1
|
Wei J, Liu C, Qin D, Ren F, Duan J, Chen T, Wu A. Targeting inflammation and gut microbiota with antibacterial therapy: Implications for central nervous system health. Ageing Res Rev 2024; 102:102544. [PMID: 39419400 DOI: 10.1016/j.arr.2024.102544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024]
Abstract
The complex symbiotic relationship between inflammation, the gut microbiota, and the central nervous system (CNS) has become a pivotal focus of contemporary biomedical research. Inflammation, as a physiological defense mechanism, plays a dual role as both a protective and pathological factor, and is intricately associated with gut microbiota homeostasis, often termed the "second brain." The gutbrain axis (GBA) exemplifies this multifaceted interaction, where gut health exerts significantly regulatory effects on CNS functions. Antibacterial therapies represent both promising and challenging strategies for modulating inflammation and gut microbiota composition to confer CNS benefits. However, while such therapies may exert positive modulatory effects on the gut microbiota, they also carry the potential to disrupt microbial equilibrium, potentially exacerbating neurological dysfunction. Recent advances have provided critical insights into the therapeutic implications of antibacterial interventions; nevertheless, the application of these therapies in the context of CNS health warrants a judicious and evidence-based approach. As research progresses, deeper investigation into the microbial-neural interface is essential to fully realize the potential of therapies targeting inflammation and the gut microbiota for CNS health. Future efforts should focus on refining antibacterial interventions to modulate the gut microbiota while minimizing disruption to microbial balance, thereby reducing risks and enhancing efficacy in CNS-related conditions. In conclusion, despite challenges, a more comprehensive understanding of the GBA, along with precise modulation through targeted antibacterial therapies, offers significant promise for advancing CNS disorder treatment. Continued research in this area will lead to innovative interventions and improved patient outcomes.
Collapse
Affiliation(s)
- Jing Wei
- Eye School of Chengdu University of TCM, Key Laboratory of Sichuan Province Ophthalmopathy Prevention & Cure and Visual Function Protection with TCM Laboratory, Retinal Image Technology and Chronic Vascular Disease Prevention & Control and Collaborative Innovation Center, Chengdu, China; School of Pharmaceutical Sciences, China-Pakistan International Science and Technology Innovation Cooperation Base for Ethnic Medicine Development in Hunan Province, Hunan University of Medicine, Huaihua 418000, China.
| | - Chunmeng Liu
- Eye School of Chengdu University of TCM, Key Laboratory of Sichuan Province Ophthalmopathy Prevention & Cure and Visual Function Protection with TCM Laboratory, Retinal Image Technology and Chronic Vascular Disease Prevention & Control and Collaborative Innovation Center, Chengdu, China.
| | - Dalian Qin
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Department of Cardiology, the Affiliated Hospital of Southwest Medical University and Key Laboratory of Medical Electrophysiology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China.
| | - Fang Ren
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chongqing Traditional Chinese Medicine Hospital, Chongqing 400021, China.
| | - Junguo Duan
- Eye School of Chengdu University of TCM, Key Laboratory of Sichuan Province Ophthalmopathy Prevention & Cure and Visual Function Protection with TCM Laboratory, Retinal Image Technology and Chronic Vascular Disease Prevention & Control and Collaborative Innovation Center, Chengdu, China.
| | - Ting Chen
- School of Pharmaceutical Sciences, China-Pakistan International Science and Technology Innovation Cooperation Base for Ethnic Medicine Development in Hunan Province, Hunan University of Medicine, Huaihua 418000, China.
| | - Anguo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Department of Cardiology, the Affiliated Hospital of Southwest Medical University and Key Laboratory of Medical Electrophysiology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China; State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| |
Collapse
|
2
|
Dill-Macky AS, Lee EN, Wertheim JA, Koss KM. Glia in tissue engineering: From biomaterial tools to transplantation. Acta Biomater 2024:S1742-7061(24)00600-7. [PMID: 39396630 DOI: 10.1016/j.actbio.2024.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 10/01/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
Glia are imperative in nearly every function of the nervous system, including neurotransmission, neuronal repair, development, immunity, and myelination. Recently, the reparative roles of glia in the central and peripheral nervous systems have been elucidated, suggesting a tremendous potential for these cells as novel treatments to central nervous system disorders. Glial cells often behave as 'double-edged swords' in neuroinflammation, ultimately deciding the life or death of resident cells. Compared to glia, neuronal cells have limited mobility, lack the ability to divide and self-renew, and are generally more delicate. Glia have been candidates for therapeutic use in many successful grafting studies, which have been largely focused on restoring myelin with Schwann cells, olfactory ensheathing glia, and oligodendrocytes with support from astrocytes. However, few therapeutics of this class have succeeded past clinical trials. Several tools and materials are being developed to understand and re-engineer these grafting concepts for greater success, such as extra cellular matrix-based scaffolds, bioactive peptides, biomolecular delivery systems, biomolecular discovery for neuroinflammatory mediation, composite microstructures such as artificial channels for cell trafficking, and graft enhanced electrical stimulation. Furthermore, advances in stem cell-derived cortical/cerebral organoid differentiation protocols have allowed for the generation of patient-derived glia comparable to those acquired from tissues requiring highly invasive procedures or are otherwise inaccessible. However, research on bioengineered tools that manipulate glial cells is nowhere near as comprehensive as that for systems of neurons and neural stem cells. This article explores the therapeutic potential of glia in transplantation with an emphasis on novel bioengineered tools for enhancement of their reparative properties. STATEMENT OF SIGNIFICANCE: Neural glia are responsible for a host of developmental, homeostatic, and reparative roles in the central nervous system but are often a major cause of tissue damage and cellular loss in insults and degenerative pathologies. Most glial grafts have employed Schwann cells for remyelination, but other glial with novel biomaterials have been employed, emphasizing their diverse functionality. Promising strategies have emerged, including neuroimmune mediation of glial scar tissues and facilitated migration and differentiation of stem cells for neural replacement. Herein, a comprehensive review of biomaterial tools for glia in transplantation is presented, highlighting Schwann cells, astrocytes, olfactory ensheating glia, oligodendrocytes, microglia, and ependymal cells.
Collapse
Affiliation(s)
- A S Dill-Macky
- Department of Surgery, University of Arizona, 1501 N Campbell Ave, Tucson, AZ 85724, United States
| | - E N Lee
- Department of Surgery, University of Arizona, 1501 N Campbell Ave, Tucson, AZ 85724, United States
| | - J A Wertheim
- Department of Surgery, University of Arizona, 1501 N Campbell Ave, Tucson, AZ 85724, United States
| | - K M Koss
- Department of Neurobiology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0625, United States; Sealy Institute for Drug Discovery, University of Texas Medical Branch, 105 11th Street Galveston, TX 77555-1110, United States.
| |
Collapse
|
3
|
Oura P, Hakkarainen A, Sajantila A. Forensic neuropathology in the past decade: a scoping literature review. Forensic Sci Med Pathol 2024; 20:724-735. [PMID: 37439948 PMCID: PMC11297074 DOI: 10.1007/s12024-023-00672-9] [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] [Accepted: 06/22/2023] [Indexed: 07/14/2023]
Abstract
While there has been notable research activity in the field of clinical neuropathology over the recent years, forensic approaches have been less frequent. This scoping literature review explored original research on forensic neuropathology over the past decade (January 1, 2010, until February 12, 2022) using the MEDLINE database. The aims were to (1) analyze the volume of research on the topic, (2) describe meta-level attributes and sample characteristics, and (3) summarize key research themes and methods. Of 5053 initial hits, 2864 fell within the target timeframe, and 122 were included in the review. Only 3-17 articles were published per year globally. Most articles originated from the Europe (39.3%) and Asia (36.1%) and were published in forensic journals (57.4%). A median sample included 57 subjects aged between 16 and 80 years. The most common research theme was traumatic intracranial injury (24.6%), followed by anatomy (12.3%) and substance abuse (11.5%). Key methods included immunotechniques (31.1%) and macroscopic observation (21.3%). Although a number of novel findings were reported, most were of preliminary nature and will require further validation. In order to reach breakthroughs and validate novel tools for routine use, more research input is urged from researchers across the world. It would be necessary to ensure appropriate sample sizes and make use of control groups.
Collapse
Affiliation(s)
- Petteri Oura
- Department of Forensic Medicine, Faculty of Medicine, University of Helsinki, P.O. Box 21, Helsinki, FI-00014, Finland.
- Forensic Medicine Unit, Finnish Institute for Health and Welfare, P.O. Box 30, Helsinki, FI-00271, Finland.
| | - Antti Hakkarainen
- Department of Forensic Medicine, Faculty of Medicine, University of Helsinki, P.O. Box 21, Helsinki, FI-00014, Finland
- Forensic Medicine Unit, Finnish Institute for Health and Welfare, P.O. Box 30, Helsinki, FI-00271, Finland
| | - Antti Sajantila
- Department of Forensic Medicine, Faculty of Medicine, University of Helsinki, P.O. Box 21, Helsinki, FI-00014, Finland
- Forensic Medicine Unit, Finnish Institute for Health and Welfare, P.O. Box 30, Helsinki, FI-00271, Finland
| |
Collapse
|
4
|
Nespoli E, Hakani M, Hein TM, May SN, Danzer K, Wirth T, Baumann B, Dimou L. Glial cells react to closed head injury in a distinct and spatiotemporally orchestrated manner. Sci Rep 2024; 14:2441. [PMID: 38286816 PMCID: PMC10825139 DOI: 10.1038/s41598-024-52337-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 01/17/2024] [Indexed: 01/31/2024] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of mortality and disability worldwide. Acute neuroinflammation is a prominent reaction after TBI and is mostly initiated by brain-resident glial cells such as microglia, NG2-glia and astrocytes. The magnitude of this reaction paves the way for long-lasting consequences such as chronic neurological pathologies, for which therapeutic options remain limited. The neuroinflammatory response to TBI is mostly studied with craniotomy-based animal models that are very robust but also rather artificial. Here, we aimed to analyze the reaction of glial cells in a highly translational but variable closed head injury (CHI) model and were able to correlate the severity of the trauma to the degree of glial response. Furthermore, we could show that the different glial cell types react in a temporally and spatially orchestrated manner in terms of morphological changes, proliferation, and cell numbers in the first 15 days after the lesion. Interestingly, NG2-glia, the only proliferating cells in the healthy brain parenchyma, divided at a rate that was correlated with the size of the injury. Our findings describe the previously uncharacterized posttraumatic response of the major brain glial cell types in CHI in order to gain a detailed understanding of the course of neuroinflammatory events; such knowledge may open novel avenues for future therapeutic approaches in TBI.
Collapse
Affiliation(s)
- Ester Nespoli
- Molecular and Translational Neuroscience, Department of Neurology, Ulm University, Ulm, Germany
| | - Marsela Hakani
- Molecular and Translational Neuroscience, Department of Neurology, Ulm University, Ulm, Germany
| | | | | | - Karin Danzer
- Department of Neurology, Ulm University, Ulm, Germany
- German Center for Neurodegenerative Diseases (DNZE), Ulm, Germany
| | - Thomas Wirth
- Institute of Physiological Chemistry, Ulm University, Ulm, Germany
| | - Bernd Baumann
- Institute of Physiological Chemistry, Ulm University, Ulm, Germany
| | - Leda Dimou
- Molecular and Translational Neuroscience, Department of Neurology, Ulm University, Ulm, Germany.
| |
Collapse
|
5
|
Pearson A, Ortiz C, Eisenbaum M, Arrate C, Browning M, Mullan M, Bachmeier C, Crawford F, Ojo JO. Deletion of PTEN in microglia ameliorates chronic neuroinflammation following repetitive mTBI. Mol Cell Neurosci 2023; 125:103855. [PMID: 37084991 DOI: 10.1016/j.mcn.2023.103855] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 03/25/2023] [Accepted: 04/17/2023] [Indexed: 04/23/2023] Open
Abstract
Traumatic brain injury is a leading cause of morbidity and mortality in adults and children in developed nations. Following the primary injury, microglia, the resident innate immune cells of the CNS, initiate several inflammatory signaling cascades and pathophysiological responses that may persist chronically; chronic neuroinflammation following TBI has been closely linked to the development of neurodegeneration and neurological dysfunction. Phosphoinositide 3-kinases (PI3Ks) are a family of lipid kinases that have been shown to regulate several key mechanisms in the inflammatory response to TBI. Increasing evidence has shown that the modulation of the PI3K/AKT signaling pathway has the potential to influence the cellular response to inflammatory stimuli. However, directly targeting PI3K signaling poses several challenges due to its regulatory role in several cell survival pathways. We have previously identified that the phosphatase and tensin homolog deleted on chromosome 10 (PTEN), the major negative regulator of PI3K/AKT signaling, is dysregulated following exposure to repetitive mild traumatic brain injury (r-mTBI). Moreover, this dysregulated PI3K/AKT signaling was correlated with chronic microglial-mediated neuroinflammation. Therefore, we interrogated microglial-specific PTEN as a therapeutic target in TBI by generating a microglial-specific, Tamoxifen inducible conditional PTEN knockout model using a CX3CR1 Cre recombinase mouse line PTENfl/fl/CX3CR1+/CreERT2 (mcg-PTENcKO), and exposed them to our 20-hit r-mTBI paradigm. Animals were treated with tamoxifen at 76 days post-last injury, and the effects of microglia PTEN deletion on immune-inflammatory responses were assessed at 90-days post last injury. We observed that the deletion of microglial PTEN ameliorated the proinflammatory response to repetitive brain trauma, not only reducing chronic microglial activation and proinflammatory cytokine production but also rescuing TBI-induced reactive astrogliosis, demonstrating that these effects extended beyond microglia alone. Additionally, we observed that the pharmacological inhibition of PTEN with BpV(HOpic) ameliorated the LPS-induced activation of microglial NFκB signaling in vitro. Together, these data provide support for the role of PTEN as a regulator of chronic neuroinflammation following repetitive mild TBI.
Collapse
Affiliation(s)
- Andrew Pearson
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL 34243, USA; The Open University, Walton Hall, Kents Hill, Milton Keynes MK7 6AA, United Kingdom.
| | - Camila Ortiz
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL 34243, USA; The Open University, Walton Hall, Kents Hill, Milton Keynes MK7 6AA, United Kingdom
| | - Max Eisenbaum
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL 34243, USA; The Open University, Walton Hall, Kents Hill, Milton Keynes MK7 6AA, United Kingdom
| | - Clara Arrate
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL 34243, USA
| | | | - Michael Mullan
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL 34243, USA; The Open University, Walton Hall, Kents Hill, Milton Keynes MK7 6AA, United Kingdom
| | - Corbin Bachmeier
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL 34243, USA; The Open University, Walton Hall, Kents Hill, Milton Keynes MK7 6AA, United Kingdom
| | - Fiona Crawford
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL 34243, USA; The Open University, Walton Hall, Kents Hill, Milton Keynes MK7 6AA, United Kingdom; James A. Haley Veterans' Hospital, 13000 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Joseph O Ojo
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL 34243, USA; The Open University, Walton Hall, Kents Hill, Milton Keynes MK7 6AA, United Kingdom
| |
Collapse
|
6
|
Shaughness MC, Pierron N, Smith AN, Byrnes KR. The Integrin Pathway Partially Mediates Stretch-Induced Deficits in Primary Rat Microglia. Mol Neurobiol 2023; 60:3396-3412. [PMID: 36856961 DOI: 10.1007/s12035-023-03291-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 02/21/2023] [Indexed: 03/02/2023]
Abstract
Stretch-injured microglia display significantly altered morphology, function and inflammatory-associated gene expression when cultured on a synthetic fibronectin substrate. However, the mechanism by which stretch induces these changes is unknown. Integrins, such as α5β1, mediate microglial attachment to fibronectin via the RGD binding peptide; following integrin ligation the integrin-associated signaling enzyme, focal adhesion kinase (FAK), autophosphorylates tyrosine residue 397 and mediates multiple downstream cellular processes. We therefore hypothesize that blocking the RGD binding/integrin pathway with a commercially available RGD peptide will mimic the stretch-induced morphological alterations and functional deficits in microglia. Further, we hypothesize that upregulation of stretch-inhibited downstream integrin signaling will reverse these effects. Using primary rat microglia, we tested the effects of RGD binding peptide and a FAK activator on cellular function and structure and response to stretch-injury. Similar to injured cells, RGD peptide administration significantly decreases media nitric oxide (NO) levels and iNOS expression and induced morphological alterations and migratory deficits. While stretch-injury and RGD peptide administration decreased phosphorylation of the tyrosine 397 residue on FAK, 20 nM of ZINC 40099027, an activator specific to the tyrosine 397 residue, rescued the stretch-induced decrease in FAK phosphorylation and ameliorated the injury-induced decrease in media NO levels, iNOS expression and inflammatory associated gene expression. Additionally, treatment alleviated morphological changes observed after stretch-injury and restored normal migratory behavior to control levels. Taken together, these data suggest that the integrin/FAK pathway partially mediates the stretch-injured phenotype in microglia, and may serve as a pathway to modulate microglial responses.
Collapse
Affiliation(s)
- Michael C Shaughness
- Neuroscience Program, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd., Bethesda, MD, 20814, USA.,Operational and Undersea Medicine Directorate (OUMD), En Route & Critical Care Department (ECD), Naval Medical Research Center (NMRC), Silver Spring, MD, USA
| | - Nathan Pierron
- F.E. Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Austin N Smith
- Neuroscience Program, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd., Bethesda, MD, 20814, USA
| | - Kimberly R Byrnes
- Neuroscience Program, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd., Bethesda, MD, 20814, USA. .,Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| |
Collapse
|
7
|
White MR, VandeVord PJ. Regional variances depict a unique glial-specific inflammatory response following closed-head injury. Front Cell Neurosci 2023; 17:1076851. [PMID: 36909284 PMCID: PMC9996631 DOI: 10.3389/fncel.2023.1076851] [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: 10/22/2022] [Accepted: 01/27/2023] [Indexed: 02/17/2023] Open
Abstract
Mild traumatic brain injuries (mTBI) constitute a significant health concern with clinical symptoms ranging from headaches to cognitive deficits. Despite the myriad of symptoms commonly reported following this injury, there is still a lack of knowledge on the various pathophysiological changes that occur. Preclinical studies are at the forefront of discovery delineating the changes that occur within this heterogeneous injury, with the emergence of translational models such as closed-head impact models allowing for further exploration of this injury mechanism. In the current study, male rats were subjected to a closed-head controlled cortical impact (cCCI), producing a concussion (mTBI). The pathological effects of this injury were then evaluated using immunoflourescence seven days following. The results exhibited a unique glial-specific inflammatory response, with both the ipsilateral and contralateral sides of the cortex and hippocampus showing pathological changes following impact. Overall these findings are consistent with glial changes reported following concussions and may contribute to subsequent symptoms.
Collapse
Affiliation(s)
- Michelle R. White
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, United States
| | - Pamela J. VandeVord
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, United States
- Salem VA Medical Center, Salem, VA, United States
| |
Collapse
|
8
|
Shao X, Liu Z, Mao S, Han L. Unraveling the Mechanobiology Underlying Traumatic Brain Injury with Advanced Technologies and Biomaterials. Adv Healthc Mater 2022; 11:e2200760. [PMID: 35841392 DOI: 10.1002/adhm.202200760] [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: 04/05/2022] [Revised: 06/27/2022] [Indexed: 01/27/2023]
Abstract
Traumatic brain injury (TBI) is a worldwide health and socioeconomic problem, associated with prolonged and complex neurological aftermaths, including a variety of functional deficits and neurodegenerative disorders. Research on the long-term effects has highlighted that TBI shall be regarded as a chronic health condition. The initiation and exacerbation of TBI involve a series of mechanical stimulations and perturbations, accompanied by mechanotransduction events within the brain tissues. Mechanobiology thus offers a unique perspective and likely promising approach to unravel the underlying molecular and biochemical mechanisms leading to neural cells dysfunction after TBI, which may contribute to the discovery of novel targets for future clinical treatment. This article investigates TBI and the subsequent brain dysfunction from a lens of neuromechanobiology. Following an introduction, the mechanobiological insights are examined into the molecular pathology of TBI, and then an overview is given of the latest research technologies to explore neuromechanobiology, with particular focus on microfluidics and biomaterials. Challenges and prospects in the current field are also discussed. Through this article, it is hoped that extensive technical innovation in biomedical devices and materials can be encouraged to advance the field of neuromechanobiology, paving potential ways for the research and rehabilitation of neurotrauma and neurological diseases.
Collapse
Affiliation(s)
- Xiaowei Shao
- Institute of Marine Science and Technology, Shandong University, Qingdao, Shandong, 266237, China.,Suzhou Research Institute, Shandong University, Suzhou, Jiangsu, 215123, China
| | - Zhongqian Liu
- Institute of Marine Science and Technology, Shandong University, Qingdao, Shandong, 266237, China
| | - Shijie Mao
- Institute of Marine Science and Technology, Shandong University, Qingdao, Shandong, 266237, China
| | - Lin Han
- Institute of Marine Science and Technology, Shandong University, Qingdao, Shandong, 266237, China
| |
Collapse
|
9
|
Shang Y, Wang Y, Guo Y, Ren L, Zhang X, Wang S, Zhang C, Cai J. Analysis of the risk of traumatic brain injury and evaluation neurogranin and myelin basic protein as potential biomarkers of traumatic brain injury in postmortem examination. Forensic Sci Med Pathol 2022; 18:288-298. [PMID: 35201602 DOI: 10.1007/s12024-022-00459-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2022] [Indexed: 12/14/2022]
Abstract
In forensic pathology, traumatic brain injury (TBI) is a frequently encountered cause of death. Unfortunately, the statistic autopsy data, risk investigation about injury patterns, and circumstances of TBI are still sparse. Estimates of survival time post-TBI and postmortem diagnosis of TBI are especially important implications in forensic medicine. Neurogranin (Ng) and myelin basic protein (MBP) represent potential biomarkers of TBI. The present study analyzed retrospectively the forensic autopsy records of TBI cases at a university center of medico-legal investigation from 2008 to 2020. Immunohistochemistry and enzyme-linked immunosorbent assays (ELISA) were used to investigate the expression changes of Ng and MBP in the cortical brain injury adjacent tissues and serum, respectively, from cases of TBI at autopsy with different survival times post-TBI. The results show that the major mechanism of death of TBI is assault, and accident was the major manner of death. Ng and MBP are mainly expressed in the cortical nerve cells and the myelin sheath, respectively. The serum levels of Ng and MBP in each TBI group were higher compared with those in the controls. The brain cortical levels of Ng and MBP decreased at first and then steadily increased with extended survival time post-TBI. The immunopositive ratios and serum concentration of Ng and MBP have shown significant differences among control group and all TBI group (p < 0.001). Collectively, the immunohistochemical analyses of Ng and MBP in human brain tissues may be useful to determine the survival time after TBI, and Ng and MBP level in the human blood specimens could be considered as a postmortem diagnostic tools of TBI in forensic practice.
Collapse
Affiliation(s)
- Yanjie Shang
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Yuxin Wang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Yadong Guo
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Lipin Ren
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Xiangyan Zhang
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Shujuan Wang
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Changquan Zhang
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, 410013, Hunan, China.
| | - Jifeng Cai
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, 410013, Hunan, China.
| |
Collapse
|
10
|
Spanos M, Shachar S, Sweeney T, Lehmann HI, Gokulnath P, Li G, Sigal GB, Nagaraj R, Bathala P, Rana F, Shah RV, Routenberg DA, Das S. Elevation of neural injury markers in patients with neurologic sequelae after hospitalization for SARS-CoV-2 infection. iScience 2022; 25:104833. [PMID: 35937088 PMCID: PMC9341164 DOI: 10.1016/j.isci.2022.104833] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 06/08/2022] [Accepted: 07/20/2022] [Indexed: 11/13/2022] Open
Abstract
Patients with SARS-CoV-2 infection (COVID-19) risk developing long-term neurologic symptoms after infection. Here, we identify biomarkers associated with neurologic sequelae one year after hospitalization for SARS-CoV-2 infection. SARS-CoV-2-positive patients were followed using post-SARS-CoV-2 online questionnaires and virtual visits. Hospitalized adults from the pre-SARS-CoV-2 era served as historical controls. 40% of hospitalized patients develop neurological sequelae in the year after recovery from acute COVID-19 infection. Age, disease severity, and COVID-19 infection itself was associated with additional risk for neurological sequelae in our cohorts. Glial fibrillary astrocytic protein (GFAP) and neurofilament light chain (NF-L) were significantly elevated in SARS-CoV-2 infection. After adjusting for age, sex, and disease severity, GFAP and NF-L remained significantly associated with longer term neurological symptoms in patients with SARS-CoV-2 infection. GFAP and NF-L warrant exploration as biomarkers for long-term neurologic complications after SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Michail Spanos
- Cardiovascular Research Center, 185 Cambridge Street, Simches 3 Massachusetts General Hospital, Boston, MA, USA
| | - Sigal Shachar
- Meso Scale Diagnostics, LLC. (MSD), Rockville, MD, USA
| | - Thadryan Sweeney
- Cardiovascular Research Center, 185 Cambridge Street, Simches 3 Massachusetts General Hospital, Boston, MA, USA
| | - H. Immo Lehmann
- Cardiovascular Research Center, 185 Cambridge Street, Simches 3 Massachusetts General Hospital, Boston, MA, USA
| | - Priyanka Gokulnath
- Cardiovascular Research Center, 185 Cambridge Street, Simches 3 Massachusetts General Hospital, Boston, MA, USA
| | - Guoping Li
- Cardiovascular Research Center, 185 Cambridge Street, Simches 3 Massachusetts General Hospital, Boston, MA, USA
| | | | | | | | - Farhan Rana
- Cardiovascular Research Center, 185 Cambridge Street, Simches 3 Massachusetts General Hospital, Boston, MA, USA
| | - Ravi V. Shah
- Cardiovascular Research Center, 185 Cambridge Street, Simches 3 Massachusetts General Hospital, Boston, MA, USA
| | | | - Saumya Das
- Cardiovascular Research Center, 185 Cambridge Street, Simches 3 Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
11
|
Mechanisms and Biomarker Potential of Extracellular Vesicles in Stroke. BIOLOGY 2022; 11:biology11081231. [PMID: 36009857 PMCID: PMC9405035 DOI: 10.3390/biology11081231] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/29/2022] [Accepted: 08/12/2022] [Indexed: 12/11/2022]
Abstract
Simple Summary A stroke occurs when there is a lack of blood flow to the brain. Stroke injures the brain and can have devastating outcomes depending on the size and location of the brain tissue affected. Currently, there are only a limited number of treatment options for stroke. Extracellular vesicles are small vesicles secreted by cells. Importantly, extracellular vesicles have specific markers indicating the cell they were released from and can pass from the brain into the blood. For these reasons, assessing extracellular vesicles in the blood may create a window into changes occurring in the brain. Assessing changes in extracellular vesicles in the blood during stroke may produce new insight into the cellular changes in the brain causing injury during stroke. This in turn may generate potential targets for the development of future treatments. We summarize what is known about changes in brain-cell-specific extracellular vesicles during stroke and stress the importance of continuing to study these changes. Abstract Stoke is a prevalent and devastating neurologic condition with limited options for therapeutic management. Since brain tissue is rarely accessible clinically, peripheral biomarkers for the central nervous system’s (CNS’s) cellular response to stroke may prove critical for increasing our understanding of stroke pathology and elucidating novel therapeutic targets. Extracellular vesicles (EVs) are cell-derived, membrane-enclosed vesicles secreted by all cell types within the CNS that can freely pass the blood-brain barrier (BBB) and contain unique markers and content linked to their cell of origin. These unique qualities make brain-derived EVs novel candidates for non-invasive blood-based biomarkers of both cell specificity and cell physiological state during the progression of stroke and recovery. While studies are continuously emerging that are assessing the therapeutic potential of EVs and profiling EV cargo, a vast minority of these studies link EV content to specific cell types. A better understanding of cell-specific EV release during the acute, subacute, and chronic stages of stroke is needed to further elucidate the cellular processes responsible for stroke pathophysiology. Herein, we outline what is known about EV release from distinct cell types of the CNS during stroke and the potential of these EVs as peripheral biomarkers for cellular function in the CNS during stroke.
Collapse
|
12
|
Bourget C, Adams KV, Morshead CM. Reduced microglia activation following metformin administration or microglia ablation is sufficient to prevent functional deficits in a mouse model of neonatal stroke. J Neuroinflammation 2022; 19:146. [PMID: 35705953 PMCID: PMC9199194 DOI: 10.1186/s12974-022-02487-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/29/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Neonatal stroke is a devastating insult that can lead to life-long impairments. In response to hypoxic-ischaemic injury, there is loss of neurons and glia as well as a neuroinflammatory response mediated by resident immune cells, including microglia and astrocytes, which can exacerbate damage. Administration of the antidiabetic drug metformin has been shown to improve functional outcomes in preclinical models of brain injury and the cellular basis for metformin-mediated recovery is unknown. Given metformin's demonstrated anti-inflammatory properties, we investigated its role in regulating the microglia activation and used a microglia ablation strategy to investigate the microglia-mediated outcomes in a mouse model of neonatal stroke. METHODS Hypoxia-ischaemia (H-I) was performed on post-natal day 8. Metformin was administered for one week, starting one day after injury. Immunohistochemistry was used to examine the spatiotemporal response of microglia and astrocytes after hypoxia-ischaemia, with or without metformin treatment. To evaluate the effects of microglia depletion after hypoxia-ischaemia, we delivered Plexxikon 5622 for 1 or 2 weeks post-injury. The regional pattern of microglia and astrocyte depletion was assessed through immunohistochemistry. Motor behaviour was assessed with the righting reflex, hindlimb suspension, grip strength and cylinder tests. RESULTS Herein, we revealed a spatiotemporally regulated response of microglia and astrocytes after hypoxia-ischaemia. Metformin treatment after hypoxia-ischaemia had no effect on microglia number and proliferation, but significantly reduced microglia activation in all regions examined, concomitant with improved behavioural outcomes in injured mice. Plexxikon 5622 treatment successfully ablated microglia, resulting in a > 90% depletion in microglia in the neonatal brain. Microglia rapidly repopulated upon treatment cessation of Plexxikon. Most interesting, microglia ablation was sufficient to reduce functional deficits after hypoxia-ischaemia, mimicking the effects of 1 week of metformin treatment post-injury. CONCLUSION These results highlight the importance of regulating the neuroinflammatory response after neonatal stroke to promote recovery.
Collapse
Affiliation(s)
- Clara Bourget
- Institute of Medical Sciences, University of Toronto, Toronto, M5S1A8, Canada
| | - Kelsey V Adams
- Institute of Medical Sciences, University of Toronto, Toronto, M5S1A8, Canada
| | - Cindi M Morshead
- Institute of Medical Sciences, University of Toronto, Toronto, M5S1A8, Canada.
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Room 1006, Toronto, ON, M5S3E1, Canada.
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, M5S1A8, Canada.
- Institute of Biomedical Engineering, University of Toronto, Toronto, M5S3E1, Canada.
| |
Collapse
|
13
|
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: 6.0] [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
|
14
|
Takata F, Nakagawa S, Matsumoto J, Dohgu S. Blood-Brain Barrier Dysfunction Amplifies the Development of Neuroinflammation: Understanding of Cellular Events in Brain Microvascular Endothelial Cells for Prevention and Treatment of BBB Dysfunction. Front Cell Neurosci 2021; 15:661838. [PMID: 34588955 PMCID: PMC8475767 DOI: 10.3389/fncel.2021.661838] [Citation(s) in RCA: 205] [Impact Index Per Article: 68.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 08/09/2021] [Indexed: 12/11/2022] Open
Abstract
Neuroinflammation is involved in the onset or progression of various neurodegenerative diseases. Initiation of neuroinflammation is triggered by endogenous substances (damage-associated molecular patterns) and/or exogenous pathogens. Activation of glial cells (microglia and astrocytes) is widely recognized as a hallmark of neuroinflammation and triggers the release of proinflammatory cytokines, leading to neurotoxicity and neuronal dysfunction. Another feature associated with neuroinflammatory diseases is impairment of the blood-brain barrier (BBB). The BBB, which is composed of brain endothelial cells connected by tight junctions, maintains brain homeostasis and protects neurons. Impairment of this barrier allows trafficking of immune cells or plasma proteins into the brain parenchyma and subsequent inflammatory processes in the brain. Besides neurons, activated glial cells also affect BBB integrity. Therefore, BBB dysfunction can amplify neuroinflammation and act as a key process in the development of neuroinflammation. BBB integrity is determined by the integration of multiple signaling pathways within brain endothelial cells through intercellular communication between brain endothelial cells and brain perivascular cells (pericytes, astrocytes, microglia, and oligodendrocytes). For prevention of BBB disruption, both cellular components, such as signaling molecules in brain endothelial cells, and non-cellular components, such as inflammatory mediators released by perivascular cells, should be considered. Thus, understanding of intracellular signaling pathways that disrupt the BBB can provide novel treatments for neurological diseases associated with neuroinflammation. In this review, we discuss current knowledge regarding the underlying mechanisms involved in BBB impairment by inflammatory mediators released by perivascular cells.
Collapse
Affiliation(s)
- Fuyuko Takata
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Shinsuke Nakagawa
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Junichi Matsumoto
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Shinya Dohgu
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| |
Collapse
|
15
|
Lier J, Streit WJ, Bechmann I. Beyond Activation: Characterizing Microglial Functional Phenotypes. Cells 2021; 10:cells10092236. [PMID: 34571885 PMCID: PMC8464670 DOI: 10.3390/cells10092236] [Citation(s) in RCA: 111] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/18/2021] [Accepted: 08/26/2021] [Indexed: 12/20/2022] Open
Abstract
Classically, the following three morphological states of microglia have been defined: ramified, amoeboid and phagocytic. While ramified cells were long regarded as “resting”, amoeboid and phagocytic microglia were viewed as “activated”. In aged human brains, a fourth, morphologically novel state has been described, i.e., dystrophic microglia, which are thought to be senescent cells. Since microglia are not replenished by blood-borne mononuclear cells under physiological circumstances, they seem to have an “expiration date” limiting their capacity to phagocytose and support neurons. Identifying factors that drive microglial aging may thus be helpful to delay the onset of neurodegenerative diseases, such as Alzheimer’s disease (AD). Recent progress in single-cell deep sequencing methods allowed for more refined differentiation and revealed regional-, age- and sex-dependent differences of the microglial population, and a growing number of studies demonstrate various expression profiles defining microglial subpopulations. Given the heterogeneity of pathologic states in the central nervous system, the need for accurately describing microglial morphology and expression patterns becomes increasingly important. Here, we review commonly used microglial markers and their fluctuations in expression in health and disease, with a focus on IBA1 low/negative microglia, which can be found in individuals with liver disease.
Collapse
Affiliation(s)
- Julia Lier
- Institute of Anatomy, University of Leipzig, 04109 Leipzig, Germany;
- Department of Neurology, University of Leipzig, 04109 Leipzig, Germany
- Correspondence:
| | - Wolfgang J. Streit
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL 32611, USA;
| | - Ingo Bechmann
- Institute of Anatomy, University of Leipzig, 04109 Leipzig, Germany;
| |
Collapse
|
16
|
Saba ES, Karout M, Nasrallah L, Kobeissy F, Darwish H, Khoury SJ. Long-term cognitive deficits after traumatic brain injury associated with microglia activation. Clin Immunol 2021; 230:108815. [PMID: 34339843 DOI: 10.1016/j.clim.2021.108815] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 12/19/2022]
Abstract
Traumatic Brain Injury (TBI) is the most prevalent of all head injuries. Microglia play an essential role in homeostasis and diseases of the central nervous system. We hypothesize that microglia may play a beneficial or detrimental role in TBI depending on their state of activation and duration. In this study, we evaluated whether TBI results in a spatiotemporal change in microglia phenotype and whether it affects sensory-motor or learning and memory functions in male C57BL/6 mice. We used a panel of neurological and behavioral tests and a multi-color flow cytometry-based data analysis followed by unsupervised clustering to evaluate isolated microglia from injured brain tissue. We characterized several microglial phenotypes and their association with cognitive deficits. TBI results in a spatiotemporal increase in activated microglia that correlated negatively with spatial learning and memory at 35 days post-injury. These observations could define therapeutic windows and accelerate translational research to improve patient outcomes.
Collapse
Affiliation(s)
- Esber S Saba
- Department of Experimental Pathology, Immunology, and Microbiology, Faculty of Medicine, American University of Beirut, Lebanon
| | - Mona Karout
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut Medical Center, Lebanon
| | - Leila Nasrallah
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut Medical Center, Lebanon
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut Medical Center, Lebanon
| | - Hala Darwish
- Nehme and Therese Tohme Multiple Sclerosis Center, Faculty of Medicine, American University of Beirut Medical Center, Lebanon; Hariri School of Nursing, American University of Beirut, Lebanon.
| | - Samia J Khoury
- Department of Experimental Pathology, Immunology, and Microbiology, Faculty of Medicine, American University of Beirut, Lebanon; Nehme and Therese Tohme Multiple Sclerosis Center, Faculty of Medicine, American University of Beirut Medical Center, Lebanon.
| |
Collapse
|
17
|
Zwirner J, Lier J, Franke H, Hammer N, Matschke J, Trautz F, Tse R, Ondruschka B. GFAP positivity in neurons following traumatic brain injuries. Int J Legal Med 2021; 135:2323-2333. [PMID: 34114049 PMCID: PMC8523453 DOI: 10.1007/s00414-021-02568-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 03/10/2021] [Indexed: 11/29/2022]
Abstract
Glial fibrillary acidic protein (GFAP) is a well-established astrocytic biomarker for the diagnosis, monitoring and outcome prediction of traumatic brain injury (TBI). Few studies stated an accumulation of neuronal GFAP that was observed in various brain pathologies, including traumatic brain injuries. As the neuronal immunopositivity for GFAP in Alzheimer patients was shown to cross-react with non-GFAP epitopes, the neuronal immunopositivity for GFAP in TBI patients should be challenged. In this study, cerebral and cerebellar tissues of 52 TBI fatalities and 17 controls were screened for immunopositivity for GFAP in neurons by means of immunohistochemistry and immunofluorescence. The results revealed that neuronal immunopositivity for GFAP is most likely a staining artefact as negative controls also revealed neuronal GFAP staining. However, the phenomenon was twice as frequent for TBI fatalities compared to non-TBI control cases (12 vs. 6%). Neuronal GFAP staining was observed in the pericontusional zone and the ipsilateral hippocampus, but was absent in the contralateral cortex of TBI cases. Immunopositivity for GFAP was significantly correlated with the survival time (r = 0.306, P = 0.015), but no correlations were found with age at death, sex nor the post-mortem interval in TBI fatalities. This study provides evidence that the TBI-associated neuronal immunopositivity for GFAP is indeed a staining artefact. However, an absence post-traumatic neuronal GFAP cannot readily be assumed. Regardless of the particular mechanism, this study revealed that the artefact/potential neuronal immunopositivity for GFAP is a global, rather than a regional brain phenomenon and might be useful for minimum TBI survival time determinations, if certain exclusion criteria are strictly respected.
Collapse
Affiliation(s)
- Johann Zwirner
- Department of Anatomy, University of Otago, Dunedin, New Zealand. .,Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany. .,Institute of Legal Medicine, University of Leipzig, Leipzig, Germany.
| | - Julia Lier
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Heike Franke
- Rudolf Boehm Institute of Pharmacology and Toxicology, University of Leipzig, Leipzig, Germany
| | - Niels Hammer
- Institute of Macroscopic and Clinical Anatomy, University of Graz, Graz, Austria.,Department of Trauma, Orthopedic and Plastic Surgery, University Hospital of Leipzig, Leipzig, Germany.,Fraunhofer Institute for Machine Tools and Forming Technology, Dresden, Germany
| | - Jakob Matschke
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Florian Trautz
- Institute of Legal Medicine, University of Leipzig, Leipzig, Germany
| | - Rexon Tse
- Department of Forensic Pathology, LabPLUS, Auckland City Hospital, Auckland, New Zealand
| | - Benjamin Ondruschka
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
18
|
Sakai K, Takata F, Yamanaka G, Yasunaga M, Hashiguchi K, Tominaga K, Itoh K, Kataoka Y, Yamauchi A, Dohgu S. Reactive pericytes in early phase are involved in glial activation and late-onset hypersusceptibility to pilocarpine-induced seizures in traumatic brain injury model mice. J Pharmacol Sci 2021; 145:155-165. [PMID: 33357774 DOI: 10.1016/j.jphs.2020.11.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 10/27/2020] [Accepted: 11/20/2020] [Indexed: 02/07/2023] Open
Abstract
In this study, among neurovascular unit (NVU) cells, we focused on pericyte reactivity in mice subjected to controlled cortical impact (CCI) to understand how traumatic brain injury (TBI) causes uncoordinated crosstalk in the NVU and alters neuronal activity. Histological analyses of brain pericytes, microglia and astrocytes were performed for up to 28 days after CCI in the injured ipsilateral hippocampus. To evaluate altered neuronal activity caused by CCI, we measured seizure susceptibility to a sub-threshold dose of pilocarpine on postoperative day 7, 14, 21 and 28. Platelet-derived growth factor receptor (PDGFR) β immunoreactivity in pericytes significantly increased from 1 h to 4 days after CCI. The expression of Iba1 and GFAP, as markers of microglia and astrocytes, respectively, increased from 4 to 28 days after CCI. The severity of seizure induced by pilocarpine gradually increased, becoming significant at 28 days after CCI. Then, we treated CCI mice with an inhibitor of PDGFR signaling, imatinib, during the postoperative day 0-4 period. Imatinib lowered seizure susceptibility to pilocarpine and suppressed microglial activation in the injured hippocampus at postoperative day 28. These findings indicate that brain pericytes with rapidly increased PDGFRβ expression may drive TBI-induced dysregulation of NVU function and brain hyperexcitability.
Collapse
Affiliation(s)
- Kenta Sakai
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, 814-0180, Japan
| | - Fuyuko Takata
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, 814-0180, Japan.
| | - Gaku Yamanaka
- Department of Pediatrics and Adolescent Medicine, Tokyo Medical University, Tokyo, 160-0023, Japan
| | - Miho Yasunaga
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, 814-0180, Japan
| | - Kana Hashiguchi
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, 814-0180, Japan
| | - Kazuki Tominaga
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, 814-0180, Japan
| | - Kouichi Itoh
- Laboratory for Pharmacotherapy and Experimental Neurology, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Kagawa, 769-2193, Japan
| | - Yasufumi Kataoka
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, 814-0180, Japan
| | - Atsushi Yamauchi
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, 814-0180, Japan
| | - Shinya Dohgu
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, 814-0180, Japan
| |
Collapse
|