1
|
Awan N, Weppner J, Kumar RG, Juengst SB, Dams-O'Connor K, Sevigny M, Zafonte RD, Walker WC, Szaflarski JP, Wagner AK. Impact of Post-Traumatic Epilepsy on Mental Health and Multidimensional Outcome and Quality of Life: An NIDILRR TBIMS Study. J Neurotrauma 2025. [PMID: 39761035 DOI: 10.1089/neu.2024.0117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025] Open
Abstract
Traumatic brain injury (TBI) and subsequent post-traumatic epilepsy (PTE) often impair daily activities and mental health (MH), which contribute to long-term TBI-related disability. PTE also affects driving capacity, which impacts functional independence, community participation, and satisfaction with life (SWL). However, studies evaluating the collective impact of PTE on multidimensional outcomes are lacking. Thus, we generated a model to investigate how PTE after moderate-to-severe (ms)TBI affects TBI-associated impairments, limits activities and participation, and influences SWL. Of 5108 participants with msTBI enrolled into the National Institute for Disability, Independent Living, and Rehabilitation Research TBI Model Systems between 2010 and 2018 and with seizure-event data available at year-1 post-TBI, 1214 had complete outcome data and 1003 had complete covariate data used for analysis. We constructed a conceptual framework illustrating hypothesized interrelationships between year-1 PTE, driving status, functional independence measure (FIM), depression and anxiety, as well as year-2 participation, and SWL. We performed univariate and multivariable linear and logistic regressions. A covariate-adjusted structural equation model (SEM), using the lavaan package (R), assessed the conceptual framework's suitability in establishing PTE links with outcomes 1-2 years post-injury. Multiple parameters were evaluated to assess SEM fit. Year-1 PTE was correlated with year-1 FIM motor (standardized coefficient, βstd = -0.112, p = 0.007) and showed a trend level association with year-1 FIM cognition (βstd = -0.070, p = 0.079). Individuals with year-1 PTE were less likely to drive independently at year 1 (βstd = -0.148, p < 0.001). In addition, FIM motor (βstd = 0.323, p < 0.001), FIM cognition (βstd = 0.181, p = 0.012), and anxiety (βstd = -0.135, p = 0.024) influenced driving status. FIM cognition was associated with year-1 depression (βstd = 0.386, p < 0.001) and year-1 anxiety (βstd = 0.396, p < 0.001), whereas year-1 FIM motor (βstd = 0.186, p = 0.003), depression (βstd = -0.322, p = 0.011), and driving status (βstd = 0.233, p < 0.001) directly affected year-2 objective life participation metrics. Moreover, year-1 depression (βstd = -0.382, p = 0.001) and year-2 participation (βstd = 0.160, p < 0.001) had direct effects on year-2 SWL. SWL was influenced indirectly by year-1 variables, including functional impairment, anxiety, and driving status-factors that impacted year-2 participation directly or indirectly, and consequently year-2 SWL, forming a complex relationship with year-1 PTE. A sensitivity analysis SEM showed that the number of MH disorders was associated with participation and SWL (p < 0.001), and this combined MH variable was directly related to driving status (p < 0.02). Developing PTE during year-1 after msTBI affects multiple aspects of life. PTE effects extend to motor and cognitive abilities, driving capabilities, and indirectly, to life participation and overall SWL. The implications underscore the crucial need for effective PTE management strategies during the first year post-TBI to minimize the adverse impact on factors influencing multidimensional year-2 participation and SWL outcomes. Addressing transportation barriers is warranted to enhance the well-being of those with PTE and msTBI, emphasizing a holistic approach. Further research is recommended for SEM validation studies, including testing causal inference pathways that might inform future prevention and treatment trials.
Collapse
Affiliation(s)
- Nabil Awan
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Statistics, School of Computer, Data & Information Sciences, University of Wisconsin - Madison, Madison, Wisconsin, USA
| | - Justin Weppner
- Department of Internal Medicine, Virginia Tech Carilion School of Medicine, Roanoke, Virginia, USA
- Department of Internal Medicine, Edward Via College of Osteopathic Medicine, Blacksburg, Virginia, USA
| | - Raj G Kumar
- Department of Rehabilitation and Human Performance, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Shannon B Juengst
- Texas Institute for Rehabilitation Research, University of Texas at Houston, Houston, Texas, USA
- Departments of Physical Medicine and Rehabilitation, University of Texas Southwestern, Dallas, Texas, USA
| | - Kristen Dams-O'Connor
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | - Ross D Zafonte
- Massachusetts General Hospital, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - William C Walker
- Department of Physical Medicine & Rehabilitation, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Jerzy P Szaflarski
- Department of Neurology, University of Alabama at Birmingham Epilepsy Center, University of Alabama, Birmingham, Alabama, USA
| | - Amy K Wagner
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Center for Neuroscience at University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
2
|
León-Rodríguez A, Grondona JM, Marín-Wong S, López-Aranda MF, López-Ávalos MD. Long-term reprogramming of primed microglia after moderate inhibition of CSF1R signaling. Glia 2025; 73:175-195. [PMID: 39448548 DOI: 10.1002/glia.24627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 10/01/2024] [Accepted: 10/07/2024] [Indexed: 10/26/2024]
Abstract
In acute neuroinflammation, microglia activate transiently, and return to a resting state later on. However, they may retain immune memory of such event, namely priming. Primed microglia are more sensitive to new stimuli and develop exacerbated responses, representing a risk factor for neurological disorders with an inflammatory component. Strategies to control the hyperactivation of microglia are, hence, of great interest. The receptor for colony stimulating factor 1 (CSF1R), expressed in myeloid cells, is essential for microglia viability, so its blockade with specific inhibitors (e.g. PLX5622) results in significant depletion of microglial population. Interestingly, upon inhibitor withdrawal, new naïve microglia repopulate the brain. Depletion-repopulation has been proposed as a strategy to reprogram microglia. However, substantial elimination of microglia is inadvisable in human therapy. To overcome such drawback, we aimed to reprogram long-term primed microglia by CSF1R partial inhibition. Microglial priming was induced in mice by acute neuroinflammation, provoked by intracerebroventricular injection of neuraminidase. After 3-weeks recovery, low-dose PLX5622 treatment was administrated for 12 days, followed by a withdrawal period of 7 weeks. Twelve hours before euthanasia, mice received a peripheral lipopolysaccharide (LPS) immune challenge, and the subsequent microglial inflammatory response was evaluated. PLX5622 provoked a 40%-50% decrease in microglial population, but basal levels were restored 7 weeks later. In the brain regions studied, hippocampus and hypothalamus, LPS induced enhanced microgliosis and inflammatory activation in neuraminidase-injected mice, while PLX5622 treatment prevented these changes. Our results suggest that PLX5622 used at low doses reverts microglial priming and, remarkably, prevents broad microglial depletion.
Collapse
Affiliation(s)
- Ana León-Rodríguez
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga-IBIMA Plataforma Bionand, Málaga, Spain
| | - Jesús M Grondona
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga-IBIMA Plataforma Bionand, Málaga, Spain
| | - Sonia Marín-Wong
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
| | - Manuel F López-Aranda
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga-IBIMA Plataforma Bionand, Málaga, Spain
| | - María D López-Ávalos
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga-IBIMA Plataforma Bionand, Málaga, Spain
| |
Collapse
|
3
|
Zhang Z, Xiao T, Hall MR, Crodian JS, Alford AK, Kimbrough A, Shi R. Temporal differential effects of post-injury alcohol consumption in a mouse model of blast-induced traumatic brain injury. Neuroscience 2024; 562:239-251. [PMID: 39369945 PMCID: PMC11769080 DOI: 10.1016/j.neuroscience.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/22/2024] [Accepted: 10/01/2024] [Indexed: 10/08/2024]
Abstract
Traumatic brain injury is a prevalent condition that affects millions worldwide with no clear understanding or effective therapeutic management available. Military soldiers have a high risk of exposure to blast-induced traumatic brain injury (bTBI). Furthermore, alcohol drinking is common in this population, and studies have shown that post-TBI alcohol exposure can result in memory loss. Hence, it is possible that alcohol could contribute to the overall pathological outcome of brain trauma. However, such a possibility has not been explored in detail. Here, we combined a mild bTBI (mbTBI) model with the drinking-in-the-dark (DID) paradigm to investigate the pathological synergy between mbTBI and alcohol consumption by examining brain oxidative stress levels and behavioral alterations in mice. The results revealed the anxiolytic and short-term memory improvement effects of post-trauma alcohol drinking examined at an early timepoint post mbTBI. However, extended alcohol drinking for up to three weeks post mbTBI impaired long-term memory and was accompanied by intensified oxidative stress in brain regions associated with memory and anxiety. These findings, as well as those from previous in vitro TBI/alcohol studies, suggest a pathological synergy of physical force and post-impact alcohol exposure. This knowledge could potentially aid in establishing guidelines for TBI victims to avoid further injury to their brains as well as to help maximize their recovery following TBI.
Collapse
Affiliation(s)
- Zaiyang Zhang
- Department of Basic Medical Sciences, College of Veterinary Medicine Purdue University, West Lafayette, IN, United States; Center for Paralysis Research, Purdue University, West Lafayette, IN, United States
| | - Tiange Xiao
- Department of Basic Medical Sciences, College of Veterinary Medicine Purdue University, West Lafayette, IN, United States
| | - Mekyna R Hall
- Department of Basic Medical Sciences, College of Veterinary Medicine Purdue University, West Lafayette, IN, United States; Center for Paralysis Research, Purdue University, West Lafayette, IN, United States
| | - Jennifer S Crodian
- Department of Basic Medical Sciences, College of Veterinary Medicine Purdue University, West Lafayette, IN, United States; Center for Paralysis Research, Purdue University, West Lafayette, IN, United States
| | - Anna K Alford
- Department of Basic Medical Sciences, College of Veterinary Medicine Purdue University, West Lafayette, IN, United States; Center for Paralysis Research, Purdue University, West Lafayette, IN, United States; Weldon School of Biomedical Engineering, College of Engineering, Purdue University, West Lafayette, IN, United States
| | - Adam Kimbrough
- Department of Basic Medical Sciences, College of Veterinary Medicine Purdue University, West Lafayette, IN, United States; Weldon School of Biomedical Engineering, College of Engineering, Purdue University, West Lafayette, IN, United States; The Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN, United States.
| | - Riyi Shi
- Department of Basic Medical Sciences, College of Veterinary Medicine Purdue University, West Lafayette, IN, United States; Center for Paralysis Research, Purdue University, West Lafayette, IN, United States; Weldon School of Biomedical Engineering, College of Engineering, Purdue University, West Lafayette, IN, United States; Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, United States.
| |
Collapse
|
4
|
Wehn AC, Khalin I, Hu S, Harapan BN, Mao X, Cheng S, Plesnila N, Terpolilli NA. Bradykinin 2 Receptors Mediate Long-Term Neurocognitive Deficits After Experimental Traumatic Brain Injury. J Neurotrauma 2024; 41:2442-2454. [PMID: 38818807 DOI: 10.1089/neu.2024.0042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2024] Open
Abstract
The kallikrein-kinin system is one of the first inflammatory pathways to be activated following traumatic brain injury (TBI) and has been shown to exacerbate brain edema formation in the acute phase through activation of bradykinin 2 receptors (B2R). However, the influence of B2R on chronic post-traumatic damage and outcome is unclear. In the current study, we assessed long-term effects of B2R-knockout (KO) after experimental TBI. B2R KO mice (heterozygous, homozygous) and wild-type (WT) littermates (n = 10/group) were subjected to controlled cortical impact (CCI) TBI. Lesion size was evaluated by magnetic resonance imaging up to 90 days after CCI. Motor and memory function were regularly assessed by Neurological Severity Score, Beam Walk, and Barnes maze test. Ninety days after TBI, brains were harvested for immunohistochemical analysis. There was no difference in cortical lesion size between B2R-deficient and WT animals 3 months after injury; however, hippocampal damage was reduced in B2R KO mice (p = 0.03). Protection of hippocampal tissue was accompanied by a significant improvement of learning and memory function 3 months after TBI (p = 0.02 WT vs. KO), whereas motor function was not influenced. Scar formation and astrogliosis were unaffected, but B2R deficiency led to a gene-dose-dependent attenuation of microglial activation and a reduction of CD45+ cells 3 months after TBI in cortex (p = 0.0003) and hippocampus (p < 0.0001). These results suggest that chronic hippocampal neurodegeneration and subsequent cognitive impairment are mediated by prolonged neuroinflammation and B2R. Inhibition of B2R may therefore represent a novel strategy to reduce long-term neurocognitive deficits after TBI.
Collapse
Affiliation(s)
- Antonia Clarissa Wehn
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Neurosurgery, LMU University Hospital, LMU Munich, Munich, Germany
| | - Igor Khalin
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Institute Blood and Brain @ Caen-Normandie (BB@C), Normandie University, Rouen, France
| | - Senbin Hu
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Biyan Nathanael Harapan
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
- Department of Neurosurgery, LMU University Hospital, LMU Munich, Munich, Germany
| | - Xiang Mao
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Neurotrauma Laboratory, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Shiqi Cheng
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Neurosurgery, The Second affiliated Hospital of Nanchang University, Nanchang, China
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Nicole A Terpolilli
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Neurosurgery, LMU University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
5
|
Zemke NR, Lee S, Mamde S, Yang B, Berchtold N, Maximiliano Garduño B, Indralingam HS, Bartosik WM, Lau PK, Dong K, Yang A, Tani Y, Chen C, Zeng Q, Ajith V, Tong L, Seng C, Li D, Wang T, Xu X, Ren B. Epigenetic and 3D genome reprogramming during the aging of human hippocampus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.14.618338. [PMID: 39463924 PMCID: PMC11507755 DOI: 10.1101/2024.10.14.618338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Age-related cognitive decline is associated with altered physiology of the hippocampus. While changes in gene expression have been observed in aging brain, the regulatory mechanisms underlying these changes remain underexplored. We generated single-nucleus gene expression, chromatin accessibility, DNA methylation, and 3D genome data from 40 human hippocampal tissues spanning adult lifespan. We observed a striking loss of astrocytes, OPC, and endothelial cells during aging, including astrocytes that play a role in regulating synapses. Microglia undergo a dramatic switch from a homeostatic state to a primed inflammatory state through DNA methylome and 3D genome reprogramming. Aged cells experience erosion of their 3D genome architecture. Our study identifies age-associated changes in cell types/states and gene regulatory features that provide insight into cognitive decline during human aging.
Collapse
Affiliation(s)
- Nathan R. Zemke
- Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine; La Jolla, CA, USA
- Center for Epigenomics, University of California, San Diego School of Medicine; La Jolla, CA, USA
| | - Seoyeon Lee
- Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine; La Jolla, CA, USA
| | - Sainath Mamde
- Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine; La Jolla, CA, USA
| | - Bing Yang
- Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine; La Jolla, CA, USA
- Center for Epigenomics, University of California, San Diego School of Medicine; La Jolla, CA, USA
| | - Nicole Berchtold
- Department of Anatomy and Neurobiology, University of California, Irvine School of Medicine; Irvine, CA, USA
- Immunis Inc, 18301 Von Karman Ave; Irvine, CA, USA
| | - B. Maximiliano Garduño
- Department of Anatomy and Neurobiology, University of California, Irvine School of Medicine; Irvine, CA, USA
| | - Hannah S. Indralingam
- Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine; La Jolla, CA, USA
- Center for Epigenomics, University of California, San Diego School of Medicine; La Jolla, CA, USA
| | - Weronika M. Bartosik
- Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine; La Jolla, CA, USA
- Center for Epigenomics, University of California, San Diego School of Medicine; La Jolla, CA, USA
| | - Pik Ki Lau
- Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine; La Jolla, CA, USA
- Center for Epigenomics, University of California, San Diego School of Medicine; La Jolla, CA, USA
| | - Keyi Dong
- Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine; La Jolla, CA, USA
- Center for Epigenomics, University of California, San Diego School of Medicine; La Jolla, CA, USA
| | - Amanda Yang
- Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine; La Jolla, CA, USA
| | - Yasmine Tani
- Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine; La Jolla, CA, USA
| | - Chumo Chen
- Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine; La Jolla, CA, USA
| | - Qiurui Zeng
- Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine; La Jolla, CA, USA
| | - Varun Ajith
- Department of Anatomy and Neurobiology, University of California, Irvine School of Medicine; Irvine, CA, USA
| | - Liqi Tong
- Department of Anatomy and Neurobiology, University of California, Irvine School of Medicine; Irvine, CA, USA
| | - Chanrung Seng
- Department of Genetics, The Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine; St. Louis, MO, USA
| | - Daofeng Li
- Department of Genetics, The Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine; St. Louis, MO, USA
| | - Ting Wang
- Department of Genetics, The Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine; St. Louis, MO, USA
- McDonnell Genome Institute, Washington University School of Medicine; St. Louis, MO, USA
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, University of California, Irvine School of Medicine; Irvine, CA, USA
- The Center for Neural Circuit Mapping, University of California; Irvine, CA, USA
| | - Bing Ren
- Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine; La Jolla, CA, USA
- Center for Epigenomics, University of California, San Diego School of Medicine; La Jolla, CA, USA
| |
Collapse
|
6
|
Wu J, Ren R, Chen T, Su LD, Tang T. Neuroimmune and neuroinflammation response for traumatic brain injury. Brain Res Bull 2024; 217:111066. [PMID: 39241894 DOI: 10.1016/j.brainresbull.2024.111066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 08/18/2024] [Accepted: 09/02/2024] [Indexed: 09/09/2024]
Abstract
Traumatic brain injury (TBI) is one of the major diseases leading to mortality and disability, causing a serious disease burden on individuals' ordinary lives as well as socioeconomics. In primary injury, neuroimmune and neuroinflammation are both responsible for the TBI. Besides, extensive and sustained injury induced by neuroimmune and neuroinflammation also prolongs the course and worsens prognosis of TBI. Therefore, this review aims to explore the role of neuroimmune, neuroinflammation and factors associated them in TBI as well as the therapies for TBI. Thus, we conducted by searching PubMed, Scopus, and Web of Science databases for articles published between 2010 and 2023. Keywords included "traumatic brain injury," "neuroimmune response," "neuroinflammation," "astrocytes," "microglia," and "NLRP3." Articles were selected based on relevance and quality of evidence. On this basis, we provide the cellular and molecular mechanisms of TBI-induced both neuroimmune and neuroinflammation response, as well as the different factors affecting them, are introduced based on physiology of TBI, which supply a clear overview in TBI-induced chain-reacting, for a better understanding of TBI and to offer more thoughts on the future therapies for TBI.
Collapse
Affiliation(s)
- Junyun Wu
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang 310009, China
| | - Reng Ren
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang 310009, China
| | - Tao Chen
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang 310009, China
| | - Li-Da Su
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang 310009, China.
| | - Tianchi Tang
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang 310009, China.
| |
Collapse
|
7
|
Adam CD, Mirzakhalili E, Gagnon KG, Cottone C, Arena JD, Ulyanova AV, Johnson VE, Wolf JA. Disrupted Hippocampal Theta-Gamma Coupling and Spike-Field Coherence Following Experimental Traumatic Brain Injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.30.596704. [PMID: 39314320 PMCID: PMC11418945 DOI: 10.1101/2024.05.30.596704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Traumatic brain injury (TBI) often results in persistent learning and memory deficits, likely due to disrupted hippocampal circuitry underlying these processes. Precise temporal control of hippocampal neuronal activity is important for memory encoding and retrieval and is supported by oscillations that dynamically organize single unit firing. Using high-density laminar electrophysiology, we discovered a loss of oscillatory power across CA1 lamina, with a profound, layer-specific reduction in theta-gamma phase amplitude coupling in injured rats. Interneurons from injured animals were less strongly entrained to theta and gamma oscillations, suggesting a mechanism for the loss of coupling, while pyramidal cells were entrained to a later phase of theta. During quiet immobility, we report decreased ripple amplitudes from injured animals during sharp-wave ripple events. These results reveal deficits in information encoding and retrieval schemes essential to cognition that likely underlie TBI-associated learning and memory impairments, and elucidate potential targets for future neuromodulation therapies.
Collapse
Affiliation(s)
- Christopher D Adam
- Center for Brain Injury and Repair, Department of Neurosurgery, University of Pennsylvania, Philadelphia, USA
| | - Ehsan Mirzakhalili
- Center for Brain Injury and Repair, Department of Neurosurgery, University of Pennsylvania, Philadelphia, USA
| | - Kimberly G Gagnon
- Center for Brain Injury and Repair, Department of Neurosurgery, University of Pennsylvania, Philadelphia, USA
| | - Carlo Cottone
- Center for Brain Injury and Repair, Department of Neurosurgery, University of Pennsylvania, Philadelphia, USA
| | - John D Arena
- Center for Brain Injury and Repair, Department of Neurosurgery, University of Pennsylvania, Philadelphia, USA
| | - Alexandra V Ulyanova
- Center for Brain Injury and Repair, Department of Neurosurgery, University of Pennsylvania, Philadelphia, USA
- Center for Neurotrauma, Neurodegeneration, and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, USA
| | - Victoria E Johnson
- Center for Brain Injury and Repair, Department of Neurosurgery, University of Pennsylvania, Philadelphia, USA
| | - John A Wolf
- Center for Brain Injury and Repair, Department of Neurosurgery, University of Pennsylvania, Philadelphia, USA
- Center for Neurotrauma, Neurodegeneration, and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, USA
| |
Collapse
|
8
|
Hasanpour-Segherlou Z, Masheghati F, Shakeri-Darzehkanani M, Hosseini-Siyanaki MR, Lucke-Wold B. Neurodegenerative Disorders in the Context of Vascular Changes after Traumatic Brain Injury. JOURNAL OF VASCULAR DISEASES 2024; 3:319-332. [DOI: 10.3390/jvd3030025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2025]
Abstract
Traumatic brain injury (TBI) results from external biomechanical forces that cause structural and physiological disturbances in the brain, leading to neuronal, axonal, and vascular damage. TBIs are predominantly mild (65%), with moderate (10%) and severe (25%) cases also prevalent. TBI significantly impacts health, increasing the risk of neurodegenerative diseases such as dementia, post injury. The initial phase of TBI involves acute disruption of the blood–brain barrier (BBB) due to vascular shear stress, leading to ischemic damage and amyloid-beta accumulation. Among the acute cerebrovascular changes after trauma are early progressive hemorrhage, micro bleeding, coagulopathy, neurovascular unit (NVU) uncoupling, changes in the BBB, changes in cerebral blood flow (CBF), and cerebral edema. The secondary phase is characterized by metabolic dysregulation and inflammation, mediated by oxidative stress and reactive oxygen species (ROS), which contribute to further neurodegeneration. The cerebrovascular changes and neuroinflammation include excitotoxicity from elevated extracellular glutamate levels, coagulopathy, NVU, immune responses, and chronic vascular changes after TBI result in neurodegeneration. Severe TBI often leads to dysfunction in organs outside the brain, which can significantly impact patient care and outcomes. The vascular component of systemic inflammation after TBI includes immune dysregulation, hemodynamic dysfunction, coagulopathy, respiratory failure, and acute kidney injury. There are differences in how men and women acquire traumatic brain injuries, how their brains respond to these injuries at the cellular and molecular levels, and in their brain repair and recovery processes. Also, the patterns of cerebrovascular dysfunction and stroke vulnerability after TBI are different in males and females based on animal studies.
Collapse
Affiliation(s)
| | | | | | | | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
9
|
Wang D, Li X, Li W, Duong T, Wang H, Kleschevnikova N, Patel HH, Breen E, Powell S, Wang S, Head BP. Nicotine inhalant via E-cigarette facilitates sensorimotor function recovery by upregulating neuronal BDNF-TrkB signalling in traumatic brain injury. Br J Pharmacol 2024; 181:3082-3097. [PMID: 38698493 DOI: 10.1111/bph.16395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 02/21/2024] [Accepted: 03/11/2024] [Indexed: 05/05/2024] Open
Abstract
BACKGROUND AND PURPOSE Traumatic brain injury (TBI) causes lifelong physical and psychological dysfunction in affected individuals. The current study investigated the effects of chronic nicotine exposure via E-cigarettes (E-cig) (vaping) on TBI-associated behavioural and biochemical changes. EXPERIMENTAL APPROACH Adult C57/BL6J male mice were subjected to controlled cortical impact (CCI) followed by daily exposure to E-cig vapour for 6 weeks. Sensorimotor functions, locomotion, and sociability were subsequently evaluated by nesting, open field, and social approach tests, respectively. Immunoblots were conducted to examine the expression of mature brain-derived neurotrophic factor (mBDNF) and associated downstream proteins (p-Erk, p-Akt). Histological analyses were performed to evaluate neuronal survival and neuroinflammation. KEY RESULTS Post-injury chronic nicotine exposure significantly improved nesting performance in CCI mice. Histological analysis revealed increased survival of cortical neurons in the perilesion cortex with chronic nicotine exposure. Immunoblots revealed that chronic nicotine exposure significantly up-regulated mBDNF, p-Erk and p-Akt expression in the perilesion cortex of CCI mice. Immunofluorescence microscopy indicated that elevated mBDNF and p-Akt expression were mainly localized within cortical neurons. Immunolabelling of Iba1 demonstrated that chronic nicotine exposure attenuated microglia-mediated neuroinflammation. CONCLUSIONS AND IMPLICATIONS Post-injury chronic nicotine exposure via vaping facilitates recovery of sensorimotor function by upregulating neuroprotective mBDNF/TrkB/Akt/Erk signalling. These findings suggest potential neuroprotective properties of nicotine despite its highly addictive nature. Thus, understanding the multifaceted effects of chronic nicotine exposure on TBI-associated symptoms is crucial for paving the way for informed and properly managed therapeutic interventions.
Collapse
Affiliation(s)
- Dongsheng Wang
- Department of Anesthesiology, VA San Diego Healthcare System, San Diego, California, USA
- Department of Anesthesiology, University of California San Diego, La Jolla, California, USA
| | - Xiaojing Li
- Department of Anesthesiology, VA San Diego Healthcare System, San Diego, California, USA
- Department of Anesthesiology, University of California San Diego, La Jolla, California, USA
| | - Wenxi Li
- Department of Anesthesiology, VA San Diego Healthcare System, San Diego, California, USA
- Department of Anesthesiology, University of California San Diego, La Jolla, California, USA
| | - Tiffany Duong
- Department of Anesthesiology, VA San Diego Healthcare System, San Diego, California, USA
- Department of Anesthesiology, University of California San Diego, La Jolla, California, USA
| | - Hongxia Wang
- Department of Anesthesiology, VA San Diego Healthcare System, San Diego, California, USA
- Department of Anesthesiology, University of California San Diego, La Jolla, California, USA
| | - Natalia Kleschevnikova
- Department of Anesthesiology, VA San Diego Healthcare System, San Diego, California, USA
- Department of Anesthesiology, University of California San Diego, La Jolla, California, USA
| | - Hemal H Patel
- Department of Anesthesiology, VA San Diego Healthcare System, San Diego, California, USA
- Department of Anesthesiology, University of California San Diego, La Jolla, California, USA
| | - Ellen Breen
- Department of Medicine, Division of Pulmonary Critical Care and Sleep Medicine, University of California San Diego, La Jolla, California, USA
| | - Susan Powell
- Research Service and Desert Pacific Mental Illness Research, Education & Clinical Center, Veterans Affairs San Diego Health System, San Diego, California, USA
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA
| | - Shanshan Wang
- Department of Anesthesiology, VA San Diego Healthcare System, San Diego, California, USA
- Department of Anesthesiology, University of California San Diego, La Jolla, California, USA
| | - Brian P Head
- Department of Anesthesiology, VA San Diego Healthcare System, San Diego, California, USA
- Department of Anesthesiology, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
10
|
Houle S, Tapp Z, Dobres S, Ahsan S, Reyes Y, Cotter C, Mitsch J, Zimomra Z, Peng J, Rowe RK, Lifshitz J, Sheridan J, Godbout J, Kokiko-Cochran ON. Sleep fragmentation after traumatic brain injury impairs behavior and conveys long-lasting impacts on neuroinflammation. Brain Behav Immun Health 2024; 38:100797. [PMID: 38803369 PMCID: PMC11128763 DOI: 10.1016/j.bbih.2024.100797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 05/12/2024] [Indexed: 05/29/2024] Open
Abstract
Traumatic brain injury (TBI) causes a prolonged inflammatory response in the central nervous system (CNS) driven by microglia. Microglial reactivity is exacerbated by stress, which often provokes sleep disturbances. We have previously shown that sleep fragmentation (SF) stress after experimental TBI increases microglial reactivity and impairs hippocampal function 30 days post-injury (DPI). The neuroimmune response is highly dynamic the first few weeks after TBI, which is also when injury induced sleep-wake deficits are detected. Therefore, we hypothesized that even a few weeks of TBI SF stress would synergize with injury induced sleep-wake deficits to promote neuroinflammation and impair outcome. Here, we investigated the effects of environmental SF in a lateral fluid percussion model of mouse TBI. Half of the mice were undisturbed, and half were exposed to 5 h of SF around the onset of the light cycle, daily, for 14 days. All mice were then undisturbed 15-30 DPI, providing a period for SF stress recovery (SF-R). Mice exposed to SF stress slept more than those in control housing 7-14 DPI and engaged in more total daily sleep bouts during the dark period. However, SF stress did not exacerbate post-TBI sleep deficits. Testing in the Morris water maze revealed sex dependent differences in spatial reference memory 9-14 DPI with males performing worse than females. Post-TBI SF stress suppressed neurogenesis-related gene expression and increased inflammatory signaling in the cortex at 14 DPI. No differences in sleep behavior were detected between groups during the SF stress recovery period 15-30 DPI. Microscopy revealed cortical and hippocampal IBA1 and CD68 percent-area increased in TBI SF-R mice 30 DPI. Additionally, neuroinflammatory gene expression was increased, and synaptogenesis-related gene expression was suppressed in TBI-SF mice 30 DPI. Finally, IPA canonical pathway analysis showed post-TBI SF impaired and delayed activation of synapse-related pathways between 14 and 30 DPI. These data show that transient SF stress after TBI impairs recovery and conveys long-lasting impacts on neuroimmune function independent of continuous sleep deficits. Together, these finding support that even limited exposure to post-TBI SF stress can have lasting impacts on cognitive recovery and regulation of the immune response to trauma.
Collapse
Affiliation(s)
- Samuel Houle
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, 43210, Columbus, OH, USA
| | - Zoe Tapp
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, 43210, Columbus, OH, USA
- Institute for Behavioral Medicine Research, Neurological Institute, The Ohio State University, 460 Medical Center Drive, 43210, Columbus, OH, USA
| | - Shannon Dobres
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, 43210, Columbus, OH, USA
| | - Sakeef Ahsan
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, 43210, Columbus, OH, USA
| | - Yvanna Reyes
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, 43210, Columbus, OH, USA
| | - Christopher Cotter
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, 43210, Columbus, OH, USA
| | - Jessica Mitsch
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, 43210, Columbus, OH, USA
| | - Zachary Zimomra
- Institute for Behavioral Medicine Research, Neurological Institute, The Ohio State University, 460 Medical Center Drive, 43210, Columbus, OH, USA
| | - Juan Peng
- Center for Biostatistics, The Ohio State University, 320-55 Lincoln Tower, 1800 Cannon Drive, 43210, Columbus, OH, USA
| | - Rachel K. Rowe
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Jonathan Lifshitz
- Phoenix VA Health Care System and University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - John Sheridan
- Institute for Behavioral Medicine Research, Neurological Institute, The Ohio State University, 460 Medical Center Drive, 43210, Columbus, OH, USA
- Division of Biosciences, College of Dentistry, The Ohio State University, 305 W. 12th Ave, 43210, Columbus, OH, USA
| | - Jonathan Godbout
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, 43210, Columbus, OH, USA
- Institute for Behavioral Medicine Research, Neurological Institute, The Ohio State University, 460 Medical Center Drive, 43210, Columbus, OH, USA
- Chronic Brain Injury Program, The Ohio State University, 190 North Oval Mall, 43210, Columbus, OH, USA
| | - Olga N. Kokiko-Cochran
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, 43210, Columbus, OH, USA
- Institute for Behavioral Medicine Research, Neurological Institute, The Ohio State University, 460 Medical Center Drive, 43210, Columbus, OH, USA
- Chronic Brain Injury Program, The Ohio State University, 190 North Oval Mall, 43210, Columbus, OH, USA
| |
Collapse
|
11
|
Ritzel RM, Li Y, Jiao Y, Doran SJ, Khan N, Henry RJ, Brunner K, Loane DJ, Faden AI, Szeto GL, Wu J. Bi-directional neuro-immune dysfunction after chronic experimental brain injury. J Neuroinflammation 2024; 21:83. [PMID: 38581043 PMCID: PMC10996305 DOI: 10.1186/s12974-024-03082-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 03/30/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUND It is well established that traumatic brain injury (TBI) causes acute and chronic alterations in systemic immune function and that systemic immune changes contribute to posttraumatic neuroinflammation and neurodegeneration. However, how TBI affects bone marrow (BM) hematopoietic stem/progenitor cells chronically and to what extent such changes may negatively impact innate immunity and neurological function has not been examined. METHODS To further understand the role of BM cell derivatives on TBI outcome, we generated BM chimeric mice by transplanting BM from chronically injured or sham (i.e., 90 days post-surgery) congenic donor mice into otherwise healthy, age-matched, irradiated CD45.2 C57BL/6 (WT) hosts. Immune changes were evaluated by flow cytometry, multiplex ELISA, and NanoString technology. Moderate-to-severe TBI was induced by controlled cortical impact injury and neurological function was measured using a battery of behavioral tests. RESULTS TBI induced chronic alterations in the transcriptome of BM lineage-c-Kit+Sca1+ (LSK+) cells in C57BL/6 mice, including modified epigenetic and senescence pathways. After 8 weeks of reconstitution, peripheral myeloid cells from TBI→WT mice showed significantly higher oxidative stress levels and reduced phagocytic activity. At eight months after reconstitution, TBI→WT chimeric mice were leukopenic, with continued alterations in phagocytosis and oxidative stress responses, as well as persistent neurological deficits. Gene expression analysis revealed BM-driven changes in neuroinflammation and neuropathology after 8 weeks and 8 months of reconstitution, respectively. Chimeric mice subjected to TBI at 8 weeks and 8 months post-reconstitution showed that longer reconstitution periods (i.e., time post-injury) were associated with increased microgliosis and leukocyte infiltration. Pre-treatment with a senolytic agent, ABT-263, significantly improved behavioral performance of aged C57BL/6 mice at baseline, although it did not attenuate neuroinflammation in the acutely injured brain. CONCLUSIONS TBI causes chronic activation and progressive dysfunction of the BM stem/progenitor cell pool, which drives long-term deficits in hematopoiesis, innate immunity, and neurological function, as well as altered sensitivity to subsequent brain injury.
Collapse
Affiliation(s)
- Rodney M Ritzel
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| | - Yun Li
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Yun Jiao
- Department of Chemical, Biochemical and Environmental Engineering, University of Maryland, Baltimore County, Baltimore, MD, 21250, USA
| | - Sarah J Doran
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Niaz Khan
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Rebecca J Henry
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Kavitha Brunner
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - David J Loane
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Alan I Faden
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Gregory L Szeto
- Department of Chemical, Biochemical and Environmental Engineering, University of Maryland, Baltimore County, Baltimore, MD, 21250, USA
| | - Junfang Wu
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
12
|
Taylor MA, Kokiko-Cochran ON. Context is key: glucocorticoid receptor and corticosteroid therapeutics in outcomes after traumatic brain injury. Front Cell Neurosci 2024; 18:1351685. [PMID: 38529007 PMCID: PMC10961349 DOI: 10.3389/fncel.2024.1351685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 02/21/2024] [Indexed: 03/27/2024] Open
Abstract
Traumatic brain injury (TBI) is a global health burden, and survivors suffer functional and psychiatric consequences that can persist long after injury. TBI induces a physiological stress response by activating the hypothalamic-pituitary-adrenal (HPA) axis, but the effects of injury on the stress response become more complex in the long term. Clinical and experimental evidence suggests long lasting dysfunction of the stress response after TBI. Additionally, pre- and post-injury stress both have negative impacts on outcome following TBI. This bidirectional relationship between stress and injury impedes recovery and exacerbates TBI-induced psychiatric and cognitive dysfunction. Previous clinical and experimental studies have explored the use of synthetic glucocorticoids as a therapeutic for stress-related TBI outcomes, but these have yielded mixed results. Furthermore, long-term steroid treatment is associated with multiple negative side effects. There is a pressing need for alternative approaches that improve stress functionality after TBI. Glucocorticoid receptor (GR) has been identified as a fundamental link between stress and immune responses, and preclinical evidence suggests GR plays an important role in microglia-mediated outcomes after TBI and other neuroinflammatory conditions. In this review, we will summarize GR-mediated stress dysfunction after TBI, highlighting the role of microglia. We will discuss recent studies which target microglial GR in the context of stress and injury, and we suggest that cell-specific GR interventions may be a promising strategy for long-term TBI pathophysiology.
Collapse
Affiliation(s)
| | - Olga N. Kokiko-Cochran
- Department of Neuroscience, Chronic Brain Injury Program, Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
13
|
Boland R, Kokiko-Cochran ON. Deplete and repeat: microglial CSF1R inhibition and traumatic brain injury. Front Cell Neurosci 2024; 18:1352790. [PMID: 38450286 PMCID: PMC10915023 DOI: 10.3389/fncel.2024.1352790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 01/25/2024] [Indexed: 03/08/2024] Open
Abstract
Traumatic brain injury (TBI) is a public health burden affecting millions of people. Sustained neuroinflammation after TBI is often associated with poor outcome. As a result, increased attention has been placed on the role of immune cells in post-injury recovery. Microglia are highly dynamic after TBI and play a key role in the post-injury neuroinflammatory response. Therefore, microglia represent a malleable post-injury target that could substantially influence long-term outcome after TBI. This review highlights the cell specific role of microglia in TBI pathophysiology. Microglia have been manipulated via genetic deletion, drug inhibition, and pharmacological depletion in various pre-clinical TBI models. Notably, colony stimulating factor 1 (CSF1) and its receptor (CSF1R) have gained much traction in recent years as a pharmacological target on microglia. CSF1R is a transmembrane tyrosine kinase receptor that is essential for microglia proliferation, differentiation, and survival. Small molecule inhibitors targeting CSF1R result in a swift and effective depletion of microglia in rodents. Moreover, discontinuation of the inhibitors is sufficient for microglia repopulation. Attention is placed on summarizing studies that incorporate CSF1R inhibition of microglia. Indeed, microglia depletion affects multiple aspects of TBI pathophysiology, including neuroinflammation, oxidative stress, and functional recovery with measurable influence on astrocytes, peripheral immune cells, and neurons. Taken together, the data highlight an important role for microglia in sustaining neuroinflammation and increasing risk of oxidative stress, which lends to neuronal damage and behavioral deficits chronically after TBI. Ultimately, the insights gained from CSF1R depletion of microglia are critical for understanding the temporospatial role that microglia develop in mediating TBI pathophysiology and recovery.
Collapse
Affiliation(s)
- Rebecca Boland
- Department of Neuroscience, College of Medicine, Chronic Brain Injury Program, Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States
| | - Olga N Kokiko-Cochran
- Department of Neuroscience, College of Medicine, Chronic Brain Injury Program, Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
14
|
Packer JM, Bray CE, Beckman NB, Wangler LM, Davis AC, Goodman EJ, Klingele NE, Godbout JP. Impaired cortical neuronal homeostasis and cognition after diffuse traumatic brain injury are dependent on microglia and type I interferon responses. Glia 2024; 72:300-321. [PMID: 37937831 PMCID: PMC10764078 DOI: 10.1002/glia.24475] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 09/14/2023] [Accepted: 09/20/2023] [Indexed: 11/09/2023]
Abstract
Neuropsychiatric complications including depression and cognitive decline develop in the years after traumatic brain injury (TBI), negatively affecting quality of life. Microglial and type 1 interferon (IFN-I) responses are associated with the transition from acute to chronic neuroinflammation after diffuse TBI in mice. Thus, the purpose of this study was to determine if impaired neuronal homeostasis and increased IFN-I responses intersected after TBI to cause cognitive impairment. Here, the RNA profile of neurons and microglia after TBI (single nucleus RNA-sequencing) with or without microglia depletion (CSF1R antagonist) was assessed 7 dpi. There was a TBI-dependent suppression of cortical neuronal homeostasis with reductions in CREB signaling, synaptogenesis, and synaptic migration and increases in RhoGDI and PTEN signaling (Ingenuity Pathway Analysis). Microglial depletion reversed 50% of TBI-induced gene changes in cortical neurons depending on subtype. Moreover, the microglial RNA signature 7 dpi was associated with increased stimulator of interferon genes (STING) activation and IFN-I responses. Therefore, we sought to reduce IFN-I signaling after TBI using STING knockout mice and a STING antagonist, chloroquine (CQ). TBI-associated cognitive deficits in novel object location and recognition (NOL/NOR) tasks at 7 and 30 dpi were STING dependent. In addition, TBI-induced STING expression, microglial morphological restructuring, inflammatory (Tnf, Cd68, Ccl2) and IFN-related (Irf3, Irf7, Ifi27) gene expression in the cortex were attenuated in STINGKO mice. CQ also reversed TBI-induced cognitive deficits and reduced TBI-induced inflammatory (Tnf, Cd68, Ccl2) and IFN (Irf7, Sting) cortical gene expression. Collectively, reducing IFN-I signaling after TBI with STING-dependent interventions attenuated the prolonged microglial activation and cognitive impairment.
Collapse
Affiliation(s)
- Jonathan M Packer
- Department of Neuroscience, The Ohio State University, Columbus, Ohio, USA
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio, USA
| | - Chelsea E Bray
- College of Medicine, The Ohio State University, Columbus, United States
| | - Nicolas B Beckman
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio, USA
| | - Lynde M Wangler
- Department of Neuroscience, The Ohio State University, Columbus, Ohio, USA
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio, USA
| | - Amara C Davis
- Department of Neuroscience, The Ohio State University, Columbus, Ohio, USA
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio, USA
| | - Ethan J Goodman
- Department of Neuroscience, The Ohio State University, Columbus, Ohio, USA
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio, USA
| | - Nathaniel E Klingele
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio, USA
| | - Jonathan P Godbout
- Department of Neuroscience, The Ohio State University, Columbus, Ohio, USA
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio, USA
- College of Medicine, The Ohio State University, Columbus, United States
- Chronic Brain Injury Program, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
15
|
Fleiss B, Gressens P. Role of Microglial Modulation in Therapies for Perinatal Brain Injuries Leading to Neurodevelopmental Disorders. ADVANCES IN NEUROBIOLOGY 2024; 37:591-606. [PMID: 39207715 DOI: 10.1007/978-3-031-55529-9_33] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Neurodevelopmental disorders (NDDs) encompass various conditions stemming from changes during brain development, typically diagnosed early in life. Examples include autism spectrum disorder, intellectual disability, cerebral palsy, seizures, dyslexia, and attention deficit hyperactivity disorder. Many NDDs are linked to perinatal events like infections, oxygen disturbances, or insults in combination. This chapter outlines the causes and effects of perinatal brain injury as they relate to microglia, along with efforts to prevent or treat such damage. We primarily discuss therapies targeting microglia modulation, focusing on those either clinically used or in advanced development, often tested in large animal models such as sheep, non-human primates, and piglets-standard translational models in perinatal medicine. Additionally, it touches on experimental studies showcasing advancements in the field.
Collapse
Affiliation(s)
- Bobbi Fleiss
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
- Université de Paris, NeuroDiderot, Inserm, Paris, France
| | - Pierre Gressens
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia.
- Université de Paris, NeuroDiderot, Inserm, Paris, France.
| |
Collapse
|
16
|
Wangler LM, Godbout JP. Microglia moonlighting after traumatic brain injury: aging and interferons influence chronic microglia reactivity. Trends Neurosci 2023; 46:926-940. [PMID: 37723009 PMCID: PMC10592045 DOI: 10.1016/j.tins.2023.08.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/11/2023] [Accepted: 08/24/2023] [Indexed: 09/20/2023]
Abstract
Most of the individuals who experience traumatic brain injury (TBI) develop neuropsychiatric and cognitive complications that negatively affect recovery and health span. Activation of multiple inflammatory pathways persists after TBI, but it is unclear how inflammation contributes to long-term behavioral and cognitive deficits. One outcome of TBI is microglial priming and subsequent hyper-reactivity to secondary stressors, injuries, or immune challenges that further augment complications. Additionally, microglia priming with aging contributes to exaggerated glial responses to TBI. One prominent inflammatory pathway, interferon (IFN) signaling, is increased after TBI and may contribute to microglial priming and subsequent reactivity. This review discusses the contributions of microglia to inflammatory processes after TBI, as well as the influence of aging and IFNs on microglia reactivity and chronic inflammation after TBI.
Collapse
Affiliation(s)
- Lynde M Wangler
- Department of Neuroscience, The Ohio State University Wexner Medical Center, 333 W 10th Ave, Columbus, OH, USA
| | - Jonathan P Godbout
- Department of Neuroscience, The Ohio State University Wexner Medical Center, 333 W 10th Ave, Columbus, OH, USA; Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, 460 Medical Center Drive, Columbus, OH, USA; Chronic Brain Injury Program, The Ohio State University, 190 North Oval Mall, Columbus, OH, USA.
| |
Collapse
|
17
|
Ritzel RM, Li Y, Jiao Y, Doran SJ, Khan N, Henry RJ, Brunner K, Loane DJ, Faden AI, Szeto GL, Wu J. The brain-bone marrow axis and its implications for chronic traumatic brain injury. RESEARCH SQUARE 2023:rs.3.rs-3356007. [PMID: 37790560 PMCID: PMC10543403 DOI: 10.21203/rs.3.rs-3356007/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Traumatic brain injury (TBI) causes acute and chronic alterations in systemic immune function which contribute to posttraumatic neuroinflammation and neurodegeneration. However, how TBI affects bone marrow (BM) hematopoietic stem/progenitor cells chronically and to what extent such changes may negatively impact innate immunity and neurological function has not been examined. To further understand the role of BM cell derivatives on TBI outcome, we generated BM chimeric mice by transplanting BM from chronically injured or sham congenic donor mice into otherwise healthy, age-matched, irradiated hosts. After 8 weeks of reconstitution, peripheral myeloid cells from TBI→WT mice showed significantly higher oxidative stress levels and reduced phagocytic activity. At eight months after reconstitution, TBI→WT chimeric mice were leukopenic, with continued alterations in phagocytosis and oxidative stress responses, as well as persistent neurological deficits. Gene expression analysis revealed BM-driven changes in neuroinflammation and neuropathology after 8 weeks and 8 months of reconstitution, respectively. Chimeric mice subjected to TBI showed that longer reconstitution periods were associated with increased microgliosis and leukocyte infiltration. Thus, TBI causes chronic activation and progressive dysfunction of the BM stem/progenitor cell pool, which drives long-term deficits in innate immunity and neurological function, as well as altered sensitivity to subsequent brain injury.
Collapse
Affiliation(s)
- Rodney M. Ritzel
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Texas, USA
| | - Yun Li
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Yun Jiao
- Department of Chemical, Biochemical and Environmental Engineering, University of Maryland, Baltimore County, Maryland, USA
| | - Sarah J. Doran
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Niaz Khan
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Rebecca J. Henry
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Kavitha Brunner
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - David J. Loane
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Alan I. Faden
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Gregory L. Szeto
- Department of Chemical, Biochemical and Environmental Engineering, University of Maryland, Baltimore County, Maryland, USA
| | - Junfang Wu
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
18
|
Tapp ZM, Ren C, Palmer K, Kumar J, Atluri RR, Fitzgerald J, Velasquez J, Godbout J, Sheridan J, Kokiko-Cochran ON. Divergent Spatial Learning, Enhanced Neuronal Transcription, and Blood-Brain Barrier Disruption Develop During Recovery from Post-Injury Sleep Fragmentation. Neurotrauma Rep 2023; 4:613-626. [PMID: 37752925 PMCID: PMC10518692 DOI: 10.1089/neur.2023.0018] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2023] Open
Abstract
Traumatic brain injury (TBI) causes pathophysiology that may significantly decrease quality of life over time. A major propagator of this response is chronic, maladaptive neuroinflammation, which can be exacerbated by stressors such as sleep fragmentation (SF). This study determined whether post-TBI SF had lasting behavioral and inflammatory effects even with a period of recovery. To test this, male and female mice received a moderate lateral fluid percussion TBI or sham surgery. Half the mice were left undisturbed, and half were exposed to daily SF for 30 days. All mice were then undisturbed between 30 and 60 days post-injury (DPI), allowing mice to recover from SF (SF-R). SF-R did not impair global Barnes maze performance. Nonetheless, TBI SF-R mice displayed retrogression in latency to reach the goal box within testing days. These nuanced behavioral changes in TBI SF-R mice were associated with enhanced expression of neuronal processing/signaling genes and indicators of blood-brain barrier (BBB) dysfunction. Aquaporin-4 (AQP4) expression, a marker of BBB integrity, was differentially altered by TBI and TBI SF-R. For example, TBI enhanced cortical AQP4 whereas TBI SF-R mice had the lowest cortical expression of perivascular AQP4, dysregulated AQP4 polarization, and the highest number of CD45+ cells in the ipsilateral cortex. Altogether, post-TBI SF caused lasting, divergent behavioral responses associated with enhanced expression of neuronal transcription and BBB disruption even after a period of recovery from SF. Understanding lasting impacts from post-TBI stressors can better inform both acute and chronic post-injury care to improve long-term outcome post-TBI.
Collapse
Affiliation(s)
- Zoe M. Tapp
- Department of Neuroscience, College of Medicine, College of Dentistry, The Ohio State University, Columbus, Ohio, USA
| | - Cindy Ren
- Department of Neuroscience, College of Medicine, College of Dentistry, The Ohio State University, Columbus, Ohio, USA
| | - Kelsey Palmer
- Department of Neuroscience, College of Medicine, College of Dentistry, The Ohio State University, Columbus, Ohio, USA
| | - Julia Kumar
- Department of Neuroscience, College of Medicine, College of Dentistry, The Ohio State University, Columbus, Ohio, USA
| | - Ravitej R. Atluri
- Department of Neuroscience, College of Medicine, College of Dentistry, The Ohio State University, Columbus, Ohio, USA
| | - Julie Fitzgerald
- Department of Neuroscience, College of Medicine, College of Dentistry, The Ohio State University, Columbus, Ohio, USA
| | - John Velasquez
- Department of Neuroscience, College of Medicine, College of Dentistry, The Ohio State University, Columbus, Ohio, USA
| | - Jonathan Godbout
- Department of Neuroscience, College of Medicine, College of Dentistry, The Ohio State University, Columbus, Ohio, USA
- Chronic Brain Injury Program, College of Dentistry, The Ohio State University, Columbus, Ohio, USA
- Institute for Behavioral Medicine Research, Neurological Institute, College of Dentistry, The Ohio State University, Columbus, Ohio, USA
| | - John Sheridan
- Department of Neuroscience, College of Medicine, College of Dentistry, The Ohio State University, Columbus, Ohio, USA
- Chronic Brain Injury Program, College of Dentistry, The Ohio State University, Columbus, Ohio, USA
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, Ohio, USA
| | - Olga N. Kokiko-Cochran
- Department of Neuroscience, College of Medicine, College of Dentistry, The Ohio State University, Columbus, Ohio, USA
- Chronic Brain Injury Program, College of Dentistry, The Ohio State University, Columbus, Ohio, USA
- Institute for Behavioral Medicine Research, Neurological Institute, College of Dentistry, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
19
|
Shi S, Zhang S, Kong L. Effects of Treatment with Probiotics on Cognitive Function and Regulatory Role of Cortisol and IL-1β in Adolescent Patients with Major Depressive Disorder. Life (Basel) 2023; 13:1829. [PMID: 37763233 PMCID: PMC10532456 DOI: 10.3390/life13091829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/14/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
The aim of this study was to investigate the effects of probiotics on cognitive function and the regulation of cortisol and IL-1β in adolescents with depression. All 180 participants were randomly assigned to a study group (treated with probiotics combined with sertraline hydrochloride) and a control group (treated with sertraline hydrochloride). The repetitive Neuropsychological State Test (RBANS) and Hamilton Depression Scale (HAMD) were administered to MDD patients. The levels of serum cortisol and IL-1β were detected using an ELISA kit. Except for speech function, factors including immediate memory, visual span, attention function, delayed memory, and the RBANS in the study group were significantly higher than those in the control group. The levels of cortisol and interleukin-1β in the study group were significantly downregulated compared to those in the control group. Except for speech function, the cortisol level was negatively correlated with the RBANS total score and other factors in the study group. Interleukin-1β was also negatively correlated with the RBANS total score and each factor score. Cortisol and interleukin-1β were predictors of the RBANS total score, which explained 46.80% of the variance. Cortisol had significant predictive effects on attention function and delayed memory, and interleukin-1β had significant predictive effects on visual span and speech function. It could be concluded that probiotics could improve cognitive function in adolescents with depression by regulating cortisol and IL-1β levels.
Collapse
Affiliation(s)
- Shaoli Shi
- Psychiatry Department, The 5th People’s Hospital of Luoyang, Luoyang 471027, China;
| | - Shuyou Zhang
- Intervention Center of Mental Crisis, No.904 Hospital, Changzhou 213003, China;
| | - Lingming Kong
- Intervention Center of Mental Crisis, No.904 Hospital, Changzhou 213003, China;
| |
Collapse
|
20
|
Sanchez K, Wu SL, Kakkar R, Darling JS, Harper CS, Fonken LK. Ovariectomy in mice primes hippocampal microglia to exacerbate behavioral sickness responses. Brain Behav Immun Health 2023; 30:100638. [PMID: 37256192 PMCID: PMC10225896 DOI: 10.1016/j.bbih.2023.100638] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 04/14/2023] [Accepted: 05/07/2023] [Indexed: 06/01/2023] Open
Abstract
Estrogens are a group of steroid hormones that promote the development and maintenance of the female reproductive system and secondary sex characteristics. Estrogens also modulate immune responses; estrogen loss at menopause increases the risk of inflammatory disorders. Elevated inflammatory responses in the brain can lead to affective behavioral changes, which are characteristic of menopause. Thus, here we examined whether loss of estrogens sensitizes microglia, the primary innate immune cell of the brain, leading to changes in affective behaviors. To test this question, adult C57BL/6 mice underwent an ovariectomy to remove endogenous estrogens and then received estradiol hormone replacement or vehicle. After a one-month recovery, mice received an immune challenge with lipopolysaccharide (LPS) or vehicle control treatment and underwent behavioral testing. Ovariectomized, saline-treated mice exhibited reduced social investigation compared to sham-operated mice. Furthermore, ovariectomized mice that received LPS exhibited an exacerbated decrease in sucrose preference, which was ameliorated by estradiol replacement. These results indicate that ovariectomy modulates affective behaviors at baseline and in response to an inflammatory challenge. Ovariectomy-related behavioral changes were associated with downregulation of Cx3cr1, a microglial receptor that limits activation, suggesting that estrogen loss can disinhibit microglia to immune stimuli. Indeed, estradiol treatment reduced ovariectomy-induced increases in Il1b and Il6 expression after an immune challenge. Changes in microglial reactivity following ovariectomy are likely subtle, as overt changes in microglial morphology (e.g., soma size and branching) were limited. Collectively, these results suggest that a lack of estrogens may allow microglia to confer exaggerated neuroimmune responses, thereby raising vulnerability to adverse affective- and sickness-related behavioral changes.
Collapse
Affiliation(s)
- Kevin Sanchez
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Sienna L. Wu
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Reha Kakkar
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Jeffrey S. Darling
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Claire S. Harper
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Laura K. Fonken
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78712, USA
| |
Collapse
|
21
|
Datta S, Lin F, Jones LD, Pingle SC, Kesari S, Ashili S. Traumatic brain injury and immunological outcomes: the double-edged killer. Future Sci OA 2023; 9:FSO864. [PMID: 37228857 PMCID: PMC10203904 DOI: 10.2144/fsoa-2023-0037] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 04/20/2023] [Indexed: 05/27/2023] Open
Abstract
Traumatic brain injury (TBI) is a significant cause of mortality and morbidity worldwide resulting from falls, car accidents, sports, and blast injuries. TBI is characterized by severe, life-threatening consequences due to neuroinflammation in the brain. Contact and collision sports lead to higher disability and death rates among young adults. Unfortunately, no therapy or drug protocol currently addresses the complex pathophysiology of TBI, leading to the long-term chronic neuroinflammatory assaults. However, the immune response plays a crucial role in tissue-level injury repair. This review aims to provide a better understanding of TBI's immunobiology and management protocols from an immunopathological perspective. It further elaborates on the risk factors, disease outcomes, and preclinical studies to design precisely targeted interventions for enhancing TBI outcomes.
Collapse
Affiliation(s)
- Souvik Datta
- Rhenix Lifesciences, 237 Arsha Apartments, Kalyan Nagar, Hyderabad, TG 500038, India
| | - Feng Lin
- CureScience, 5820 Oberlin Drive #202, San Diego, CA 92121, USA
| | | | | | - Santosh Kesari
- Saint John's Cancer Institute, Santa Monica, CA 90404, USA
| | | |
Collapse
|
22
|
Sansone F, Pellegrino GM, Caronni A, Bonazza F, Vegni E, Lué A, Bocci T, Pipolo C, Giusti G, Di Filippo P, Di Pillo S, Chiarelli F, Sferrazza Papa GF, Attanasi M. Long COVID in Children: A Multidisciplinary Review. Diagnostics (Basel) 2023; 13:1990. [PMID: 37370884 DOI: 10.3390/diagnostics13121990] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/01/2023] [Accepted: 06/01/2023] [Indexed: 06/29/2023] Open
Abstract
Long COVID syndrome has emerged as a long-lasting consequence of acute SARS-CoV-2 infection in adults. In addition, children may be affected by Long COVID, with potential clinical issues in different fields, including problems in school performance and daily activities. Yet, the pathophysiologic bases of Long COVID in children are largely unknown, and it is difficult to predict who will develop the syndrome. In this multidisciplinary clinical review, we summarise the latest scientific data regarding Long COVID and its impact on children. Special attention is given to diagnostic tests, in order to help the physicians to find potential disease markers and quantify impairment. Specifically, we assess the respiratory, upper airways, cardiac, neurologic and motor and psychological aspects. Finally, we also propose a multidisciplinary clinical approach.
Collapse
Affiliation(s)
| | | | - Antonio Caronni
- Department of Neurorehabilitation Sciences, IRCCS Istituto Auxologico Italiano, Ospedale San Luca, 20122 Milan, Italy
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy
| | - Federica Bonazza
- Department of Health Sciences, Clinical Psychology, University of Milan, Via di Rudinì 8, 20142 Milan, Italy
| | - Elena Vegni
- Department of Health Sciences, Clinical Psychology, University of Milan, Via di Rudinì 8, 20142 Milan, Italy
- Unit of Clinical Psychology, San Paolo Hospital, ASST Santi Paolo e Carlo, Via di Rudinì 8, 20142 Milan, Italy
| | - Alberto Lué
- Service of Digestive Diseases, University Clinic Hospital Lozano Blesa, IIS Aragón, 50009 Zaragoza, Spain
| | - Tommaso Bocci
- Department of Health Sciences, University of Milan, 20146 Milan, Italy
- Clinical Neurology Unit, Department of Health Sciences, "Azienda Socio-Sanitaria Territoriale Santi Paolo e Carlo", University of Milan, 20146 Milan, Italy
| | - Carlotta Pipolo
- Department of Health Sciences, Otorhinolaryngology Department, ASST Santi Paolo e Carlo, University of Milan, 20142 Milan, Italy
| | - Giuliano Giusti
- Paediatric Cardiology Unit, Niguarda Hospital, 20162 Milan, Italy
| | - Paola Di Filippo
- Paediatric Allergy and Pulmonology Unit, Department of Paediatrics, University of Chieti-Pescara, 66100 Chieti, Italy
| | - Sabrina Di Pillo
- Paediatric Allergy and Pulmonology Unit, Department of Paediatrics, University of Chieti-Pescara, 66100 Chieti, Italy
| | - Francesco Chiarelli
- Paediatric Allergy and Pulmonology Unit, Department of Paediatrics, University of Chieti-Pescara, 66100 Chieti, Italy
| | | | - Marina Attanasi
- Paediatric Allergy and Pulmonology Unit, Department of Paediatrics, University of Chieti-Pescara, 66100 Chieti, Italy
| |
Collapse
|
23
|
Law LM, Griffiths DR, Lifshitz J. Peg Forest Rehabilitation - A novel spatial navigation based cognitive rehabilitation paradigm for experimental neurotrauma. Behav Brain Res 2023; 443:114355. [PMID: 36801425 PMCID: PMC10883691 DOI: 10.1016/j.bbr.2023.114355] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 01/18/2023] [Accepted: 02/16/2023] [Indexed: 02/19/2023]
Abstract
Traumatic brain injury (TBI) results from mechanical forces applied to the head. Ensuing cascades of complex pathophysiology transition the injury event into a disease process. The enduring constellation of emotional, somatic, and cognitive impairments degrade quality of life for the millions of TBI survivors suffering from long-term neurological symptoms. Rehabilitation strategies have reported mixed results, as most have not focused on specific symptomatology or explored cellular processes. The current experiments evaluated a novel cognitive rehabilitation paradigm for brain-injured and uninjured rats. The arena is a plastic floor with a cartesian grid of holes for plastic dowels to create new environments with the rearrangement of threaded pegs. Rats received either two weeks of Peg Forest rehabilitation (PFR) or open field exposure starting at 7 days post-injury; or one week starting at either day 7 or 14 post-injury; or served as caged controls. Cognitive performance was assessed on a battery of novel object tasks at 28 days post-injury. The results revealed that two weeks of PFR was required to prevent the onset of cognitive impairments, while one week of PFR was insufficient regardless of when rehabilitation was initiated after injury. Further assessment of the task showed that novel daily arrangements of the environment were required to impart the cognitive performance benefits, as exposure to a static arrangement of pegs for PFR each day did not improve cognitive performance. The results indicate that PFR prevents the onset of cognitive disorders following acquired a mild to moderate brain injury, and potentially other neurological conditions.
Collapse
Affiliation(s)
- L Matthew Law
- Phoenix Veterans Affairs Health Care System, Phoenix, AZ, United States; University of Arizona College of Medicine, Phoenix, AZ, United States; BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States.
| | - Daniel R Griffiths
- Phoenix Veterans Affairs Health Care System, Phoenix, AZ, United States; University of Arizona College of Medicine, Phoenix, AZ, United States; BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States
| | - Jonathan Lifshitz
- Phoenix Veterans Affairs Health Care System, Phoenix, AZ, United States; University of Arizona College of Medicine, Phoenix, AZ, United States; BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States
| |
Collapse
|
24
|
Wangler LM, Bray CE, Packer JM, Tapp ZM, Davis AC, O'Neil SM, Baetz K, Ouviña M, Witzel M, Godbout JP. Amplified Gliosis and Interferon-Associated Inflammation in the Aging Brain following Diffuse Traumatic Brain Injury. J Neurosci 2022; 42:9082-9096. [PMID: 36257689 PMCID: PMC9732830 DOI: 10.1523/jneurosci.1377-22.2022] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/07/2022] [Accepted: 09/12/2022] [Indexed: 02/08/2023] Open
Abstract
Traumatic brain injury (TBI) is associated with chronic psychiatric complications and increased risk for development of neurodegenerative pathology. Aged individuals account for most TBI-related hospitalizations and deaths. Nonetheless, neurobiological mechanisms that underlie worsened functional outcomes after TBI in the elderly remain unclear. Therefore, this study aimed to identify pathways that govern differential responses to TBI with age. Here, adult (2 months of age) and aged (16-18 months of age) male C57BL/6 mice were subjected to diffuse brain injury (midline fluid percussion), and cognition, gliosis, and neuroinflammation were determined 7 or 30 d postinjury (dpi). Cognitive impairment was evident 7 dpi, independent of age. There was enhanced morphologic restructuring of microglia and astrocytes 7 dpi in the cortex and hippocampus of aged mice compared with adults. Transcriptional analysis revealed robust age-dependent amplification of cytokine/chemokine, complement, innate immune, and interferon-associated inflammatory gene expression in the cortex 7 dpi. Ingenuity pathway analysis of the transcriptional data showed that type I interferon (IFN) signaling was significantly enhanced in the aged brain after TBI compared with adults. Age prolonged inflammatory signaling and microgliosis 30 dpi with an increased presence of rod microglia. Based on these results, a STING (stimulator of interferon genes) agonist, DMXAA, was used to determine whether augmenting IFN signaling worsened cortical inflammation and gliosis after TBI. DMXAA-treated Adult-TBI mice showed comparable expression of myriad genes that were overexpressed in the cortex of Aged-TBI mice, including Irf7, Clec7a, Cxcl10, and Ccl5 Overall, diffuse TBI promoted amplified IFN signaling in aged mice, resulting in extended inflammation and gliosis.SIGNIFICANCE STATEMENT Elderly individuals are at higher risk of complications following traumatic brain injury (TBI). Individuals >70 years old have the highest rates of TBI-related hospitalization, neurodegenerative pathology, and death. Although inflammation has been linked with poor outcomes in aging, the specific biological pathways driving worsened outcomes after TBI in aging remain undefined. In this study, we identify amplified interferon-associated inflammation and gliosis in aged mice following TBI that was associated with persistent inflammatory gene expression and microglial morphologic diversity 30 dpi. STING (stimulator of interferon genes) agonist DMXAA was used to demonstrate a causal link between augmented interferon signaling and worsened neuroinflammation after TBI. Therefore, interferon signaling may represent a therapeutic target to reduce inflammation-associated complications following TBI.
Collapse
Affiliation(s)
- Lynde M Wangler
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, Ohio 43210
- Institute for Behavioral Medicine Research, Wexner Medical Center, The Ohio State University, Columbus, Ohio 43210
| | - Chelsea E Bray
- Institute for Behavioral Medicine Research, Wexner Medical Center, The Ohio State University, Columbus, Ohio 43210
- College of Medicine, The Ohio State University, Columbus, Ohio 43210
| | - Jonathan M Packer
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, Ohio 43210
- Institute for Behavioral Medicine Research, Wexner Medical Center, The Ohio State University, Columbus, Ohio 43210
| | - Zoe M Tapp
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, Ohio 43210
- Institute for Behavioral Medicine Research, Wexner Medical Center, The Ohio State University, Columbus, Ohio 43210
| | - Amara C Davis
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, Ohio 43210
- Institute for Behavioral Medicine Research, Wexner Medical Center, The Ohio State University, Columbus, Ohio 43210
| | - Shane M O'Neil
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, Ohio 43210
- Institute for Behavioral Medicine Research, Wexner Medical Center, The Ohio State University, Columbus, Ohio 43210
| | - Kara Baetz
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, Ohio 43210
| | - Michelle Ouviña
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, Ohio 43210
| | - Mollie Witzel
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, Ohio 43210
| | - Jonathan P Godbout
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, Ohio 43210
- Institute for Behavioral Medicine Research, Wexner Medical Center, The Ohio State University, Columbus, Ohio 43210
- Chronic Brain Injury Program, The Ohio State University, Columbus, Ohio 43210
- College of Medicine, The Ohio State University, Columbus, Ohio 43210
| |
Collapse
|
25
|
Huo X, Xu X, Li M, Xiao L, Wang Y, Li W, Wang C, Sun T. Effectiveness of antiseizure medications therapy in preventing seizures in brain injury patients: A network meta-analysis. Front Pharmacol 2022; 13:1001363. [PMID: 36188582 PMCID: PMC9521202 DOI: 10.3389/fphar.2022.1001363] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 08/30/2022] [Indexed: 12/03/2022] Open
Abstract
Purpose: To explore the effectiveness of different anti-seizure medications in preventing early and late post-traumatic epilepsy (PTE). The efficacy, treatment-related side-effects, and mortality of the different treatments were compared using a ranking model to identify the optimal treatment. Methods: A comprehensive literature search was performed using Pubmed, Medline, Embase, and Cochrane library databases. All relevant published articles up to 10 March 2022 were evaluated. The quality of the extracted data was assessed using either the Cochrane risk of bias tool or the Newcastle-Ottawa scale. The primary outcome measures were early or late post-traumatic seizures. The secondary outcome measures were mortality, treatment-related adverse effects, length of hospital stay, and length of stay within the intensive care unit (ICU). Results: A total of seven randomized controlled trials and 18 non-randomized controlled trials were included in this network meta-analysis. The trials included six interventions: Phenytoin (PHT)+phenobarbital (PB), levetiracetam (LEV), PHT, PHT-LEV, lacosamide (LCM), and valproate (VPA). All interventions except VPA significantly reduced the rate of early PTE in TBI patients compared with the placebo. Seven studies reported the impact of four treatments (PHT + PB, LEV, PHT, VPA) on late seizures and showed a significant reduction in the incidence of late seizures in patients with TBI compared with placebo. The impact of PHT, LEV, and VPA on mortality was reported in nine studies. PHT had no impact on mortality, but patients treated with both LEV and VPA had higher mortality than those treated with placebo. The treatment-related adverse effects of LEV, PHT, and LCM were reported in five studies. LEV and PHT had higher treatment-related adverse effects incidence than placebo, while LCM had no effect on treatment related-adverse effects. Conclusion: LEV and PHT prevented early and late PTE. PHT also reduced the mortality rate in patients with TBI. Both LEV and PHT had higher treatment-related adverse effects compared with placebo. However, LEV had a slightly lower incidence of treatment-related adverse effects when compared with PHT. Compared with PHT, LEV did not reduce the length of hospital stay but shortened the length of ICU stays. Therefore, based on the findings of this meta-analysis, we speculate that LEV is the best treatment option for TBI patients. However, further high-quality randomized controlled trials are required to confirm these findings.
Collapse
Affiliation(s)
- Xianhao Huo
- Neurosurgery Department, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Cerebrocranial Disease, Ningxia Medical University, Yinchuan, China
- Clinical Medical College, Ningxia Medical University, Yinchuan, China
| | - Xingguo Xu
- Neurosurgery Department, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Mei Li
- Ningxia Key Laboratory of Cerebrocranial Disease, Ningxia Medical University, Yinchuan, China
- Clinical Medical College, Ningxia Medical University, Yinchuan, China
| | - Lifei Xiao
- Neurosurgery Department, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Cerebrocranial Disease, Ningxia Medical University, Yinchuan, China
- Clinical Medical College, Ningxia Medical University, Yinchuan, China
| | - Yangyang Wang
- Ningxia Key Laboratory of Cerebrocranial Disease, Ningxia Medical University, Yinchuan, China
- Clinical Medical College, Ningxia Medical University, Yinchuan, China
- Neurosurgery Department, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Wenchao Li
- Ningxia Key Laboratory of Cerebrocranial Disease, Ningxia Medical University, Yinchuan, China
- Clinical Medical College, Ningxia Medical University, Yinchuan, China
- Neurosurgery Department, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Chaofan Wang
- Neurosurgery Department, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Cerebrocranial Disease, Ningxia Medical University, Yinchuan, China
- Clinical Medical College, Ningxia Medical University, Yinchuan, China
| | - Tao Sun
- Neurosurgery Department, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Cerebrocranial Disease, Ningxia Medical University, Yinchuan, China
- *Correspondence: Tao Sun,
| |
Collapse
|
26
|
Fernández‐Arjona MDM, León‐Rodríguez A, Grondona JM, López‐Ávalos MD. Long-term priming of hypothalamic microglia is associated with energy balance disturbances under diet-induced obesity. Glia 2022; 70:1734-1761. [PMID: 35603807 PMCID: PMC9540536 DOI: 10.1002/glia.24217] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/13/2022] [Accepted: 05/06/2022] [Indexed: 12/16/2022]
Abstract
Exposure of microglia to an inflammatory environment may lead to their priming and exacerbated response to future inflammatory stimuli. Here we aimed to explore hypothalamic microglia priming and its consequences on energy balance regulation. A model of intracerebroventricular administration of neuraminidase (NA, which is present in various pathogens such as influenza virus) was used to induce acute neuroinflammation. Evidences of primed microglia were observed 3 months after NA injection, namely (1) a heightened response of microglia located in the hypothalamic arcuate nucleus after an in vivo inflammatory challenge (high fat diet [HFD] feeding for 10 days), and (2) an enhanced response of microglia isolated from NA-treated mice and challenged in vitro to LPS. On the other hand, the consequences of a previous NA-induced neuroinflammation were further evaluated in an alternative inflammatory and hypercaloric scenario, such as the obesity generated by continued HDF feeding. Compared with sham-injected mice, NA-treated mice showed increased food intake and, surprisingly, reduced body weight. Besides, NA-treated mice had enhanced microgliosis (evidenced by increased number and reactive morphology of microglia) and a reduced population of POMC neurons in the basal hypothalamus. Thus, a single acute neuroinflammatory event may elicit a sustained state of priming in microglial cells, and in particular those located in the hypothalamus, with consequences in hypothalamic cytoarchitecture and its regulatory function upon nutritional challenges.
Collapse
Affiliation(s)
- María del Mar Fernández‐Arjona
- Instituto de Investigación Biomédica de Málaga‐IBIMAMálagaSpain
- Grupo de investigación en Neuropsicofarmacología, Laboratorio de Medicina RegenerativaHospital Regional Universitario de MálagaMálagaSpain
| | - Ana León‐Rodríguez
- Instituto de Investigación Biomédica de Málaga‐IBIMAMálagaSpain
- Departamento de Biología Celular, Genética y Fisiología, Facultad de CienciasUniversidad de Málaga, Campus de TeatinosMálagaSpain
| | - Jesús M. Grondona
- Instituto de Investigación Biomédica de Málaga‐IBIMAMálagaSpain
- Departamento de Biología Celular, Genética y Fisiología, Facultad de CienciasUniversidad de Málaga, Campus de TeatinosMálagaSpain
| | - María D. López‐Ávalos
- Instituto de Investigación Biomédica de Málaga‐IBIMAMálagaSpain
- Departamento de Biología Celular, Genética y Fisiología, Facultad de CienciasUniversidad de Málaga, Campus de TeatinosMálagaSpain
| |
Collapse
|
27
|
Levison SW, Rocha-Ferreira E, Kim BH, Hagberg H, Fleiss B, Gressens P, Dobrowolski R. Mechanisms of Tertiary Neurodegeneration after Neonatal Hypoxic-Ischemic Brain Damage. PEDIATRIC MEDICINE (HONG KONG, CHINA) 2022; 5:28. [PMID: 37601279 PMCID: PMC10438849 DOI: 10.21037/pm-20-104] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Neonatal encephalopathy linked to hypoxia-ischemia (H-I) which is regarded as the most important neurological problem of the newborn, can lead to a spectrum of adverse neurodevelopmental outcomes such as cerebral palsy, epilepsy, hyperactivity, cognitive impairment and learning difficulties. There have been numerous reviews that have focused on the epidemiology, diagnosis and treatment of neonatal H-I; however, a topic that is less often considered is the extent to which the injury might worsen over time, which is the focus of this review. Similarly, there have been numerous reviews that have focused on mechanisms that contribute to the acute or subacute injury; however, there is a tertiary phase of recovery that can be defined by cellular and molecular changes that occur many weeks and months after brain injury and this topic has not been the focus of any review for over a decade. Therefore, in this article we review both the clinical and pre-clinical data that show that tertiary neurodegeneration is a significant contributor to the final outcome, especially after mild to moderate injuries. We discuss the contributing roles of apoptosis, necroptosis, autophagy, protein homeostasis, inflammation, microgliosis and astrogliosis. We also review the limited number of studies that have shown that significant neuroprotection and preservation of neurological function can be achieved administering drugs during the period of tertiary neurodegeneration. As the tertiary phase of neurodegeneration is a stage when interventions are eminently feasible, it is our hope that this review will stimulate a new focus on this stage of recovery towards the goal of producing new treatment options for neonatal hypoxic-ischemic encephalopathy.
Collapse
Affiliation(s)
- Steven W. Levison
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers University, New Jersey Medical School, Cancer Center, 205 South Orange Avenue, Newark, NJ 07103, USA
| | - Eridan Rocha-Ferreira
- Centre of Perinatal Medicine & Health, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Brian H. Kim
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers University, New Jersey Medical School, Cancer Center, 205 South Orange Avenue, Newark, NJ 07103, USA
| | - Henrik Hagberg
- Centre of Perinatal Medicine & Health, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Centre for the Developing Brain, Division of Imaging Sciences and Biomedical Engineering, King’s College London, King’s Health Partners, St. Thomas’ Hospital, London, SE1 7EH, UK
| | - Bobbi Fleiss
- Centre for the Developing Brain, Division of Imaging Sciences and Biomedical Engineering, King’s College London, King’s Health Partners, St. Thomas’ Hospital, London, SE1 7EH, UK
- Université de Paris, NeuroDiderot, Inserm, F-75019 Paris, France
- School of Health and Biomedical Sciences, RMIT University, Bundoora, 3083, VIC, Australia
| | - Pierre Gressens
- Centre for the Developing Brain, Division of Imaging Sciences and Biomedical Engineering, King’s College London, King’s Health Partners, St. Thomas’ Hospital, London, SE1 7EH, UK
- Université de Paris, NeuroDiderot, Inserm, F-75019 Paris, France
| | | |
Collapse
|
28
|
Lu X, Xue Z, Qian Y, Wei S, Qiao Y, Zhang W, Lu H. Changes in intestinal microflora and its metabolites underlie the cognitive impairment in preterm rats. Front Cell Infect Microbiol 2022; 12:945851. [PMID: 36061856 PMCID: PMC9437323 DOI: 10.3389/fcimb.2022.945851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 07/29/2022] [Indexed: 11/25/2022] Open
Abstract
Background The brain development of preterm infants is easily affected by various adverse extrauterine factors and complications, resulting in abnormal neurological and cognitive development. Recent studies have found that there is a significant correlation between intestinal microbial changes and cognitive behavior. Nevertheless, the correlation between the cognitive impairment and abnormal changes of intestinal microflora in the preterm newborn has been rarely elucidated. Aim To analyze the differences of fecal intestinal flora, short chain fatty acids (SCFAs) and microbiota-gut-brain axis (MGBA)-related serum factors between preterm birth with and without cognitive impairment. Methods Healthy female rats (body weight 410 ± 40 g) of 16-17 days of gestation were selected for the establishment of preterm cognitive impairment model and screened by Morris water maze navigation experiments. The pathological change of rat hippocampus was confirmed by HE staining. The abundance of fecal intestinal microflora was determined by 16sRNA sequencing, while the contents of fecal SCFAs were examined by gas chromatography. Results Compared with the control group, the cognitive impairment group had decreased abundance and diversity of intestinal microflora and increased abundance of Proteobacteria at the level of phylum. While the abundances of Alistipes, Bacteroides, Prevotella, and Lactobacillus decreased significantly at the level of order, family, and genus, the abundances of Staphylococcaceae, Enterococci, Psychrobacter, and Oligella increased significantly. Moreover, the levels of total SCFAs and acetic acid in the disease group were significantly lower. The fecal abundance of acetic acid was positively correlated with that of Lactobacillaceae or Peptostreptococcaceae, and negatively correlated with that of Aerococcaceae, and Alcaligenaceae in disease rats. Furthermore, cognitive impairment caused significantly decreased levels of 5-HT, GABA, and BDNF, and increased levels of GR, CRH, IL-6, and TNF-α in rat blood. Conclusion Alterations in intestinal microflora structure and the abundances of SCFAs contributed substantially to the cognitive impairment in preterm rats, which was associated with significant changes in MGBA-related soluble factors.
Collapse
Affiliation(s)
- Xiang Lu
- Pediatrics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Zhengyang Xue
- Pediatrics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yu Qian
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Shanjie Wei
- Pediatrics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yu Qiao
- Pediatrics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Wen Zhang
- Pediatrics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- School of Medicine, Jiangsu University, Zhenjiang, China
- *Correspondence: Wen Zhang, ; Hongyan Lu,
| | - Hongyan Lu
- Pediatrics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- *Correspondence: Wen Zhang, ; Hongyan Lu,
| |
Collapse
|
29
|
Fernández-Arjona MDM, León-Rodríguez A, Grondona JM, López-Ávalos MD. Microbial neuraminidase induces TLR4-dependent long-term immune priming in the brain. Front Cell Neurosci 2022; 16:945229. [PMID: 35966200 PMCID: PMC9366060 DOI: 10.3389/fncel.2022.945229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/06/2022] [Indexed: 11/13/2022] Open
Abstract
Innate immune memory explains the plasticity of immune responses after repeated immune stimulation, leading to either enhanced or suppressed immune responses. This process has been extensively reported in peripheral immune cells and also, although modestly, in the brain. Here we explored two relevant aspects of brain immune priming: its persistence over time and its dependence on TLR receptors. For this purpose, we used an experimental paradigm consisting in applying two inflammatory stimuli three months apart. Wild type, toll-like receptor (TLR) 4 and TLR2 mutant strains were used. The priming stimulus was the intracerebroventricular injection of neuraminidase (an enzyme that is present in various pathogens able to provoke brain infections), which triggers an acute inflammatory process in the brain. The second stimulus was the intraperitoneal injection of lipopolysaccharide (a TLR4 ligand) or Pam3CSK4 (a TLR2 ligand). One day after the second inflammatory challenge the immune response in the brain was examined. In wild type mice, microglial and astroglial density, as well as the expression of 4 out of 5 pro-inflammatory genes studied (TNFα, IL1β, Gal-3, and NLRP3), were increased in mice that received the double stimulus compared to those exposed only to the second one, which were initially injected with saline instead of neuraminidase. Such enhanced response suggests immune training in the brain, which lasts at least 3 months. On the other hand, TLR2 mutants under the same experimental design displayed an enhanced immune response quite similar to that of wild type mice. However, in TLR4 mutant mice the response after the second immune challenge was largely dampened, indicating the pivotal role of this receptor in the establishment of immune priming. Our results demonstrate that neuraminidase-induced inflammation primes an enhanced immune response in the brain to a subsequent immune challenge, immune training that endures and that is largely dependent on TLR4 receptor.
Collapse
Affiliation(s)
- María del Mar Fernández-Arjona
- Laboratorio de Medicina Regenerativa, Grupo de investigación en Neuropsicofarmacología, Hospital Regional Universitario de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain
| | - Ana León-Rodríguez
- Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain
- Laboratorio de Fisiología Animal, Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
| | - Jesús M. Grondona
- Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain
- Laboratorio de Fisiología Animal, Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
| | - María Dolores López-Ávalos
- Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain
- Laboratorio de Fisiología Animal, Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- *Correspondence: María Dolores López-Ávalos
| |
Collapse
|
30
|
Anxiety-like behavior and microglial activation in the amygdala after acute neuroinflammation induced by microbial neuraminidase. Sci Rep 2022; 12:11581. [PMID: 35803999 PMCID: PMC9270343 DOI: 10.1038/s41598-022-15617-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 06/27/2022] [Indexed: 11/08/2022] Open
Abstract
Short-term behavioral alterations are associated with infection and aid the recovery from sickness. However, concerns have raised that sustained behavioral disturbances after acute neuroinflammation could relate to neurological diseases in the long run. We aimed to explore medium- and long-term behavioral disturbances after acute neuroinflammation in rats, using a model based on the intracerebroventricular administration of the enzyme neuraminidase (NA), which is part of some pathogenic bacteria and viruses. Neurological and behavioral assessments were performed 2 and 10 weeks after the injection of NA, and neuroinflammation was evaluated by gene expression and histology. No alterations were observed regarding basic neurological functions or locomotor capacity in NA-injected rats. However, they showed a reduction in unsupported rearing, and increased grooming and freezing behaviors, which indicate anxiety-like behavior. A principal component analysis including a larger set of parameters further supported such anxiety-like behavior. The anxiety profile was observed 2 weeks after NA-injection, but not after 10 weeks. Concomitantly, the amygdala presented increased number of microglial cells showing a morphologic bias towards an activated state. A similar but subtler tendency was observed in hypothalamic microglia located in the paraventricular nucleus. Also, in the hypothalamus the pattern recognition receptor toll-like receptor 4 (TLR4) was slightly overexpressed 2 weeks after NA injection. These results demonstrate that NA-induced neuroinflammation provokes anxiety-like behavior in the medium term, which disappears with time. Concurrent microgliosis in the amygdala could explain such behavior. Further experiments should aim to explore subtle but long-lasting alterations observed 10 weeks after NA injection, both in amygdala and hypothalamus, as well as mild behavioral changes.
Collapse
|
31
|
Tapp ZM, Cornelius S, Oberster A, Kumar JE, Atluri R, Witcher KG, Oliver B, Bray C, Velasquez J, Zhao F, Peng J, Sheridan J, Askwith C, Godbout JP, Kokiko-Cochran ON. Sleep fragmentation engages stress-responsive circuitry, enhances inflammation and compromises hippocampal function following traumatic brain injury. Exp Neurol 2022; 353:114058. [PMID: 35358498 PMCID: PMC9068267 DOI: 10.1016/j.expneurol.2022.114058] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 03/04/2022] [Accepted: 03/24/2022] [Indexed: 02/08/2023]
Abstract
Traumatic brain injury (TBI) impairs the ability to restore homeostasis in response to stress, indicating hypothalamic-pituitary-adrenal (HPA)-axis dysfunction. Many stressors result in sleep disturbances, thus mechanical sleep fragmentation (SF) provides a physiologically relevant approach to study the effects of stress after injury. We hypothesize SF stress engages the dysregulated HPA-axis after TBI to exacerbate post-injury neuroinflammation and compromise recovery. To test this, male and female mice were given moderate lateral fluid percussion TBI or sham-injury and left undisturbed or exposed to daily, transient SF for 7- or 30-days post-injury (DPI). Post-TBI SF increases cortical expression of interferon- and stress-associated genes characterized by inhibition of the upstream regulator NR3C1 that encodes glucocorticoid receptor (GR). Moreover, post-TBI SF increases neuronal activity in the hippocampus, a key intersection of the stress-immune axes. By 30 DPI, TBI SF enhances cortical microgliosis and increases expression of pro-inflammatory glial signaling genes characterized by persistent inhibition of the NR3C1 upstream regulator. Within the hippocampus, post-TBI SF exaggerates microgliosis and decreases CA1 neuronal activity. Downstream of the hippocampus, post-injury SF suppresses neuronal activity in the hypothalamic paraventricular nucleus indicating decreased HPA-axis reactivity. Direct application of GR agonist, dexamethasone, to the CA1 at 30 DPI increases GR activity in TBI animals, but not sham animals, indicating differential GR-mediated hippocampal action. Electrophysiological assessment revealed TBI and SF induces deficits in Schaffer collateral long-term potentiation associated with impaired acquisition of trace fear conditioning, reflecting dorsal hippocampal-dependent cognitive deficits. Together these data demonstrate that post-injury SF engages the dysfunctional post-injury HPA-axis, enhances inflammation, and compromises hippocampal function. Therefore, external stressors that disrupt sleep have an integral role in mediating outcome after brain injury.
Collapse
Affiliation(s)
- Zoe M Tapp
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, Columbus, OH 43210, USA.
| | - Sydney Cornelius
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, Columbus, OH 43210, USA.
| | - Alexa Oberster
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, Columbus, OH 43210, USA
| | - Julia E Kumar
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, Columbus, OH 43210, USA.
| | - Ravitej Atluri
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, Columbus, OH 43210, USA.
| | - Kristina G Witcher
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, Columbus, OH 43210, USA.
| | - Braedan Oliver
- Institute for Behavioral Medicine Research, Neurological Institute, The Ohio State University, 460 Medical Center Drive, Columbus, OH 43210, USA.
| | - Chelsea Bray
- Institute for Behavioral Medicine Research, Neurological Institute, The Ohio State University, 460 Medical Center Drive, Columbus, OH 43210, USA.
| | - John Velasquez
- Institute for Behavioral Medicine Research, Neurological Institute, The Ohio State University, 460 Medical Center Drive, Columbus, OH 43210, USA.
| | - Fangli Zhao
- Institute for Behavioral Medicine Research, Neurological Institute, The Ohio State University, 460 Medical Center Drive, Columbus, OH 43210, USA.
| | - Juan Peng
- Center for Biostatistics, The Ohio State University, 320-55 Lincoln Tower, 1800 Cannon Drive, Columbus, OH 43210, USA.
| | - John Sheridan
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, Columbus, OH 43210, USA; Institute for Behavioral Medicine Research, Neurological Institute, The Ohio State University, 460 Medical Center Drive, Columbus, OH 43210, USA; Division of Biosciences, College of Dentistry, The Ohio State University, 305 W. 12(th) Ave, Columbus, OH 43210, USA.
| | - Candice Askwith
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, Columbus, OH 43210, USA.
| | - Jonathan P Godbout
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, Columbus, OH 43210, USA; Institute for Behavioral Medicine Research, Neurological Institute, The Ohio State University, 460 Medical Center Drive, Columbus, OH 43210, USA.
| | - Olga N Kokiko-Cochran
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, Columbus, OH 43210, USA; Institute for Behavioral Medicine Research, Neurological Institute, The Ohio State University, 460 Medical Center Drive, Columbus, OH 43210, USA.
| |
Collapse
|
32
|
Wang H, Huang Q, Zhang Z, Ji J, Sun T, Wang D. Transient post-operative overexpression of CXCR2 on monocytes of traumatic brain injury patients drives monocyte chemotaxis toward cerebrospinal fluid and enhances monocyte-mediated immunogenic cell death of neurons in vitro. J Neuroinflammation 2022; 19:171. [PMID: 35768823 PMCID: PMC9245242 DOI: 10.1186/s12974-022-02535-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 06/20/2022] [Indexed: 11/23/2022] Open
Abstract
Background After traumatic brain injury (TBI), peripheral monocytes infiltrate into the central nervous system due to disruption of the blood–brain barrier, and play an important role in neuroinflammation. However, the mechanisms regulating the movement and function of peripheral monocytes after TBI have not been fully investigated. Methods TBI patients who underwent surgery at our hospital were recruited. CXCR2 expression in CD14+ monocytes from peripheral blood and cerebrospinal fluid (CSF) of TBI patients around surgery was analyzed by flow cytometry and compared with that of patients who suffered TBI 2–24 months prior and underwent cranioplasty. In vitro, serum or CSF from TBI/non-TBI patients were used to treat peripheral monocytes isolated from healthy volunteers to evaluate their effect on CXCR2 expression. Transwell experiments were performed to analyze the role of CXCR2 in monocyte chemotaxis toward the CSF. The role of CXCR2 in monocyte-mediated immunogenic cell death (ICD) of nerve cells was explored in an indirect co-culture system. Results Transient CXCR2 upregulation in monocytes from the peripheral blood and CSF of TBI patients was detected soon after surgery and was associated with unfavorable outcomes. TBI serum and CSF promoted CXCR2 expression in monocytes, and dexamethasone reversed this effect. Peripheral monocytes from TBI patients showed enhanced chemotaxis toward the CSF and increased inflammatory cytokine secretion. The CXCR2 antagonist SB225002 decreased monocyte chemotaxis toward TBI CSF, and lowered pro-inflammatory cytokine secretion in monocytes treated with TBI serum. SB225002 also relieved ICD in nerve cells co-cultured with TBI serum-treated monocytes. Conclusions CXCR2 is transiently overexpressed in the peripheral monocytes of TBI patients post-surgery, and drives peripheral monocyte chemotaxis toward CSF and monocyte-mediated ICD of nerve cells. Therefore, CXCR2 may be a target for monocyte-based therapies for TBI. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02535-6.
Collapse
Affiliation(s)
- Huayang Wang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, China
| | - Qibing Huang
- Department of Neurosurgical Intensive Care Unit, Qilu Hospital of Shandong University, Jinan, China
| | - Zhijie Zhang
- Department of Ultrasound, Shandong Maternal and Child Health Hospital, Jinan, China
| | - Jian Ji
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, China
| | - Tao Sun
- Department of Neurosurgery, Qilu Hospital of Shandong University, # 107 Wenhuaxi Road, Jinan, Shandong, China
| | - Donghai Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University, # 107 Wenhuaxi Road, Jinan, Shandong, China.
| |
Collapse
|
33
|
Ganne A, Balasubramaniam M, Griffin WST, Shmookler Reis RJ, Ayyadevara S. Glial Fibrillary Acidic Protein: A Biomarker and Drug Target for Alzheimer’s Disease. Pharmaceutics 2022; 14:pharmaceutics14071354. [PMID: 35890250 PMCID: PMC9322874 DOI: 10.3390/pharmaceutics14071354] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 02/05/2023] Open
Abstract
Glial fibrillary acidic protein (GFAP) is an intermediate filament structural protein involved in cytoskeleton assembly and integrity, expressed in high abundance in activated glial cells. GFAP is neuroprotective, as knockout mice are hypersensitive to traumatic brain injury. GFAP in cerebrospinal fluid is a biomarker of Alzheimer’s disease (AD), dementia with Lewy bodies, and frontotemporal dementia (FTD). Here, we present novel evidence that GFAP is markedly overexpressed and differentially phosphorylated in AD hippocampus, especially in AD with the apolipoprotein E [ε4, ε4] genotype, relative to age-matched controls (AMCs). Kinases that phosphorylate GFAP are upregulated in AD relative to AMC. A knockdown of these kinases in SH-SY5Y-APPSw human neuroblastoma cells reduced amyloid accrual and lowered protein aggregation and associated behavioral traits in C. elegans models of polyglutamine aggregation (as observed in Huntington’s disease) and of Alzheimer’s-like amyloid formation. In silico screening of the ChemBridge structural library identified a small molecule, MSR1, with stable and specific binding to GFAP. Both MSR1 exposure and GF AP-specific RNAi knockdown reduce aggregation with remarkably high concordance of aggregate proteins depleted. These data imply that GFAP and its phosphorylation play key roles in neuropathic aggregate accrual and provide valuable new biomarkers, as well as novel therapeutic targets to alleviate, delay, or prevent AD.
Collapse
Affiliation(s)
- Akshatha Ganne
- Central Arkansas Veterans Healthcare Service, Little Rock, AR 72205, USA; (A.G.); (M.B.); (W.S.T.G.)
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | | | - W. Sue T. Griffin
- Central Arkansas Veterans Healthcare Service, Little Rock, AR 72205, USA; (A.G.); (M.B.); (W.S.T.G.)
- BioInformatics Program, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Robert J. Shmookler Reis
- Central Arkansas Veterans Healthcare Service, Little Rock, AR 72205, USA; (A.G.); (M.B.); (W.S.T.G.)
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- BioInformatics Program, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Biochemistry & Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Correspondence: (R.J.S.R.); (S.A.); Tel.: +1-501-526-5820 (R.J.S.R.); +1-501-526-7282 (S.A.)
| | - Srinivas Ayyadevara
- Central Arkansas Veterans Healthcare Service, Little Rock, AR 72205, USA; (A.G.); (M.B.); (W.S.T.G.)
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- BioInformatics Program, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Correspondence: (R.J.S.R.); (S.A.); Tel.: +1-501-526-5820 (R.J.S.R.); +1-501-526-7282 (S.A.)
| |
Collapse
|
34
|
Bray CE, Witcher KG, Adekunle-Adegbite D, Ouvina M, Witzel M, Hans E, Tapp ZM, Packer J, Goodman E, Zhao F, Chunchai T, O'Neil S, Chattipakorn SC, Sheridan J, Kokiko-Cochran ON, Askwith C, Godbout JP. Chronic Cortical Inflammation, Cognitive Impairment, and Immune Reactivity Associated with Diffuse Brain Injury Are Ameliorated by Forced Turnover of Microglia. J Neurosci 2022; 42:4215-4228. [PMID: 35440489 PMCID: PMC9121837 DOI: 10.1523/jneurosci.1910-21.2022] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 03/04/2022] [Accepted: 03/07/2022] [Indexed: 02/08/2023] Open
Abstract
Traumatic brain injury (TBI) is associated with an increased risk of cognitive, psychiatric, and neurodegenerative complications that may develop after injury. Increased microglial reactivity following TBI may underlie chronic neuroinflammation, neuropathology, and exaggerated responses to immune challenges. Therefore, the goal of this study was to force turnover of trauma-associated microglia that develop after diffuse TBI and determine whether this alleviated chronic inflammation, improved functional recovery and attenuated reduced immune reactivity to lipopolysaccharide (LPS) challenge. Male mice received a midline fluid percussion injury (mFPI) and 7 d later were subjected to a forced microglia turnover paradigm using CSF1R antagonism (PLX5622). At 30 d postinjury (dpi), cortical gene expression, dendritic complexity, myelin content, neuronal connectivity, cognition, and immune reactivity were assessed. Myriad neuropathology-related genes were increased 30 dpi in the cortex, and 90% of these gene changes were reversed by microglial turnover. Reduced neuronal connectivity was evident 30 dpi and these deficits were attenuated by microglial turnover. TBI-associated dendritic remodeling and myelin alterations, however, remained 30 dpi independent of microglial turnover. In assessments of functional recovery, increased depressive-like behavior, and cognitive impairment 30 dpi were ameliorated by microglia turnover. To investigate microglial priming and reactivity 30 dpi, mice were injected intraperitoneally with LPS. This immune challenge caused prolonged lethargy, sickness behavior, and microglial reactivity in the TBI mice. These extended complications with LPS in TBI mice were prevented by microglia turnover. Collectively, microglial turnover 7 dpi alleviated behavioral and cognitive impairments associated with microglial priming and immune reactivity 30 dpi.SIGNIFICANCE STATEMENT A striking feature of traumatic brain injury (TBI), even mild injuries, is that over 70% of individuals have long-term neuropsychiatric complications. Chronic inflammatory processes are implicated in the pathology of these complications and these issues can be exaggerated by immune challenge. Therefore, our goal was to force the turnover of microglia 7 d after TBI. This subacute 7 d postinjury (dpi) time point is a critical transitional period in the shift toward chronic inflammatory processes and microglia priming. This forced microglia turnover intervention in mice attenuated the deficits in behavior and cognition 30 dpi. Moreover, microglia priming and immune reactivity after TBI were also reduced with microglia turnover. Therefore, microglia represent therapeutic targets after TBI to reduce persistent neuroinflammation and improve recovery.
Collapse
Affiliation(s)
- Chelsea E Bray
- Department of Neuroscience, The Ohio State University, Columbus, Ohio 43210
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio 43210
| | - Kristina G Witcher
- Department of Neuroscience, The Ohio State University, Columbus, Ohio 43210
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio 43210
| | | | - Michelle Ouvina
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio 43210
| | - Mollie Witzel
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio 43210
| | - Emma Hans
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio 43210
| | - Zoe M Tapp
- Department of Neuroscience, The Ohio State University, Columbus, Ohio 43210
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio 43210
| | - Jonathan Packer
- Department of Neuroscience, The Ohio State University, Columbus, Ohio 43210
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio 43210
| | - Ethan Goodman
- Department of Neuroscience, The Ohio State University, Columbus, Ohio 43210
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio 43210
| | - Fangli Zhao
- Department of Neuroscience, The Ohio State University, Columbus, Ohio 43210
| | - Titikorn Chunchai
- Neurophysiology unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Shane O'Neil
- Department of Neuroscience, The Ohio State University, Columbus, Ohio 43210
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio 43210
| | - Siriporn C Chattipakorn
- Neurophysiology unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - John Sheridan
- Department of Neuroscience, The Ohio State University, Columbus, Ohio 43210
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio 43210
| | - Olga N Kokiko-Cochran
- Department of Neuroscience, The Ohio State University, Columbus, Ohio 43210
- Chronic Brain Injury Program, The Ohio State University, Columbus, Ohio 43210
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio 43210
| | - Candice Askwith
- Department of Neuroscience, The Ohio State University, Columbus, Ohio 43210
| | - Jonathan P Godbout
- Department of Neuroscience, The Ohio State University, Columbus, Ohio 43210
- Chronic Brain Injury Program, The Ohio State University, Columbus, Ohio 43210
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio 43210
| |
Collapse
|
35
|
Salmani H, Hosseini M, Nabi MM, Samadi-Noshahr Z, Baghcheghi Y, Sadeghi M. Exacerbated immune response of the brain to peripheral immune challenge in post-septic mice. Brain Res Bull 2022; 185:74-85. [PMID: 35523357 DOI: 10.1016/j.brainresbull.2022.04.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 04/24/2022] [Accepted: 04/26/2022] [Indexed: 11/02/2022]
Abstract
BACKGROUND Mounting evidence indicates that sepsis can induce long-lasting brain dysfunction. Recently, it has been proposed that the brain may become more sensitive to systemic inflammation if microglial cells are already primed. Microglial priming has been demonstrated in aging, traumatic brain injury, and neurodegenerative diseases. There is evidence suggesting that systemic inflammation may also prime microglia. This study aimed to investigate the brain's response to a second immune challenge in sepsis survivors and the possible role of microglial priming. METHODS Adult BALB/c mice were intraperitoneally (ip) injected with 5 mg/kg lipopolysaccharide (LPS) for sepsis induction. One month later, mice received a second immune challenge (LPS, 0.33 mg/kg). A cohort of mice was sacrificed 2 h post-LPS injection to measure inflammatory mediators mRNA expression. The second cohort of mice was tested on a battery of behavioral tests and then sacrificed, and brain tissues were removed for biochemical analyses. RESULTS Results showed that in septic mice, secondary LPS challenge induced heightened neuroinflammation compared to the control mice, as evident by a significant increase of IL-1β, TNF-α, and iNOS mRNA expression. In the immunochallenged septic mice, the anti-inflammatory cytokine IL-10 expression was also significantly increased compared to the control mice. Sepsis induction significantly disrupted the recognition ability in the novel object recognition, but the second immune challenge had no significant effect. However, immunochallenged septic mice exhibited more anxiety-like behavior in the marble burying task and intensive depressive-like behavior in the forced swim test. Additionally, the second immune challenge reduced arginase-1 levels in septic but not control mice. On the other hand, CIITA levels were increased more significantly in the LPS injected control mice compared to septic mice. Neither sepsis nor the second immune challenge significantly affected inhibitory avoidance behavior and Aβ1-42 levels in brain tissue. CONCLUSION Our finding suggests that low-grade immune challenge can induce exacerbated behavioral change and exaggerated inflammatory response in the brain of post-septic mice.
Collapse
Affiliation(s)
- Hossein Salmani
- Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran.
| | - Mahmoud Hosseini
- Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mohammad Mahdi Nabi
- Mashhad Branch, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Mashhad, Iran; Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran.
| | | | - Yousef Baghcheghi
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran.
| | - Mostafa Sadeghi
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran.
| |
Collapse
|
36
|
Giordano KR, Law LM, Henderson J, Rowe RK, Lifshitz J. Time Course of Remote Neuropathology Following Diffuse Traumatic Brain Injury in the Male Rat. Exp Neurobiol 2022; 31:105-115. [PMID: 35673999 PMCID: PMC9194637 DOI: 10.5607/en21027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 12/15/2021] [Accepted: 04/12/2022] [Indexed: 11/19/2022] Open
Abstract
Traumatic brain injury (TBI) can affect different regions throughout the brain. Regions near the site of impact are the most vulnerable to injury. However, damage to distal regions occurs. We investigated progressive neuropathology in the dorsal hippocampus (near the impact) and cerebellum (distal to the impact) after diffuse TBI. Adult male rats were subjected to midline fluid percussion injury or sham injury. Brain tissue was stained by the amino cupric silver stain. Neuropathology was quantified in sub-regions of the dorsal hippocampus at 1, 7, and 28 days post-injury (DPI) and coronal cerebellar sections at 1, 2, and 7 DPI. The highest observed neuropathology in the dentate gyrus occurred at 7 DPI which attenuated by 28 DPI, whereas the highest observed neuropathology was at 1 DPI in the CA3 region. There was no significant neuropathology in the CA1 region at any time point. Neuropathology was increased at 7 DPI in the cerebellum compared to shams and stripes of pathology were observed in the molecular layer perpendicular to the cerebellar cortical surface. Together these data show that diffuse TBI can result in neuropathology across the brain. By describing the time course of pathology in response to TBI, it is possible to build the temporal profile of disease progression.
Collapse
Affiliation(s)
- Katherine R Giordano
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ 85013, USA.,Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ 85004, USA.,Phoenix Veterans Affairs Health Care System, Phoenix, AZ 85012, USA
| | - L Matthew Law
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ 85013, USA.,Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ 85004, USA.,Phoenix Veterans Affairs Health Care System, Phoenix, AZ 85012, USA
| | - Jordan Henderson
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ 85013, USA.,Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ 85004, USA
| | - Rachel K Rowe
- Department of Integrative Physiology, University of Colorado, Boulder, CO 80309, USA
| | - Jonathan Lifshitz
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ 85013, USA.,Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ 85004, USA.,Phoenix Veterans Affairs Health Care System, Phoenix, AZ 85012, USA
| |
Collapse
|
37
|
Role of Inflammation in Traumatic Brain Injury-Associated Risk for Neuropsychiatric Disorders: State of the Evidence and Where Do We Go From Here. Biol Psychiatry 2022; 91:438-448. [PMID: 34955170 DOI: 10.1016/j.biopsych.2021.11.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 10/01/2021] [Accepted: 11/02/2021] [Indexed: 02/06/2023]
Abstract
In the past decade, there has been an increasing awareness that traumatic brain injury (TBI) and concussion substantially increase the risk for developing psychiatric disorders. Even mild TBI increases the risk for depression and anxiety disorders such as posttraumatic stress disorder by two- to threefold, predisposing patients to further functional impairment. This strong epidemiological link supports examination of potential mechanisms driving neuropsychiatric symptom development after TBI. One potential mechanism for increased neuropsychiatric symptoms after TBI is via inflammatory processes, as central nervous system inflammation can last years after initial injury. There is emerging preliminary evidence that TBI patients with posttraumatic stress disorder or depression exhibit increased central and peripheral inflammatory markers compared with TBI patients without these comorbidities. Growing evidence has demonstrated that immune signaling in animals plays an integral role in depressive- and anxiety-like behaviors after severe stress or brain injury. In this review, we will 1) discuss current evidence for chronic inflammation after TBI in the development of neuropsychiatric symptoms, 2) highlight potential microglial activation and cytokine signaling contributions, and 3) discuss potential promise and pitfalls for immune-targeted interventions and biomarker strategies to identify and treat TBI patients with immune-related neuropsychiatric symptoms.
Collapse
|
38
|
Srinivasan G, Brafman DA. The Emergence of Model Systems to Investigate the Link Between Traumatic Brain Injury and Alzheimer's Disease. Front Aging Neurosci 2022; 13:813544. [PMID: 35211003 PMCID: PMC8862182 DOI: 10.3389/fnagi.2021.813544] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 12/20/2021] [Indexed: 12/12/2022] Open
Abstract
Numerous epidemiological studies have demonstrated that individuals who have sustained a traumatic brain injury (TBI) have an elevated risk for developing Alzheimer's disease and Alzheimer's-related dementias (AD/ADRD). Despite these connections, the underlying mechanisms by which TBI induces AD-related pathology, neuronal dysfunction, and cognitive decline have yet to be elucidated. In this review, we will discuss the various in vivo and in vitro models that are being employed to provide more definite mechanistic relationships between TBI-induced mechanical injury and AD-related phenotypes. In particular, we will highlight the strengths and weaknesses of each of these model systems as it relates to advancing the understanding of the mechanisms that lead to TBI-induced AD onset and progression as well as providing platforms to evaluate potential therapies. Finally, we will discuss how emerging methods including the use of human induced pluripotent stem cell (hiPSC)-derived cultures and genome engineering technologies can be employed to generate better models of TBI-induced AD.
Collapse
Affiliation(s)
| | - David A. Brafman
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, United States
| |
Collapse
|
39
|
Innate immune stimulation prevents the development of anxiety-like behaviors in chronically stressed mice. Neuropharmacology 2022; 207:108950. [PMID: 35074304 DOI: 10.1016/j.neuropharm.2022.108950] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 12/30/2021] [Accepted: 01/12/2022] [Indexed: 12/19/2022]
Abstract
Anxiety is a common psychological disease which can induce severe social burdens. Searching methods that prevent the onset of anxiety is of great significance for ameliorating the social and individual problems induced by this type of disease. In this study, we investigated how innate immune pre-stimulation influences the anxiety-like behaviors in chronically stressed mice. Our results showed that a single injection of an innate immune stimulant lipopolysaccharide (LPS) at the dose of 50, 100, and 500 μg/kg 1 day before stress exposure prevented chronic social defeat stress (CSDS)-induced anxiety-like behaviors in mice. A single injection of LPS (100 μg/kg) 5 days before stress exposure produced similar preventive effects on CSDS-induced anxiety-like behaviors, while similar effects were not observed at the condition of 10-days interval between LPS injection and stress exposure. A second LPS injection 10 days after the first LPS injection or a 4 × LPS injection 10 days before stress exposure also prevented CSDS-induced anxiety-like behaviors. Moreover, a single injection of LPS (100 μg/kg) 1 day before stress exposure prevented the production of pro-inflammatory cytokines in the hippocampus and prefrontal cortex of CSDS mice. Suppression of innate immune stimulation by minocycline pretreatment simultaneously abrogated the preventive effect of LPS pre-injection (100 μg/kg) on CSDS-induced anxiety-like behaviors and pro-inflammatory cytokine production in the brain. Our results demonstrated that the pre-stimulation of the innate immune system can prevent the development of anxiety-like behaviors and the progression of the neuroinflammatory responses in the brain in chronically stressed mice.
Collapse
|
40
|
Green TRF, Murphy SM, Ortiz JB, Rowe RK. Age-At-Injury Influences the Glial Response to Traumatic Brain Injury in the Cortex of Male Juvenile Rats. Front Neurol 2022; 12:804139. [PMID: 35111130 PMCID: PMC8802670 DOI: 10.3389/fneur.2021.804139] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/14/2021] [Indexed: 11/29/2022] Open
Abstract
Few translational studies have examined how age-at-injury affects the glial response to traumatic brain injury (TBI). We hypothesized that rats injured at post-natal day (PND) 17 would exhibit a greater glial response, that would persist into early adulthood, compared to rats injured at PND35. PND17 and PND35 rats (n = 75) received a mild to moderate midline fluid percussion injury or sham surgery. In three cortical regions [peri-injury, primary somatosensory barrel field (S1BF), perirhinal], we investigated the glial response relative to age-at-injury (PND17 or PND35), time post-injury (2 hours, 1 day, 7 days, 25 days, or 43 days), and post-natal age, such that rats injured at PND17 or PND35 were compared at the same post-natal-age (e.g., PND17 + 25D post-injury = PND42; PND35 + 7D post-injury = PND42). We measured Iba1 positive microglia cells (area, perimeter) and quantified their activation status using skeletal analysis (branch length/cell, mean processes/cell, cell abundance). GFAP expression was examined using immunohistochemistry and pixel analysis. Data were analyzed using Bayesian multivariate multi-level models. Independent of age-at-injury, TBI activated microglia (shorter branches, fewer processes) in the S1BF and perirhinal cortex with more microglia in all regions compared to uninjured shams. TBI-induced microglial activation (shorter branches) was sustained in the S1BF into early adulthood (PND60). Overall, PND17 injured rats had more microglial activation in the perirhinal cortex than PND35 injured rats. Activation was not confounded by age-dependent cell size changes, and microglial cell body sizes were similar between PND17 and PND35 rats. There were no differences in astrocyte GFAP expression. Increased microglial activation in PND17 brain-injured rats suggests that TBI upregulates the glial response at discrete stages of development. Age-at-injury and aging with an injury are translationally important because experiencing a TBI at an early age may trigger an exaggerated glial response.
Collapse
Affiliation(s)
- Tabitha R. F. Green
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
| | - Sean M. Murphy
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
| | - J. Bryce Ortiz
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
- Phoenix Veterans Affairs (VA) Health Care System, Phoenix, AZ, United States
| | - Rachel K. Rowe
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
- Department of Integrative Physiology, University of Colorado, Boulder, CO, United States
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States
| |
Collapse
|
41
|
Peripheral Infection after Traumatic Brain Injury Augments Excitability in the Perilesional Cortex and Dentate Gyrus. Biomedicines 2021; 9:biomedicines9121946. [PMID: 34944762 PMCID: PMC8698476 DOI: 10.3390/biomedicines9121946] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/15/2021] [Accepted: 12/17/2021] [Indexed: 12/16/2022] Open
Abstract
Peripheral infections occur in up to 28% of patients with traumatic brain injury (TBI), which is a major etiology for structural epilepsies. We hypothesized that infection occurring after TBI acts as a “second hit” and facilitates post-traumatic epileptogenesis. Adult male Sprague–Dawley rats were subjected to lateral fluid-percussion injury or sham-operation. At 8 weeks post-injury, rats were treated with lipopolysaccharide (LPS, 5 mg/kg) to mimic Gram-negative peripheral infection. T2-weighted magnetic resonance imaging was used to detect the cortical lesion type (small focal inflammatory [TBIFI] vs. large cavity-forming [TBICF]). Spontaneous seizures were detected with video-electroencephalography, and seizure susceptibility was determined by the pentylenetetrazole (PTZ) test. Post-PTZ neuronal activation was assessed using c-Fos immunohistochemistry. LPS treatment increased the percentage of rats with PTZ-induced seizures among animals with TBIFI lesions (p < 0.05). It also increased the cumulative duration of PTZ-induced seizures (p < 0.01), particularly in the TBIFI group (p < 0.05). The number of c-Fos immunopositive cells was higher in the perilesional cortex of injured animals compared with sham-operated animals (p < 0.05), particularly in the TBI-LPS group (p < 0.05). LPS treatment increased the percentage of injured rats with bilateral c-Fos staining in the dentate gyrus (p < 0.05), particularly in the TBIFI group (p < 0.05). Our findings demonstrate that peripheral infection after TBI increases PTZ-induced seizure susceptibility and neuronal activation in the perilesional cortex and bilaterally in the dentate gyrus, particularly in animals with prolonged perilesional T2 enhancement. Our data suggest that treatment of infections and reduction of post-injury neuro-inflammation are important components of the treatment regimen aiming at preventing epileptogenesis after TBI.
Collapse
|
42
|
Mastorakos P, Russo MV, Zhou T, Johnson K, McGavern DB. Antimicrobial immunity impedes CNS vascular repair following brain injury. Nat Immunol 2021; 22:1280-1293. [PMID: 34556874 PMCID: PMC8488012 DOI: 10.1038/s41590-021-01012-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 07/27/2021] [Indexed: 12/14/2022]
Abstract
Traumatic brain injury (TBI) and cerebrovascular injury are leading causes of disability and mortality worldwide. Systemic infections often accompany these disorders and can worsen outcomes. Recovery after brain injury depends on innate immunity, but the effect of infections on this process is not well understood. Here, we demonstrate that systemically introduced microorganisms and microbial products interfered with meningeal vascular repair after TBI in a type I interferon (IFN-I)-dependent manner, with sequential infections promoting chronic disrepair. Mechanistically, we discovered that MDA5-dependent detection of an arenavirus encountered after TBI disrupted pro-angiogenic myeloid cell programming via induction of IFN-I signaling. Systemic viral infection similarly blocked restorative angiogenesis in the brain parenchyma after intracranial hemorrhage, leading to chronic IFN-I signaling, blood-brain barrier leakage and a failure to restore cognitive-motor function. Our findings reveal a common immunological mechanism by which systemic infections deviate reparative programming after central nervous system injury and offer a new therapeutic target to improve recovery.
Collapse
Affiliation(s)
- Panagiotis Mastorakos
- Viral Immunology & Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.,Department of Surgical Neurology, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Matthew V Russo
- Viral Immunology & Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Tianzan Zhou
- Viral Immunology & Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Kory Johnson
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Dorian B McGavern
- Viral Immunology & Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
43
|
Abstract
Interleukin-1 (IL-1) is an inflammatory cytokine that has been shown to modulate neuronal signaling in homeostasis and diseases. In homeostasis, IL-1 regulates sleep and memory formation, whereas in diseases, IL-1 impairs memory and alters affect. Interestingly, IL-1 can cause long-lasting changes in behavior, suggesting IL-1 can alter neuroplasticity. The neuroplastic effects of IL-1 are mediated via its cognate receptor, Interleukin-1 Type 1 Receptor (IL-1R1), and are dependent on the distribution and cell type(s) of IL-1R1 expression. Recent reports found that IL-1R1 expression is restricted to discrete subpopulations of neurons, astrocytes, and endothelial cells and suggest IL-1 can influence neural circuits directly through neuronal IL-1R1 or indirectly via non-neuronal IL-1R1. In this review, we analyzed multiple mechanisms by which IL-1/IL-1R1 signaling might impact neuroplasticity based upon the most up-to-date literature and provided potential explanations to clarify discrepant and confusing findings reported in the past.
Collapse
Affiliation(s)
- Daniel P. Nemeth
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
- Department of Biomedical Science, Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Ning Quan
- Department of Biomedical Science, Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| |
Collapse
|
44
|
McDonald SJ, Sharkey JM, Sun M, Kaukas LM, Shultz SR, Turner RJ, Leonard AV, Brady RD, Corrigan F. Beyond the Brain: Peripheral Interactions after Traumatic Brain Injury. J Neurotrauma 2021; 37:770-781. [PMID: 32041478 DOI: 10.1089/neu.2019.6885] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability, and there are currently no pharmacological treatments known to improve patient outcomes. Unquestionably, contributing toward a lack of effective treatments is the highly complex and heterogenous nature of TBI. In this review, we highlight the recent surge of research that has demonstrated various central interactions with the periphery as a potential major contributor toward this heterogeneity and, in particular, the breadth of research from Australia. We describe the growing evidence of how extracranial factors, such as polytrauma and infection, can significantly alter TBI neuropathology. In addition, we highlight how dysregulation of the autonomic nervous system and the systemic inflammatory response induced by TBI can have profound pathophysiological effects on peripheral organs, such as the heart, lung, gastrointestinal tract, liver, kidney, spleen, and bone. Collectively, this review firmly establishes TBI as a systemic condition. Further, the central and peripheral interactions that can occur after TBI must be further explored and accounted for in the ongoing search for effective treatments.
Collapse
Affiliation(s)
- Stuart J McDonald
- Department Neuroscience, Monash University, Melbourne, Victoria, Australia.,Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Jessica M Sharkey
- Discipline of Anatomy and Pathology, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Mujun Sun
- Department Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Lola M Kaukas
- School of Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Sandy R Shultz
- Department Neuroscience, Monash University, Melbourne, Victoria, Australia.,Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Renee J Turner
- Discipline of Anatomy and Pathology, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Anna V Leonard
- Discipline of Anatomy and Pathology, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Rhys D Brady
- Department Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Frances Corrigan
- School of Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| |
Collapse
|
45
|
Hanscom M, Loane DJ, Shea-Donohue T. Brain-gut axis dysfunction in the pathogenesis of traumatic brain injury. J Clin Invest 2021; 131:143777. [PMID: 34128471 PMCID: PMC8203445 DOI: 10.1172/jci143777] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Traumatic brain injury (TBI) is a chronic and progressive disease, and management requires an understanding of both the primary neurological injury and the secondary sequelae that affect peripheral organs, including the gastrointestinal (GI) tract. The brain-gut axis is composed of bidirectional pathways through which TBI-induced neuroinflammation and neurodegeneration impact gut function. The resulting TBI-induced dysautonomia and systemic inflammation contribute to the secondary GI events, including dysmotility and increased mucosal permeability. These effects shape, and are shaped by, changes in microbiota composition and activation of resident and recruited immune cells. Microbial products and immune cell mediators in turn modulate brain-gut activity. Importantly, secondary enteric inflammatory challenges prolong systemic inflammation and worsen TBI-induced neuropathology and neurobehavioral deficits. The importance of brain-gut communication in maintaining GI homeostasis highlights it as a viable therapeutic target for TBI. Currently, treatments directed toward dysautonomia, dysbiosis, and/or systemic inflammation offer the most promise.
Collapse
Affiliation(s)
- Marie Hanscom
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - David J. Loane
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Terez Shea-Donohue
- Division of Digestive Diseases and Nutrition, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| |
Collapse
|
46
|
Postolache TT, Wadhawan A, Can A, Lowry CA, Woodbury M, Makkar H, Hoisington AJ, Scott AJ, Potocki E, Benros ME, Stiller JW. Inflammation in Traumatic Brain Injury. J Alzheimers Dis 2021; 74:1-28. [PMID: 32176646 DOI: 10.3233/jad-191150] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
There is an increasing evidence that inflammation contributes to clinical and functional outcomes in traumatic brain injury (TBI). Many successful target-engaging, lesion-reducing, symptom-alleviating, and function-improving interventions in animal models of TBI have failed to show efficacy in clinical trials. Timing and immunological context are paramount for the direction, quality, and intensity of immune responses to TBI and the resulting neuroanatomical, clinical, and functional course. We present components of the immune system implicated in TBI, potential immune targets, and target-engaging interventions. The main objective of our article is to point toward modifiable molecular and cellular mechanisms that may modify the outcomes in TBI, and contribute to increasing the translational value of interventions that have been identified in animal models of TBI.
Collapse
Affiliation(s)
- Teodor T Postolache
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.,Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 19, Aurora, CO, USA.,Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, CO, USA.,Mental Illness Research, Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 5, VA Capitol Health Care Network, Baltimore, MD, USA
| | - Abhishek Wadhawan
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.,Saint Elizabeths Hospital, Department of Psychiatry, Washington, DC, USA
| | - Adem Can
- School of Medicine, University of Maryland Baltimore, Baltimore, MD, USA
| | - Christopher A Lowry
- Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 19, Aurora, CO, USA.,Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, CO, USA.,Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA.,Department of Physical Medicine and Rehabilitation and Center for Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Margaret Woodbury
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.,VA Maryland Healthcare System, Baltimore VA Medical Center, Baltimore, MD, USA
| | - Hina Makkar
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Andrew J Hoisington
- Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 19, Aurora, CO, USA.,Systems Engineering and Management, Air Force Institute of Technology, Wright-Patterson AFB, OH, USA
| | - Alison J Scott
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, MD, USA
| | - Eileen Potocki
- VA Maryland Healthcare System, Baltimore VA Medical Center, Baltimore, MD, USA
| | - Michael E Benros
- Copenhagen Research Center for Mental Health-CORE, Mental Health Centre Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - John W Stiller
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.,Maryland State Athletic Commission, Baltimore, MD, USA.,Saint Elizabeths Hospital, Neurology Consultation Services, Washington, DC, USA
| |
Collapse
|
47
|
Hosseini M, Salmani H, Baghcheghi Y. Losartan improved hippocampal long-term potentiation impairment induced by repeated LPS injection in rats. Physiol Rep 2021; 9:e14874. [PMID: 34042283 PMCID: PMC8157761 DOI: 10.14814/phy2.14874] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 04/19/2021] [Accepted: 04/23/2021] [Indexed: 11/24/2022] Open
Abstract
Cognitive impairment has been known as a common consequence of brain inflammation. Long-term potentiation (LTP), the generally accepted cellular mechanism for memory formation in the mammalian brain, has been shown to be suppressed by inflammation. Studies have shown that angiotensin II (Ang II) through the Ang II type 1 receptor (AT1R) has a role in brain and peripheral immune system communication and brain inflammation. Here, the effect of AT1R blockade on hippocampal LTP in rats undergoing repeated lipopolysaccharide (LPS) injection was investigated. Rats received intraperitoneal (ip) injections of LPS (250 μg kg-1 day-1 ) for seven days. Treatment with losartan (ip; 3 mg kg-1 day-1 ) was started 3 days before LPS injection and continued during the LPS injections. Rats were anesthetized, and field excitatory postsynaptic potential (fEPSP) was recorded from the stratum radiatum of the CA1 area of the hippocampus in response to stimulation of the Schaffer collateral pathway. Results showed that LTP was suppressed in the LPS-injected rats as no significant differences were found in the fEPSP slope and amplitude before and after the LTP induction. AT1R blockade by losartan restored fEPSP to the control levels. These findings indicate that Ang II, through AT1R, has a role in LTP suppression induced by systemic inflammation.
Collapse
Affiliation(s)
- Mahmoud Hosseini
- Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research CenterMashhad University of Medical SciencesMashhadIran
- Neuroscience Research CenterMashhad University of Medical SciencesMashhadIran
| | - Hossein Salmani
- Applied Biomedical Research CenterMashhad University of Medical SciencesMashhadIran
| | - Yousef Baghcheghi
- Student Research CommitteeJiroft University of Medical SciencesJiroftIran
| |
Collapse
|
48
|
Kaukas L, Krieg J, Collins-Praino L, Corrigan F. Effects of Remote Immune Activation on Performance in the 5-Choice Serial Reaction Time Task Following Mild Traumatic Brain Injury in Adolescence. Front Behav Neurosci 2021; 15:659679. [PMID: 33867953 PMCID: PMC8046921 DOI: 10.3389/fnbeh.2021.659679] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/12/2021] [Indexed: 11/17/2022] Open
Abstract
In adult pre-clinical models, traumatic brain injury (TBI) has been shown to prime microglia, exaggerating the central inflammatory response to an acute immune challenge, worsening depressive-like behavior, and enhancing cognitive deficits. Whether this phenomenon exists following mTBI during adolescence has yet to be explored, with age at injury potentially altering the inflammatory response. Furthermore, to date, studies have predominantly examined hippocampal-dependent learning domains, although pre-frontal cortex-driven functions, including attention, motivation, and impulsivity, are significantly affected by both adolescent TBI and acute inflammatory stimuli. As such, the current study examined the effects of a single acute peripheral dose of LPS (0.33 mg/kg) given in adulthood following mTBI in mid-adolescence in male Sprague–Dawley rats on performance in the 5-choice serial reaction time task (5-CSRTT). Only previously injured animals given LPS showed an increase in omissions and reward collection latency on the 5-CSRTT, with no effect noted in sham animals given LPS. This is suggestive of impaired motivation and a prolonged central inflammatory response to LPS administration in these animals. Indeed, morphological analysis of myeloid cells within the pre-frontal cortex, via IBA1 immunohistochemistry, found that injured animals administered LPS had an increase in complexity in IBA1+ve cells, an effect that was seen to a lesser extent in sham animals. These findings suggest that there may be ongoing alterations in the effects of acute inflammatory stimuli that are driven, in part by increased reactivity of microglial cells.
Collapse
Affiliation(s)
- Lola Kaukas
- Head Injury Laboratory, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Justin Krieg
- Head Injury Laboratory, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Lyndsey Collins-Praino
- Head Injury Laboratory, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Frances Corrigan
- Head Injury Laboratory, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
49
|
Sharma R, Zamani A, Dill LK, Sun M, Chu E, Robinson MJ, O'Brien TJ, Shultz SR, Semple BD. A systemic immune challenge to model hospital-acquired infections independently regulates immune responses after pediatric traumatic brain injury. J Neuroinflammation 2021; 18:72. [PMID: 33731173 PMCID: PMC7968166 DOI: 10.1186/s12974-021-02114-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 02/16/2021] [Indexed: 11/10/2022] Open
Abstract
Background Traumatic brain injury (TBI) is a major cause of disability in young children, yet the factors contributing to poor outcomes in this population are not well understood. TBI patients are highly susceptible to nosocomial infections, which are mostly acquired within the first week of hospitalization, and such infections may modify TBI pathobiology and recovery. In this study, we hypothesized that a peripheral immune challenge such as lipopolysaccharide (LPS)—mimicking a hospital-acquired infection—would worsen outcomes after experimental pediatric TBI, by perpetuating the inflammatory immune response. Methods Three-week-old male mice received either a moderate controlled cortical impact or sham surgery, followed by a single LPS dose (1 mg/kg i.p.) or vehicle (0.9% saline) at 4 days post-surgery, then analysis at 5 or 8 days post-injury (i.e., 1 or 4 days post-LPS). Results LPS-treated mice exhibited a time-dependent reduction in general activity and social investigation, and increased anxiety, alongside substantial body weight loss, indicating transient sickness behaviors. Spleen-to-body weight ratios were also increased in LPS-treated mice, indicative of persistent activation of adaptive immunity at 4 days post-LPS. TBI + LPS mice showed an impaired trajectory of weight gain post-LPS, reflecting a synergistic effect of TBI and the LPS-induced immune challenge. Flow cytometry analysis demonstrated innate immune cell activation in blood, brain, and spleen post-LPS; however, this was not potentiated by TBI. Cytokine protein levels in serum, and gene expression levels in the brain, were altered in response to LPS but not TBI across the time course. Immunofluorescence analysis of brain sections revealed increased glia reactivity due to injury, but no additive effect of LPS was observed. Conclusions Together, we found that a transient, infection-like systemic challenge had widespread effects on the brain and immune system, but these were not synergistic with prior TBI in pediatric mice. These findings provide novel insight into the potential influence of a secondary immune challenge to the injured pediatric brain, with future studies needed to elucidate the chronic effects of this two-hit insult. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02114-1.
Collapse
Affiliation(s)
- Rishabh Sharma
- Department of Neuroscience, Central Clinical School, Monash University, Level 6, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Akram Zamani
- Department of Neuroscience, Central Clinical School, Monash University, Level 6, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Larissa K Dill
- Department of Neuroscience, Central Clinical School, Monash University, Level 6, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia.,Department of Neurology, Alfred Health, Prahran, VIC, Australia
| | - Mujun Sun
- Department of Neuroscience, Central Clinical School, Monash University, Level 6, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Erskine Chu
- Department of Neuroscience, Central Clinical School, Monash University, Level 6, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Marcus J Robinson
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Central Clinical School, Monash University, Level 6, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia.,Department of Neurology, Alfred Health, Prahran, VIC, Australia.,Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, VIC, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Level 6, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia.,Department of Neurology, Alfred Health, Prahran, VIC, Australia.,Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, VIC, Australia
| | - Bridgette D Semple
- Department of Neuroscience, Central Clinical School, Monash University, Level 6, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia. .,Department of Neurology, Alfred Health, Prahran, VIC, Australia. .,Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
50
|
Zhang LM, Zhang DX, Zheng WC, Hu JS, Fu L, Li Y, Xin Y, Wang XP. CORM-3 exerts a neuroprotective effect in a rodent model of traumatic brain injury via the bidirectional gut-brain interactions. Exp Neurol 2021; 341:113683. [PMID: 33711325 DOI: 10.1016/j.expneurol.2021.113683] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 02/12/2021] [Accepted: 03/02/2021] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Traumatic brain injury (TBI) induced the gastrointestinal inflammation that is associated with TBI-related morbidity and mortality. Carbon monoxide-releasing molecule (CORM)-3 is a water-soluble exogenous carbon monoxide that exerts protective effects against inflammation-induced pyroptosis. We investigated the gastrointestinal inflammation in a rodent model of traumatic brain injury (TBI) with subsequent hemorrhagic shock and resuscitation (HSR), as well as effects of CORM-3 using an intestinal injection on both gut and brain. METHODS Following exposure to TBI plus HSR, rats were administrated with CORM-3 (8 mg/kg) through an intestinal injection after resuscitation immediately. The pathological changes and pyroptosis in the gut were measured at 24 h and 30 day post-trauma. We also assessed the intestinal and cortical CO content, as well as IL-1β and IL-18 levels in the serum within 48 h after trauma. We then explored pathological changes in the ventromedial prefrontal cortex (vmPFC) and neurological behavior deficits on 30 day post-trauma. RESULTS After TBI + HSR exposure, CORM-3-treated rats presented significantly decreased pyroptosis, more CO content in the jejunum, and lower IL-1β, IL-18 levels in the serum at 24 h after trauma. Moreover, the rats treated with CORM-3 exerted ameliorated jejunal and vmPFC injury, enhanced learning/memory ability and exploratory activity, improved anxiety-like behaviors than the TBI + HSR-treated rats on 30 day post-trauma. CONCLUSION These experimental data demonstrated and bidirectional gut-brain interactions after TBI, anti-inflammatory effects of CORM-3, which may improve late outcomes after brain injury.
Collapse
Affiliation(s)
- Li-Min Zhang
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, China.
| | - Dong-Xue Zhang
- Department of Gerontology, Cangzhou Central Hospital, Cangzhou, China
| | - Wei-Chao Zheng
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, China
| | - Jin-Shu Hu
- Clinical Laboratory, Cangzhou Central Hospital, Cangzhou, China
| | - Lan Fu
- Department of Radiodiagnosis, Cangzhou Central Hospital, Cangzhou, China
| | - Yan Li
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, China
| | - Yue Xin
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, China
| | - Xu-Peng Wang
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, China
| |
Collapse
|