 11-12-month-old KI mice for 14 days, and levels of proinflammatory cytokines IL-1β, TNFα, and IL-6 measured in homogenates of mouse cortex using ELISA. Glial markers IBA1, CD45, CD68, and GFAP were assessed by immunohistochemistry. Microglia and amyloid plaques were quantified by immunofluorescence staining followed by confocal imaging. Levels of soluble and insoluble of Aβ40 and Aβ42 were measured by ELISA. The studies of in vivo pharmacodynamic effects on markers of neuroinflammation were complemented by mechanistic studies in the murine microglia BV2 cell line, using live cell imaging techniques to monitor proliferation, migration, and phagocytosis activities. RESULTS Intervention with MW150 in KI mice during the established therapeutic time window attenuated the increased levels of IL-1β and TNFα but not IL-6. MW150 treatment also increased the IBA1+ microglia within a 15 μm radius of the amyloid plaques, without significantly affecting overall microglia or plaque volume. Levels of IBA1, CD45, CD68, GFAP, and Aβ40 and Aβ42 were not affected by MW150 treatment. MW150 did not significantly alter microglial migration, proliferation, or phagocytosis in BV2 cells. CONCLUSIONS Our results demonstrate that MW150 at an efficacious dose can selectively modulate neuroinflammatory responses associated with pathology progression without pan-suppression of normal physiological functions of microglia.
Collapse
Affiliation(s)
- Zhengqiu Zhou
- Sanders-Brown Center on Aging, University of Kentucky, 800 S. Limestone Street, Lexington, KY, USA
| | - Adam D Bachstetter
- Sanders-Brown Center on Aging, University of Kentucky, 800 S. Limestone Street, Lexington, KY, USA.,Spinal Cord and Brain Injury Research Center, University of Kentucky, 741 S. Limestone Street, Lexington, KY, USA.,Department of Neuroscience, University of Kentucky, 800 Rose Street, Lexington, KY, USA
| | - Claudia B Späni
- Sanders-Brown Center on Aging, University of Kentucky, 800 S. Limestone Street, Lexington, KY, USA
| | - Saktimayee M Roy
- Department of Pharmacology, Northwestern University, 303 E Chicago Ave, Chicago, IL, USA
| | - D Martin Watterson
- Department of Pharmacology, Northwestern University, 303 E Chicago Ave, Chicago, IL, USA
| | - Linda J Van Eldik
- Sanders-Brown Center on Aging, University of Kentucky, 800 S. Limestone Street, Lexington, KY, USA. .,Spinal Cord and Brain Injury Research Center, University of Kentucky, 741 S. Limestone Street, Lexington, KY, USA. .,Department of Neuroscience, University of Kentucky, 800 Rose Street, Lexington, KY, USA.
| |
Collapse
|
30
|
Andreasson KI, Bachstetter AD, Colonna M, Ginhoux F, Holmes C, Lamb B, Landreth G, Lee DC, Low D, Lynch MA, Monsonego A, O’Banion MK, Pekny M, Puschmann T, Russek-Blum N, Sandusky LA, Selenica MLB, Takata K, Teeling J, Town T, Van Eldik LJ, Russek-Blum N, Monsonego A, Low D, Takata K, Ginhoux F, Town T, O’Banion MK, Lamb B, Colonna M, Landreth G, Andreasson KI, Sandusky LA, Selenica MLB, Lee DC, Holmes C, Teeling J, Lynch MA, Van Eldik LJ, Bachstetter AD, Pekny M, Puschmann T. Targeting innate immunity for neurodegenerative disorders of the central nervous system. J Neurochem 2016; 138:653-93. [PMID: 27248001 PMCID: PMC5433264 DOI: 10.1111/jnc.13667] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 04/01/2016] [Accepted: 04/30/2016] [Indexed: 12/21/2022]
Abstract
Neuroinflammation is critically involved in numerous neurodegenerative diseases, and key signaling steps of innate immune activation hence represent promising therapeutic targets. This mini review series originated from the 4th Venusberg Meeting on Neuroinflammation held in Bonn, Germany, 7-9th May 2015, presenting updates on innate immunity in acute brain injury and chronic neurodegenerative disorders, such as traumatic brain injury and Alzheimer disease, on the role of astrocytes and microglia, as well as technical developments that may help elucidate neuroinflammatory mechanisms and establish clinical relevance. In this meeting report, a brief overview of physiological and pathological microglia morphology is followed by a synopsis on PGE2 receptors, insights into the role of arginine metabolism and further relevant aspects of neuroinflammation in various clinical settings, and concluded by a presentation of technical challenges and solutions when working with microglia and astrocyte cultures. Microglial ontogeny and induced pluripotent stem cell-derived microglia, advances of TREM2 signaling, and the cytokine paradox in Alzheimer's disease are further contributions to this article. Neuroinflammation is critically involved in numerous neurodegenerative diseases, and key signaling steps of innate immune activation hence represent promising therapeutic targets. This mini review series originated from the 4th Venusberg Meeting on Neuroinflammation held in Bonn, Germany, 7-9th May 2015, presenting updates on innate immunity in acute brain injury and chronic neurodegenerative disorders, such as traumatic brain injury and Alzheimer's disease, on the role of astrocytes and microglia, as well as technical developments that may help elucidate neuroinflammatory mechanisms and establish clinical relevance. In this meeting report, a brief overview on physiological and pathological microglia morphology is followed by a synopsis on PGE2 receptors, insights into the role of arginine metabolism and further relevant aspects of neuroinflammation in various clinical settings, and concluded by a presentation of technical challenges and solutions when working with microglia cultures. Microglial ontogeny and induced pluripotent stem cell-derived microglia, advances of TREM2 signaling, and the cytokine paradox in Alzheimer's disease are further contributions to this article.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Niva Russek-Blum
- The Dead Sea and Arava Science Center, Central Arava Branch, Yair Station, Hazeva, Israel
| | - Alon Monsonego
- The Shraga Segal Dept. of Microbiology, Immunology and Genetics, The Faculty of Health Sciences: The National Institute of Biotechnology in the Negev, and Zlotowski Center for Neuroscience, Ben-Gurion University, Beer-Sheva 84105, Israel
| | - Donovan Low
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Kazuyuki Takata
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
- Department of Clinical and Translational Physiology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Terrence Town
- Departments of Physiology and Biophysics, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089,
| | - M. Kerry O’Banion
- Departments of Neuroscience and Neurology, Del Monte Neuromedicine Institute, University of Rochester School of Medicine & Dentistry, Rochester, NY 14642,
| | - Bruce Lamb
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH 44106
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Gary Landreth
- Department of Neurosciences, Case Western Reserve University 44106
| | - Katrin I. Andreasson
- Department of Neurology and Neurological Sciences, Stanford Neuroscience Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Leslie A. Sandusky
- USF Health Byrd Alzheimer’s Institute, Tampa, FL 33613
- College of Pharmacy & Pharmaceutical Sciences, Tampa, FL 33613
| | - Maj-Linda B. Selenica
- USF Health Byrd Alzheimer’s Institute, Tampa, FL 33613
- College of Pharmacy & Pharmaceutical Sciences, Tampa, FL 33613
| | - Daniel C. Lee
- USF Health Byrd Alzheimer’s Institute, Tampa, FL 33613
- College of Pharmacy & Pharmaceutical Sciences, Tampa, FL 33613
| | - Clive Holmes
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Tremona Road, Southampton, SO16 7YD, United Kingdom
| | - Jessica Teeling
- Centre for Biological Sciences, University of Southampton, Southampton General Hospital, Tremona Road, Southampton, SO16 7YD, United Kingdom
| | | | | | | | - Milos Pekny
- Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, SE-405 30 Gothenburg, Sweden
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
- Hunter Medical Research Institute, University of Newcastle, New South Wales, Australia
| | - Till Puschmann
- Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, SE-405 30 Gothenburg, Sweden
| |
Collapse
|
31
|
Sordillo PP, Sordillo LA, Helson L. Bifunctional role of pro-inflammatory cytokines after traumatic brain injury. Brain Inj 2016; 30:1043-53. [DOI: 10.3109/02699052.2016.1163618] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
32
|
Khalin I, Jamari NLA, Razak NBA, Hasain ZB, Nor MABM, Zainudin MHBA, Omar AB, Alyautdin R. A mouse model of weight-drop closed head injury: emphasis on cognitive and neurological deficiency. Neural Regen Res 2016; 11:630-5. [PMID: 27212925 PMCID: PMC4870921 DOI: 10.4103/1673-5374.180749] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability in individuals worldwide. Producing a clinically relevant TBI model in small-sized animals remains fairly challenging. For good screening of potential therapeutics, which are effective in the treatment of TBI, animal models of TBI should be established and standardized. In this study, we established mouse models of closed head injury using the Shohami weight-drop method with some modifications concerning cognitive deficiency assessment and provided a detailed description of the severe TBI animal model. We found that 250 g falling weight from 2 cm height produced severe closed head injury in C57BL/6 male mice. Cognitive disorders in mice with severe closed head injury could be detected using passive avoidance test on day 7 after injury. Findings from this study indicate that weight-drop injury animal models are suitable for further screening of brain neuroprotectants and potentially are similar to those seen in human TBI.
Collapse
Affiliation(s)
- Igor Khalin
- Faculty of Medicine and Defence Health, National Defence University of Malaysia, Kem Sungai Besi, Kuala Lumpur, Malaysia
| | - Nor Laili Azua Jamari
- Chemistry Department, Centre for Defence Foundation Studies, National Defence University of Malaysia, Kem Sungai Besi, Kuala Lumpur, Malaysia
| | - Nadiawati Bt Abdul Razak
- Faculty of Medicine and Defence Health, National Defence University of Malaysia, Kem Sungai Besi, Kuala Lumpur, Malaysia
| | - Zubaidah Bt Hasain
- Faculty of Medicine and Defence Health, National Defence University of Malaysia, Kem Sungai Besi, Kuala Lumpur, Malaysia
| | - Mohd Asri Bin Mohd Nor
- Department of Civil Engineering, Faculty of Engineering, National Defence University of Malaysia, Kem Sungai Besi, Kuala Lumpur, Malaysia
| | - Mohd Hakimi Bin Ahmad Zainudin
- Centre for Research and Innovation Management, National Defence University of Malaysia, Kem Sungai Besi, Kuala Lumpur, Malaysia
| | - Ainsah Bt Omar
- Faculty of Medicine and Defence Health, National Defence University of Malaysia, Kem Sungai Besi, Kuala Lumpur, Malaysia
| | - Renad Alyautdin
- Scientific Centre for Expertise of Medical Application Products, Petrovsky Blvd, Moscow, Russia
| |
Collapse
|
33
|
Serum Metabolic Profiling Reveals Altered Metabolic Pathways in Patients with Post-traumatic Cognitive Impairments. Sci Rep 2016; 6:21320. [PMID: 26883691 PMCID: PMC4756382 DOI: 10.1038/srep21320] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 01/21/2016] [Indexed: 12/18/2022] Open
Abstract
Cognitive impairment, the leading cause of traumatic brain injury (TBI)-related disability, adversely affects the quality of life of TBI patients, and exacts a personal and economic cost that is difficult to quantify. The underlying pathophysiological mechanism is currently unknown, and an effective treatment of the disease has not yet been identified. This study aimed to advance our understanding of the mechanism of disease pathogenesis; thus, metabolomics based on gas chromatography/mass spectrometry (GC-MS), coupled with multivariate and univariate statistical methods were used to identify potential biomarkers and the associated metabolic pathways of post-TBI cognitive impairment. A biomarker panel consisting of nine serum metabolites (serine, pyroglutamic acid, phenylalanine, galactose, palmitic acid, arachidonic acid, linoleic acid, citric acid, and 2,3,4-trihydroxybutyrate) was identified to be able to discriminate between TBI patients with cognitive impairment, TBI patients without cognitive impairment and healthy controls. Furthermore, associations between these metabolite markers and the metabolism of amino acids, lipids and carbohydrates were identified. In conclusion, our study is the first to identify several serum metabolite markers and investigate the altered metabolic pathway that is associated with post-TBI cognitive impairment. These markers appear to be suitable for further investigation of the disease mechanisms of post-TBI cognitive impairment.
Collapse
|
34
|
Bachstetter AD, Zhou Z, Rowe RK, Xing B, Goulding DS, Conley AN, Sompol P, Meier S, Abisambra JF, Lifshitz J, Watterson DM, Van Eldik LJ. MW151 Inhibited IL-1β Levels after Traumatic Brain Injury with No Effect on Microglia Physiological Responses. PLoS One 2016; 11:e0149451. [PMID: 26871438 PMCID: PMC4752278 DOI: 10.1371/journal.pone.0149451] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 01/31/2016] [Indexed: 12/26/2022] Open
Abstract
A prevailing neuroinflammation hypothesis is that increased production of proinflammatory cytokines contributes to progressive neuropathology, secondary to the primary damage caused by a traumatic brain injury (TBI). In support of the hypothesis, post-injury interventions that inhibit the proinflammatory cytokine surge can attenuate the progressive pathology. However, other post-injury neuroinflammatory responses are key to endogenous recovery responses. Therefore, it is critical that pharmacological attenuation of detrimental or dysregulated neuroinflammatory processes avoid pan-suppression of inflammation. MW151 is a CNS-penetrant, small molecule experimental therapeutic that restores injury- or disease-induced overproduction of proinflammatory cytokines towards homeostasis without immunosuppression. Post-injury administration of MW151 in a closed head injury model of mild TBI suppressed acute cytokine up-regulation and downstream cognitive impairment. Here, we report results from a diffuse brain injury model in mice using midline fluid percussion. Low dose (0.5–5.0 mg/kg) administration of MW151 suppresses interleukin-1 beta (IL-1β) levels in the cortex while sparing reactive microglia and astrocyte responses. To probe molecular mechanisms, we used live cell imaging of the BV-2 microglia cell line to demonstrate that MW151 does not affect proliferation, migration, or phagocytosis of the cells. Our results provide insight into the roles of glial responses to brain injury and indicate the feasibility of using appropriate dosing for selective therapeutic modulation of injurious IL-1β increases while sparing other glial responses to injury.
Collapse
Affiliation(s)
- Adam D. Bachstetter
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States of America
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky, Lexington, Kentucky, United States of America
- Department of Anatomy and Neurobiology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Zhengqiu Zhou
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States of America
| | - Rachel K. Rowe
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky, Lexington, Kentucky, United States of America
- Department of Anatomy and Neurobiology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Bin Xing
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States of America
| | - Danielle S. Goulding
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States of America
| | - Alyssa N. Conley
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States of America
| | - Pradoldej Sompol
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States of America
| | - Shelby Meier
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States of America
| | - Jose F. Abisambra
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States of America
- Department of Physiology, University of Kentucky, University of Kentucky, Lexington, Kentucky, United States of America
| | - Jonathan Lifshitz
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky, Lexington, Kentucky, United States of America
- Department of Anatomy and Neurobiology, University of Kentucky, Lexington, Kentucky, United States of America
| | - D. Martin Watterson
- Department of Pharmacology, Northwestern University, Chicago, Illinois, United States of America
| | - Linda J. Van Eldik
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States of America
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky, Lexington, Kentucky, United States of America
- Department of Anatomy and Neurobiology, University of Kentucky, Lexington, Kentucky, United States of America
- * E-mail:
| |
Collapse
|
35
|
Eslami M, Sayyah M, Soleimani M, Alizadeh L, Hadjighassem M. Lipopolysaccharide preconditioning prevents acceleration of kindling epileptogenesis induced by traumatic brain injury. J Neuroimmunol 2015; 289:143-151. [PMID: 26616884 DOI: 10.1016/j.jneuroim.2015.11.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 10/31/2015] [Accepted: 11/03/2015] [Indexed: 02/08/2023]
Abstract
10-20% of symptomatic epilepsies are post-traumatic. We examined effect of LPS preconditioning on epileptogenesis after controlled cortical impact (CCI). LPS (0.01, 0.1 and 0.5 mg/kg) was injected i.p. to rats 5 days before induction of CCI to parieto-temporal cortex. Kindling started 24h after CCI by i.p. injection of 30 mg/kg of pentylenetetrazole every other day until manifestation of 3 consecutive generalized seizures. CCI injury accelerated the rate of kindled seizures acquisition. LPS (0.1 and 0.5 mg/kg) prevented the acceleration of kindling. LPS preconditioning significantly decreased IL-1β and TNF-α over-expression and the number of damaged neurons in the hippocampus of traumatic rats.
Collapse
Affiliation(s)
- Mansoureh Eslami
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran; Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran; Department of Basic Sciences, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Sayyah
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran.
| | - Mansoureh Soleimani
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | - Mahmoudreza Hadjighassem
- Brain and Spinal cord injury Research Center, Neuroscience Institute, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran; Department of Neuroscience, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
36
|
Closed head injury in an age-related Alzheimer mouse model leads to an altered neuroinflammatory response and persistent cognitive impairment. J Neurosci 2015; 35:6554-69. [PMID: 25904805 DOI: 10.1523/jneurosci.0291-15.2015] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Epidemiological studies have associated increased risk of Alzheimer's disease (AD)-related clinical symptoms with a medical history of head injury. Currently, little is known about pathophysiology mechanisms linked to this association. Persistent neuroinflammation is one outcome observed in patients after a single head injury. Neuroinflammation is also present early in relevant brain regions during AD pathology progression. In addition, previous mechanistic studies in animal models link neuroinflammation as a contributor to neuropathology and cognitive impairment in traumatic brain injury (TBI) or AD-related models. Therefore, we explored the potential interplay of neuroinflammatory responses in TBI and AD by analysis of the temporal neuroinflammatory changes after TBI in an AD model, the APP/PS1 knock-in (KI) mouse. Discrete temporal aspects of astrocyte, cytokine, and chemokine responses in the injured KI mice were delayed compared with the injured wild-type mice, with a peak neuroinflammatory response in the injured KI mice occurring at 7 d after injury. The neuroinflammatory responses were more persistent in the injured KI mice, leading to a chronic neuroinflammation. At late time points after injury, KI mice exhibited a significant impairment in radial arm water maze performance compared with sham KI mice or injured wild-type mice. Intervention with a small-molecule experimental therapeutic (MW151) that selectively attenuates proinflammatory cytokine production yielded improved cognitive behavior outcomes, consistent with a link between neuroinflammatory responses and altered risk for AD-associated pathology changes with head injury.
Collapse
|