201
|
Konishi H, Kiyama H. Microglial TREM2/DAP12 Signaling: A Double-Edged Sword in Neural Diseases. Front Cell Neurosci 2018; 12:206. [PMID: 30127720 PMCID: PMC6087757 DOI: 10.3389/fncel.2018.00206] [Citation(s) in RCA: 185] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 06/25/2018] [Indexed: 12/21/2022] Open
Abstract
Microglia are activated after neuronal injury and in neurodegenerative diseases, and trigger neuroinflammation in the central nervous system (CNS). Microglia-derived neuroinflammation has both beneficial and detrimental effects on neurons. Because the timing and magnitude of microglial activation is thought to be a critical determinant of neuronal fate, understanding the molecular mechanisms underlying microglial activation is required to enable establishment of microglia-targeted therapies for neural diseases. Plasma membrane receptors play primary roles as activators of microglia and in this review, we focus on a receptor complex involving triggering receptor expressed on myeloid cells 2 (TREM2) and DNAX-activating protein of 12 kDa (DAP12), both of which are causative genes for Nasu-Hakola disease, a dementia with bone cysts. Recent transcriptome approaches demonstrated TREM2/DAP12 signaling as the principal regulator that transforms microglia from a homeostatic to a neural disease-associated state. Furthermore, animal model studies revealed critical roles for TREM2/DAP12 in the regulation of microglial activity, including survival, phagocytosis, and cytokine production, not only in Alzheimer's disease but also in other neural diseases, such as Parkinson's disease, demyelinating disease, ischemia, and peripheral nerve injury. Intriguingly, while TREM2/DAP12-mediated microglial activation is detrimental for some diseases, including peripheral nerve injury, it is beneficial for other diseases. As the role of activated microglia differs among disease models, TREM2/DAP12 signaling may result in different outcomes in different diseases. In this review we discuss recent perspectives on the role of TREM2/DAP12 in microglia and their contribution to neural diseases.
Collapse
Affiliation(s)
- Hiroyuki Konishi
- Department of Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Hiroshi Kiyama
- Department of Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya University, Nagoya, Japan
| |
Collapse
|
202
|
Shi H, Hua X, Kong D, Stein D, Hua F. Role of Toll-like receptor mediated signaling in traumatic brain injury. Neuropharmacology 2018; 145:259-267. [PMID: 30075158 DOI: 10.1016/j.neuropharm.2018.07.022] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 07/04/2018] [Accepted: 07/18/2018] [Indexed: 12/13/2022]
Abstract
The mechanisms underlying secondary brain damage following traumatic brain injury (TBI) remain unclear. A great many studies have demonstrated that inflammatory cascades contribute to brain damage through the activation of immune/inflammatory responses, including the increased release of cytokines and chemokines, and the recruitment of leukocytes. The cells and tissues damaged by primary mechanical injury release a number of endogenous factors acting as damage-associated molecular patterns (DAMPs), which initiate and perpetuate noninfectious inflammatory responses through transduction signaling pathways. Toll-like receptors (TLRs) are a transmembrane receptor family that can recognize the specific DAMPs released from damaged cells and recruit a set of adaptors leading to the activation of downstream kinases and nuclear factors which regulate the expression of inflammatory genes. The activation of inflammatory responses mediated by TLR signaling is closely associated with brain tissue damage and neurological dysfunction following TBI. TLRs and their downstream protein kinases may be potential targets for the treatment of TBI. Modulation of TLR-mediated signaling may attenuate brain damage and improve TBI outcome. In this review, we briefly discuss the role of TLR-mediated signaling in TBI and the new treatments targeting TLR signaling. This article is part of the Special Issue entitled "Novel Treatments for Traumatic Brain Injury".
Collapse
Affiliation(s)
- Hongjuan Shi
- Department of Neurology, The Affiliated Hospital, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Xiaodong Hua
- Augusta University/University of Georgia Medical Partnership, Athens, GA, 30606, USA; Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Delian Kong
- Department of Neurology, The Affiliated Hospital, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Donald Stein
- Brain Research Laboratory, Department of Emergency Medicine, Emory University School of Medicine, Atlanta, GA, 30032, USA
| | - Fang Hua
- Department of Neurology, The Affiliated Hospital, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China; Key Laboratory of Anesthesiology of Jiangsu Province, Xuzhou, 221002, China.
| |
Collapse
|
203
|
Meng X, Li N, Zhang Y, Fan D, Yang C, Li H, Guo D, Pan S. Beneficial Effect of β-Elemene Alone and in Combination with Hyperbaric Oxygen in Traumatic Brain Injury by Inflammatory Pathway. Transl Neurosci 2018; 9:33-37. [PMID: 29992051 PMCID: PMC6034101 DOI: 10.1515/tnsci-2018-0007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 03/09/2018] [Indexed: 11/25/2022] Open
Abstract
Background Present study evaluates the neuroprotective effect of β-elemene alone and in combination with hyperbaric oxygen (HO) in traumatic brain injury (TBI). Methodology TBI was induced by dropping a weight from a specific height. All the animals were separated in to five groups (n=20) like control group; TBI group; β-elemene treated group which receives β-elemene (100 mg/kg, i.p.) half an hour after the injury; HO group which receives hyperbaric oxygen therapy and β-elemene + HO group which receives β-elemene (100 mg/kg, i.p.) half an hour after the injury and hyperbaric oxygen therapy. Neurological function was assessed to evaluate the effect of β-elemene in TBI rats. Thereafter level of inflammatory cytokines and expression of protein of inflammatory pathway was assessed in the brain tissues of TBI rats. In addition TUNEL assay was also done for the determination apoptosis in neuronal cells. Result Data of the report reveals that β-elemene alone and in combination with hyperbaric oxygen (HO) significantly decreases the neurological score Compared to TBI group. Moreover level of inflammatory cytokines and expression of LTR4 and casepase 3 significantly decrease and increase in the expression of IkB in β-elemene alone and in combination with hyperbaric oxygen (HO) treated group compared to TBI group. Data of TUNEL assay also reveals that β-elemene treated group shows significant decrease in the TUNEL positive cells and apoptosis index compared to TBI group. Conclusion Thus present study concludes the neuroprotective effect of β-elemene against TBI and it shows synergistic effect on TBI when treated with HO.
Collapse
Affiliation(s)
- Xiangen Meng
- Department of Hyperbric Oxygen, Navy General Hospital, Beijing, 100048, P.R. China
| | - Na Li
- Department of Hyperbric Oxygen, Navy General Hospital, Beijing, 100048, P.R. China
| | - Yu Zhang
- Department of Hyperbric Oxygen, Navy General Hospital, Beijing, 100048, P.R. China
| | - Danfeng Fan
- Department of Hyperbric Oxygen, Navy General Hospital, Beijing, 100048, P.R. China
| | - Chen Yang
- Department of Hyperbric Oxygen, Navy General Hospital, Beijing, 100048, P.R. China
| | - Hang Li
- Department of Hyperbric Oxygen, Navy General Hospital, Beijing, 100048, P.R. China
| | - Dazhi Guo
- Department of Hyperbric Oxygen, Navy General Hospital, Beijing, 100048, P.R. China
| | - Shuyi Pan
- Department of Hyperbric Oxygen, Navy General Hospital, Beijing, 100048, P.R. China
| |
Collapse
|
204
|
Haber M, Amyot F, Kenney K, Meredith-Duliba T, Moore C, Silverman E, Podell J, Chou YY, Pham DL, Butman J, Lu H, Diaz-Arrastia R, Sandsmark D. Vascular Abnormalities within Normal Appearing Tissue in Chronic Traumatic Brain Injury. J Neurotrauma 2018; 35:2250-2258. [PMID: 29609518 DOI: 10.1089/neu.2018.5684] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
Abstract
Magnetic resonance imaging (MRI) is a powerful tool for visualizing traumatic brain injury(TBI)-related lesions. Trauma-induced encephalomalacia is frequently identified by its hyperintense appearance on fluid-attenuated inversion recovery (FLAIR) sequences. In addition to parenchymal lesions, TBI commonly results in cerebral microvascular injury, but its anatomical relationship to parenchymal encephalomalacia is not well characterized. The current study utilized a multi-modal MRI protocol to assess microstructural tissue integrity (by mean diffusivity [MD] and fractional aniosotropy [FA]) and altered vascular function (by cerebral blood flow [CBF] and cerebral vascular reactivity [CVR]) within regions of visible encephalomalacia and normal appearing tissue in 27 chronic TBI (minimum 6 months post-injury) subjects. Fifteen subjects had visible encephalomalacias whereas 12 did not have evident lesions on MRI. Imaging from 14 age-matched healthy volunteers were used as controls. CBF was assessed by arterial spin labeling (ASL) and CVR by measuring the change in blood-oxygen-level-dependent (BOLD) MRI during a hypercapnia challenge. There was a significant reduction in FA, CBF, and CVR with a complementary increase in MD within regions of FLAIR-visible encephalomalacia (p < 0.05 for all comparisons). In normal-appearing brain regions, only CVR was significantly reduced relative to controls (p < 0.05). These findings indicate that vascular dysfunction represents a TBI endophenotype that is distinct from structural injury detected using conventional MRI, may be present even in the absence of visible structural injury, and persists long after trauma. CVR may serve as a useful diagnostic and pharmacodynamic imaging biomarker of traumatic microvascular injury.
Collapse
Affiliation(s)
- Margalit Haber
- 1 Department of Neurology, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania
| | - Franck Amyot
- 6 National Intrepid Center of Excellence, Walter Reed National Military Medical Center, Bethesda, Maryland
| | - Kimbra Kenney
- 2 Department of Neurology, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - Tawny Meredith-Duliba
- 1 Department of Neurology, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania
| | - Carol Moore
- 2 Department of Neurology, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - Erika Silverman
- 1 Department of Neurology, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania
| | - Jamie Podell
- 1 Department of Neurology, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania
| | - Yi-Yu Chou
- 3 Center for Neuroscience and Regenerative Medicine , Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland
| | - Dzung L Pham
- 3 Center for Neuroscience and Regenerative Medicine , Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland
| | - John Butman
- 4 National Institutes of Health , Clinical Center, Radiology and Imaging Sciences, Bethesda, Maryland
| | - Hanzhang Lu
- 5 Department of Radiology, Johns Hopkins University Baltimore , Maryland
| | - Ramon Diaz-Arrastia
- 1 Department of Neurology, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania
| | - Danielle Sandsmark
- 1 Department of Neurology, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania
| |
Collapse
|
205
|
Wang W, Zinsmaier AK, Firestone E, Lin R, Yatskievych TA, Yang S, Zhang J, Bao S. Blocking Tumor Necrosis Factor-Alpha Expression Prevents Blast-Induced Excitatory/Inhibitory Synaptic Imbalance and Parvalbumin-Positive Interneuron Loss in the Hippocampus. J Neurotrauma 2018; 35:2306-2316. [PMID: 29649942 DOI: 10.1089/neu.2018.5688] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Traumatic brain injury (TBI) is a major cause of neurological disorder and death in civilian and military populations. It comprises two components-direct injury from the traumatic impact and secondary injury from ensuing neural inflammatory responses. Blocking tumor necrosis factor-alpha (TNF-α), a central regulator of neural inflammation, has been shown to improve functional recovery after TBI. However, the mechanisms underlying those therapeutic effects are still poorly understood. Here, we examined effects of 3,6'-dithiothalidomide (dTT), a potentially therapeutic TNF-α inhibitor, in mice with blast-induced TBI. We found that blast exposure resulted in elevated expression of TNF-α, activation of microglial cells, enhanced excitatory synaptic transmission, reduced inhibitory synaptic transmission, and a loss of parvalbumin-positive (PV+) inhibitory interneurons. Administration of dTT for 5 days after the blast exposure completely suppressed blast-induced increases in TNF-α transcription, largely reversed blasted-induced synaptic changes, and prevented PV+ neuron loss. However, blocking TNF-α expression by dTT failed to mitigate blast-induced microglial activation in the hippocampus, as evidenced by their non-ramified morphology. These results indicate that TNF-α plays a major role in modulating neuronal functions in blast-induced TBI and that it is a potential target for treatment of TBI-related brain disorders.
Collapse
Affiliation(s)
- Weihua Wang
- 1 Department of Physiology, College of Medicine, University of Arizona , Tucson, Arizona
| | - Alexander K Zinsmaier
- 1 Department of Physiology, College of Medicine, University of Arizona , Tucson, Arizona
| | - Ethan Firestone
- 2 Department of Otolaryngology-Head and Neck Surgery and Department of Communication Sciences and Disorders, School of Medicine, Wayne State University , Detroit, Michigan
| | - Ruizhu Lin
- 1 Department of Physiology, College of Medicine, University of Arizona , Tucson, Arizona.,3 Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University , Guangzhou, China
| | - Tatiana A Yatskievych
- 1 Department of Physiology, College of Medicine, University of Arizona , Tucson, Arizona
| | - Sungchil Yang
- 4 Department of Biomedical Sciences, City University of Hong Kong , Kowloon, Hong Kong, China
| | - Jinsheng Zhang
- 2 Department of Otolaryngology-Head and Neck Surgery and Department of Communication Sciences and Disorders, School of Medicine, Wayne State University , Detroit, Michigan
| | - Shaowen Bao
- 1 Department of Physiology, College of Medicine, University of Arizona , Tucson, Arizona
| |
Collapse
|
206
|
Abstract
Traumatic brain injury (TBI) is a widely recognized risk factor for neurodegenerative disease. The purpose of this review is to provide an update on the state of the science related to injury cascades in TBI-related neurodegeneration. Acute and chronic pathological outcomes of TBI are similar to those seen in several neurodegenerative conditions, suggesting common linking pathways. Initial research described severe TBI patients with post-mortem identification of abnormal proteins, such as amyloid deposits. History of mild TBI (mTBI) is less consistently associated with heightened risk of neurodegenerative outcomes, but specific populations with complicated medical histories and comorbidities may be more susceptible. Our understanding of a pathological signature associated with repetitive mTBI and/or subclinical brain trauma advanced significantly over the past decade, and is now commonly referred to as chronic traumatic encephalopathy. We discuss hypotheses linking TBI to neurodegenerative disease, and the importance of considering factors like injury severity, timing of injury (early life versus older age), injury frequency, and repetitive subclinical brain trauma when extrapolating results from current literature to certain populations. We describe the challenges to obtaining the data necessary for accurate epidemiological research and determination of true risk magnitude, and note the importance of developing treatment-based approaches to risk mitigation.
Collapse
Affiliation(s)
- Steven T DeKosky
- a Departments of Neurology and Neuroscience , McKnight Brain Institute, University of Florida , Gainesville , FL , USA
| | - Breton M Asken
- b Department of Clinical and Health Psychology, Neuropsychology , College of Public Health and Health Professions, University of Florida , Gainesville FL , USA
| |
Collapse
|
207
|
Xiong Y, Mahmood A, Chopp M. Current understanding of neuroinflammation after traumatic brain injury and cell-based therapeutic opportunities. Chin J Traumatol 2018; 21:137-151. [PMID: 29764704 PMCID: PMC6034172 DOI: 10.1016/j.cjtee.2018.02.003] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 03/02/2018] [Accepted: 03/05/2018] [Indexed: 02/04/2023] Open
Abstract
Traumatic brain injury (TBI) remains a major cause of death and disability worldwide. Increasing evidence indicates that TBI is an important risk factor for neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, and chronic traumatic encephalopathy. Despite improved supportive and rehabilitative care of TBI patients, unfortunately, all late phase clinical trials in TBI have yet to yield a safe and effective neuroprotective treatment. The disappointing clinical trials may be attributed to variability in treatment approaches and heterogeneity of the population of TBI patients as well as a race against time to prevent or reduce inexorable cell death. TBI is not just an acute event but a chronic disease. Among many mechanisms involved in secondary injury after TBI, emerging preclinical studies indicate that posttraumatic prolonged and progressive neuroinflammation is associated with neurodegeneration which may be treatable long after the initiating brain injury. This review provides an overview of recent understanding of neuroinflammation in TBI and preclinical cell-based therapies that target neuroinflammation and promote functional recovery after TBI.
Collapse
Affiliation(s)
- Ye Xiong
- Department of Neurosurgery Henry Ford Health System, 2799 West Grand Boulevard, Detroit, MI, 48202, USA.
| | - Asim Mahmood
- Department of Neurosurgery Henry Ford Health System, 2799 West Grand Boulevard, Detroit, MI, 48202, USA
| | - Michael Chopp
- Department of Neurology, Henry Ford Health System, 2799 West Grand Boulevard, Detroit, MI, 48202, USA; Department of Physics, Oakland University, Rochester, MI, 48309, USA
| |
Collapse
|
208
|
Autologous white blood cell infusion for trauma, brain trauma, stroke and select immune dysfunction co-morbidities: A promising and timely proposal? Med Hypotheses 2018; 117:7-15. [PMID: 30077201 DOI: 10.1016/j.mehy.2018.05.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 04/23/2018] [Accepted: 05/14/2018] [Indexed: 11/21/2022]
Abstract
All traumas suppress the immune system, resulting in higher morbidity and mortality. Infections, poor nutritional status, chronic illness, fatigue, therapies or procedures performed during and after transport also negatively affect the immune system. Large populations are impacted by trauma worldwide and suffer enormous costs in both direct and indirect expenditures from physical, psychological and functional losses. Most therapies and studies of trauma, brain trauma, stroke, immune suppression and their co-morbidities do not address nor discuss methods that promote immune system resuscitation or efficacy to support its role in post-trauma healing and rehabilitation. These omissions present an opportunity for using autologous stored naïve (unexposed to the current trauma and co-morbidities) white blood cell infusions (autologous white blood cell infusion) (AWBCI) to supplement treatment of most traumas, trauma-associated infections, other co-morbidities and immune suppression derived problems in order to improve the global standard of trauma care. We hypothesize to give the traumatized patients back their own immune system that has been 'stored' in some fashion, either cryogenically or just after or during the trauma event [surgery, etc for example]. We emphasize that other treatments should not be replaced - rather we suggest AWBCI as concurrent therapy. We present focused select animal and human studies as proofs of concept to arrive at and support our therapeutic suggestion and hypotheses, flowing historically from donor white blood cell therapy [DLI] to close cohort white blood cell therapy to autologous white blood cell infusion [AWBCI]. We integrate the concept of personalized medicine from an evidence-based framework while maintaining scientific rigor and statistical proof as a basis of our hypotheses.
Collapse
|
209
|
Rowe RK, Harrison JL, Zhang H, Bachstetter AD, Hesson DP, O'Hara BF, Greene MI, Lifshitz J. Novel TNF receptor-1 inhibitors identified as potential therapeutic candidates for traumatic brain injury. J Neuroinflammation 2018; 15:154. [PMID: 29789012 PMCID: PMC5964690 DOI: 10.1186/s12974-018-1200-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 05/13/2018] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) begins with the application of mechanical force to the head or brain, which initiates systemic and cellular processes that are hallmarks of the disease. The pathological cascade of secondary injury processes, including inflammation, can exacerbate brain injury-induced morbidities and thus represents a plausible target for pharmaceutical therapies. We have pioneered research on post-traumatic sleep, identifying that injury-induced sleep lasting for 6 h in brain-injured mice coincides with increased cortical levels of inflammatory cytokines, including tumor necrosis factor (TNF). Here, we apply post-traumatic sleep as a physiological bio-indicator of inflammation. We hypothesized the efficacy of novel TNF receptor (TNF-R) inhibitors could be screened using post-traumatic sleep and that these novel compounds would improve functional recovery following diffuse TBI in the mouse. METHODS Three inhibitors of TNF-R activation were synthesized based on the structure of previously reported TNF CIAM inhibitor F002, which lodges into a defined TNFR1 cavity at the TNF-binding interface, and screened for in vitro efficacy of TNF pathway inhibition (IκB phosphorylation). Compounds were screened for in vivo efficacy in modulating post-traumatic sleep. Compounds were then tested for efficacy in improving functional recovery and verification of cellular mechanism. RESULTS Brain-injured mice treated with Compound 7 (C7) or SGT11 slept significantly less than those treated with vehicle, suggesting a therapeutic potential to target neuroinflammation. SGT11 restored cognitive, sensorimotor, and neurological function. C7 and SGT11 significantly decreased cortical inflammatory cytokines 3 h post-TBI. CONCLUSIONS Using sleep as a bio-indicator of TNF-R-dependent neuroinflammation, we identified C7 and SGT11 as potential therapeutic candidates for TBI.
Collapse
Affiliation(s)
- Rachel K Rowe
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA. .,Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA. .,Phoenix Veteran Affairs Healthcare System, Phoenix, AZ, USA.
| | - Jordan L Harrison
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Hongtao Zhang
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Adam D Bachstetter
- Sanders-Brown Center on Aging, Spinal Cord and Brain Injury Research Center, and Department of Neuroscience, University of Kentucky, Lexington, KY, USA
| | - David P Hesson
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Bruce F O'Hara
- Department of Biology, University of Kentucky, Lexington, KY, USA
| | - Mark I Greene
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jonathan Lifshitz
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA.,Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA.,Phoenix Veteran Affairs Healthcare System, Phoenix, AZ, USA.,Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| |
Collapse
|
210
|
Abstract
Mild traumatic brain injury (mTBI) represents a significant public healthcare concern, accounting for the majority of all head injuries. While symptoms are generally transient, some patients go on to experience long-term cognitive impairments and additional mild impacts can result in exacerbated and persisting negative outcomes. To date, studies using a range of experimental models have reported chronic behavioral deficits in the presence of axonal injury and inflammation following repeated mTBI; assessments of oxidative stress and myelin pathology have thus far been limited. However, some models employed induced acute focal damage more suggestive of moderate–severe brain injury and are therefore not relevant to repeated mTBI. Given that the nature of mechanical loading in TBI is implicated in downstream pathophysiological changes, the mechanisms of damage and chronic consequences of single and repeated closed-head mTBI remain to be fully elucidated. This review covers literature on potential mechanisms of damage following repeated mTBI, integrating known mechanisms of pathology underlying moderate–severe TBIs, with recent studies on adult rodent models relevant to direct impact injuries rather than blast-induced damage. Pathology associated with excitotoxicity and cerebral blood flow-metabolism uncoupling, oxidative stress, cell death, blood-brain barrier dysfunction, astrocyte reactivity, microglial activation, diffuse axonal injury, and dysmyelination is discussed, followed by a summary of functional deficits and preclinical assessments of therapeutic strategies. Comprehensive characterization of the pathology underlying delayed and persisting deficits following repeated mTBI is likely to facilitate further development of therapeutic strategies to limit long-term sequelae.
Collapse
Affiliation(s)
- Brooke Fehily
- 1 Experimental and Regenerative Neurosciences, School of Biological sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Melinda Fitzgerald
- 1 Experimental and Regenerative Neurosciences, School of Biological sciences, The University of Western Australia, Perth, Western Australia, Australia.,2 Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia.,3 Perron Institute for Neurological and Translational Science, Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia
| |
Collapse
|
211
|
Lee CJ, Felix ER, Levitt RC, Eddy C, Vanner EA, Feuer WJ, Sarantopoulos CD, Galor A. Traumatic brain injury, dry eye and comorbid pain diagnoses in US veterans. Br J Ophthalmol 2018; 102:667-673. [PMID: 28844048 DOI: 10.1136/bjophthalmol-2017-310509] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 07/16/2017] [Accepted: 07/30/2017] [Indexed: 01/12/2023]
Abstract
AIMS The purpose of the study is to evaluate the relationship between dry eye (DE) and pain diagnoses in US veterans with and without traumatic brain injury (TBI). METHODS Retrospective cohort study of veterans who were seen in the Veterans Administration Hospital (VA) between 1 January 2010 and 31 December 2014. Veterans were separated into two groups by the presence or absence of an International Classification of Diseases, Ninth Revision diagnosis of TBI and assessed for DE and other comorbidities. A dendrogram was used to investigate the linkage between TBI, DE, chronic pain and other comorbid conditions. RESULTS Of the 3 265 894 veterans seen during the 5-year period, 3.97% carried a diagnosis of TBI. Veterans with TBI were more likely to have a diagnosis of DE compared with their counterparts without TBI (37.2% vs 29.1%, p<0.0005). The association was stronger between TBI and ocular pain (OR 3.08; 95% CI 3.03 to 3.13) compared with tear film dysfunction (OR 1.09; 95% CI 1.07 to 1.10). Those with TBI were also about twice as likely to have a diagnosis of chronic pain, headache, depression or post-traumatic stress disorder compared with their counterparts without TBI. Cluster analysis of TBI, DE and pain diagnoses of interest revealed that central pain syndrome, cluster headache, sicca syndrome, keratoconjunctivitis sicca and late effect of injury to the nervous system (as can be seen after TBI) were all closely clustered together. CONCLUSIONS DE and pain disorders occur at higher frequencies in patients with a diagnosis of TBI, suggesting a common underlying pathophysiology.
Collapse
Affiliation(s)
- Charity J Lee
- Department of Ophthalmology, Miami VA Medical Center, Miami, Florida, USA.,Bascom Palmer Eye Institute, University of Miami, Miami, Florida, USA
| | - Elizabeth R Felix
- Department of Ophthalmology, Miami VA Medical Center, Miami, Florida, USA.,Department of Physical Medicine & Rehabilitation, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Roy C Levitt
- Department of Ophthalmology, Miami VA Medical Center, Miami, Florida, USA.,Department of Anesthesiology, Perioperative Medicine and Pain Management, University of Miami, Miller School of Medicine, Miami, Florida, USA.,Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, Florida, USA.,Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Christopher Eddy
- Department of Anesthesiology, Perioperative Medicine and Pain Management, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | | | - William J Feuer
- Bascom Palmer Eye Institute, University of Miami, Miami, Florida, USA
| | - Constantine D Sarantopoulos
- Department of Anesthesiology, Perioperative Medicine and Pain Management, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Anat Galor
- Department of Ophthalmology, Miami VA Medical Center, Miami, Florida, USA.,Bascom Palmer Eye Institute, University of Miami, Miami, Florida, USA
| |
Collapse
|
212
|
Hogan SR, Phan JH, Alvarado-Velez M, Wang MD, Bellamkonda RV, Fernández FM, LaPlaca MC. Discovery of Lipidome Alterations Following Traumatic Brain Injury via High-Resolution Metabolomics. J Proteome Res 2018; 17:2131-2143. [PMID: 29671324 DOI: 10.1021/acs.jproteome.8b00068] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Traumatic brain injury (TBI) can occur across wide segments of the population, presenting in a heterogeneous manner that makes diagnosis inconsistent and management challenging. Biomarkers offer the potential to objectively identify injury status, severity, and phenotype by measuring the relative concentrations of endogenous molecules in readily accessible biofluids. Through a data-driven, discovery approach, novel biomarker candidates for TBI were identified in the serum lipidome of adult male Sprague-Dawley rats in the first week following moderate controlled cortical impact (CCI). Serum samples were analyzed in positive and negative modes by ultraperformance liquid chromatography-mass spectrometry (UPLC-MS). A predictive panel for the classification of injured and uninjured sera samples, consisting of 26 dysregulated species belonging to a variety of lipid classes, was developed with a cross-validated accuracy of 85.3% using omniClassifier software to optimize feature selection. Polyunsaturated fatty acids (PUFAs) and PUFA-containing diacylglycerols were found to be upregulated in sera from injured rats, while changes in sphingolipids and other membrane phospholipids were also observed, many of which map to known secondary injury pathways. Overall, the identified biomarker panel offers viable molecular candidates representing lipids that may readily cross the blood-brain barrier (BBB) and aid in the understanding of TBI pathophysiology.
Collapse
Affiliation(s)
- Scott R Hogan
- School of Chemistry and Biochemistry , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
| | - John H Phan
- Wallace H Coulter Department of Biomedical Engineering , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
| | - Melissa Alvarado-Velez
- Wallace H Coulter Department of Biomedical Engineering , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
| | - May Dongmei Wang
- Wallace H Coulter Department of Biomedical Engineering , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
| | - Ravi V Bellamkonda
- Wallace H Coulter Department of Biomedical Engineering , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
| | - Facundo M Fernández
- School of Chemistry and Biochemistry , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
| | - Michelle C LaPlaca
- Wallace H Coulter Department of Biomedical Engineering , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
| |
Collapse
|
213
|
Liu B, He Z, Wang J, Xin Z, Wang J, Li F, Fu Y. Taraxasterol Inhibits LPS-Induced Inflammatory Response in BV2 Microglia Cells by Activating LXRα. Front Pharmacol 2018; 9:278. [PMID: 29670526 PMCID: PMC5893773 DOI: 10.3389/fphar.2018.00278] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 03/12/2018] [Indexed: 12/20/2022] Open
Abstract
Neuroinflammation plays a critical role in the development of neurodegenerative diseases. Taraxasterol, a pentacyclic-triterpene isolated from Taraxacum officinale, has been reported to have anti-inflammatory effect. The aim of this study was to investigate the anti-inflammatory effects and mechanism of taraxasterol in LPS-stimulated BV2 microglia cells. BV2 microglia cells were treated with taraxasterol 12 h before LPS stimulation. The effects of taraxasterol on LPS-induced TNF-α and IL-1β production were detected by ELISA. The effects of taraxasterol on LXRα, ABCA1, TLR4, and NF-κB expression were detected by western blot analysis. The results showed that taraxasterol dose-dependently inhibited LPS-induced TNF-α and IL-1β production and NF-κB activation. Taraxasterol also disrupted the formation of lipid rafts and inhibited translocation of TLR4 into lipid rafts. Furthermore, taraxasterol was found to activate LXRα-ABCA1 signaling pathway which induces cholesterol efflux from cells. In addition, our results showed that the anti-inflammatory effect of taraxasterol was attenuated by transfection with LXRα siRNA. In conclusion, these results suggested that taraxasterol inhibits LPS-induced inflammatory response in BV2 microglia cells by activating LXRα-ABCA1 signaling pathway.
Collapse
Affiliation(s)
- Bin Liu
- Cardiovascular Disease Center, First Hospital of Jilin University, Changchun, China
| | - Zhaoqi He
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jingjing Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Zhuoyuan Xin
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, China
| | - Jiaxin Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Fan Li
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, China
| | - Yunhe Fu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, China.,Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, China
| |
Collapse
|
214
|
Morley WA. Environmental Subconcussive Injury, Axonal Injury, and Chronic Traumatic Encephalopathy. Front Neurol 2018; 9:166. [PMID: 29636723 PMCID: PMC5880887 DOI: 10.3389/fneur.2018.00166] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 03/05/2018] [Indexed: 12/14/2022] Open
Abstract
Brain injury occurs in two phases: the initial injury itself and a secondary cascade of precise immune-based neurochemical events. The secondary phase is typically functional in nature and characterized by delayed axonal injury with more axonal disconnections occurring than in the initial phase. Axonal injury occurs across the spectrum of disease severity, with subconcussive injury, especially when repetitive, now considered capable of producing significant neurological damage consistent with axonal injury seen in clinically evident concussion, despite no observable symptoms. This review is the first to introduce the concept of environmental subconcussive injury (ESCI) and sets out how secondary brain damage from ESCI once past the juncture of microglial activation appears to follow the same neuron-damaging pathway as secondary brain damage from conventional brain injury. The immune response associated with ESCI is strikingly similar to that mounted after conventional concussion. Specifically, microglial activation is followed closely by glutamate and calcium flux, excitotoxicity, reactive oxygen species and reactive nitrogen species (RNS) generation, lipid peroxidation, and mitochondrial dysfunction and energy crisis. ESCI damage also occurs in two phases, with the primary damage coming from microbiome injury (due to microbiome-altering events) and secondary damage (axonal injury) from progressive secondary neurochemical events. The concept of ESCI and the underlying mechanisms have profound implications for the understanding of chronic traumatic encephalopathy (CTE) etiology because it has previously been suggested that repetitive axonal injury may be the primary CTE pathogenesis in susceptible individuals and it is best correlated with lifetime brain trauma load. Taken together, it appears that susceptibility to brain injury and downstream neurodegenerative diseases, such as CTE, can be conceptualized as a continuum of brain resilience. At one end is optimal resilience, capable of launching effective responses to injury with spontaneous recovery, and at the other end is diminished resilience with a compromised ability to respond and/or heal appropriately. Modulating factors such as one's total cumulative and synergistic brain trauma load, bioavailability of key nutrients needed for proper functioning of restorative metabolic pathways (specifically those involved in the deactivation and clearance of metabolic by-products of brain injury) are key to ultimately determining one's brain resilience.
Collapse
|
215
|
Dong W, Yang B, Wang L, Li B, Guo X, Zhang M, Jiang Z, Fu J, Pi J, Guan D, Zhao R. Curcumin plays neuroprotective roles against traumatic brain injury partly via Nrf2 signaling. Toxicol Appl Pharmacol 2018; 346:28-36. [PMID: 29571711 DOI: 10.1016/j.taap.2018.03.020] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 03/15/2018] [Accepted: 03/19/2018] [Indexed: 12/28/2022]
Abstract
Traumatic brain injury (TBI), which leads to high mortality and morbidity, is a prominent public health problem worldwide with no effective treatment. Curcumin has been shown to be beneficial for neuroprotection in vivo and in vitro, but the underlying mechanism remains unclear. This study determined whether the neuroprotective role of curcumin in mouse TBI is dependent on the NF-E2-related factor (Nrf2) pathway. The Feeney weight-drop contusion model was used to mimic TBI. Curcumin was administered intraperitoneally 15 min after TBI induction, and brains were collected at 24 h after TBI. The levels of Nrf2 and its downstream genes (Hmox-1, Nqo1, Gclm, and Gclc) were detected by Western blot and qRT-PCR at 24 h after TBI. In addition, edema, oxidative damage, cell apoptosis and inflammatory reactions were evaluated in wild type (WT) and Nrf2-knockout (Nrf2-KO) mice to explore the role of Nrf2 signaling after curcumin treatment. In wild type mice, curcumin treatment resulted in reduced ipsilateral cortex injury, neutrophil infiltration, and microglia activation, improving neuron survival against TBI-induced apoptosis and degeneration. These effects were accompanied by increased expression and nuclear translocation of Nrf2, and enhanced expression of antioxidant enzymes. However, Nrf2 deletion attenuated the neuroprotective effects of curcumin in Nrf2-KO mice after TBI. These findings demonstrated that curcumin effects on TBI are associated with the activation the Nrf2 pathway, providing novel insights into the neuroprotective role of Nrf2 and the potential therapeutic use of curcumin for TBI.
Collapse
Affiliation(s)
- Wenwen Dong
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China
| | - Bei Yang
- Department of Histology and Embryology, College of Basic Medical Sciences, China Medical University, Shenyang 110122, China
| | - Linlin Wang
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China
| | - Bingxuan Li
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China
| | - Xiangshen Guo
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China
| | - Miao Zhang
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China
| | - Zhenfei Jiang
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China
| | - Jingqi Fu
- Program of Environmental Toxicology, School of Public Health, China Medical Univeristy, Shenyang 110122, China
| | - Jingbo Pi
- Program of Environmental Toxicology, School of Public Health, China Medical Univeristy, Shenyang 110122, China
| | - Dawei Guan
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China.
| | - Rui Zhao
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China.
| |
Collapse
|
216
|
Bajwa NM, Kesavan C, Mohan S. Long-term Consequences of Traumatic Brain Injury in Bone Metabolism. Front Neurol 2018; 9:115. [PMID: 29556212 PMCID: PMC5845384 DOI: 10.3389/fneur.2018.00115] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 02/15/2018] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) leads to long-term cognitive, behavioral, affective deficits, and increase neurodegenerative diseases. It is only in recent years that there is growing awareness that TBI even in its milder form poses long-term health consequences to not only the brain but to other organ systems. Also, the concept that hormonal signals and neural circuits that originate in the hypothalamus play key roles in regulating skeletal system is gaining recognition based on recent mouse genetic studies. Accordingly, many TBI patients have also presented with hormonal dysfunction, increased skeletal fragility, and increased risk of skeletal diseases. Research from animal models suggests that TBI may exacerbate the activation and inactivation of molecular pathways leading to changes in both osteogenesis and bone destruction. TBI has also been found to induce the formation of heterotopic ossification and increased callus formation at sites of muscle or fracture injury through increased vascularization and activation of systemic factors. Recent studies also suggest that the disruption of endocrine factors and neuropeptides caused by TBI may induce adverse skeletal effects. This review will discuss the long-term consequences of TBI on the skeletal system and TBI-induced signaling pathways that contribute to the formation of ectopic bone, altered fracture healing, and reduced bone mass.
Collapse
Affiliation(s)
- Nikita M Bajwa
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, CA, United States
| | - Chandrasekhar Kesavan
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, CA, United States.,Department of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Subburaman Mohan
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, CA, United States.,Department of Medicine, Loma Linda University, Loma Linda, CA, United States.,Department of Orthopedic Surgery, Loma Linda University, Loma Linda, CA, United States
| |
Collapse
|
217
|
Wang X, Lan YL, Xing JS, Lan XQ, Wang LT, Zhang B. Alantolactone plays neuroprotective roles in traumatic brain injury in rats via anti-inflammatory, anti-oxidative and anti-apoptosis pathways. Am J Transl Res 2018; 10:368-380. [PMID: 29511431 PMCID: PMC5835802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 01/06/2018] [Indexed: 06/08/2023]
Abstract
Traumatic brain injury (TBI) is a common disease associated with a high rate of morbidity and mortality. Secondary brain injury following TBI triggers pathological, physiological, and biological reactions that lead to neurological dysfunctions. Alantolactone (ATL) is a well-known Chinese medicine that possesses strong anti-inflammatory properties, but its role in TBI remains poorly understood. The objective of this study was to evaluate the protective effect of ATL in a rat model of controlled cortical impact (CCI). We observed the neurological scores, brain water content, oxidative stress, neuroinflammation and apoptosis by performing an enzyme-linked immunosorbent assay, western blotting, quantitative real-time reverse transcription polymerase chain reaction (RT-qPCR), immunohistochemical (IHC) staining and other methods after CCI. The neurological scores, brain water content, levels of oxidative stress and inflammatory cytokines, and apoptosis index were markedly decreased following the ATL treatment in rats after TBI. Moreover, the antioxidant and anti-inflammatory effects of ATL in TBI may be partially mediated by inhibition of the NF-κB pathway and suppression of Cyclooxygenase 2 (COX-2). In addition, ATL attenuated TBI-induced neuronal apoptosis by suppressing the cytochrome c/caspase-dependent apoptotic pathway. Thus, ATL could exert neuroprotection in rats in a TBI model. Importantly, ATL has great potential in the clinical treatment of TBI.
Collapse
Affiliation(s)
- Xun Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical UniversityDalian 116023, China
- Department of Neurosurgery, The Third People’s Hospital of Dalian, Non-Directly Affiliated Hospital of Dalian Medical UniversityDalian 116033, China
- Department of Pharmacy, Dalian Medical UniversityDalian 116044, China
| | - Yu-Long Lan
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical UniversityDalian 116023, China
- Department of Pharmacy, Dalian Medical UniversityDalian 116044, China
- Department of Physiology, Dalian Medical UniversityDalian 116044, China
| | - Jin-Shan Xing
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical UniversityDalian 116023, China
- Department of Pharmacy, Dalian Medical UniversityDalian 116044, China
| | - Xiao-Qiang Lan
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical UniversityDalian 116023, China
| | - Li-Tao Wang
- Department of Neurosurgery, The Third People’s Hospital of Dalian, Non-Directly Affiliated Hospital of Dalian Medical UniversityDalian 116033, China
| | - Bo Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical UniversityDalian 116023, China
| |
Collapse
|
218
|
Truettner JS, Bramlett HM, Dietrich WD. Hyperthermia and Mild Traumatic Brain Injury: Effects on Inflammation and the Cerebral Vasculature. J Neurotrauma 2018; 35:940-952. [PMID: 29108477 DOI: 10.1089/neu.2017.5303] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Mild traumatic brain injury (mTBI) or concussion represents the majority of brain trauma in the United States. The pathophysiology of mTBI is complex and may include both focal and diffuse injury patterns. In addition to altered circuit dysfunction and traumatic axonal injury (TAI), chronic neuroinflammation has also been implicated in the pathophysiology of mTBI. Recently, our laboratory has reported the detrimental effects of mild hyperthermic mTBI in terms of worsening histopathological and behavioral outcomes. To clarify the role of temperature-sensitive neuroinflammatory processes on these consequences, we evaluated the effects of elevated brain temperature (39°C) on altered microglia/macrophage phenotype patterns after mTBI, changes in leukocyte recruitment, and TAI. Sprague-Dawley male rats underwent mild parasagittal fluid-percussion injury under normothermic (37°C) or hyperthermic (39°C) conditions. Cortical and hippocampal regions were analyzed using several cellular and molecular outcome measures. At 24 h, the ratio of iNOS-positive (M1 type phenotype) to arginase-positive (M2 type phenotype) cells after hyperthermic mTBI showed an increase compared with normothermia by flow cytometry. Inflammatory response gene arrays also demonstrated a significant increase in several classes of pro-inflammatory genes with hyperthermia treatment over normothermia. The injury-induced expression of chemokine ligand 2 (Ccl2) and alpha-2-macroglobulin were also increased with hyperthermic mTBI. With western blot analysis, an increase in CD18 and intercellular cell adhesion molecule-1 (ICAM-1) with hyperthermia and a significant increase in Iba1 reactive microglia are reported in the cerebral cortex. Together, these results demonstrate significant differences in the cellular and molecular consequences of raised brain temperature at the time of mTBI. The observed polarization toward a M1-phenotype with mild hyperthermia would be expected to augment chronic inflammatory cascades, sustained functional deficits, and increased vulnerability to secondary insults. Mild elevations in brain temperature may contribute to the more severe and longer lasting consequences of mTBI or concussion reported in some patients.
Collapse
Affiliation(s)
- Jessie S Truettner
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida
| | - Helen M Bramlett
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida
| | - W Dalton Dietrich
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
219
|
Wang KK, Yang Z, Zhu T, Shi Y, Rubenstein R, Tyndall JA, Manley GT. An update on diagnostic and prognostic biomarkers for traumatic brain injury. Expert Rev Mol Diagn 2018; 18:165-180. [PMID: 29338452 PMCID: PMC6359936 DOI: 10.1080/14737159.2018.1428089] [Citation(s) in RCA: 305] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Traumatic brain injury (TBI) is a major worldwide neurological disorder of epidemic proportions. To date, there are still no FDA-approved therapies to treat any forms of TBI. Encouragingly, there are emerging data showing that biofluid-based TBI biomarker tests have the potential to diagnose the presence of TBI of different severities including concussion, and to predict outcome. Areas covered: The authors provide an update on the current knowledge of TBI biomarkers, including protein biomarkers for neuronal cell body injury (UCH-L1, NSE), astroglial injury (GFAP, S100B), neuronal cell death (αII-spectrin breakdown products), axonal injury (NF proteins), white matter injury (MBP), post-injury neurodegeneration (total Tau and phospho-Tau), post-injury autoimmune response (brain antigen-targeting autoantibodies), and other emerging non-protein biomarkers. The authors discuss biomarker evidence in TBI diagnosis, outcome prognosis and possible identification of post-TBI neurodegernative diseases (e.g. chronic traumatic encephalopathy and Alzheimer's disease), and as theranostic tools in pre-clinical and clinical settings. Expert commentary: A spectrum of biomarkers is now at or near the stage of formal clinical validation of their diagnostic and prognostic utilities in the management of TBI of varied severities including concussions. TBI biomarkers could serve as a theranostic tool in facilitating drug development and treatment monitoring.
Collapse
Affiliation(s)
- Kevin K Wang
- a Program for Neurotrauma, Neuroproteomics & Biomarkers Research, Departments of Emergency Medicine, Psychiatry, Neuroscience and Chemistry , University of Florida , Gainesville , Florida , USA
| | - Zhihui Yang
- a Program for Neurotrauma, Neuroproteomics & Biomarkers Research, Departments of Emergency Medicine, Psychiatry, Neuroscience and Chemistry , University of Florida , Gainesville , Florida , USA
| | - Tian Zhu
- a Program for Neurotrauma, Neuroproteomics & Biomarkers Research, Departments of Emergency Medicine, Psychiatry, Neuroscience and Chemistry , University of Florida , Gainesville , Florida , USA
| | - Yuan Shi
- b Department Of Pediatrics, Daping Hospital, Chongqing , Third Military Medical University , Chongqing , China
| | - Richard Rubenstein
- c Laboratory of Neurodegenerative Diseases and CNS Biomarker Discovery, Departments of Neurology and Physiology/Pharmacology , SUNY Downstate Medical Center , Brooklyn , NY , USA
| | - J Adrian Tyndall
- d Department of Emergency Medicine , University of Florida , Gainesville , Florida , USA
| | - Geoff T Manley
- e Brain and Spinal Injury Center , San Francisco General Hospital , San Francisco , CA , USA
- f Department of Neurological Surgery , University of California, San Francisco , San Francisco , CA , USA
| |
Collapse
|
220
|
Gao J, Grill RJ, Dunn TJ, Bedi S, Labastida JA, Hetz RA, Xue H, Thonhoff JR, DeWitt DS, Prough DS, Cox CS, Wu P. Human Neural Stem Cell Transplantation-Mediated Alteration of Microglial/Macrophage Phenotypes after Traumatic Brain Injury. Cell Transplant 2018; 25:1863-1877. [PMID: 26980267 DOI: 10.3727/096368916x691150] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Neural stem cells (NSCs) promote recovery from brain trauma, but neuronal replacement is unlikely the sole underlying mechanism. We hypothesize that grafted NSCs enhance neural repair at least partially through modulating the host immune response after traumatic brain injury (TBI). C57BL/6 mice were intracerebrally injected with primed human NSCs (hNSCs) or vehicle 24 h after a severe controlled cortical impact injury. Six days after transplantation, brain tissues were collected for Western blot and immunohistochemical analyses. Observations included indicators of microglia/macrophage activation, M1 and M2 phenotypes, axonal injury detected by amyloid precursor protein (APP), lesion size, and the fate of grafted hNSCs. Animals receiving hNSC transplantation did not show significant decreases of brain lesion volumes compared to transplantation procedures with vehicle alone, but did show significantly reduced injury-dependent accumulation of APP. Furthermore, intracerebral transplantation of hNSCs reduced microglial activation as shown by a diminished intensity of Iba1 immunostaining and a transition of microglia/macrophages toward the M2 anti-inflammatory phenotype. The latter was represented by an increase in the brain M2/M1 ratio and increases of M2 microglial proteins. These phenotypic switches were accompanied by the increased expression of anti-inflammatory interleukin-4 receptor α and decreased proinflammatory interferon-γ receptor β. Finally, grafted hNSCs mainly differentiated into neurons and were phagocytized by either M1 or M2 microglia/macrophages. Thus, intracerebral transplantation of primed hNSCs efficiently leads host microglia/macrophages toward an anti-inflammatory phenotype that presumably contributes to stem cell-mediated neuroprotective effects after severe TBI in mice.
Collapse
Affiliation(s)
- Junling Gao
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Raymond J Grill
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Tiffany J Dunn
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Supinder Bedi
- Department of Pediatric Surgery, University of Texas Medical School at Houston, Houston, TX, USA
| | - Javier Allende Labastida
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Robert A Hetz
- Department of Pediatric Surgery, University of Texas Medical School at Houston, Houston, TX, USA
| | - Hasen Xue
- Department of Pediatric Surgery, University of Texas Medical School at Houston, Houston, TX, USA
| | - Jason R Thonhoff
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Douglas S DeWitt
- Department of Anesthesiology, University of Texas Medical Branch at Galveston, TX, USA
| | - Donald S Prough
- Department of Anesthesiology, University of Texas Medical Branch at Galveston, TX, USA
| | - Charles S Cox
- Department of Pediatric Surgery, University of Texas Medical School at Houston, Houston, TX, USA
| | - Ping Wu
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch at Galveston, Galveston, TX, USA.,Beijing Institute for Brain Disorders, Beijing, P.R. China
| |
Collapse
|
221
|
George N, Geller HM. Extracellular matrix and traumatic brain injury. J Neurosci Res 2018; 96:573-588. [PMID: 29344975 DOI: 10.1002/jnr.24151] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 07/21/2017] [Accepted: 08/14/2017] [Indexed: 12/27/2022]
Abstract
The brain extracellular matrix (ECM) plays a crucial role in both the developing and adult brain by providing structural support and mediating cell-cell interactions. In this review, we focus on the major constituents of the ECM and how they function in both normal and injured brain, and summarize the changes in the composition of the ECM as well as how these changes either promote or inhibit recovery of function following traumatic brain injury (TBI). Modulation of ECM composition to facilitates neuronal survival, regeneration and axonal outgrowth is a potential therapeutic target for TBI treatment.
Collapse
Affiliation(s)
- Naijil George
- Laboratory of Developmental Neurobiology, Cell Biology and Physiology Center, NHLBI, NIH, Bethesda, MD, 20892-1603, USA
| | - Herbert M Geller
- Laboratory of Developmental Neurobiology, Cell Biology and Physiology Center, NHLBI, NIH, Bethesda, MD, 20892-1603, USA
| |
Collapse
|
222
|
Bhatti J, Nascimento B, Akhtar U, Rhind SG, Tien H, Nathens A, da Luz LT. Systematic Review of Human and Animal Studies Examining the Efficacy and Safety of N-Acetylcysteine (NAC) and N-Acetylcysteine Amide (NACA) in Traumatic Brain Injury: Impact on Neurofunctional Outcome and Biomarkers of Oxidative Stress and Inflammation. Front Neurol 2018; 8:744. [PMID: 29387038 PMCID: PMC5776005 DOI: 10.3389/fneur.2017.00744] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 12/22/2017] [Indexed: 12/22/2022] Open
Abstract
Background No new therapies for traumatic brain injury (TBI) have been officially translated into current practice. At the tissue and cellular level, both inflammatory and oxidative processes may be exacerbated post-injury and contribute to further brain damage. N-acetylcysteine (NAC) has the potential to downregulate both processes. This review focuses on the potential neuroprotective utility of NAC and N-acetylcysteine amide (NACA) post-TBI. Methods Medline, Embase, Cochrane Library, and ClinicalTrials.gov were searched up to July 2017. Studies that examined clinical and laboratory effects of NAC and NACA post-TBI in human and animal studies were included. Risk of bias was assessed in human and animal studies according to the design of each study (randomized or not). The primary outcome assessed was the effect of NAC/NACA treatment on functional outcome, while secondary outcomes included the impact on biomarkers of inflammation and oxidation. Due to the clinical and methodological heterogeneity observed across studies, no meta-analyses were conducted. Results Our analyses revealed only three human trials, including two randomized controlled trials (RCTs) and 20 animal studies conducted using standardized animal models of brain injury. The two RCTs reported improvement in the functional outcome post-NAC/NACA administration. Overall, the evidence from animal studies is more robust and demonstrated substantial improvement of cognition and psychomotor performance following NAC/NACA use. Animal studies also reported significantly more cortical sparing, reduced apoptosis, and lower levels of biomarkers of inflammation and oxidative stress. No safety concerns were reported in any of the studies included in this analysis. Conclusion Evidence from the animal literature demonstrates a robust association for the prophylactic application of NAC and NACA post-TBI with improved neurofunctional outcomes and downregulation of inflammatory and oxidative stress markers at the tissue level. While a growing body of scientific literature suggests putative beneficial effects of NAC/NACA treatment for TBI, the lack of well-designed and controlled clinical investigations, evaluating therapeutic outcomes, prognostic biomarkers, and safety profiles, limits definitive interpretation and recommendations for its application in humans at this time.
Collapse
Affiliation(s)
- Junaid Bhatti
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Barto Nascimento
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Umbreen Akhtar
- Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Shawn G Rhind
- Defense Research and Development Canada (DRDC), Toronto Research Centre, Toronto, ON, Canada
| | - Homer Tien
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Avery Nathens
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Luis Teodoro da Luz
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
223
|
Taylor AN, Tio DL, Paydar A, Sutton RL. Sex Differences in Thermal, Stress, and Inflammatory Responses to Minocycline Administration in Rats with Traumatic Brain Injury. J Neurotrauma 2018; 35:630-638. [PMID: 29179648 DOI: 10.1089/neu.2017.5238] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Persistent inflammation, mediated in part by increases in cytokines, is a hallmark of traumatlc brain injury (TBI). Minocycline has been shown to inhibit post-TBI neuroinflammation in male rats and mice, but has not been tested in females. Here, we studied sex differences in thermal, stress, and inflammatory responses to TBI and minocycline. Female rats were ovariectomized under isoflurane anesthesia at 33-36 days of age. At 45-55 days of age, male and female rats were implanted intraperitoneally (i.p.) with calibrated transmitters for monitoring body temperature. Moderate cortical contusion injury (CCI) or sham surgery was performed when the rats attained 60-70 days of age. One hour after surgery, rats were injected i.p. with minocycline (50 mg/kg) or saline (0.3 mL); injections were repeated once daily for the next 3 days. At 28 days after CCI or sham surgery, 30 min restraint stress was initiated and blood samples were obtained by tail venipuncture before the onset of restraint and at 30, 60, and 90 min after stress onset. At 35 days after CCI or sham surgery, rats were decapitated and blood was collected for corticosterone (CORT) and cytokine analysis. The brains were removed and ipsilateral cortical tissue and hippocampus were dissected and subsequently assayed for interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α. Hyperthermia occurred during days 1-6 post-CCI in male rats, but only on the day of CCI in female rats, and minocycline prevented its occurrence in both sexes. Minocycline facilitated suppression of the CORT response to restraint stress in both sexes. In females, but not males, hippocampal IL-6 content increased post-CCI compared with sham-injured controls, whereas IL-1β content was augmented by minocycline. Hippocampal TNF-α was unaffected by CCI and minocycline. These results demonstrate sex differences in immediate thermal and long-lasting stress and cytokine responses to CCI, and only short-term protective effects of minocycline on hyperthermia.
Collapse
Affiliation(s)
- Anna N Taylor
- 1 Department of Neurobiology, David Geffen School of Medicine at UCLA , Los Angeles, California
| | - Delia L Tio
- 1 Department of Neurobiology, David Geffen School of Medicine at UCLA , Los Angeles, California
| | - Afshin Paydar
- 2 Department of Neurosurgery, David Geffen School of Medicine at UCLA , Los Angeles, California
| | - Richard L Sutton
- 2 Department of Neurosurgery, David Geffen School of Medicine at UCLA , Los Angeles, California
| |
Collapse
|
224
|
Ondruschka B, Schuch S, Pohlers D, Franke H, Dreßler J. Acute phase response after fatal traumatic brain injury. Int J Legal Med 2018; 132:531-539. [PMID: 29306988 DOI: 10.1007/s00414-017-1768-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 12/15/2017] [Indexed: 12/20/2022]
Abstract
An inflammatory response occurring after fatal traumatic brain injury (TBI) initiates time-dependent cascades of acute phase response. This may offer the potential to monitor postmortem biomarker levels of several pro-inflammatory cytokines to gain information about the cause of death and the trauma survival time. Cerebrospinal fluid (CSF) and serum samples were collected from forensic autopsies of 95 adult cadavers after postmortem intervals up to 6 days. The cases were divided according to their cause of death into fatal TBI (n = 46) with different survival times and age- and gender-matching non-TBI fatalities as controls (n = 49). Quantitative marker levels of interleukin-6 (IL-6), ferritin, soluble tumor necrosis factor receptor type 1, C-reactive protein, and lactate dehydrogenase were analyzed using immunoassays. Standardized statistical tests were performed to differentiate causes of death and survival time of TBI cases. The CSF IL-6, ferritin, and LDH levels after TBI were significantly higher than those in the controls (p < 0.001). Only serum IL-6 values showed comparable differences (p < 0.05). Both CSF and serum ferritin levels were discriminative between early and delayed death after TBI (p < 0.05). There were partly distinctive correlations between marker levels in both fluids with rising values after longer survival. There were up to moderate correlation between the marker levels and the postmortem interval due to postmortem hemolysis. However, neither CSF nor serum level ranges were affected by the age or gender of the subjects. This study is the first to measure all five proteins systematically in postmortem trauma cases. Ferritin and IL-6 proved themselves to be interesting postmortem biomarkers to provide specific information on the injury pattern and the survival time of traumatic fatalities. Such forensic investigations could serve as inexpensive and fast laboratory tests.
Collapse
Affiliation(s)
- Benjamin Ondruschka
- Institute of Legal Medicine, Medical Faculty University of Leipzig, Johannisallee 28, 04103, Leipzig, Germany.
| | - Sandra Schuch
- Institute of Legal Medicine, Medical Faculty University of Leipzig, Johannisallee 28, 04103, Leipzig, Germany
| | - Dirk Pohlers
- Center of Diagnostics GmbH, Klinikum Chemnitz, Chemnitz, Germany
| | - Heike Franke
- Rudolf Boehm Institute of Pharmacology and Toxicology, Medical Faculty University of Leipzig, Leipzig, Germany
| | - Jan Dreßler
- Institute of Legal Medicine, Medical Faculty University of Leipzig, Johannisallee 28, 04103, Leipzig, Germany
| |
Collapse
|
225
|
McNeela AM, Bernick C, Hines RM, Hines DJ. TSPO regulation in reactive gliotic diseases. J Neurosci Res 2018; 96:978-988. [PMID: 29315754 DOI: 10.1002/jnr.24212] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 11/29/2017] [Accepted: 12/06/2017] [Indexed: 12/19/2022]
Abstract
The brain is the most metabolically active organ in the body. This high metabolic demand is apparent in that 60% of the brain is comprised of mitochondria-enriched cells. A disruption of the brain's ability to meet this immense metabolic demand is central to the pathogenesis of a multitude of neurological disorders, which range from depression to Alzheimer's disease. Central to these pathologies are glial signaling and energy metabolism cascades regulating apoptosis and inflammation. Thus, diseases causing inflammation and disruption of metabolism can be correlated with glial reactivity. Acutely, reactive gliosis provides a mechanism for limiting the progression of a disease. Following chronic activation, the ability of reactive gliosis to limit disease progression decreases and, in some cases, transitions into a harmful state. The necessity for a noninvasive biomarker of disease in the brain has linked reactive gliosis with an upregulation of translocator protein (TSPO). TSPO is an 18kDa protein that is both a therapeutic target for multiple acute and chronic neuroinflammatory diseases and the leading biomarker for Alzheimer's disease. Although a central function of TSPO is not well known, the protein was named for its ability to translocate cholesterol. Increased TSPO expression is an indicator of disrupted metabolic activity and increased reactive oxygen production. The changes in TSPO expression levels both temporally and spatially relate to the pathogenesis of stroke, Alzheimer's disease, traumatic brain injury, and depression. Therefore, research into the basic function and potential therapeutics targeting TSPO will have broad implications for many diseases of the brain.
Collapse
Affiliation(s)
- Adam M McNeela
- Department of Psychology, University of Nevada Las Vegas, Las Vegas, NV
| | - Charles Bernick
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV
| | - Rochelle M Hines
- Department of Psychology, University of Nevada Las Vegas, Las Vegas, NV
| | - Dustin J Hines
- Department of Psychology, University of Nevada Las Vegas, Las Vegas, NV
| |
Collapse
|
226
|
Chandel S, Gupta SK, Medhi B. Epileptogenesis following experimentally induced traumatic brain injury - a systematic review. Rev Neurosci 2018; 27:329-46. [PMID: 26581067 DOI: 10.1515/revneuro-2015-0050] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 10/21/2015] [Indexed: 12/20/2022]
Abstract
Traumatic brain injury (TBI) is a complex neurotrauma in civilian life and the battlefield with a broad spectrum of symptoms, long-term neuropsychological disability, as well as mortality worldwide. Posttraumatic epilepsy (PTE) is a common outcome of TBI with unknown mechanisms, followed by posttraumatic epileptogenesis. There are numerous rodent models of TBI available with varying pathomechanisms of head injury similar to human TBI, but there is no evidence for an adequate TBI model that can properly mimic all aspects of clinical TBI and the first successive spontaneous focal seizures follow a single episode of neurotrauma with respect to epileptogenesis. This review aims to provide current information regarding the various experimental animal models of TBI relevant to clinical TBI. Mossy fiber sprouting, loss of dentate hilar neurons along with recurrent seizures, and epileptic discharge similar to human PTE have been studied in fluid percussion injury, weight-drop injury, and cortical impact models, but further refinement of animal models and functional test is warranted to better understand the underlying pathophysiology of posttraumatic epileptogenesis. A multifaceted research approach in TBI model may lead to exploration of the potential treatment measures, which are a major challenge to the research community and drug developers. With respect to clinical setting, proper patient data collection, improved clinical trials with advancement in drug delivery strategies, blood-brain barrier permeability, and proper monitoring of level and effects of target drug are also important.
Collapse
|
227
|
Kosari-Nasab M, Shokouhi G, Ghorbanihaghjo A, Abbasi MM, Salari AA. Anxiolytic- and antidepressant-like effects of Silymarin compared to diazepam and fluoxetine in a mouse model of mild traumatic brain injury. Toxicol Appl Pharmacol 2018; 338:159-173. [DOI: 10.1016/j.taap.2017.11.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 11/15/2017] [Accepted: 11/17/2017] [Indexed: 12/31/2022]
|
228
|
Cook S, Hung V, Duncan KA. Crosstalk between Estrogen Withdrawal and NFκB Signaling following Penetrating Brain Injury. Neuroimmunomodulation 2018; 25:193-200. [PMID: 30423555 DOI: 10.1159/000493506] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 09/04/2018] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES Characterized by neuroinflammation, traumatic brain injury (TBI) induces neuropathological changes and cognitive deficits. Estrogens are neuroprotective by increasing cell survival and this increase is mediated by a decrease in neuroinflammation. To further explore the relationship between estrogens, brain injury, and neuroinflammation, we examined the expression of the IKK/NFκB complex. The IKK/NFκB complex is a pleiotropic regulator of many cellular signaling pathways linked to inflammation, as well as three major cytokines (IL-1β, IL-6, and TNF-α). We hypothesized that NFκB expression would be upregulated following injury and that this increase would be exacerbated when circulating estrogens were decreased with fadrozole (aromatase inhibitor). METHODS Using adult zebra finches, we first determined the expression of major components of the NFκB complex (NFκB, IκB-α, and IκB-β) following injury using qPCR. Next, male and female finches were collected at 2 time points (2 or 24 h after injury) and brain tissue was analyzed to determine whether NFκB expression was differentially expressed in males and females at either time point. Finally, we examined how the expression of NFκB changed when estrogen levels were decreased immediately after injury. RESULTS Our study documented an increase in the expression of the major components of the NFκB complex (NFκB, IκB-α, and IκB-β) following injury. Decreasing estrogen levels resulted in a surprising decrease in the NFκB complex studied here. DISCUSSION These data further expand the model of how estrogens and other steroid hormones interact with the inflammatory pathways following injury and may prove beneficial when developing therapies for treatment of TBI.
Collapse
Affiliation(s)
- Samarah Cook
- Program in Neuroscience and Behavior, Vassar College, Poughkeepsie, New York, USA
| | - Vanessa Hung
- Program in Neuroscience and Behavior, Vassar College, Poughkeepsie, New York, USA
| | - Kelli A Duncan
- Program in Neuroscience and Behavior, Vassar College, Poughkeepsie, New York, USA,
- Department of Biology, Vassar College, Poughkeepsie, New York, USA,
| |
Collapse
|
229
|
Gincberg G, Shohami E, Trembovler V, Alexandrovich AG, Lazarovici P, Elchalal U. Nerve growth factor plays a role in the neurotherapeutic effect of a CD45 + pan-hematopoietic subpopulation derived from human umbilical cord blood in a traumatic brain injury model. Cytotherapy 2017; 20:245-261. [PMID: 29274773 DOI: 10.1016/j.jcyt.2017.11.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Revised: 10/09/2017] [Accepted: 11/14/2017] [Indexed: 01/09/2023]
Abstract
BACKGROUND AIMS Human umbilical cord blood (HUCB) is an important source of stem cells for therapy of hematopoietic disorders and is a potential therapy for various neurological disorders, including traumatic brain injury (TBI). The expression of nerve growth factor (NGF) and its receptors TrkA, p75NTR and α9β1 integrin on an HUCB CD45+ pan-hematopoietic subpopulation was investigated in the context of its neurotherapeutic potential after TBI. METHODS NGF and its receptors were detected on CD45+ cells by reverse transcriptase polymerase chain reaction, flow cytometry analysis and confocal microscopy. CD45+ cells were stimulated by TBI brain extracts, and NGF levels were measured by enzyme-linked immunosorbent assay. TBI mice were divided into six groups for xenogeneic intravenous transplantation, 1 day post-trauma, with 1 × 106 CD45+ cells untreated or treated with the anti-NGF neutralizing antibody K252a, a TrkA antagonist; VLO5, an α9β1 disintegrin; or negative (vehicle) and positive (NGF) controls. RESULTS The HUCB CD45+ subpopulation constitutively expresses NGF and its receptors, mainly TrkA and p75NTR and minor levels of α9β1. In vitro experiments provided evidence that trauma-related mediators from brain extracts of TBI mice induced release of NGF from HUCB CD45+ cell cultures. HUCB CD45+ cells induced a neurotherapeutic effect in TBI mice, abrogated by cell treatment with either anti-NGF antibody or K252a, but not VLO5. CONCLUSIONS These findings strengthen the role of NGF and its TrkA receptor in the HUCB CD45+ subpopulation's neurotherapeutic effect. The presence of neurotrophin receptors in the HUCB CD45+ pan-hematopoietic subpopulation may explain the neuroprotective effect of cord blood in therapy of a variety of neurological disorders.
Collapse
Affiliation(s)
- Galit Gincberg
- School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Esther Shohami
- School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Victoria Trembovler
- School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Alexander G Alexandrovich
- School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Philip Lazarovici
- School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Uriel Elchalal
- Department of Obstetrics and Gynecology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
230
|
Woodcock TM, Frugier T, Nguyen TT, Semple BD, Bye N, Massara M, Savino B, Besio R, Sobacchi C, Locati M, Morganti-Kossmann MC. The scavenging chemokine receptor ACKR2 has a significant impact on acute mortality rate and early lesion development after traumatic brain injury. PLoS One 2017; 12:e0188305. [PMID: 29176798 PMCID: PMC5703564 DOI: 10.1371/journal.pone.0188305] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 11/04/2017] [Indexed: 12/17/2022] Open
Abstract
The atypical chemokine receptor ACKR2 promotes resolution of acute inflammation by operating as a scavenger receptor for inflammatory CC chemokines in several experimental models of inflammatory disorders, however its role in the brain remains unclear. Based on our previous reports of increased expression of inflammatory chemokines and their corresponding receptors following traumatic brain injury (TBI), we hypothesised that ACKR2 modulates neuroinflammation following brain trauma and that its deletion exacerbates cellular inflammation and chemokine production. We demonstrate increased CCL2 and ACKR2 mRNA expression in post-mortem human brain, whereby ACKR2 mRNA levels correlated with later times post-TBI. This data is consistent with the transient upregulation of ACKR2 observed in mouse brain after closed head injury (CHI). As compared to WT animals, ACKR2-/- mice showed a higher mortality rate after CHI, while the neurological outcome in surviving mice was similar. At day 1 post-injury, ACKR2-/- mice displayed aggravated lesion volume and no differences in CCL2 expression and macrophage recruitment relative to WT mice. Reciprocal regulation of ACKR2 and CCL2 expression was explored in cultured astrocytes, which are recognized as the major source of CCL2 and also express ACKR2. ACKR2 mRNA increased as early as 2 hours after an inflammatory challenge in WT astrocytes. As expected, CCL2 expression also dramatically increased at 4 hours in WT astrocytes but was significantly lower in ACKR2-/- astrocytes, possibly indicating a co-regulation of CCL2 and ACKR2 in these cells. Conversely, in vivo, CCL2 mRNA/protein levels were increased similarly in ACKR2-/- and WT brains at 4 and 12 hours after CHI, in line with the lack of differences in cerebral macrophage recruitment and neurological recovery. In conclusion, ACKR2 is induced after TBI and has a significant impact on mortality and lesion development acutely following CHI, while its role in chemokine expression, macrophage activation, brain pathology, and neurological recovery at later time-points is minor. Concordant to evidence in multiple sclerosis experimental models, our data corroborate a distinct role for ACKR2 in cerebral inflammatory processes compared to its reported functions in peripheral tissues.
Collapse
MESH Headings
- Animals
- Astrocytes/metabolism
- Astrocytes/pathology
- Bone and Bones/pathology
- Brain/metabolism
- Brain/pathology
- Brain/physiopathology
- Brain Injuries, Traumatic/genetics
- Brain Injuries, Traumatic/metabolism
- Brain Injuries, Traumatic/mortality
- Brain Injuries, Traumatic/physiopathology
- Cells, Cultured
- Chemokine CCL2/genetics
- Chemokine CCL2/metabolism
- Gene Deletion
- Humans
- Inflammation/pathology
- Macrophages/metabolism
- Macrophages/pathology
- Male
- Mice, Inbred C57BL
- Mortality
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Chemokine/genetics
- Receptors, Chemokine/metabolism
- Recovery of Function
- Up-Regulation/genetics
Collapse
Affiliation(s)
- Thomas M. Woodcock
- National Trauma Research Institute, The Alfred Hospital, Melbourne, Australia
- Department of Surgery, Monash University, Melbourne, Australia
| | - Tony Frugier
- Department of Pharmacology and Therapeutics School of Biomedical Sciences, The University of Melbourne, Melboune, Australia
| | - Tan Thanh Nguyen
- National Trauma Research Institute, The Alfred Hospital, Melbourne, Australia
- Department of Surgery, Monash University, Melbourne, Australia
| | - Bridgette Deanne Semple
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Melbourne, Australia
| | - Nicole Bye
- Division of Pharmacy, School of Medicine, University of Tasmania, Hobart, Australia
| | - Matteo Massara
- Humanitas Clinical and Research Center, Rozzano, Italy
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Benedetta Savino
- Humanitas Clinical and Research Center, Rozzano, Italy
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Roberta Besio
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Cristina Sobacchi
- Humanitas Clinical and Research Center, Rozzano, Italy
- Istituto di Ricerca Genetica e Biomedica Milan Unit, National Research Council, Milan, Italy
| | - Massimo Locati
- Humanitas Clinical and Research Center, Rozzano, Italy
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
- * E-mail: (MCMK); (ML)
| | - Maria Cristina Morganti-Kossmann
- Department of Epidemiology and Preventive Medicine, and Australian New Zealand Intensive Care Research Centre, Monash University, Melbourne, Australia
- Barrow Neurological Institute, Department of Child Health, University of Arizona, Phoenix, AZ, United States of America
- * E-mail: (MCMK); (ML)
| |
Collapse
|
231
|
Hui B, Zhang L, Zhou Q, Hui L. Pristimerin Inhibits LPS-Triggered Neurotoxicity in BV-2 Microglia Cells Through Modulating IRAK1/TRAF6/TAK1-Mediated NF-κB and AP-1 Signaling Pathways In Vitro. Neurotox Res 2017; 33:268-283. [PMID: 29119451 DOI: 10.1007/s12640-017-9837-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 09/24/2017] [Accepted: 10/20/2017] [Indexed: 12/25/2022]
Abstract
Microglia plays a prominent role in the brain's inflammatory response to injury or infection by migrating to affected locations and secreting inflammatory molecules. However, hyperactivated microglial is neurotoxic and plays critical roles in the pathogenesis of neurodegenerative diseases. Pristimerin, a naturally occurring triterpenoid, possesses antitumor, antioxidant, and anti-inflammatory activities. However, the effect and the molecular mechanism of pristimerin against lipopolysaccharide (LPS)-induced neurotoxicity in microglia remain to be revealed. In the present study, using BV-2 microglial cultures, we investigated whether pristimerin modifies neurotoxicity after LPS stimulation and which intracellular pathways are involved in the effect of pristimerin. Here we show that pristimerin markedly suppressed the release of Regulated on Activation, Normal T Expressed and Secreted (RANTES), transforming growth factor-β1 (TGF-β1), IL-6, tumor necrosis factor-α (TNF-α), and nitric oxide (NO). Pristimerin also significantly inhibited migration of BV-2 microglia and alleviated the death of neuron-like PC12 cell induced by the conditioned medium from LPS-activated BV-2 microglial cells. Moreover, pristimerin reduced the expression and interaction of TNF Receptor-Associated Factor 6 (TRAF6) and Interleukin-1 Receptor-Associated Kinases (IRAK1), limiting TGF-beta activating kinase 1 (TAK1) activation, and resulting in an inhibition of IKKα/β/NF-κB and MKK7/JNK/AP-1 signaling pathway in LPS-activated BV-2 microglia. Taken together, the anti-neurotoxicity action of pristimerin is mediated through the inhibition of TRAF6/IRAK1/TAK1 interaction as well as the related pathways: IKKα/β/NF-κB and MKK7/JNK/AP-1 signaling pathways. These findings may suggest that pristimerin might serve as a new therapeutic agent for treating hyperactivated microglial induced neurodegenerative diseases.
Collapse
Affiliation(s)
- Bin Hui
- College of Pharmacy, Shanghai University of Medical & Health Sciences, Shanghai, China
| | - Liping Zhang
- Department of Emergency Medicine, Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Qinhua Zhou
- College of Pharmacy, Shanghai University of Medical & Health Sciences, Shanghai, China. .,Department of Pharmacology, College of Medicine, Jiaxing University, Jiaxing, China.
| | - Ling Hui
- Center for Experimental Medicine, Lanzhou Military Command, Lanzhou General Hospital, Lanzhou, Gansu, China
| |
Collapse
|
232
|
Kaplan GB, Leite-Morris KA, Wang L, Rumbika KK, Heinrichs SC, Zeng X, Wu L, Arena DT, Teng YD. Pathophysiological Bases of Comorbidity: Traumatic Brain Injury and Post-Traumatic Stress Disorder. J Neurotrauma 2017; 35:210-225. [PMID: 29017388 DOI: 10.1089/neu.2016.4953] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The high rates of traumatic brain injury (TBI) and post-traumatic stress disorder (PTSD) diagnoses encountered in recent years by the United States Veterans Affairs Healthcare System have increased public awareness and research investigation into these conditions. In this review, we analyze the neural mechanisms underlying the TBI/PTSD comorbidity. TBI and PTSD present with common neuropsychiatric symptoms including anxiety, irritability, insomnia, personality changes, and memory problems, and this overlap complicates diagnostic differentiation. Interestingly, both TBI and PTSD can be produced by overlapping pathophysiological changes that disrupt neural connections termed the "connectome." The neural disruptions shared by PTSD and TBI and the comorbid condition include asymmetrical white matter tract abnormalities and gray matter changes in the basolateral amygdala, hippocampus, and prefrontal cortex. These neural circuitry dysfunctions result in behavioral changes that include executive function and memory impairments, fear retention, fear extinction deficiencies, and other disturbances. Pathophysiological etiologies can be identified using experimental models of TBI, such as fluid percussion or blast injuries, and for PTSD, using models of fear conditioning, retention, and extinction. In both TBI and PTSD, there are discernible signs of neuroinflammation, excitotoxicity, and oxidative damage. These disturbances produce neuronal death and degeneration, axonal injury, and dendritic spine dysregulation and changes in neuronal morphology. In laboratory studies, various forms of pharmacological or psychological treatments are capable of reversing these detrimental processes and promoting axonal repair, dendritic remodeling, and neurocircuitry reorganization, resulting in behavioral and cognitive functional enhancements. Based on these mechanisms, novel neurorestorative therapeutics using anti-inflammatory, antioxidant, and anticonvulsant agents may promote better outcomes for comorbid TBI and PTSD.
Collapse
Affiliation(s)
- Gary B Kaplan
- 1 Mental Health Service , VA Boston Healthcare System, Brockton, Massachusetts.,2 Department of Psychiatry, Boston University School of Medicine , Boston, Massachusetts.,3 Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine , Boston, Massachusetts
| | - Kimberly A Leite-Morris
- 2 Department of Psychiatry, Boston University School of Medicine , Boston, Massachusetts.,3 Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine , Boston, Massachusetts.,4 Research Service, VA Boston Healthcare System , Jamaica Plain, Massachusetts
| | - Lei Wang
- 5 Division of Spinal Cord Injury Research, VA Boston Healthcare System , West Roxbury, Massachusetts.,6 Departments of Physical Medicine and Rehabilitation and Neurosurgery, Harvard Medical School , Boston, Massachusetts
| | - Kendra K Rumbika
- 7 Research Service, VA Boston Healthcare System , West Roxbury, Massachusetts
| | - Stephen C Heinrichs
- 7 Research Service, VA Boston Healthcare System , West Roxbury, Massachusetts
| | - Xiang Zeng
- 5 Division of Spinal Cord Injury Research, VA Boston Healthcare System , West Roxbury, Massachusetts.,6 Departments of Physical Medicine and Rehabilitation and Neurosurgery, Harvard Medical School , Boston, Massachusetts
| | - Liquan Wu
- 5 Division of Spinal Cord Injury Research, VA Boston Healthcare System , West Roxbury, Massachusetts.,6 Departments of Physical Medicine and Rehabilitation and Neurosurgery, Harvard Medical School , Boston, Massachusetts
| | - Danielle T Arena
- 7 Research Service, VA Boston Healthcare System , West Roxbury, Massachusetts
| | - Yang D Teng
- 5 Division of Spinal Cord Injury Research, VA Boston Healthcare System , West Roxbury, Massachusetts.,6 Departments of Physical Medicine and Rehabilitation and Neurosurgery, Harvard Medical School , Boston, Massachusetts
| |
Collapse
|
233
|
Thelin EP, Hall CE, Gupta K, Carpenter KLH, Chandran S, Hutchinson PJ, Patani R, Helmy A. Elucidating Pro-Inflammatory Cytokine Responses after Traumatic Brain Injury in a Human Stem Cell Model. J Neurotrauma 2017; 35:341-352. [PMID: 28978285 PMCID: PMC5784793 DOI: 10.1089/neu.2017.5155] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Cytokine mediated inflammation likely plays an important role in secondary pathology after traumatic brain injury (TBI). The aim of this study was to elucidate secondary cytokine responses in an in vitro enriched (>80%) human stem cell-derived neuronal model. We exposed neuronal cultures to pre-determined and clinically relevant pathophysiological levels of tumor necrosis factor-α (TNF), interleukin-6 (IL-6) and interleukin-1β (IL-1β), shown to be present in the inflammatory aftermath of TBI. Data from this reductionist human model were then compared with our in vivo data. Human embryonic stem cell (hESC)-derived neurons were exposed to recombinant TNF (1–10,000 pg/mL), IL-1β (1–10,000 pg/mL), and IL-6 (0.1–1000 ng/mL). After 1, 24, and 72 h, culture supernatant was sampled and analyzed using a human cytokine/chemokine 42-plex Milliplex kit on the Luminex platform. The culture secretome revealed both a dose- and/or time-dependent release of cytokines. The IL-6 and TNF exposure each resulted in significantly increased levels of >10 cytokines over time, while IL-1β increased the level of C-X-C motif chemokine 10 (CXCL10/IP10) alone. Importantly, these patterns are consistent with our in vivo (human) TBI data, thus validating our human stem cell-derived neuronal platform as a clinically useful reductionist model. Our data cumulatively suggest that IL-6 and TNF have direct actions, while the action of IL-1β on human neurons likely occurs indirectly through inflammatory cells. The hESC-derived neurons provide a valuable platform to model cytokine mediated inflammation and can provide important insights into the mechanisms of neuroinflammation after TBI.
Collapse
Affiliation(s)
- Eric Peter Thelin
- 1 Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge , Cambridge, United Kingdom .,2 Department of Clinical Neuroscience, Karolinska Institutet , Stockholm, Sweden
| | - Claire E Hall
- 3 Department of Molecular Neuroscience, Institute of Neurology, University College London , London, United Kingdom
| | - Kunal Gupta
- 4 Department of Neurological Surgery, Oregon Health & Science University , Portland, Oregon
| | - Keri L H Carpenter
- 1 Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge , Cambridge, United Kingdom .,5 Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge , Cambridge, United Kingdom
| | - Siddharthan Chandran
- 6 Centre for Clinical Brain Sciences, University of Edinburgh , Edinburgh, United Kingdom
| | - Peter J Hutchinson
- 1 Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge , Cambridge, United Kingdom .,5 Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge , Cambridge, United Kingdom
| | - Rickie Patani
- 3 Department of Molecular Neuroscience, Institute of Neurology, University College London , London, United Kingdom .,7 The Francis Crick Institute , London, United Kingdom
| | - Adel Helmy
- 1 Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge , Cambridge, United Kingdom
| |
Collapse
|
234
|
Attiq A, Jalil J, Husain K. Annonaceae: Breaking the Wall of Inflammation. Front Pharmacol 2017; 8:752. [PMID: 29104539 PMCID: PMC5654839 DOI: 10.3389/fphar.2017.00752] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 10/03/2017] [Indexed: 12/12/2022] Open
Abstract
Inventories of tropical forests have listed Annonaceae as one of the most diverse plant families. For centuries, it is employed in traditional medicines to cure various pathological conditions including snakebite, analgesic, astringent, diarrhea, dysentery, arthritis pain, rheumatism, neuralgia, and weight loss etc. Phytochemical analysis of Annonaceae family have reported the occurrence of alkaloids, flavonoids, triterpenes, diterpenes and diterpene flavone glycosides, sterols, lignans, and annonaceous acetogenin characteristically affiliated with Annonaceae sp. Numerous past studies have underlined the pleotropic pharmacological activities of the crude extracts and isolated compounds from Annonaceae species. This review is an effort to abridge the ethnobotany, morphology, phytochemistry, toxicity, and particularly focusing on the anti-inflammatory activity of the Annonaceae species.
Collapse
Affiliation(s)
- Ali Attiq
- Drug and Herbal Research Centre, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Juriyati Jalil
- Drug and Herbal Research Centre, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Khairana Husain
- Drug and Herbal Research Centre, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
235
|
Schimmel SJ, Acosta S, Lozano D. Neuroinflammation in traumatic brain injury: A chronic response to an acute injury. Brain Circ 2017; 3:135-142. [PMID: 30276315 PMCID: PMC6057689 DOI: 10.4103/bc.bc_18_17] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 09/02/2017] [Accepted: 09/05/2017] [Indexed: 02/06/2023] Open
Abstract
Every year, approximately 1.4 million US citizens visit emergency rooms for traumatic brain injuries. Formerly known as an acute injury, chronic neurodegenerative symptoms such as compromised motor skills, decreased cognitive abilities, and emotional and behavioral changes have caused the scientific community to consider chronic aspects of the disorder. The injury causing impact prompts multiple cell death processes, starting with neuronal necrosis, and progressing to various secondary cell death mechanisms. Secondary cell death mechanisms, including excitotoxicity, oxidative stress, mitochondrial dysfunction, blood-brain barrier disruption, and inflammation accompany chronic traumatic brain injury (TBI) and often contribute to long-term disabilities. One hallmark of both acute and chronic TBI is neuroinflammation. In acute stages, neuroinflammation is beneficial and stimulates an anti-inflammatory response to the damage. Conversely, in chronic TBI, excessive inflammation stimulates the aforementioned secondary cell death. Converting inflammatory cells from pro-inflammatory to anti-inflammatory may expand the therapeutic window for treating TBI, as inflammation plays a role in all stages of the injury. By expanding current research on the role of inflammation in TBI, treatment options and clinical outcomes for afflicted individuals may improve. This paper is a review article. Referred literature in this paper has been listed in the references section. The data sets supporting the conclusions of this article are available online by searching various databases, including PubMed. Some original points in this article come from the laboratory practice in our research center and the authors' experiences.
Collapse
Affiliation(s)
| | - Sandra Acosta
- Center of Excellence for Aging and Brain, Tampa, FL, USA
| | - Diego Lozano
- School of Medicine, University of Miami School of Medicine, Miami, FL, USA
| |
Collapse
|
236
|
Polysaccharide Hydrogels Support the Long-Term Viability of Encapsulated Human Mesenchymal Stem Cells and Their Ability to Secrete Immunomodulatory Factors. Stem Cells Int 2017; 2017:9303598. [PMID: 29158741 PMCID: PMC5660815 DOI: 10.1155/2017/9303598] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 08/03/2017] [Accepted: 08/08/2017] [Indexed: 01/06/2023] Open
Abstract
While therapeutically interesting, the injection of MSCs suffers major limitations including cell death upon injection and a massive leakage outside the injection site. We proposed to entrap MSCs within spherical particles derived from alginate, as a control, or from silanized hydroxypropyl methylcellulose (Si-HPMC). We developed water in an oil dispersion method to produce small Si-HPMC particles with an average size of about 68 μm. We evidenced a faster diffusion of fluorescein isothiocyanate-dextran in Si-HPMC particles than in alginate ones. Human adipose-derived MSCs (hASC) were encapsulated either in alginate or in Si-HPMC, and the cellularized particles were cultured for up to 1 month. Both alginate and Si-HPMC particles supported cell survival, and the average number of encapsulated hASC per alginate and Si-HPMC particle (7102 and 5100, resp.) did not significantly change. The stimulation of encapsulated hASC with proinflammatory cytokines resulted in the production of IDO, PGE2, and HGF whose concentration was always higher when cells were encapsulated in Si-HPMC particles than in alginate ones. We have demonstrated that Si-HPMC and alginate particles support hASC viability and the maintenance of their ability to secrete therapeutic factors.
Collapse
|
237
|
Neuroimmunology of Traumatic Brain Injury: Time for a Paradigm Shift. Neuron 2017; 95:1246-1265. [PMID: 28910616 DOI: 10.1016/j.neuron.2017.07.010] [Citation(s) in RCA: 467] [Impact Index Per Article: 66.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 07/07/2017] [Accepted: 07/10/2017] [Indexed: 12/14/2022]
Abstract
Traumatic brain injury (TBI) is a leading cause of morbidity and disability, with a considerable socioeconomic burden. Heterogeneity of pathoanatomical subtypes and diversity in the pathogenesis and extent of injury contribute to differences in the course and outcome of TBI. Following the primary injury, extensive and lasting damage is sustained through a complex cascade of events referred to as "secondary injury." Neuroinflammation is proposed as an important manipulable aspect of secondary injury in animal and human studies. Because neuroinflammation can be detrimental or beneficial, before developing immunomodulatory therapies, it is necessary to better understand the timing and complexity of the immune responses that follow TBI. With a rapidly increasing body of literature, there is a need for a clear summary of TBI neuroimmunology. This review presents our current understanding of the immune response to TBI in a chronological and compartment-based manner, highlighting early changes in gene expression and initial signaling pathways that lead to activation of innate and adaptive immunity. Based on recent advances in our understanding of innate immune cell activation, we propose a new paradigm to study innate immune cells following TBI that moves away from the existing M1/M2 classification of activation states toward a stimulus- and disease-specific understanding of polarization state based on transcriptomic and proteomic profiling.
Collapse
|
238
|
Zou Y, Xiong JB, Ma K, Wang AZ, Qian KJ. Rac2 deficiency attenuates CCl 4-induced liver injury through suppressing inflammation and oxidative stress. Biomed Pharmacother 2017; 94:140-149. [PMID: 28759751 DOI: 10.1016/j.biopha.2017.07.074] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 06/25/2017] [Accepted: 07/17/2017] [Indexed: 12/19/2022] Open
Abstract
Oxidative stress is a leading cause to liver injury. Rac2 is a Ras-associated guanosine triphosphatase, an important molecule modulating a large number of cells and involved in the regulation of reactive oxygen species (ROS). For the study described here, we supposed that Rac2 knockout protects mice against CCl4-induced acute liver injury. We found that Rac2 expressed highly in CCl4-induced liver tissues. CCl4-treated Rac2 knockout (Rac2-/-) mice had reduced CD24 levels and steatosis. In addition, CCl4-induced high expression of pro-inflammatory cytokines and chemokine were reversed by Rac2 deficiency compared to CCl4-treated wild type (WT) mice. We also found that fibrosis-related signals of MMP-9, MMP-2 and TGF-β1 were also down-regulated in Rac2 knockout mice induced by CCl4. Significantly, oxidative stress induced by CCl4 was also suppressed owing to the lack of Rac2, evidenced by enhanced superoxide dismutase (SOD) activity, and reduced malondialdehyde (MDA) levels, superoxide radical, H2O2, xanthine oxidase (XO), xanthine dehydrogenase (XDH) and XO/XDH ratio. Moreover, c-Jun N-terminal protein kinase mitogen-activated protein kinases (JNK MAPK) was activated by CCl4, which was reversed in the liver of Rac2-/- mice through western blot and immunohistochemical analysis. In vitro, endotoxin (LPS) was treated to hepatocytes isolated from WT mice and Rac2-/- mice. The data further confirmed the role of Rac2 deficiency suppressed pro-inflammatory cytokines and chemokine, as well as fibrosis-related signals. Of note, production of ROS induced by LPS was reduced in Rac2-/- cells, accompanied with enhanced SOD1, SOD2 and reduced XO and phosphorylated-JNK expressions. Our results indicated that Rac2 played an essential role in acute liver injury induced by CCl4, providing the compelling information of the effects of Rac2 on liver injury, and revealing a novel regulatory mechanism for acute liver injury.
Collapse
Affiliation(s)
- Yan Zou
- Department of Intensive Care Medicine, Shanghai Jiaotong University Affiliated Sixth People's Hospital, No. 222 Huanhuxisan Road, Pudong, Shanghai 201306, China
| | - Ji-Bin Xiong
- Department of Hyperbaric Oxygen Therapy, Shanghai Jiaotong University Affiliated Sixth People's Hospital, No. 222 Huanhuxisan Road, Pudong, Shanghai 201306, China
| | - Ke Ma
- Department of Emergency Medicine, Shanghai Jiaotong University Affiliated Sixth People's Hospital, No. 222 Huanhuxisan Road, Pudong, Shanghai 201306, China
| | - Ai-Zhong Wang
- Department of Anesthesiology, Shanghai Jiaotong University Affiliated Sixth People's Hospital, No. 222 Huanhuxisan Road, Pudong, Shanghai 201306, China
| | - Ke-Jian Qian
- Department of Intensive Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China.
| |
Collapse
|
239
|
Anthonymuthu TS, Kenny EM, Amoscato AA, Lewis J, Kochanek PM, Kagan VE, Bayır H. Global assessment of oxidized free fatty acids in brain reveals an enzymatic predominance to oxidative signaling after trauma. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2601-2613. [PMID: 28347845 PMCID: PMC5612836 DOI: 10.1016/j.bbadis.2017.03.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 03/15/2017] [Accepted: 03/23/2017] [Indexed: 12/31/2022]
Abstract
Traumatic brain injury (TBI) is a major health problem associated with significant morbidity and mortality. The pathophysiology of TBI is complex involving signaling through multiple cascades, including lipid peroxidation. Oxidized free fatty acids, a prominent product of lipid peroxidation, are potent cellular mediators involved in induction and resolution of inflammation and modulation of vasomotor tone. While previous studies have assessed lipid peroxidation after TBI, to our knowledge no studies have used a systematic approach to quantify the global oxidative changes in free fatty acids. In this study, we identified and quantified 244 free fatty acid oxidation products using a newly developed global liquid chromatography tandem-mass spectrometry (LC-MS/MS) method. This methodology was used to follow the time course of these lipid species in the contusional cortex of our pediatric rat model of TBI. We show that oxidation peaked at 1h after controlled cortical impact and was progressively attenuated at 4 and 24h time points. While enzymatic and non-enzymatic pathways were activated at 1h post-TBI, enzymatic lipid peroxidation was the predominant mechanism with 15-lipoxygenase (LOX) contributing to the majority of total oxidized fatty acid content. Pro-inflammatory lipid mediators were significantly increased at 1 and 4h after TBI with return to basal levels by 24h. Anti-inflammatory lipid mediators remained significantly increased across all three time points, indicating an elevated and sustained anti-inflammatory response following TBI.
Collapse
Affiliation(s)
- Tamil S Anthonymuthu
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15261, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15224, United States; Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA 15219, United States
| | - Elizabeth M Kenny
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15261, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15224, United States; Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA 15219, United States
| | - Andrew A Amoscato
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15219, United States
| | - Jesse Lewis
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15261, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15224, United States; Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA 15219, United States
| | - Patrick M Kochanek
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15261, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15224, United States; Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, PA 15224, United States
| | - Valerian E Kagan
- Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA 15219, United States; Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15219, United States
| | - Hülya Bayır
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15261, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15224, United States; Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA 15219, United States; Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15219, United States; Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, PA 15224, United States.
| |
Collapse
|
240
|
Esculentoside A inhibits LPS-induced BV2 microglia activation through activating PPAR-γ. Eur J Pharmacol 2017; 813:61-65. [DOI: 10.1016/j.ejphar.2017.07.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 07/14/2017] [Accepted: 07/17/2017] [Indexed: 12/11/2022]
|
241
|
Park G, Lee SH, Oh DS, Kim YU. Melatonin inhibits neuronal dysfunction-associated with neuroinflammation by atopic psychological stress in NC/Nga atopic-like mouse models. J Pineal Res 2017; 63. [PMID: 28500766 DOI: 10.1111/jpi.12420] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 05/05/2017] [Indexed: 01/03/2023]
Abstract
Atopic dermatitis (AD), also known as atopic eczema, is chronic pruritic skin disease. AD can increase psychological stress as well, increasing glucocorticoid release and exacerbating the associated symptoms. Chronic glucocorticoid elevation disturbs neuroendocrine signaling and can induce neuroinflammation, neurotoxicity, and cognitive impairment; however, it is unclear whether AD-related psychological stress elevates glucocorticoids enough to cause neuronal damage. Therefore, we assessed the effects of AD-induced stress in a mouse AD model. AD-related psychological stress increased astroglial and microglial activation, neuroinflammatory cytokine expression, and markers of neuronal loss. Notably, melatonin administration inhibited the development of skin lesions, scratching behavior, and serum IgE levels in the model mice, and additionally caused a significant reduction in corticotropin-releasing hormone responsiveness, and a significant reduction in neuronal damage. Finally, we produced similar results in a corticosterone-induced AD-like skin model. This is the first study to demonstrate that AD-related psychological stress increases neuroendocrine dysfunction, exacerbates neuroinflammation, and potentially accelerates other neurodegenerative disease states.
Collapse
Affiliation(s)
- Gunhyuk Park
- The K-herb Research Center, Korea Institute of Oriental Medicine, Daejeon, Korea
| | - Seung Hoon Lee
- The K-herb Research Center, Korea Institute of Oriental Medicine, Daejeon, Korea
| | - Dal-Seok Oh
- The K-herb Research Center, Korea Institute of Oriental Medicine, Daejeon, Korea
| | - Yong-Ung Kim
- Department of Pharmaceutical Engineering, College of Biomedical Science, Daegu Haany University, Gyeongsan, Korea
| |
Collapse
|
242
|
Anti-inflammatory and immunomodulatory mechanisms of atorvastatin in a murine model of traumatic brain injury. J Neuroinflammation 2017; 14:167. [PMID: 28835272 PMCID: PMC5569493 DOI: 10.1186/s12974-017-0934-2] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 08/07/2017] [Indexed: 12/25/2022] Open
Abstract
Background Neuroinflammation is an important secondary injury mechanism that has dual beneficial and detrimental roles in the pathophysiology of traumatic brain injury (TBI). Compelling data indicate that statins, a group of lipid-lowering drugs, also have extensive immunomodulatory and anti-inflammatory properties. Among statins, atorvastatin has been demonstrated as a neuroprotective agent in experimental TBI; however, there is a lack of evidence regarding its effects on neuroinflammation during the acute phase of TBI. The current study aimed to evaluate the effects of atorvastatin therapy on modulating the immune reaction, and to explore the possible involvement of peripheral leukocyte invasion and microglia/macrophage polarization in the acute period post-TBI. Methods C57BL/6 mice were subjected to TBI using a controlled cortical impact (CCI) device. Either atorvastatin or vehicle saline was administered orally starting 1 h post-TBI for three consecutive days. Short-term neurological deficits were evaluated using the modified neurological severity score (mNSS) and Rota-rod. Brain-invading leukocyte subpopulations were analyzed by flow cytometry and immunohistochemistry. Pro- and anti-inflammatory cytokines and chemokines were examined using enzyme-linked immunosorbent assay (ELISA). Markers of classically activated (M1) and alternatively activated (M2) microglia/macrophages were then determined by quantitative real-time PCR (qRT-PCR) and flow cytometry. Neuronal apoptosis was identified by double staining of terminal deoxynucleotidyl transferase-dUTP nick end labeling (TUNEL) staining and immunofluorescence labeling for neuronal nuclei (NeuN). Results Acute treatment with atorvastatin at doses of 1 mg/kg/day significantly reduced neuronal apoptosis and improved behavioral deficits. Invasions of T cells, neutrophils and natural killer (NK) cells were attenuated profoundly after atorvastatin therapy, as was the production of pro-inflammatory cytokines (IFN-γ and IL-6) and chemokines (RANTES and IP-10). Notably, atorvastatin treatment significantly increased the proportion of regulatory T cells (Tregs) in both the peripheral spleen and brain, and at the same time, increased their main effector cytokines IL-10 and TGF-β1. We also found that atorvastatin significantly attenuated total microglia/macrophage activation but augmented the M2/M1 ratio by both inhibiting M1 polarization and enhancing M2 polarization. Conclusions Our data demonstrated that acute atorvastatin administration could modulate post-TBI neuroinflammation effectively, via a mechanism that involves altering peripheral leukocyte invasion and the alternative polarization of microglia/macrophages.
Collapse
|
243
|
Jung Y, Ahn SH, Park SH, Choi YH. Effect of glucose level on chemical hypoxia- and hydrogen peroxide-induced chemokine expression in human glioblastoma cell lines. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2017; 21:509-518. [PMID: 28883755 PMCID: PMC5587601 DOI: 10.4196/kjpp.2017.21.5.509] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 04/30/2017] [Accepted: 06/01/2017] [Indexed: 01/21/2023]
Abstract
Glioblastoma multiforme (GBM) is the most common primary intracranial tumor in adults and has poor prognosis. The GBM-specific tumor microenvironment (TME) plays a crucial role in tumor progression, immune escape, local invasion, and metastasis of GBM. Here, we demonstrate that hypoxia, reactive oxygen species (ROS), and differential concentration of glucose influence the expression of cytokines and chemokines, such as IL-6, IL-8, and IP-10, in human glial cell lines. Treatment with cobalt chloride (CoCl2) and hydrogen peroxide (H2O2) significantly increased the expression levels of IL-6, IL-8, and IP-10 in a dose-dependent manner in CRT-MG and U251-MG astroglioma cells, but not in microglia cells. However, we found strikingly different patterns of expression of cytokines and chemokines between H2O2-treated CRT-MG cells cultured in low- and high-glucose medium. These results suggest that astroglioma and microglia cells exhibit distinct patterns of cytokine and chemokine expression in response to CoCl2 and H2O2 treatment, and different concentrations of glucose influence this expression under either hypoxic or oxidant-enriched conditions.
Collapse
Affiliation(s)
- Yieun Jung
- Department of Physiology, Ewha Womans University School of Medicine, Seoul 07985, Korea.,Tissue Injury Defense Research Center, Ewha Womans University School of Medicine, Seoul 07985, Korea
| | - So-Hee Ahn
- Department of Physiology, Ewha Womans University School of Medicine, Seoul 07985, Korea.,Tissue Injury Defense Research Center, Ewha Womans University School of Medicine, Seoul 07985, Korea
| | - Sang Hui Park
- Department of Pathology, Ewha Womans University School of Medicine, Seoul 07985, Korea
| | - Youn-Hee Choi
- Department of Physiology, Ewha Womans University School of Medicine, Seoul 07985, Korea.,Tissue Injury Defense Research Center, Ewha Womans University School of Medicine, Seoul 07985, Korea
| |
Collapse
|
244
|
Abstract
Traumatic brain injury (TBI) constitutes a heterogeneous condition that affects the most complex organ of the human body. It is commonly classified by its location as focal injury (e.g. epidural hematoma) and diffuse injury (e.g. diffuse axonal shearing injury) as well as by primary and secondary tissue injury. Accordingly, direct mechanical force causes the primary insult. The tissue damage occurring afterwards is subsumed under the term secondary brain damage. Some of these processes are overlapping and include in the early phase local cerebral ischemia resulting in excitotoxicity, which together with the triggered neuroinflammatory cascade causes the formation of cerebral edema and ultimately increased intracranial pressure once the intracranial compliance is exhausted. In survivors the long-term sequelae of the late stage include seizures caused by synaptic reorganization (incidence depending on the severity of TBI), persistent neuroinflammation promoting further neurodegeneration and increased risk for Alzheimer's disease probably because of TBI-related protein misfolding (tauopathy). Acute phase biomarkers of TBI should ideally originate from the injured brain. They should help distinguish disease severity and predict morbidity and mortality; however, the most commonly used biomarkers (S-100β and neurone-specific enolase) show a low specificity. In theory their successors (i. e. GFAP, pNF-H) seem more specific; however, these "new kids on the block" still need to be thoroughly investigated in large scale studies.
Collapse
Affiliation(s)
- D Lahner
- Ludwig Boltzmann Institut für experimentelle und klinische Traumatologie, Donaueschingenstraße 13, 1200, Wien, Österreich
| | - G Fritsch
- Paracelsus Medizinische Universität Salzburg, Strubergasse 21, 5020, Salzburg, Österreich. .,AUVA-Unfallkrankenhaus Lorenz Böhler, Donaueschingenstraße 13, 1200, Wien, Österreich.
| |
Collapse
|
245
|
Truettner JS, Bramlett HM, Dietrich WD. Posttraumatic therapeutic hypothermia alters microglial and macrophage polarization toward a beneficial phenotype. J Cereb Blood Flow Metab 2017; 37:2952-2962. [PMID: 27864465 PMCID: PMC5536802 DOI: 10.1177/0271678x16680003] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Posttraumatic inflammatory processes contribute to pathological and reparative processes observed after traumatic brain injury (TBI). Recent findings have emphasized that these divergent effects result from subsets of proinflammatory (M1) or anti-inflammatory (M2) microglia and macrophages. Therapeutic hypothermia has been tested in preclinical and clinical models of TBI to limit secondary injury mechanisms including proinflammatory processes. This study evaluated the effects of posttraumatic hypothermia (PTH) on phenotype patterns of microglia/macrophages. Sprague-Dawley rats underwent moderate fluid percussion brain injury with normothermia (37℃) or hypothermia (33℃). Cortical and hippocampal regions were analyzed using flow cytometry and reverse transcription-polymerase chain reaction (RT-PCR) at several periods after injury. Compared to normothermia, PTH attenuated infiltrating cortical macrophages positive for CD11b+ and CD45high. At 24 h, the ratio of iNOS+ (M1) to arginase+ (M2) cells after hypothermia showed a decrease compared to normothermia. RT-PCR of M1-associated genes including iNOS and IL-1β was significantly reduced with hypothermia while M2-associated genes including arginase and CD163 were significantly increased compared to normothermic conditions. The injury-induced increased expression of the chemokine Ccl2 was also reduced with PTH. These studies provide a link between temperature-sensitive alterations in macrophage/microglia activation and polarization toward a M2 phenotype that could be permissive for cell survival and repair.
Collapse
Affiliation(s)
- Jessie S Truettner
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, USA
| | - Helen M Bramlett
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, USA
| | - W Dalton Dietrich
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, USA
| |
Collapse
|
246
|
de Castro MRT, Ferreira APDO, Busanello GL, da Silva LRH, da Silveira Junior MEP, Fiorin FDS, Arrifano G, Crespo-López ME, Barcelos RP, Cuevas MJ, Bresciani G, González-Gallego J, Fighera MR, Royes LFF. Previous physical exercise alters the hepatic profile of oxidative-inflammatory status and limits the secondary brain damage induced by severe traumatic brain injury in rats. J Physiol 2017; 595:6023-6044. [PMID: 28726269 DOI: 10.1113/jp273933] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 06/19/2017] [Indexed: 12/20/2022] Open
Abstract
KEY POINTS An early inflammatory response and oxidative stress are implicated in the signal transduction that alters both hepatic redox status and mitochondrial function after traumatic brain injury (TBI). Peripheral oxidative/inflammatory responses contribute to neuronal dysfunction after TBI Exercise training alters the profile of oxidative-inflammatory status in liver and protects against acute hyperglycaemia and a cerebral inflammatory response after TBI. Approaches such as exercise training, which attenuates neuronal damage after TBI, may have therapeutic potential through modulation of responses by metabolic organs. The vulnerability of the body to oxidative/inflammatory in TBI is significantly enhanced in sedentary compared to physically active counterparts. ABSTRACT Although systemic responses have been described after traumatic brain injury (TBI), little is known regarding potential interactions between brain and peripheral organs after neuronal injury. Accordingly, we aimed to investigate whether a peripheral oxidative/inflammatory response contributes to neuronal dysfunction after TBI, as well as the prophylactic role of exercise training. Animals were submitted to fluid percussion injury after 6 weeks of swimming training. Previous exercise training increased mRNA expression of X receptor alpha and ATP-binding cassette transporter, and decreased inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), tumor necrosis factor (TNF)-α and interleukin (IL)-6 expression per se in liver. Interestingly, exercise training protected against hepatic inflammation (COX-2, iNOS, TNF-α and IL-6), oxidative stress (decreases in non-protein sulfhydryl and glutathione, as well as increases in 2',7'-dichlorofluorescein diacetate oxidation and protein carbonyl), which altered hepatic redox status (increases in myeloperoxidase and superoxide dismutase activity, as well as inhibition of catalase activity) mitochondrial function (decreases in methyl-tetrazolium and Δψ, as well as inhibition of citrate synthase activity) and ion gradient homeostasis (inhibition of Na+ ,K+ -ATPase activity inhibition) when analysed 24 h after TBI. Previous exercise training also protected against dysglycaemia, impaired hepatic signalling (increase in phosphorylated c-Jun NH2-terminal kinase, phosphorylated decreases in insulin receptor substrate and phosphorylated AKT expression), high levels of circulating and neuronal cytokines, the opening of the blood-brain barrier, neutrophil infiltration and Na+ ,K+ -ATPase activity inhibition in the ipsilateral cortex after TBI. Moreover, the impairment of protein function, neurobehavioural (neuromotor dysfunction and spatial learning) disability and hippocampal cell damage in sedentary rats suggests that exercise training also modulates peripheral oxidative/inflammatory pathways in TBI, which corroborates the ever increasing evidence regarding health-related outcomes with respect to a physically active lifestyle.
Collapse
Affiliation(s)
- Mauro Robson Torres de Castro
- Programa de Pós-graduação em Educação Física.,Centro de Educação Física e Desportos, Laboratório de Bioquímica do Exercício
| | | | - Guilherme Lago Busanello
- Programa de Pós-graduação em Educação Física.,Centro de Educação Física e Desportos, Laboratório de Bioquímica do Exercício
| | | | | | - Fernando da Silva Fiorin
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Gabriela Arrifano
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas (ICB), Universidade Federal do Pará (UFPA), Belém, Brazil
| | - Maria Elena Crespo-López
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas (ICB), Universidade Federal do Pará (UFPA), Belém, Brazil
| | - Rômulo Pillon Barcelos
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - María J Cuevas
- Institute of Biomedicine (IBIOMED) and Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), University of León, León, Spain
| | - Guilherme Bresciani
- Escuela de Educación Física, Pontificia Universidad Católica de Valparaiso (PUCV), Valparaiso, Chile
| | - Javier González-Gallego
- Institute of Biomedicine (IBIOMED) and Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), University of León, León, Spain
| | - Michele Rechia Fighera
- Programa de Pós-graduação em Educação Física.,Centro de Educação Física e Desportos, Laboratório de Bioquímica do Exercício.,Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Luiz Fernando Freire Royes
- Programa de Pós-graduação em Educação Física.,Centro de Educação Física e Desportos, Laboratório de Bioquímica do Exercício.,Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, Brazil
| |
Collapse
|
247
|
Ziebell JM, Ray-Jones H, Lifshitz J. Nogo presence is inversely associated with shifts in cortical microglial morphology following experimental diffuse brain injury. Neuroscience 2017; 359:209-223. [PMID: 28736137 DOI: 10.1016/j.neuroscience.2017.07.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 06/28/2017] [Accepted: 07/12/2017] [Indexed: 01/08/2023]
Abstract
Diffuse traumatic brain injury (TBI) initiates secondary pathology, including inflammation and reduced myelination. Considering these injury-related pathologies, the many states of activated microglia as demonstrated by differing morphologies would form, migrate, and function in and through fields of growth-inhibitory myelin byproduct, specifically Nogo. Here we evaluate the relationship between inflammation and reduced myelin antigenicity in the wake of diffuse TBI and present the hypothesis that the Nogo-66 receptor antagonist peptide NEP(1-40) would reverse the injury-induced shift in distribution of microglia morphologies by limiting myelin-based inhibition. Adult male rats were subjected to midline fluid percussion sham or brain injury. At 2h, 6h, 1d, 2d, 7d, and 21d post-injury, immunohistochemical staining was analyzed in sensory cortex (S1BF) for myelin antigens (myelin basic protein; MBP and CNPase), microglia morphology (ionized calcium-binding adapter protein; Iba1), Nogo receptor and Nogo. Pronounced reduction in myelin antigenicity was evident transiently at 1d post-injury, as evidenced by decreased MBP and CNPase staining, as well as loss of white matter organization, compared to sham and later injury time points. Concomitant with reduced myelin antigenicity, injury shifted microglia morphology from the predominantly ramified morphology observed in sham-injured cortex to hyper-ramified, activated, fully activated, or rod. Changes in microglial morphology were evident as early as 2h post-injury, and remained at least until day 21. Additional cohorts of uninjured and brain-injured animals received vehicle or drug (NEP(1-40), i.p., 15min and 19h post-injury) and brains were collected at 2h, 6h, 1d, 2d, or 7d post-injury. NEP(1-40) administration further shifted distributions of microglia away from an injury-induced activated morphology toward greater proportions of rod and macrophage-like morphologies compared to vehicle-treated. By 7d post-injury, no differences in the distributions of microglia were noted between vehicle and NEP(1-40). This study begins to link secondary pathologies of white matter damage and inflammation after diffuse TBI. In the injured brain, secondary pathologies co-occur and likely interact, with consequences for neuronal circuit disruption leading to neurological symptoms.
Collapse
Affiliation(s)
- Jenna M Ziebell
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA; Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA.
| | - Helen Ray-Jones
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA; Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA; Department of Biology and Biochemistry, University of Bath, Bath, England, UK
| | - Jonathan Lifshitz
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA; Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA; VA Healthcare System, Phoenix, AZ, USA; Psychology, Arizona State University, Tempe, AZ, USA
| |
Collapse
|
248
|
Weil ZM, Karelina K. Traumatic Brain Injuries during Development: Implications for Alcohol Abuse. Front Behav Neurosci 2017; 11:135. [PMID: 28775682 PMCID: PMC5517445 DOI: 10.3389/fnbeh.2017.00135] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 07/07/2017] [Indexed: 11/13/2022] Open
Abstract
Traumatic brain injuries are strongly related to alcohol intoxication as by some estimates half or more of all brain injuries involve at least one intoxicated individual. Additionally, there is mounting evidence that traumatic brain injuries can themselves serve as independent risk factors for the development of alcohol use disorders, particularly when injury occurs during juvenile or adolescent development. Here, we will review the epidemiological and experimental evidence for this phenomenon and discuss potential psychosocial mediators including attenuation of negative affect and impaired decision making as well as neurochemical mediators including disruption in the glutamatergic, GABAergic, and dopaminergic signaling pathways and increases in inflammation.
Collapse
Affiliation(s)
- Zachary M Weil
- Behavioral Neuroendocrinology Group, Department of Neuroscience, Center for Brain and Spinal Cord Repair, Ohio State University Wexner Medical CenterColumbus, OH, United States
| | - Kate Karelina
- Behavioral Neuroendocrinology Group, Department of Neuroscience, Center for Brain and Spinal Cord Repair, Ohio State University Wexner Medical CenterColumbus, OH, United States
| |
Collapse
|
249
|
de Lemos L, Junyent F, Camins A, Castro-Torres RD, Folch J, Olloquequi J, Beas-Zarate C, Verdaguer E, Auladell C. Neuroprotective Effects of the Absence of JNK1 or JNK3 Isoforms on Kainic Acid-Induced Temporal Lobe Epilepsy-Like Symptoms. Mol Neurobiol 2017; 55:4437-4452. [PMID: 28664455 DOI: 10.1007/s12035-017-0669-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 06/20/2017] [Indexed: 12/16/2022]
Abstract
The activation of c-Jun-N-terminal kinases (JNK) pathway has been largely associated with the pathogenesis and the neuronal death that occur in neurodegenerative diseases. Altogether, this justifies why JNKs have become a focus of screens for new therapeutic strategies. The aim of the present study was to identify the role of the different JNK isoforms (JNK1, JNK2, and JNK3) in apoptosis and inflammation after induction of brain damage. To address this aim, we induced excitotoxicity in wild-type and JNK knockout mice (jnk1 -/- , jnk2 -/- , and jnk3 -/- ) via an intraperitoneal injection of kainic acid, an agonist of glutamic-kainate-receptors, that induce status epilepticus.Each group of animals was divided into two treatments: a single intraperitoneal dose of saline solution, used as a control, and a single intraperitoneal dose (30 mg/kg) of kainic acid. Our results reported a significant decrease in neuronal degeneration in the hippocampus of jnk1 -/- and jnk3 -/- mice after kainic acid treatment, together with reduced or unaltered expression of several apoptotic genes compared to WT treated mice. In addition, both jnk1 -/- and jnk3 -/- mice exhibited a reduction in glial reactivity, as shown by the lower expression of inflammatory genes and a reduction of JNK phosphorylation. In addition, in jnk3 -/- mice, the c-Jun phosphorylation was also diminished.Collectively, these findings provide compelling evidence that the absence of JNK1 or JNK3 isoforms confers neuroprotection against neuronal damage induced by KA and evidence, for the first time, the implication of JNK1 in excitotoxicity. Accordingly, JNK1 and/or JNK3 are promising targets for the prevention of cell death and inflammation during epileptogenesis.
Collapse
Affiliation(s)
- Luisa de Lemos
- Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia, Universitat de Barcelona, Avda Diagonal 641, E-08028, Barcelona, Spain.,Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, 2780-156, Oeiras, Portugal
| | - Felix Junyent
- Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia, Universitat de Barcelona, Avda Diagonal 641, E-08028, Barcelona, Spain
| | - Antoni Camins
- Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia, Universitat de Barcelona, Avda Diagonal 641, E-08028, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Neuroscience Institute, University of Barcelona, Barcelona, Spain
| | - Rubén Darío Castro-Torres
- Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia, Universitat de Barcelona, Avda Diagonal 641, E-08028, Barcelona, Spain.,Laboratorio de Regeneración Neural, Departamento de Biología Celular y Molecular, CUCBA, Universidad de Guadalajara, Guadalajara, Mexico
| | - Jaume Folch
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Unitat de Bioquímica, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Tarragona, Spain
| | - Jordi Olloquequi
- Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Talca, Chile
| | - Carlos Beas-Zarate
- Laboratorio de Regeneración Neural, Departamento de Biología Celular y Molecular, CUCBA, Universidad de Guadalajara, Guadalajara, Mexico
| | - Ester Verdaguer
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Neuroscience Institute, University of Barcelona, Barcelona, Spain.,Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Carme Auladell
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain. .,Neuroscience Institute, University of Barcelona, Barcelona, Spain. .,Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
250
|
Donat CK, Scott G, Gentleman SM, Sastre M. Microglial Activation in Traumatic Brain Injury. Front Aging Neurosci 2017; 9:208. [PMID: 28701948 PMCID: PMC5487478 DOI: 10.3389/fnagi.2017.00208] [Citation(s) in RCA: 282] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 06/12/2017] [Indexed: 12/15/2022] Open
Abstract
Microglia have a variety of functions in the brain, including synaptic pruning, CNS repair and mediating the immune response against peripheral infection. Microglia rapidly become activated in response to CNS damage. Depending on the nature of the stimulus, microglia can take a number of activation states, which correspond to altered microglia morphology, gene expression and function. It has been reported that early microglia activation following traumatic brain injury (TBI) may contribute to the restoration of homeostasis in the brain. On the other hand, if they remain chronically activated, such cells display a classically activated phenotype, releasing pro-inflammatory molecules, resulting in further tissue damage and contributing potentially to neurodegeneration. However, new evidence suggests that this classification is over-simplistic and the balance of activation states can vary at different points. In this article, we review the role of microglia in TBI, analyzing their distribution, morphology and functional phenotype over time in animal models and in humans. Animal studies have allowed genetic and pharmacological manipulations of microglia activation, in order to define their role. In addition, we describe investigations on the in vivo imaging of microglia using translocator protein (TSPO) PET and autoradiography, showing that microglial activation can occur in regions far remote from sites of focal injuries, in humans and animal models of TBI. Finally, we outline some novel potential therapeutic approaches that prime microglia/macrophages toward the beneficial restorative microglial phenotype after TBI.
Collapse
Affiliation(s)
| | | | | | - Magdalena Sastre
- Division of Brain Sciences, Department of Medicine, Imperial College LondonLondon, United Kingdom
| |
Collapse
|