1
|
Dyhrfort P, Lindblad C, Widgren A, Virhammar J, Piehl F, Bergquist J, Al Nimer F, Rostami E. Deciphering Proteomic Expression in Inflammatory Disorders: A Mass Spectrometry Exploration Comparing Infectious, Noninfectious, and Traumatic Brain Injuries in Human Cerebrospinal Fluid. Neurotrauma Rep 2024; 5:857-873. [PMID: 39391051 PMCID: PMC11462427 DOI: 10.1089/neur.2024.0050] [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: 10/12/2024] Open
Abstract
The central nervous system (CNS) evokes a complex inflammatory response to injury. Inflammatory cascades are present in traumatic, infectious, and noninfectious disorders affecting the brain. It contains a mixture of pro- and anti-inflammatory reactions involving well-known proteins, but also numerous proteins less explored in these processes. The aim of this study was to explore the distinct inflammatory response in traumatic brain injury (TBI) compared with other CNS injuries by utilization of mass-spectrometry. In total, 56 patients had their cerebrospinal fluid (CSF) analyzed with the use of mass-spectrometry. Among these, CSF was collected via an external ventricular drain (EVD) from n = 21 patients with acute TBI. The resulting protein findings were then compared with CSF obtained by lumbar puncture from n = 14 patients with noninfectious CNS disorders comprising relapsing-remitting multiple sclerosis, anti-N-methyl-d-aspartate-receptor encephalitis, acute disseminated encephalomyelitis, and n = 14 patients with progressive multifocal leukoencephalopathy, herpes simplex encephalitis, and other types of viral meningitis. We also utilized n = 7 healthy controls (HCs). In the comparison between TBI and noninfectious inflammatory CNS disorders, concentrations of 55 proteins significantly differed between the groups. Among them, 23 and 32 proteins were up- and downregulated, respectively, in the TBI group. No proteins were uniquely identified in either group. In the comparison of TBI and HC, 51 proteins were significantly different, with 24 and 27 proteins being up- and downregulated, respectively, in TBI. Two proteins (fibrinogen gamma chain and transketolase) were uniquely identified in all samples of the TBI group. Also in the last comparison, TBI versus infectious inflammatory CNS disorders, 51 proteins differed between the two groups, with 19 and 32 proteins being up- and downregulated, respectively, in TBI, and no unique proteins being identified. Due to large discrepancies between the groups compared, the following proteins were selected for further deeper analysis among those being differentially regulated: APOE, CFB, CHGA, CHI3L1, C3, FCGBP, FGA, GSN, IGFBP7, LRG1, SERPINA3, SOD3, and TTR. We found distinct proteomic profiles in the CSF of TBI patients compared with HC and different disease controls, indicating a specific interplay between inflammatory factors, metabolic response, and cell integrity. In relation to primarily infectious or inflammatory disorders, unique inflammatory pathways seem to be engaged, and could potentially serve as future treatment targets.
Collapse
Affiliation(s)
- Philip Dyhrfort
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Caroline Lindblad
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- Department of Neurosurgery, Uppsala University Hospital, Uppsala, Sweden
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Neurosciences, Addenbrooke’s Hospital, Cambridge University, Turku, Finland
| | - Anna Widgren
- Department of Chemistry—BMC, Analytical Chemistry and Neurochemistry, Uppsala University, Uppsala, Sweden
| | - Johan Virhammar
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- Department of Neurology, Uppsala University Hospital, Uppsala, Sweden
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Center for Neurology, Academic Specialist Center, Stockholm, Sweden
| | - Jonas Bergquist
- Department of Chemistry—BMC, Analytical Chemistry and Neurochemistry, Uppsala University, Uppsala, Sweden
| | - Faiez Al Nimer
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Center for Neurology, Academic Specialist Center, Stockholm, Sweden
| | - Elham Rostami
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- Department of Neurosurgery, Uppsala University Hospital, Uppsala, Sweden
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
2
|
Smith CA, Carpenter KLH, Hutchinson PJ, Smielewski P, Helmy A. Candidate neuroinflammatory markers of cerebral autoregulation dysfunction in human acute brain injury. J Cereb Blood Flow Metab 2023; 43:1237-1253. [PMID: 37132274 PMCID: PMC10369156 DOI: 10.1177/0271678x231171991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 02/27/2023] [Accepted: 03/31/2023] [Indexed: 05/04/2023]
Abstract
The loss of cerebral autoregulation (CA) is a common and detrimental secondary injury mechanism following acute brain injury and has been associated with worse morbidity and mortality. However patient outcomes have not as yet been conclusively proven to have improved as a result of CA-directed therapy. While CA monitoring has been used to modify CPP targets, this approach cannot work if the impairment of CA is not simply related to CPP but involves other underlying mechanisms and triggers, which at present are largely unknown. Neuroinflammation, particularly inflammation affecting the cerebral vasculature, is an important cascade that occurs following acute injury. We hypothesise that disturbances to the cerebral vasculature can affect the regulation of CBF, and hence the vascular inflammatory pathways could be a putative mechanism that causes CA dysfunction. This review provides a brief overview of CA, and its impairment following brain injury. We discuss candidate vascular and endothelial markers and what is known about their link to disturbance of the CBF and autoregulation. We focus on human traumatic brain injury (TBI) and subarachnoid haemorrhage (SAH), with supporting evidence from animal work and applicability to wider neurologic diseases.
Collapse
Affiliation(s)
- Claudia A Smith
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Keri LH Carpenter
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Peter J Hutchinson
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Peter Smielewski
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Adel Helmy
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| |
Collapse
|
3
|
Brown LN, Barth JL, Jafri S, Rumschlag JA, Jenkins TR, Atkinson C, Lang H. Complement factor B is essential for the proper function of the peripheral auditory system. Front Neurol 2023; 14:1214408. [PMID: 37560455 PMCID: PMC10408708 DOI: 10.3389/fneur.2023.1214408] [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: 04/29/2023] [Accepted: 07/05/2023] [Indexed: 08/11/2023] Open
Abstract
Sensorineural hearing loss is associated with dysfunction of cochlear cells. Although immune cells play a critical role in maintaining the inner ear microenvironment, the precise immune-related molecular mechanisms underlying the pathophysiology of hearing loss remain unclear. The complement cascade contributes to the regulation of immune cell activity. Additionally, activation of the complement cascade can lead to the cellular opsonization of cells and pathogens, resulting in their engulfment and elimination by phagocytes. Complement factor B (fB) is an essential activator protein in the alternative complement pathway, and variations in the fB gene are associated with age-related macular degeneration. Here we show that mice of both sexes deficient in fB functional alleles (fB-/-) demonstrate progressive hearing impairment. Transcriptomic analysis of auditory nerves from adult mice detected 706 genes that were significantly differentially expressed between fB-/- and wild-type control animals, including genes related to the extracellular matrix and neural development processes. Additionally, a subset of differentially expressed genes was related to myelin function and neural crest development. Histological and immunohistochemical investigations revealed pathological alterations in auditory nerve myelin sheathes of fB-/- mice. Pathological alterations were also seen in the stria vascularis of the cochlear lateral wall in these mice. Our results implicate fB as an integral regulator of myelin maintenance and stria vascularis integrity, underscoring the importance of understanding the involvement of immune signaling pathways in sensorineural hearing loss.
Collapse
Affiliation(s)
- LaShardai N. Brown
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Jeremy L. Barth
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States
| | - Shabih Jafri
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Jeffrey A. Rumschlag
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Tyreek R. Jenkins
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Carl Atkinson
- Division of Pulmonary Medicine, University of Florida, Gainesville, FL, United States
| | - Hainan Lang
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
4
|
Yang Z, Nunn MA, Le TD, Simovic MO, Edsall PR, Liu B, Barr JL, Lund BJ, Hill-Pryor CD, Pusateri AE, Cancio LC, Li Y. Immunopathology of terminal complement activation and complement C5 blockade creating a pro-survival and organ-protective phenotype in trauma. Br J Pharmacol 2023; 180:422-440. [PMID: 36251578 PMCID: PMC10100417 DOI: 10.1111/bph.15970] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 06/24/2022] [Accepted: 09/17/2022] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND AND PURPOSE Traumatic haemorrhage (TH) is the leading cause of potentially preventable deaths that occur during the prehospital phase of care. No effective pharmacological therapeutics are available for critical TH patients yet. Here, we identify terminal complement activation (TCA) as a therapeutic target in combat casualties and evaluate the efficacy of a TCA inhibitor (nomacopan) on organ damage and survival in vivo. EXPERIMENTAL APPROACH Complement activation products and cytokines were analysed in plasma from 54 combat casualties. The correlations between activated complement pathway(s) and the clinical outcomes in trauma patients were assessed. Nomacopan was administered to rats subjected to lethal TH (blast injury and haemorrhagic shock). Effects of nomacopan on TH were determined using survival rate, organ damage, physiological parameters, and laboratory profiles. KEY RESULTS Early TCA was associated with systemic inflammatory responses and clinical outcomes in this trauma cohort. Lethal TH in the untreated rats induced early TCA that correlated with the severity of tissue damage and mortality. The addition of nomacopan to a damage-control resuscitation (DCR) protocol significantly inhibited TCA, decreased local and systemic inflammatory responses, improved haemodynamics and metabolism, attenuated tissue and organ damage, and increased survival. CONCLUSION AND IMPLICATIONS Previous findings of our and other groups revealed that early TCA represents a rational therapeutic target for trauma patients. Nomacopan as a pro-survival and organ-protective drug, could emerge as a promising adjunct to DCR that may significantly reduce the morbidity and mortality in severe TH patients while awaiting transport to critical care facilities.
Collapse
Affiliation(s)
- Zhangsheng Yang
- US Army Institute of Surgical Research, JBSA-Fort Sam Houston, San Antonio, Texas, USA
| | | | - Tuan D Le
- US Army Institute of Surgical Research, JBSA-Fort Sam Houston, San Antonio, Texas, USA
| | - Milomir O Simovic
- US Army Institute of Surgical Research, JBSA-Fort Sam Houston, San Antonio, Texas, USA.,The Geneva Foundation, Tacoma, Washington, USA
| | - Peter R Edsall
- US Army Institute of Surgical Research, JBSA-Fort Sam Houston, San Antonio, Texas, USA
| | - Bin Liu
- US Army Institute of Surgical Research, JBSA-Fort Sam Houston, San Antonio, Texas, USA
| | - Johnny L Barr
- US Army Institute of Surgical Research, JBSA-Fort Sam Houston, San Antonio, Texas, USA
| | - Brian J Lund
- 59th Medical Wing Operational Medicine, JBSA-Fort Sam Houston, San Antonio, Texas, USA
| | | | - Anthony E Pusateri
- Naval Medical Research Unit San Antonio, JBSA-Fort Sam Houston, San Antonio, Texas, USA
| | - Leopoldo C Cancio
- US Army Institute of Surgical Research, JBSA-Fort Sam Houston, San Antonio, Texas, USA
| | - Yansong Li
- US Army Institute of Surgical Research, JBSA-Fort Sam Houston, San Antonio, Texas, USA.,The Geneva Foundation, Tacoma, Washington, USA
| |
Collapse
|
5
|
van Erp IAM, Michailidou I, van Essen TA, van der Jagt M, Moojen W, Peul WC, Baas F, Fluiter K. Tackling Neuroinflammation After Traumatic Brain Injury: Complement Inhibition as a Therapy for Secondary Injury. Neurotherapeutics 2023; 20:284-303. [PMID: 36222978 PMCID: PMC10119357 DOI: 10.1007/s13311-022-01306-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/14/2022] [Indexed: 11/30/2022] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of mortality, sensorimotor morbidity, and neurocognitive disability. Neuroinflammation is one of the key drivers causing secondary brain injury after TBI. Therefore, attenuation of the inflammatory response is a potential therapeutic goal. This review summarizes the most important neuroinflammatory pathophysiology resulting from TBI and the clinical trials performed to attenuate neuroinflammation. Studies show that non-selective attenuation of the inflammatory response, in the early phase after TBI, might be detrimental and that there is a gap in the literature regarding pharmacological trials targeting specific pathways. The complement system and its crosstalk with the coagulation system play an important role in the pathophysiology of secondary brain injury after TBI. Therefore, regaining control over the complement cascades by inhibiting overshooting activation might constitute useful therapy. Activation of the complement cascade is an early component of neuroinflammation, making it a potential target to mitigate neuroinflammation in TBI. Therefore, we have described pathophysiological aspects of complement inhibition and summarized animal studies targeting the complement system in TBI. We also present the first clinical trial aimed at inhibition of complement activation in the early days after brain injury to reduce the risk of morbidity and mortality following severe TBI.
Collapse
Affiliation(s)
- Inge A M van Erp
- University Neurosurgical Center Holland, Leiden University Medical Center, Haaglanden Medical Center and HaGa Hospital, Leiden and The Hague, Albinusdreef 2, J-11-R-83, 2333 ZA, Leiden, The Netherlands.
| | - Iliana Michailidou
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Thomas A van Essen
- University Neurosurgical Center Holland, Leiden University Medical Center, Haaglanden Medical Center and HaGa Hospital, Leiden and The Hague, Albinusdreef 2, J-11-R-83, 2333 ZA, Leiden, The Netherlands
| | - Mathieu van der Jagt
- Department of Intensive Care Adults, Erasmus MC - University Medical Center, Rotterdam, The Netherlands
| | - Wouter Moojen
- University Neurosurgical Center Holland, Leiden University Medical Center, Haaglanden Medical Center and HaGa Hospital, Leiden and The Hague, Albinusdreef 2, J-11-R-83, 2333 ZA, Leiden, The Netherlands
| | - Wilco C Peul
- University Neurosurgical Center Holland, Leiden University Medical Center, Haaglanden Medical Center and HaGa Hospital, Leiden and The Hague, Albinusdreef 2, J-11-R-83, 2333 ZA, Leiden, The Netherlands
| | - Frank Baas
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Kees Fluiter
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
6
|
Alshareef M, Mallah K, Vasas T, Alawieh A, Borucki D, Couch C, Cutrone J, Shope C, Eskandari R, Tomlinson S. A Role of Complement in the Pathogenic Sequelae of Mouse Neonatal Germinal Matrix Hemorrhage. Int J Mol Sci 2022; 23:2943. [PMID: 35328364 PMCID: PMC8954718 DOI: 10.3390/ijms23062943] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/02/2022] [Accepted: 03/06/2022] [Indexed: 11/29/2022] Open
Abstract
Germinal matrix hemorrhage (GMH) is a devastating disease of infancy that results in intraventricular hemorrhage, post-hemorrhagic hydrocephalus (PHH), periventricular leukomalacia, and neurocognitive deficits. There are no curative treatments and limited surgical options. We developed and characterized a mouse model of GMH based on the injection of collagenase into the subventricular zone of post-natal pups and utilized the model to investigate the role of complement in PHH development. The site-targeted complement inhibitor CR2Crry, which binds deposited C3 complement activation products, localized specifically in the brain following its systemic administration after GMH. Compared to vehicle, CR2Crry treatment reduced PHH and lesion size, which was accompanied by decreased perilesional complement deposition, decreased astrocytosis and microgliosis, and the preservation of dendritic and neuronal density. Complement inhibition also improved survival and weight gain, and it improved motor performance and cognitive outcomes measured in adolescence. The progression to PHH, neuronal loss, and associated behavioral deficits was linked to the microglial phagocytosis of complement opsonized neurons, which was reversed with CR2Crry treatment. Thus, complement plays an important role in the pathological sequelae of GMH, and complement inhibition represents a novel therapeutic approach to reduce the disease progression of a condition for which there is currently no treatment outside of surgical intervention.
Collapse
Affiliation(s)
- Mohammed Alshareef
- Department of Neurological Surgery, Medical University of South Carolina, 301 CSB, Charleston, SC 29425, USA;
| | - Khalil Mallah
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB 204, MSC 504, Charleston, SC 29425, USA; (K.M.); (C.C.)
| | - Tyler Vasas
- College of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA; (T.V.); (D.B.); (J.C.); (C.S.)
| | - Ali Alawieh
- Department of Neurological Surgery, Emory University School of Medicine, Atlanta, GA 30322, USA;
| | - Davis Borucki
- College of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA; (T.V.); (D.B.); (J.C.); (C.S.)
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Christine Couch
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB 204, MSC 504, Charleston, SC 29425, USA; (K.M.); (C.C.)
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Jonathan Cutrone
- College of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA; (T.V.); (D.B.); (J.C.); (C.S.)
| | - Chelsea Shope
- College of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA; (T.V.); (D.B.); (J.C.); (C.S.)
| | - Ramin Eskandari
- Department of Neurological Surgery, Medical University of South Carolina, 301 CSB, Charleston, SC 29425, USA;
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB 204, MSC 504, Charleston, SC 29425, USA; (K.M.); (C.C.)
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB 204, MSC 504, Charleston, SC 29425, USA; (K.M.); (C.C.)
- Ralph Johnson VA Medical Center, Charleston, SC 29401, USA
| |
Collapse
|
7
|
van Erp IAM, van Essen TA, Fluiter K, van Zwet E, van Vliet P, Baas F, Haitsma I, Verbaan D, Coert B, de Ruiter GCW, Moojen WA, van der Jagt M, Peul WC. Safety and efficacy of C1-inhibitor in traumatic brain injury (CIAO@TBI): study protocol for a randomized, placebo-controlled, multi-center trial. Trials 2021; 22:874. [PMID: 34863258 PMCID: PMC8642972 DOI: 10.1186/s13063-021-05833-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 11/15/2021] [Indexed: 01/21/2023] Open
Abstract
Background Traumatic brain injury (TBI) is a major cause of death and disability across all ages. After the primary impact, the pathophysiologic process of secondary brain injury consists of a neuroinflammation response that critically leads to irreversible brain damage in the first days after the trauma. A key catalyst in this inflammatory process is the complement system. Inhibiting the complement system could therefore be a therapeutic target in TBI. Objective To study the safety and efficacy of C1-inhibitor (C1-INH) compared to placebo in patients with TBI. By temporarily blocking the complement system, we hypothesize a decrease in the posttraumatic neuroinflammatory response resulting in a less unfavorable clinical outcome for TBI patients. Methods CIAO@TBI is a multicenter, randomized, blinded, phase II placebo-controlled trial. Adult TBI patients with GCS < 13 requiring intracranial pressure (ICP) monitoring will be randomized, using block randomization, within 12 h after trauma to one dose 6000 IU C1-INH or placebo. A total of 106 patients will be included, and follow-up will occur up to 12 months. The primary endpoints are (1) Therapy Intensity Level (TIL) Scale, (2) Glasgow Outcome Scale-Extended (GOSE) at 6 months, and (3) complication rate during hospitalization. Outcomes will be determined by a trial nurse blinded for the treatment allocation. Analyses will be conducted in an intention-to-treat analysis. Discussion We expect that C1-INH administration will be safe and potentially effective to improve clinical outcomes by reducing neuroinflammation in TBI patients. Trial registration ClinicalTrials.gov NCT04489160. Registered on 27 July 2020. EudraCT 2020-000140-58 Supplementary Information The online version contains supplementary material available at 10.1186/s13063-021-05833-1.
Collapse
Affiliation(s)
- Inge A M van Erp
- University Neurosurgical Center Holland, Leiden University Medical Center, Haaglanden Medical Center and Haga Teaching Hospital, Albinusdreef 2, J-11-R-83, 2333 ZA, Leiden, Hague, The Netherlands.
| | - Thomas A van Essen
- University Neurosurgical Center Holland, Leiden University Medical Center, Haaglanden Medical Center and Haga Teaching Hospital, Albinusdreef 2, J-11-R-83, 2333 ZA, Leiden, Hague, The Netherlands
| | - Kees Fluiter
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Erik van Zwet
- Department of Biomedical Data Science, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter van Vliet
- Department of Intensive Care, Haaglanden Medical Center, The Hague, The Netherlands
| | - Frank Baas
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Iain Haitsma
- Department of Neurosurgery, Erasmus MC - University Medical Center, Rotterdam, The Netherlands
| | - Dagmar Verbaan
- Neurosurgical Center Amsterdam, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Bert Coert
- Neurosurgical Center Amsterdam, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Godard C W de Ruiter
- University Neurosurgical Center Holland, Leiden University Medical Center, Haaglanden Medical Center and Haga Teaching Hospital, Albinusdreef 2, J-11-R-83, 2333 ZA, Leiden, Hague, The Netherlands
| | - Wouter A Moojen
- University Neurosurgical Center Holland, Leiden University Medical Center, Haaglanden Medical Center and Haga Teaching Hospital, Albinusdreef 2, J-11-R-83, 2333 ZA, Leiden, Hague, The Netherlands
| | - Mathieu van der Jagt
- Department of Intensive Care Adults, Erasmus MC - University Medical Center, Rotterdam, The Netherlands
| | - Wilco C Peul
- University Neurosurgical Center Holland, Leiden University Medical Center, Haaglanden Medical Center and Haga Teaching Hospital, Albinusdreef 2, J-11-R-83, 2333 ZA, Leiden, Hague, The Netherlands
| |
Collapse
|
8
|
Chen M, Edwards SR, Reutens DC. Complement in the Development of Post-Traumatic Epilepsy: Prospects for Drug Repurposing. J Neurotrauma 2021; 37:692-705. [PMID: 32000582 DOI: 10.1089/neu.2019.6942] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Targeting neuroinflammation is a novel frontier in the prevention and treatment of epilepsy. A substantial body of evidence supports a key role for neuroinflammation in epileptogenesis, the pathological process that leads to the development and progression of spontaneous recurrent epileptic seizures. It is also well recognized that traumatic brain injury (TBI) induces a vigorous neuroinflammatory response and that a significant proportion of patients with TBI suffer from debilitating post-traumatic epilepsy. The complement system is a potent effector of innate immunity and a significant contributor to secondary tissue damage and to epileptogenesis following central nervous system injury. Several therapeutic agents targeting the complement system are already on the market to treat other central nervous system disorders or are well advanced in their development. The purpose of this review is to summarize findings on complement activation in experimental TBI and epilepsy models, highlighting the potential of drug repurposing in the development of therapeutics to ameliorate post-traumatic epileptogenesis.
Collapse
Affiliation(s)
- Min Chen
- Center for Advanced Imaging, University of Queensland, St. Lucia, Queensland, Australia
| | - Stephen R Edwards
- Center for Advanced Imaging, University of Queensland, St. Lucia, Queensland, Australia
| | - David C Reutens
- Center for Advanced Imaging, University of Queensland, St. Lucia, Queensland, Australia
| |
Collapse
|
9
|
Toutonji A, Mandava M, Guglietta S, Tomlinson S. Chronic complement dysregulation drives neuroinflammation after traumatic brain injury: a transcriptomic study. Acta Neuropathol Commun 2021; 9:126. [PMID: 34281628 PMCID: PMC8287781 DOI: 10.1186/s40478-021-01226-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 07/07/2021] [Indexed: 02/07/2023] Open
Abstract
Activation of the complement system propagates neuroinflammation and brain damage early and chronically after traumatic brain injury (TBI). The complement system is complex and comprises more than 50 components, many of which remain to be characterized in the normal and injured brain. Moreover, complement therapeutic studies have focused on a limited number of histopathological outcomes, which while informative, do not assess the effect of complement inhibition on neuroprotection and inflammation in a comprehensive manner. Using high throughput gene expression technology (NanoString), we simultaneously analyzed complement gene expression profiles with other neuroinflammatory pathway genes at different time points after TBI. We additionally assessed the effects of complement inhibition on neuropathological processes. Analyses of neuroinflammatory genes were performed at days 3, 7, and 28 post injury in male C57BL/6 mice following a controlled cortical impact injury. We also characterized the expression of 59 complement genes at similar time points, and also at 1- and 2-years post injury. Overall, TBI upregulated the expression of markers of astrogliosis, immune cell activation, and cellular stress, and downregulated the expression of neuronal and synaptic markers from day 3 through 28 post injury. Moreover, TBI upregulated gene expression across most complement activation and effector pathways, with an early emphasis on classical pathway genes and with continued upregulation of C2, C3 and C4 expression 2 years post injury. Treatment using the targeted complement inhibitor, CR2-Crry, significantly ameliorated TBI-induced transcriptomic changes at all time points. Nevertheless, some immune and synaptic genes remained dysregulated with CR2-Crry treatment, suggesting adjuvant anti-inflammatory and neurotropic therapy may confer additional neuroprotection. In addition to characterizing complement gene expression in the normal and aging brain, our results demonstrate broad and chronic dysregulation of the complement system after TBI, and strengthen the view that the complement system is an attractive target for TBI therapy.
Collapse
Affiliation(s)
- Amer Toutonji
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB 204, MSC 504, Charleston, SC 29425 USA
| | - Mamatha Mandava
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB 204, MSC 504, Charleston, SC 29425 USA
| | - Silvia Guglietta
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB 204, MSC 504, Charleston, SC 29425 USA
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB 204, MSC 504, Charleston, SC 29425 USA
- Ralph Johnson VA Medical Center, Charleston, SC 29401 USA
| |
Collapse
|
10
|
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
|
11
|
Ziabska K, Ziemka-Nalecz M, Pawelec P, Sypecka J, Zalewska T. Aberrant Complement System Activation in Neurological Disorders. Int J Mol Sci 2021; 22:4675. [PMID: 33925147 PMCID: PMC8125564 DOI: 10.3390/ijms22094675] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/22/2021] [Accepted: 04/26/2021] [Indexed: 12/13/2022] Open
Abstract
The complement system is an assembly of proteins that collectively participate in the functions of the healthy and diseased brain. The complement system plays an important role in the maintenance of uninjured (healthy) brain homeostasis, contributing to the clearance of invading pathogens and apoptotic cells, and limiting the inflammatory immune response. However, overactivation or underregulation of the entire complement cascade within the brain may lead to neuronal damage and disturbances in brain function. During the last decade, there has been a growing interest in the role that this cascading pathway plays in the neuropathology of a diverse array of brain disorders (e.g., acute neurotraumatic insult, chronic neurodegenerative diseases, and psychiatric disturbances) in which interruption of neuronal homeostasis triggers complement activation. Dysfunction of the complement promotes a disease-specific response that may have either beneficial or detrimental effects. Despite recent advances, the explicit link between complement component regulation and brain disorders remains unclear. Therefore, a comprehensible understanding of such relationships at different stages of diseases could provide new insight into potential therapeutic targets to ameliorate or slow progression of currently intractable disorders in the nervous system. Hence, the aim of this review is to provide a summary of the literature on the emerging role of the complement system in certain brain disorders.
Collapse
Affiliation(s)
| | | | | | | | - Teresa Zalewska
- Mossakowski Medical Research Centre, NeuroRepair Department, Polish Academy of Sciences, 5 Pawinskiego Street, 02-106 Warsaw, Poland; (K.Z.); (M.Z.-N.); (P.P.); (J.S.)
| |
Collapse
|
12
|
Garred P, Tenner AJ, Mollnes TE. Therapeutic Targeting of the Complement System: From Rare Diseases to Pandemics. Pharmacol Rev 2021; 73:792-827. [PMID: 33687995 PMCID: PMC7956994 DOI: 10.1124/pharmrev.120.000072] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The complement system was discovered at the end of the 19th century as a heat-labile plasma component that "complemented" the antibodies in killing microbes, hence the name "complement." Complement is also part of the innate immune system, protecting the host by recognition of pathogen-associated molecular patterns. However, complement is multifunctional far beyond infectious defense. It contributes to organ development, such as sculpting neuron synapses, promoting tissue regeneration and repair, and rapidly engaging and synergizing with a number of processes, including hemostasis leading to thromboinflammation. Complement is a double-edged sword. Although it usually protects the host, it may cause tissue damage when dysregulated or overactivated, such as in the systemic inflammatory reaction seen in trauma and sepsis and severe coronavirus disease 2019 (COVID-19). Damage-associated molecular patterns generated during ischemia-reperfusion injuries (myocardial infarction, stroke, and transplant dysfunction) and in chronic neurologic and rheumatic disease activate complement, thereby increasing damaging inflammation. Despite the long list of diseases with potential for ameliorating complement modulation, only a few rare diseases are approved for clinical treatment targeting complement. Those currently being efficiently treated include paroxysmal nocturnal hemoglobinuria, atypical hemolytic-uremic syndrome, myasthenia gravis, and neuromyelitis optica spectrum disorders. Rare diseases, unfortunately, preclude robust clinical trials. The increasing evidence for complement as a pathogenetic driver in many more common diseases suggests an opportunity for future complement therapy, which, however, requires robust clinical trials; one ongoing example is COVID-19 disease. The current review aims to discuss complement in disease pathogenesis and discuss future pharmacological strategies to treat these diseases with complement-targeted therapies. SIGNIFICANCE STATEMENT: The complement system is the host's defense friend by protecting it from invading pathogens, promoting tissue repair, and maintaining homeostasis. Complement is a double-edged sword, since when dysregulated or overactivated it becomes the host's enemy, leading to tissue damage, organ failure, and, in worst case, death. A number of acute and chronic diseases are candidates for pharmacological treatment to avoid complement-dependent damage, ranging from the well established treatment for rare diseases to possible future treatment of large patient groups like the pandemic coronavirus disease 2019.
Collapse
Affiliation(s)
- Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Rigshospitalet, Copenhagen, Denmark, and Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (P.G.); Departments of Molecular Biology and Biochemistry, Neurobiology and Behavior, and Pathology and Laboratory Medicine, University of California, Irvine, California (A.J.T.); and Research Laboratory, Nordland Hospital, Bodø, Norway, Faculty of Health Sciences, K.G. Jebsen TREC, University of Tromsø, Tromsø, Norway (T.E.M.); Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway (T.E.M.); and Department of Immunology, Oslo University Hospital and University of Oslo, Oslo, Norway (T.E.M.)
| | - Andrea J Tenner
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Rigshospitalet, Copenhagen, Denmark, and Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (P.G.); Departments of Molecular Biology and Biochemistry, Neurobiology and Behavior, and Pathology and Laboratory Medicine, University of California, Irvine, California (A.J.T.); and Research Laboratory, Nordland Hospital, Bodø, Norway, Faculty of Health Sciences, K.G. Jebsen TREC, University of Tromsø, Tromsø, Norway (T.E.M.); Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway (T.E.M.); and Department of Immunology, Oslo University Hospital and University of Oslo, Oslo, Norway (T.E.M.)
| | - Tom E Mollnes
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Rigshospitalet, Copenhagen, Denmark, and Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (P.G.); Departments of Molecular Biology and Biochemistry, Neurobiology and Behavior, and Pathology and Laboratory Medicine, University of California, Irvine, California (A.J.T.); and Research Laboratory, Nordland Hospital, Bodø, Norway, Faculty of Health Sciences, K.G. Jebsen TREC, University of Tromsø, Tromsø, Norway (T.E.M.); Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway (T.E.M.); and Department of Immunology, Oslo University Hospital and University of Oslo, Oslo, Norway (T.E.M.)
| |
Collapse
|
13
|
Verboon LN, Patel HC, Greenhalgh AD. The Immune System's Role in the Consequences of Mild Traumatic Brain Injury (Concussion). Front Immunol 2021; 12:620698. [PMID: 33679762 PMCID: PMC7928307 DOI: 10.3389/fimmu.2021.620698] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 01/25/2021] [Indexed: 12/14/2022] Open
Abstract
Mild traumatic brain injury (mild TBI), often referred to as concussion, is the most common form of TBI and affects millions of people each year. A history of mild TBI increases the risk of developing emotional and neurocognitive disorders later in life that can impact on day to day living. These include anxiety and depression, as well as neurodegenerative conditions such as chronic traumatic encephalopathy (CTE) and Alzheimer's disease (AD). Actions of brain resident or peripherally recruited immune cells are proposed to be key regulators across these diseases and mood disorders. Here, we will assess the impact of mild TBI on brain and patient health, and evaluate the recent evidence for immune cell involvement in its pathogenesis.
Collapse
Affiliation(s)
- Laura N. Verboon
- Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, The University of Manchester, Manchester, United Kingdom
| | - Hiren C. Patel
- Division of Cardiovascular Sciences, Salford Royal National Health Service Foundation Trust, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
- Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance National Health Service Group, University of Manchester, Manchester, United Kingdom
| | - Andrew D. Greenhalgh
- Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, The University of Manchester, Manchester, United Kingdom
- Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance National Health Service Group, University of Manchester, Manchester, United Kingdom
- Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
14
|
Schartz ND, Tenner AJ. The good, the bad, and the opportunities of the complement system in neurodegenerative disease. J Neuroinflammation 2020; 17:354. [PMID: 33239010 PMCID: PMC7690210 DOI: 10.1186/s12974-020-02024-8] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 11/04/2020] [Indexed: 02/06/2023] Open
Abstract
The complement cascade is a critical effector mechanism of the innate immune system that contributes to the rapid clearance of pathogens and dead or dying cells, as well as contributing to the extent and limit of the inflammatory immune response. In addition, some of the early components of this cascade have been clearly shown to play a beneficial role in synapse elimination during the development of the nervous system, although excessive complement-mediated synaptic pruning in the adult or injured brain may be detrimental in multiple neurogenerative disorders. While many of these later studies have been in mouse models, observations consistent with this notion have been reported in human postmortem examination of brain tissue. Increasing awareness of distinct roles of C1q, the initial recognition component of the classical complement pathway, that are independent of the rest of the complement cascade, as well as the relationship with other signaling pathways of inflammation (in the periphery as well as the central nervous system), highlights the need for a thorough understanding of these molecular entities and pathways to facilitate successful therapeutic design, including target identification, disease stage for treatment, and delivery in specific neurologic disorders. Here, we review the evidence for both beneficial and detrimental effects of complement components and activation products in multiple neurodegenerative disorders. Evidence for requisite co-factors for the diverse consequences are reviewed, as well as the recent studies that support the possibility of successful pharmacological approaches to suppress excessive and detrimental complement-mediated chronic inflammation, while preserving beneficial effects of complement components, to slow the progression of neurodegenerative disease.
Collapse
Affiliation(s)
- Nicole D. Schartz
- Department of Molecular Biology and Biochemistry, University of California Irvine, 3205 McGaugh Hall, Irvine, CA 92697 USA
| | - Andrea J. Tenner
- Department of Molecular Biology and Biochemistry, University of California Irvine, 3205 McGaugh Hall, Irvine, CA 92697 USA
- Department of Neurobiology and Behavior, University of California Irvine, 3205 McGaugh Hall, Irvine, CA 92697 USA
- Department of Pathology and Laboratory Medicine, University of California Irvine, 3205 McGaugh Hall, Irvine, CA 92697 USA
| |
Collapse
|
15
|
Role of ABT888, a Novel Poly(ADP-Ribose) Polymerase (PARP) Inhibitor in Countering Autophagy and Apoptotic Processes Associated to Spinal Cord Injury. Mol Neurobiol 2020; 57:4394-4407. [PMID: 32729104 DOI: 10.1007/s12035-020-02033-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/22/2020] [Indexed: 02/04/2023]
Abstract
Accidents are the cause of some 50 deaths per 100,000 population each year; some 3% of these are from traumatic spinal cord injury (SCI), a damage that causes temporary or permanent motor deficits, often leading to permanent neurological alterations. The activation of poly(ADP-ribose) polymerase (PARP) as DNA damage response, together with autophagy and apoptosis processes contributes to the secondary injury processes seen after SCI. Thus, in the present study, a mouse compression model of SCI was used to determine whether the treatment with ABT888, as PARP-1/2 inhibitor, could restore the neuronal damage induced by SCI. Mice were orally administered with ABT888 (at a dose of 25 mg/kg) 1 h and 6 h after SCI induction. Histological analysis, myeloperoxidase (MPO) activity, and Basso Mouse scale (BMS) were performed. The expression of autophagy-related proteins and apoptosis-inducing factors was quantified in the cytosolic fraction from spinal cord tissue collected after 24 h after SCI. TUNEL assay was performed in SCI-tissues 24 h after damage. ABT888 treatment significantly reduced histological damage and neutrophilic infiltration, improving motor skills. PARP-1/2 inhibition by ABT888 slowed cell death, decreasing autophagy-activation proteins. These results showed that ABT888, inhibiting PARP-1/2 activity, through a reduction in the apoptosis-autophagy machinery, plays a protective role after SCI, suggesting a new insight into the potential application of ABT888 as novel candidate in SCI therapies.
Collapse
|
16
|
Soluble terminal complement activation fragment sC5b-9: a new serum biomarker for traumatic brain injury? Eur J Trauma Emerg Surg 2020; 47:1491-1497. [DOI: 10.1007/s00068-020-01407-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 05/18/2020] [Indexed: 12/19/2022]
|
17
|
Anderson J, Patel M, Forenzo D, Ai X, Cai C, Wade Q, Risman R, Cai L. A novel mouse model for the study of endogenous neural stem and progenitor cells after traumatic brain injury. Exp Neurol 2020; 325:113119. [PMID: 31751572 PMCID: PMC10885014 DOI: 10.1016/j.expneurol.2019.113119] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 10/23/2019] [Accepted: 11/16/2019] [Indexed: 11/29/2022]
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability in the US. Neural stem/progenitor cells (NSPCs) persist in the adult brain and represent a potential cell source for tissue regeneration and wound healing after injury. The Notch signaling pathway is critical for embryonic development and adult brain injury response. However, the specific role of Notch signaling in the injured brain is not well characterized. Our previous study has established a Notch1CR2-GFP reporter mouse line in which the Notch1CR2 enhancer directs GFP expression in NSPCs and their progeny. In this study, we performed closed head injury (CHI) in the Notch1CR2-GFP mice to study the response of injury-activated NSPCs. We show that CHI induces neuroinflammation, cell death, and the expression of typical TBI markers (e.g., ApoE, Il1b, and Tau), validating the animal model. In addition, CHI induces cell proliferation in GFP+ cells expressing NSPC markers, e.g., Notch1 and Nestin. A significant higher percentage of GFP+ astrocytes and GABAergic neurons was observed in the injured brain, with no significant change in oligodendrocyte lineage between the CHI and sham animal groups. Since injury is known to activate astrogliosis, our results suggest that injury-induced GFP+ NSPCs preferentially differentiate into GABAergic neurons. Our study establishes that Notch1CR2-GFP transgenic mouse is a useful tool for the study of NSPC behavior in vivo after TBI. Unveiling the potential of NSPCs response to TBI (e.g., proliferation and differentiation) will identify new therapeutic strategy for the treatment of brain trauma.
Collapse
Affiliation(s)
- Jeremy Anderson
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, United States of America
| | - Misaal Patel
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, United States of America
| | - Dylan Forenzo
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, United States of America
| | - Xin Ai
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, United States of America
| | - Catherine Cai
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, United States of America
| | - Quinn Wade
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, United States of America
| | - Rebecca Risman
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, United States of America
| | - Li Cai
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, United States of America.
| |
Collapse
|
18
|
Dinet V, Petry KG, Badaut J. Brain-Immune Interactions and Neuroinflammation After Traumatic Brain Injury. Front Neurosci 2019; 13:1178. [PMID: 31780883 PMCID: PMC6861304 DOI: 10.3389/fnins.2019.01178] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 10/18/2019] [Indexed: 01/04/2023] Open
Abstract
Traumatic brain injury (TBI) is the principal cause of death and disability in children and young adults. Clinical and preclinical research efforts have been carried out to understand the acute, life-threatening pathophysiological events happening after TBI. In the past few years, however, it was recognized that TBI causes significant morbidity weeks, months, or years after the initial injury, thereby contributing substantially to the overall burden of TBI and the decrease of life expectancy in these patients. Long-lasting sequels of TBI include cognitive decline/dementia, sensory-motor dysfunction, and psychiatric disorders, and most important for patients is the need for socio-economic rehabilitation affecting their quality of life. Cerebrovascular alterations have been described during the first week after TBI for direct consequence development of neuroinflammatory process in relation to brain edema. Within the brain-immune interactions, the complement system, which is a family of blood and cell surface proteins, participates in the pathophysiology process. In fact, the complement system is part of the primary defense and clearance component of innate and adaptive immune response. In this review, the complement activation after TBI will be described in relation to the activation of the microglia and astrocytes as well as the blood-brain barrier dysfunction during the first week after the injury. Considering the neuroinflammatory activity as a causal element of neurological handicaps, some major parallel lines of complement activity in multiple sclerosis and Alzheimer pathologies with regard to cognitive impairment will be discussed for chronic TBI. A better understanding of the role of complement activation could facilitate the development of new therapeutic approaches for TBI.
Collapse
Affiliation(s)
- Virginie Dinet
- INSERM U1029, Angiogenesis and Neuroinflammation Group, University of Bordeaux, Bordeaux, France
| | - Klaus G. Petry
- INSERM U1029, Angiogenesis and Neuroinflammation Group, University of Bordeaux, Bordeaux, France
| | - Jerome Badaut
- CNRS UMR 5287, INCIA, Brain molecular Imaging Team, University of Bordeaux, Bordeaux, France
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States
| |
Collapse
|
19
|
Lee JD, Coulthard LG, Woodruff TM. Complement dysregulation in the central nervous system during development and disease. Semin Immunol 2019; 45:101340. [PMID: 31708347 DOI: 10.1016/j.smim.2019.101340] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 10/15/2019] [Accepted: 10/24/2019] [Indexed: 12/14/2022]
Abstract
The complement cascade is an important arm of the immune system that plays a key role in protecting the central nervous system (CNS) from infection. Recently, it has also become clear that complement proteins have fundamental roles in the developing and aging CNS that are distinct from their roles in immunity. During neurodevelopment, complement signalling is involved in diverse processes including neural tube closure, neural progenitor proliferation and differentiation, neuronal migration, and synaptic pruning. In acute neurotrauma and ischamic brain injury, complement drives inflammation and neuronal death, but also neuroprotection and regeneration. In diseases of the aging CNS including dementias and motor neuron disease, chronic complement activation is associated with glial activation, and synapse and neuron loss. Proper regulation of complement is thus essential to allow for an appropriately developed CNS and prevention of excessive damage following neurotrauma or during neurodegeneration. This review provides a comprehensive overview of the evidence for functional roles of complement in brain formation, and its dysregulation during acute and chronic disease. We also provide working models for how complement can lead to neurodevelopmental disorders such as schizophrenia and autism, and either protect, or propagate neurodegenerative diseases including Alzheimer's disease and amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- John D Lee
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Liam G Coulthard
- Royal Brisbane and Women's Hospital, Herston, Australia; School of Clinical Medicine, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Trent M Woodruff
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Australia.
| |
Collapse
|
20
|
Bodnar CN, Roberts KN, Higgins EK, Bachstetter AD. A Systematic Review of Closed Head Injury Models of Mild Traumatic Brain Injury in Mice and Rats. J Neurotrauma 2019; 36:1683-1706. [PMID: 30661454 PMCID: PMC6555186 DOI: 10.1089/neu.2018.6127] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Mild TBI (mTBI) is a significant health concern. Animal models of mTBI are essential for understanding mechanisms, and pathological outcomes, as well as to test therapeutic interventions. A variety of closed head models of mTBI that incorporate different aspects (i.e., biomechanics) of the mTBI have been reported. The aim of the current review was to compile a comprehensive list of the closed head mTBI rodent models, along with the common data elements, and outcomes, with the goal to summarize the current state of the field. Publications were identified from a search of PubMed and Web of Science and screened for eligibility following PRISMA guidelines. Articles were included that were closed head injuries in which the authors classified the injury as mild in rats or mice. Injury model and animal-specific common data elements, as well as behavioral and histological outcomes, were collected and compiled from a total of 402 articles. Our results outline the wide variety of methods used to model mTBI. We also discovered that female rodents and both young and aged animals are under-represented in experimental mTBI studies. Our findings will aid in providing context comparing the injury models and provide a starting point for the selection of the most appropriate model of mTBI to address a specific hypothesis. We believe this review will be a useful starting place for determining what has been done and what knowledge is missing in the field to reduce the burden of mTBI.
Collapse
Affiliation(s)
- Colleen N. Bodnar
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky
| | - Kelly N. Roberts
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky
| | - Emma K. Higgins
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky
| | - Adam D. Bachstetter
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
21
|
De Blasio D, Fumagalli S, Orsini F, Neglia L, Perego C, Ortolano F, Zanier ER, Picetti E, Locatelli M, Stocchetti N, Longhi L, Garred P, De Simoni MG. Human brain trauma severity is associated with lectin complement pathway activation. J Cereb Blood Flow Metab 2019; 39:794-807. [PMID: 29425056 PMCID: PMC6501516 DOI: 10.1177/0271678x18758881] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/03/2018] [Accepted: 01/15/2018] [Indexed: 12/25/2022]
Abstract
We explored the involvement of the lectin pathway of complement in post-traumatic brain injury (TBI) pathophysiology in humans. Brain samples were obtained from 28 patients who had undergone therapeutic contusion removal, within 12 h (early) or from >12 h until five days (late) from injury, and from five non-TBI patients. Imaging analysis indicated that lectin pathway initiator molecules (MBL, ficolin-1, ficolin-2 and ficolin-3), the key enzymes MASP-2 and MASP-3, and the downstream complement components (C3 fragments and TCC) were present inside and outside brain vessels in all contusions. Only ficolin-1 was found in the parenchyma of non-TBI tissues. Immunoassays in brain homogenates showed that MBL, ficolin-2 and ficolin-3 increased in TBI compared to non-TBI (2.0, 2.2 and 6.0-times) samples. MASP-2 increased with subarachnoid hemorrhage and abnormal pupil reactivity, two indicators of structural and functional damage. C3 fragments and TCC increased, respectively, by 3.5 - and 4.0-fold in TBI compared to non-TBI tissue and significantly correlated with MBL, ficolin-2, ficolin-3, MASP-2 and MASP-3 levels in the homogenates. In conclusion, we show for the first time the direct presence of lectin pathway components in human cerebral contusions and their association with injury severity, suggesting a central role for the lectin pathway in the post-traumatic pathophysiology of human TBI.
Collapse
Affiliation(s)
- Daiana De Blasio
- IRCCS-Istituto di Ricerche
Farmacologiche Mario Negri, Milano, Italy
| | - Stefano Fumagalli
- IRCCS-Istituto di Ricerche
Farmacologiche Mario Negri, Milano, Italy
| | - Franca Orsini
- IRCCS-Istituto di Ricerche
Farmacologiche Mario Negri, Milano, Italy
| | - Laura Neglia
- IRCCS-Istituto di Ricerche
Farmacologiche Mario Negri, Milano, Italy
| | - Carlo Perego
- IRCCS-Istituto di Ricerche
Farmacologiche Mario Negri, Milano, Italy
| | - Fabrizio Ortolano
- Department of Anesthesia and Critical
Care Medicine, Fondazione IRCCS Ca' Granda- Ospedale Maggiore Policlinico, Milano,
Italy
| | - Elisa R Zanier
- IRCCS-Istituto di Ricerche
Farmacologiche Mario Negri, Milano, Italy
| | - Edoardo Picetti
- Division of Anesthesia and Intensive
Care, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Marco Locatelli
- Department of Neurosurgery, Fondazione
IRCCS Ca' Granda-Ospedale Maggiore Policlinico, Milano, Italy
| | - Nino Stocchetti
- Department of Anesthesia and Critical
Care Medicine, Fondazione IRCCS Ca' Granda- Ospedale Maggiore Policlinico, Milano,
Italy
- Department of Physiopathology and
Transplantation, Milan University, Milan, Italy
| | - Luca Longhi
- Department of Anesthesia and Critical
Care Medicine, Neurosurgical Intensive Care Unit, Azienda Socio Sanitaria
Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Peter Garred
- Laboratory of Molecular Medicine,
Department of Clinical Immunology, Rigshospitalet Faculty of Medical and Health
Sciences, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
22
|
Carpanini SM, Torvell M, Morgan BP. Therapeutic Inhibition of the Complement System in Diseases of the Central Nervous System. Front Immunol 2019; 10:362. [PMID: 30886620 PMCID: PMC6409326 DOI: 10.3389/fimmu.2019.00362] [Citation(s) in RCA: 140] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 02/12/2019] [Indexed: 12/14/2022] Open
Abstract
The complement system plays critical roles in development, homeostasis, and regeneration in the central nervous system (CNS) throughout life; however, complement dysregulation in the CNS can lead to damage and disease. Complement proteins, regulators, and receptors are widely expressed throughout the CNS and, in many cases, are upregulated in disease. Genetic and epidemiological studies, cerebrospinal fluid (CSF) and plasma biomarker measurements and pathological analysis of post-mortem tissues have all implicated complement in multiple CNS diseases including multiple sclerosis (MS), neuromyelitis optica (NMO), neurotrauma, stroke, amyotrophic lateral sclerosis (ALS), Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD). Given this body of evidence implicating complement in diverse brain diseases, manipulating complement in the brain is an attractive prospect; however, the blood-brain barrier (BBB), critical to protect the brain from potentially harmful agents in the circulation, is also impermeable to current complement-targeting therapeutics, making drug design much more challenging. For example, antibody therapeutics administered systemically are essentially excluded from the brain. Recent protocols have utilized "Trojan horse" techniques to transport therapeutics across the BBB or used osmotic shock or ultrasound to temporarily disrupt the BBB. Most research to date exploring the impact of complement inhibition on CNS diseases has been in animal models, and some of these studies have generated convincing data; for example, in models of MS, NMO, and stroke. There have been a few recent clinical trials of available anti-complement drugs in CNS diseases associated with BBB impairment, for example the use of the anti-C5 monoclonal antibody (mAb) eculizumab in NMO, but for most CNS diseases there have been no human trials of anti-complement therapies. Here we will review the evidence implicating complement in diverse CNS disorders, from acute, such as traumatic brain or spine injury, to chronic, including demyelinating, neuroinflammatory, and neurodegenerative diseases. We will discuss the particular problems of drug access into the CNS and explore ways in which anti-complement therapies might be tailored for CNS disease.
Collapse
Affiliation(s)
- Sarah M Carpanini
- UK Dementia Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Megan Torvell
- UK Dementia Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Bryan Paul Morgan
- UK Dementia Research Institute, Cardiff University, Cardiff, United Kingdom.,Division of Infection and Immunity, School of Medicine, Systems Immunity Research Institute, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
23
|
Casili G, Campolo M, Paterniti I, Lanza M, Filippone A, Cuzzocrea S, Esposito E. Dimethyl Fumarate Attenuates Neuroinflammation and Neurobehavioral Deficits Induced by Experimental Traumatic Brain Injury. J Neurotrauma 2018; 35:1437-1451. [DOI: 10.1089/neu.2017.5260] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Affiliation(s)
- Giovanna Casili
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Michela Campolo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Irene Paterniti
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Marika Lanza
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Alessia Filippone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
- Department of Pharmacological and Physiological Science, Saint Louis University, St. Louis, Missouri
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| |
Collapse
|
24
|
Bao W, He F, Gao J, Meng F, Zou H, Luo B. Alpha-1-antitrypsin: a novel predictor for long-term recovery of chronic disorder of consciousness. Expert Rev Mol Diagn 2018; 18:307-313. [PMID: 29419340 DOI: 10.1080/14737159.2018.1438192] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND The aim of this manuscript was to explore the molecular basis and identify novel biomarkers for the diagnosis and prognosis of patients with chronic disorder of consciousness. METHODS A coupled isobaric tag for relative and absolute quantitation-based approach was used to screen differentially expressed proteins (DEPs) between patients with chronic disorder of consciousness and healthy individuals. Candidate proteins were identified and measured. The Coma Recovery Scale-Revised (CRS-R) score was used to quantify the severity, and long-term recovery was assessed by Glasgow Outcome Scale (GOS). RESULTS Between patients and controls, a total of 77 DEPs were identified. Based on the DEPs, a network containing 50 nodes and 207 edges was built, and alpha-1-antitrypsin was marked as the hub protein. The results indicated that alpha-1-antitrypsin correlated with the CRS-R score with a correlation coefficient of 0.631, and an outcome at 12 months (8.5 ± 2.1 ng/ml in patients with GOS 1-2 vs. 6.8 ± 1.6 ng/ml in those with GOS 3-5, p = 0.002). CONCLUSIONS The data confirm the diagnostic and prognostic potential of alpha-1-antitrypsin in chronic disorder of consciousness, which may contribute to the development of novel therapeutic agents.
Collapse
Affiliation(s)
- Wangxiao Bao
- a Department of Neurology, First Affiliated Hospital, Collaborative Innovation Center for Brain Science , Zhejiang University School of Medicine , Hangzhou , China
| | - Fangping He
- a Department of Neurology, First Affiliated Hospital, Collaborative Innovation Center for Brain Science , Zhejiang University School of Medicine , Hangzhou , China
| | - Jian Gao
- b Department of Rehabilitation , Hangzhou Hospital of Zhejiang CAPR , Hangzhou , China
| | - Fanxia Meng
- a Department of Neurology, First Affiliated Hospital, Collaborative Innovation Center for Brain Science , Zhejiang University School of Medicine , Hangzhou , China
| | - Hai Zou
- c Department of Cardiology , Zhejiang Provincial People's Hospital , Hangzhou , PR China.,d People's Hospital of Hangzhou Medical College , Hangzhou , China
| | - Benyan Luo
- a Department of Neurology, First Affiliated Hospital, Collaborative Innovation Center for Brain Science , Zhejiang University School of Medicine , Hangzhou , China
| |
Collapse
|
25
|
Identifying the Role of Complement in Triggering Neuroinflammation after Traumatic Brain Injury. J Neurosci 2018; 38:2519-2532. [PMID: 29437855 DOI: 10.1523/jneurosci.2197-17.2018] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 01/03/2018] [Accepted: 01/29/2018] [Indexed: 12/28/2022] Open
Abstract
The complement system is implicated in promoting acute secondary injury after traumatic brain injury (TBI), but its role in chronic post-traumatic neuropathology remains unclear. Using various injury-site targeted complement inhibitors that block different complement pathways and activation products, we investigated how complement is involved in neurodegeneration and chronic neuroinflammation after TBI in a clinically relevant setting of complement inhibition. The current paradigm is that complement propagates post-TBI neuropathology predominantly through the terminal membrane attack complex (MAC), but the focus has been on acute outcomes. Following controlled cortical impact in adult male mice, we demonstrate that although inhibition of the MAC (with CR2-CD59) reduces acute deficits, inhibition of C3 activation is required to prevent chronic inflammation and ongoing neuronal loss. Activation of C3 triggered a sustained degenerative mechanism of microglial and astrocyte activation, reduced dendritic and synaptic density, and inhibited neuroblast migration several weeks after TBI. Moreover, inhibiting all complement pathways (with CR2-Crry), or only the alternative complement pathway (with CR2-fH), provided similar and significant improvements in chronic histological, cognitive, and functional recovery, indicating a key role for the alternative pathway in propagating chronic post-TBI pathology. Although we confirm a role for the MAC in acute neuronal loss after TBI, this study shows that upstream products of complement activation generated predominantly via the alternative pathway propagate chronic neuroinflammation, thus challenging the current concept that the MAC represents a therapeutic target for treating TBI. A humanized version of CR2fH has been shown to be safe and non-immunogenic in clinical trials.SIGNIFICANCE STATEMENT Complement, and specifically the terminal membrane attack complex, has been implicated in secondary injury and neuronal loss after TBI. However, we demonstrate here that upstream complement activation products, generated predominantly via the alternative pathway, are responsible for propagating chronic inflammation and injury following CCI. Chronic inflammatory microgliosis is triggered by sustained complement activation after CCI, and is associated with chronic loss of neurons, dendrites and synapses, a process that continues to occur even 30 d after initial impact. Acute and injury-site targeted inhibition of the alternative pathway significantly improves chronic outcomes, and together these findings modify the conceptual paradigm for targeting the complement system to treat TBI.
Collapse
|
26
|
Hammad A, Westacott L, Zaben M. The role of the complement system in traumatic brain injury: a review. J Neuroinflammation 2018; 15:24. [PMID: 29357880 PMCID: PMC5778697 DOI: 10.1186/s12974-018-1066-z] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 01/15/2018] [Indexed: 02/08/2023] Open
Abstract
Traumatic brain injury (TBI) is an important cause of disability and mortality in the western world. While the initial injury sustained results in damage, it is the subsequent secondary cascade that is thought to be the significant determinant of subsequent outcomes. The changes associated with the secondary injury do not become irreversible until some time after the start of the cascade. This may present a window of opportunity for therapeutic interventions aiming to improve outcomes subsequent to TBI. A prominent contributor to the secondary injury is a multifaceted inflammatory reaction. The complement system plays a notable role in this inflammatory reaction; however, it has often been overlooked in the context of TBI secondary injury. The complement system has homeostatic functions in the uninjured central nervous system (CNS), playing a part in neurodevelopment as well as having protective functions in the fully developed CNS, including protection from infection and inflammation. In the context of CNS injury, it can have a number of deleterious effects, evidence for which primarily comes not only from animal models but also, to a lesser extent, from human post-mortem studies. In stark contrast to this, complement may also promote neurogenesis and plasticity subsequent to CNS injury. This review aims to explore the role of the complement system in TBI secondary injury, by examining evidence from both clinical and animal studies. We examine whether specific complement activation pathways play more prominent roles in TBI than others. We also explore the potential role of complement in post-TBI neuroprotection and CNS repair/regeneration. Finally, we highlight the therapeutic potential of targeting the complement system in the context of TBI and point out certain areas on which future research is needed.
Collapse
Affiliation(s)
- Adnan Hammad
- School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Laura Westacott
- Neuroscience and Mental Health Research Institute (NMHRI), School of Medicine, Cardiff University, Room 4FT 80E, 4th Floor, Heath Park, Cardiff, CF14 4XN UK
| | - Malik Zaben
- Neuroscience and Mental Health Research Institute (NMHRI), School of Medicine, Cardiff University, Room 4FT 80E, 4th Floor, Heath Park, Cardiff, CF14 4XN UK
| |
Collapse
|
27
|
Roselli F, Karasu E, Volpe C, Huber-Lang M. Medusa's Head: The Complement System in Traumatic Brain and Spinal Cord Injury. J Neurotrauma 2017; 35:226-240. [PMID: 28816089 DOI: 10.1089/neu.2017.5168] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Traumatic brain injury (TBI) and spinal cord injury (SCI) are critical medical conditions and a public health problem for which limited therapeutic options are available. The complement cascade is activated after TBI and SCI, and the resulting effects have been investigated in gene-knockout and pharmacological models. Multiple experimental studies support a net detrimental role of C3 and C5 activation in the early stages of TBI and SCI. Less firm experimental evidence suggests that, downstream of C3/C5, effector mechanisms, including the generation of membrane-activated complex and direct damage to membranes and neutrophils infiltration, may bring about the direct damage of central nervous system tissue and enhancement of neuroinflammation. The role of upstream classical, alternative, or extrinsic complement activation cascades remains unclear. Although several issues remain to be investigated, current evidence supports the investigation of a number of complement-targeting agents targeting C3 or C5, such as eculizumab, for repurposing in TBI and SCI treatment.
Collapse
Affiliation(s)
- Francesco Roselli
- 1 Department of Neurology, Medical School, University of Ulm , Ulm, Germany
- 2 Department of Anatomy and Cell Biology, Medical School, University of Ulm , Ulm, Germany
| | - Ebru Karasu
- 3 Institute of Clinical and Experimental Trauma-Immunology, Medical School, University of Ulm , Ulm, Germany
| | - Clara Volpe
- 1 Department of Neurology, Medical School, University of Ulm , Ulm, Germany
| | - Markus Huber-Lang
- 3 Institute of Clinical and Experimental Trauma-Immunology, Medical School, University of Ulm , Ulm, Germany
| |
Collapse
|
28
|
Neuroimmunology of Traumatic Brain Injury: Time for a Paradigm Shift. Neuron 2017; 95:1246-1265. [PMID: 28910616 DOI: 10.1016/j.neuron.2017.07.010] [Citation(s) in RCA: 474] [Impact Index Per Article: 59.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 07/07/2017] [Accepted: 07/10/2017] [Indexed: 12/14/2022]
Abstract
Traumatic brain injury (TBI) is a leading cause of morbidity and disability, with a considerable socioeconomic burden. Heterogeneity of pathoanatomical subtypes and diversity in the pathogenesis and extent of injury contribute to differences in the course and outcome of TBI. Following the primary injury, extensive and lasting damage is sustained through a complex cascade of events referred to as "secondary injury." Neuroinflammation is proposed as an important manipulable aspect of secondary injury in animal and human studies. Because neuroinflammation can be detrimental or beneficial, before developing immunomodulatory therapies, it is necessary to better understand the timing and complexity of the immune responses that follow TBI. With a rapidly increasing body of literature, there is a need for a clear summary of TBI neuroimmunology. This review presents our current understanding of the immune response to TBI in a chronological and compartment-based manner, highlighting early changes in gene expression and initial signaling pathways that lead to activation of innate and adaptive immunity. Based on recent advances in our understanding of innate immune cell activation, we propose a new paradigm to study innate immune cells following TBI that moves away from the existing M1/M2 classification of activation states toward a stimulus- and disease-specific understanding of polarization state based on transcriptomic and proteomic profiling.
Collapse
|
29
|
Bergdolt S, Kovtun A, Hägele Y, Liedert A, Schinke T, Amling M, Huber-Lang M, Ignatius A. Osteoblast-specific overexpression of complement receptor C5aR1 impairs fracture healing. PLoS One 2017; 12:e0179512. [PMID: 28614388 PMCID: PMC5470759 DOI: 10.1371/journal.pone.0179512] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 05/30/2017] [Indexed: 01/08/2023] Open
Abstract
The anaphylatoxin receptor C5aR1 plays an important role not only in innate immune responses, but also in bone metabolism and fracture healing, being highly expressed on immune and bone cells, including osteoblasts and osteoclasts. C5aR1 induces osteoblast migration, cytokine generation and osteoclastogenesis, however, the exact role of C5aR1-mediated signaling in osteoblasts is not entirely known. Therefore, we hypothesized that osteoblasts are essential target cells for C5a and that fracture healing should be disturbed in mice with an osteoblast-specific C5aR1 overexpression (Col1a1-C5aR1). Osteoblast activity in vitro, bone phenotype and fracture healing after isolated osteotomy and after combined osteotomy with additional thoracic trauma were analyzed. The systemic and local inflammatory reactions were analyzed by determining C5a and IL-6 concentrations in blood, bronchoalveolar lavage fluid and fracture callus and the recruitment of immune cells. In vitro, osteoblast proliferation and differentiation were similar to wildtype cells, and phosphorylation of p38 and expression of IL-6 and RANKL were increased in osteoblasts derived from Col1a1-C5aR1 mice. Bone phenotype and the inflammatory reaction were unaffected in Col1a1-C5aR1 mice. Fracture healing was significantly impaired as demonstrated by significantly reduced bone content, bone mineral density and flexural rigidity, possibly due to significantly increased osteoclast numbers. C5aR1 signaling in osteoblasts might possibly affect RANKL/OPG balance, leading to increased bone resorption. Additional trauma significantly impaired fracture healing, particularly in Col1a1-C5aR1 mice. In conclusion, the data indicate that C5aR1 signaling in osteoblasts plays a detrimental role in bone regeneration after fracture.
Collapse
Affiliation(s)
- Stephanie Bergdolt
- Institute of Orthopedic Research and Biomechanics, University of Ulm, Ulm, Germany
| | - Anna Kovtun
- Institute of Orthopedic Research and Biomechanics, University of Ulm, Ulm, Germany
| | - Yvonne Hägele
- Institute of Orthopedic Research and Biomechanics, University of Ulm, Ulm, Germany
| | - Astrid Liedert
- Institute of Orthopedic Research and Biomechanics, University of Ulm, Ulm, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Traumaimmunology, University of Ulm, Ulm, Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, University of Ulm, Ulm, Germany
- * E-mail:
| |
Collapse
|
30
|
Biggins PJC, Brennan FH, Taylor SM, Woodruff TM, Ruitenberg MJ. The Alternative Receptor for Complement Component 5a, C5aR2, Conveys Neuroprotection in Traumatic Spinal Cord Injury. J Neurotrauma 2017; 34:2075-2085. [PMID: 28173736 DOI: 10.1089/neu.2016.4701] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
This study investigated the role of the alternative receptor for complement activation fragment C5a, C5aR2, in secondary inflammatory pathology after contusive spinal cord injury (SCI) in mice. C5ar2-/- mice exhibited decreased intraparenchymal tumor necrosis factor alpha and interleukin-6 acutely post-injury, but these reductions did not translate into improved outcomes. We show that loss of C5aR2 leads to increased lesion volumes, reduced myelin sparing, and significantly worsened recovery from SCI in C5ar2-/- animals compared to wild-type (WT) controls. Loss of C5aR2 did not alter leukocyte mobilization from the bone marrow in response to SCI, and neutrophil recruitment/presence at the lesion site was also not different between genotypes. Acute treatment of SCI mice with the selective C5aR1 antagonist, PMX205, improved SCI outcomes, compared to vehicle controls, and, importantly, fully alleviated the worsened recovery of C5ar2-/- mice compared to their WT counterparts. Collectively, these findings indicate that C5aR2 is neuroprotective and a novel target to restrain injurious C5a signaling after a major neurotraumatic event.
Collapse
Affiliation(s)
- Patrick J C Biggins
- 1 School of Biomedical Sciences, The University of Queensland , Brisbane, Australia
| | - Faith H Brennan
- 1 School of Biomedical Sciences, The University of Queensland , Brisbane, Australia
| | - Stephen M Taylor
- 1 School of Biomedical Sciences, The University of Queensland , Brisbane, Australia
| | - Trent M Woodruff
- 1 School of Biomedical Sciences, The University of Queensland , Brisbane, Australia
| | - Marc J Ruitenberg
- 1 School of Biomedical Sciences, The University of Queensland , Brisbane, Australia .,2 Queensland Brain Institute, The University of Queensland , Brisbane, Australia .,3 Trauma, Critical Care and Recovery, Brisbane Diamantina Health Partners , Brisbane, Australia
| |
Collapse
|
31
|
Hess C, Kemper C. Complement-Mediated Regulation of Metabolism and Basic Cellular Processes. Immunity 2016; 45:240-54. [PMID: 27533012 PMCID: PMC5019180 DOI: 10.1016/j.immuni.2016.08.003] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 07/25/2016] [Accepted: 08/01/2016] [Indexed: 02/07/2023]
Abstract
Complement is well appreciated as a critical arm of innate immunity. It is required for the removal of invading pathogens and works by directly destroying them through the activation of innate and adaptive immune cells. However, complement activation and function is not confined to the extracellular space but also occurs within cells. Recent work indicates that complement activation regulates key metabolic pathways and thus can impact fundamental cellular processes, such as survival, proliferation, and autophagy. Newly identified functions of complement include a key role in shaping metabolic reprogramming, which underlies T cell effector differentiation, and a role as a nexus for interactions with other effector systems, in particular the inflammasome and Notch transcription-factor networks. This review focuses on the contributions of complement to basic processes of the cell, in particular the integration of complement with cellular metabolism and the potential implications in infection and other disease settings.
Collapse
Affiliation(s)
- Christoph Hess
- Department of Biomedicine, Immunobiology, University of Basel, 20 Hebelstrasse, 4031 Basel, Switzerland.
| | - Claudia Kemper
- Division of Transplant Immunology and Mucosal Biology, Medical Reseaerch Council Centre for Transplantation, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK; Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
32
|
Huber-Lang M, Gebhard F, Schmidt CQ, Palmer A, Denk S, Wiegner R. Complement therapeutic strategies in trauma, hemorrhagic shock and systemic inflammation – closing Pandora’s box? Semin Immunol 2016; 28:278-84. [DOI: 10.1016/j.smim.2016.04.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 04/19/2016] [Accepted: 04/20/2016] [Indexed: 12/21/2022]
|
33
|
Bahia El Idrissi N, Bosch S, Ramaglia V, Aronica E, Baas F, Troost D. Complement activation at the motor end-plates in amyotrophic lateral sclerosis. J Neuroinflammation 2016; 13:72. [PMID: 27056040 PMCID: PMC4823861 DOI: 10.1186/s12974-016-0538-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 03/31/2016] [Indexed: 02/07/2023] Open
Abstract
Background Amyotrophic lateral sclerosis (ALS) is a fatal progressive neurodegenerative disease with no available therapy. Components of the innate immune system are activated in the spinal cord and central nervous system of ALS patients. Studies in the SOD1G93A mouse show deposition of C1q and C3/C3b at the motor end-plate before neurological symptoms are apparent, suggesting that complement activation precedes neurodegeneration in this model. To obtain a better understanding of the role of complement at the motor end-plates in human ALS pathology, we analyzed post-mortem tissue of ALS donors for complement activation and its regulators. Methods Post-mortem intercostal muscle biopsies were collected at autopsy from ALS (n = 11) and control (n = 6) donors. The samples were analyzed for C1q, membrane attack complex (MAC), CD55, and CD59 on the motor end-plates, using immunofluorescence or immunohistochemistry. Results Here, we show that complement activation products and regulators are deposited on the motor end-plates of ALS patients. C1q co-localized with neurofilament in the intercostal muscle of ALS donors and was absent in controls (P = 0.001). In addition, C1q was found deposited on the motor end-plates in the intercostal muscle. MAC was also found deposited on motor end-plates that were innervated by nerves in the intercostal muscle of ALS donors but not in controls (P = 0.001). High levels of the regulators CD55 and CD59 were detected at the motor end-plates of ALS donors but not in controls, suggesting an attempt to counteract complement activation and prevent MAC deposition on the end-plates before they are lost. Conclusions This study provides evidence that complement activation products are deposited on innervated motor end-plates in the intercostal muscle of ALS donors, indicating that complement activation may precede end-plate denervation in human ALS. This study adds to the understanding of ALS pathology in man and identifies complement as a potential modifier of the disease process. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0538-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nawal Bahia El Idrissi
- Department of Genome Analysis, Academic Medical Center, Amsterdam, 1105 AZ, The Netherlands
| | - Sanne Bosch
- Department of Genome Analysis, Academic Medical Center, Amsterdam, 1105 AZ, The Netherlands
| | - Valeria Ramaglia
- Department of Genome Analysis, Academic Medical Center, Amsterdam, 1105 AZ, The Netherlands
| | - Eleonora Aronica
- Department of Neuropathology, Academic Medical Center, Amsterdam, 1105 AZ, The Netherlands
| | - Frank Baas
- Department of Genome Analysis, Academic Medical Center, Amsterdam, 1105 AZ, The Netherlands.
| | - Dirk Troost
- Department of Neuropathology, Academic Medical Center, Amsterdam, 1105 AZ, The Netherlands
| |
Collapse
|
34
|
Brennan FH, Lee JD, Ruitenberg MJ, Woodruff TM. Therapeutic targeting of complement to modify disease course and improve outcomes in neurological conditions. Semin Immunol 2016; 28:292-308. [PMID: 27049459 DOI: 10.1016/j.smim.2016.03.015] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 03/17/2016] [Accepted: 03/18/2016] [Indexed: 12/14/2022]
Abstract
The recognition that complement proteins are abundantly present and can have pathological roles in neurological conditions offers broad scope for therapeutic intervention. Accordingly, an increasing number of experimental investigations have explored the potential of harnessing the unique activation pathways, proteases, receptors, complexes, and natural inhibitors of complement, to mitigate pathology in acute neurotrauma and chronic neurodegenerative diseases. Here, we review mechanisms of complement activation in the central nervous system (CNS), and explore the effects of complement inhibition in cerebral ischemic-reperfusion injury, traumatic brain injury, spinal cord injury, Alzheimer's disease, amyotrophic lateral sclerosis, Parkinson's disease and Huntington's disease. We consider the challenges and opportunities arising from these studies. As complement therapies approach clinical translation, we provide perspectives on how promising complement-targeted therapeutics could become part of novel and effective future treatment options to improve outcomes in the initiation and progression stages of these debilitating CNS disorders.
Collapse
Affiliation(s)
- Faith H Brennan
- School of Biomedical Sciences, The University of Queensland, Brisbane 4072, Australia
| | - John D Lee
- School of Biomedical Sciences, The University of Queensland, Brisbane 4072, Australia
| | - Marc J Ruitenberg
- School of Biomedical Sciences, The University of Queensland, Brisbane 4072, Australia; Queensland Brain Institute, The University of Queensland, Brisbane 4072, Australia; Trauma, Critical Care and Recovery, Brisbane Diamantina Health Partners, The University of Queensland, Brisbane 4072, Australia
| | - Trent M Woodruff
- School of Biomedical Sciences, The University of Queensland, Brisbane 4072, Australia.
| |
Collapse
|
35
|
Rich MC, Keene CN, Neher MD, Johnson K, Yu ZX, Ganivet A, Holers VM, Stahel PF. Site-targeted complement inhibition by a complement receptor 2-conjugated inhibitor (mTT30) ameliorates post-injury neuropathology in mouse brains. Neurosci Lett 2016; 617:188-94. [PMID: 26892188 DOI: 10.1016/j.neulet.2016.02.025] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Revised: 02/10/2016] [Accepted: 02/12/2016] [Indexed: 11/26/2022]
Abstract
Intracerebral complement activation after severe traumatic brain injury (TBI) leads to a cascade of neuroinflammatory pathological sequelae that propagate host-mediated secondary brain injury and adverse outcomes. There are currently no specific pharmacological agents on the market to prevent or mitigate the development of secondary cerebral insults after TBI. A novel chimeric CR2-fH compound (mTT30) provides targeted inhibition of the alternative complement pathway at the site of tissue injury. This experimental study was designed to test the neuroprotective effects of mTT30 in a mouse model of closed head injury. The administration of 500 μg mTT30 i.v. at 1 h, 4 h and 24 h after head injury attenuated complement C3 deposition in injured brains, reduced the extent of neuronal cell death, and decreased post-injury microglial activation, compared to vehicle-injected placebo controls. These data imply that site-targeted alternative pathway complement inhibition may represent a new promising therapeutic avenue for the future management of severe TBI.
Collapse
Affiliation(s)
- Megan C Rich
- Department of Orthopaedic Surgery, Denver Health Medical Center and University of Colorado School of Medicine, Denver, CO 80204, USA
| | - Chesleigh N Keene
- Department of Orthopaedic Surgery, Denver Health Medical Center and University of Colorado School of Medicine, Denver, CO 80204, USA
| | - Miriam D Neher
- Department of Orthopaedic Surgery, Denver Health Medical Center and University of Colorado School of Medicine, Denver, CO 80204, USA
| | | | - Zhao-Xue Yu
- Alexion Pharmaceuticals, Cheshire, CT 06410, USA
| | - Antoine Ganivet
- Department of Orthopaedic Surgery, Denver Health Medical Center and University of Colorado School of Medicine, Denver, CO 80204, USA
| | - V Michael Holers
- Department of Medicine and Department of Immunology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Philip F Stahel
- Department of Orthopaedic Surgery, Denver Health Medical Center and University of Colorado School of Medicine, Denver, CO 80204, USA; Department of Neurosurgery, University of Colorado School of Medicine, Denver, CO 80204, USA.
| |
Collapse
|
36
|
An anticomplement agent that homes to the damaged brain and promotes recovery after traumatic brain injury in mice. Proc Natl Acad Sci U S A 2015; 112:14319-24. [PMID: 26578778 DOI: 10.1073/pnas.1513698112] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Activation of complement is a key determinant of neuropathology and disability after traumatic brain injury (TBI), and inhibition is neuroprotective. However, systemic complement is essential to fight infections, a critical complication of TBI. We describe a targeted complement inhibitor, comprising complement receptor of the Ig superfamily (CRIg) fused with complement regulator CD59a, designed to inhibit membrane attack complex (MAC) assembly at sites of C3b/iC3b deposition. CRIg and CD59a were linked via the IgG2a hinge, yielding CD59-2a-CRIg dimer with increased iC3b/C3b binding avidity and MAC inhibitory activity. CD59-2a-CRIg inhibited MAC formation and prevented complement-mediated lysis in vitro. CD59-2a-CRIg dimer bound C3b-coated surfaces with submicromolar affinity (KD). In experimental TBI, CD59-2a-CRIg administered posttrauma homed to sites of injury and significantly reduced MAC deposition, microglial accumulation, mitochondrial stress, and axonal damage and enhanced neurologic recovery compared with placebo controls. CD59-2a-CRIg inhibited MAC-induced inflammasome activation and IL-1β production in microglia. Given the important anti-infection roles of complement opsonization, site-targeted inhibition of MAC should be considered to promote recovery postneurotrauma.
Collapse
|
37
|
Surgucheva I, He S, Rich MC, Sharma R, Ninkina NN, Stahel PF, Surguchov A. Role of synucleins in traumatic brain injury — an experimental in vitro and in vivo study in mice. Mol Cell Neurosci 2015; 63:114-23. [PMID: 25447944 DOI: 10.1016/j.mcn.2014.10.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 10/16/2014] [Accepted: 10/23/2014] [Indexed: 11/25/2022] Open
Abstract
Synucleins are small prone to aggregate proteins associated with several neurodegenerative diseases (NDDs), however their role in traumatic brain injury (TBI) is an emerging area of investigation. Using in vitro scratch injury model and in vivo mouse weight-drop model we have found that the injury causes alterations in the expression and localization of synucleins near the damaged area. Before injury, α-synuclein is diffused in the cytoplasm of neurons and γ-synuclein is both in the cytoplasm and nucleus of oligodendrocytes. After the scratch injury of the mixed neuronal and glial culture, α-synuclein forms punctate structures in the cytoplasm of neurons and γ-synuclein is almost completely localized to the nucleus of the oligodendrocytes. Furthermore, the amount of post-translationally modified Met38-oxidized γ-synuclein is increased 3.8 fold 24 h after the scratch. α- and γ-synuclein containing cells increased in the initially cell free scratch zone up to 24 h after the scratch.Intracellular expression and localization of synucleins are also changed in a mouse model of focal closed head injury, using a standardized weight drop device. γ-Synuclein goes from diffuse to punctate staining in a piriform cortex near the amygdala, which may reflect the first steps in the formation of deposits/inclusions. Surprisingly, oxidized γ-synuclein co-localizes with cofilin-actin rods in the thalamus, which are absent in all other regions of the brain. These structures reach their peak amounts 7 days after injury. The changes in γ-synuclein localization are accompanied by injury-induced alterations in the morphology of both astrocytes and neurons.
Collapse
|
38
|
Is plasma C3 and C4 levels useful in young cerebral ischemic stroke patients? Associations with prognosis at 3 months. J Thromb Thrombolysis 2014; 39:209-14. [DOI: 10.1007/s11239-014-1100-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
39
|
Fluiter K, Opperhuizen AL, Morgan BP, Baas F, Ramaglia V. Inhibition of the membrane attack complex of the complement system reduces secondary neuroaxonal loss and promotes neurologic recovery after traumatic brain injury in mice. THE JOURNAL OF IMMUNOLOGY 2014; 192:2339-48. [PMID: 24489093 DOI: 10.4049/jimmunol.1302793] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Traumatic brain injury (TBI) is the leading cause of disability and death in young adults. The secondary neuroinflammation and neuronal damage that follows the primary mechanical injury is an important cause of disability in affected people. The membrane attack complex (MAC) of the complement system is detected in the traumatized brain early after TBI; however, its role in the pathology and neurologic outcome of TBI has not yet been investigated. We generated a C6 antisense oligonucleotide that blocks MAC formation by inhibiting C6, and we compared its therapeutic effect to that of Ornithodoros moubata complement inhibitor (OmCI), a known inhibitor of C5 activation that blocks generation of the anaphylatoxin C5a and C5b, an essential component of MAC. Severe closed head injury in mice induced abundant MAC deposition in the brain. Treatment with C6 antisense reduced C6 synthesis (85%) and serum levels (90%), and inhibited MAC deposition in the injured brain (91-96%). Treatment also reduced accumulation of microglia/macrophages (50-88%), neuronal apoptosis, axonal loss and weight loss (54-93%), and enhanced neurologic performance (84-92%) compared with placebo-treated controls after injury. These data provide the first evidence, to our knowledge, that inhibition of MAC formation in otherwise complement-sufficient animals reduces neuropathology and promotes neurologic recovery after TBI. Given the importance of maintaining a functional complement opsonization system to fight infections, a critical complication in TBI patients, inhibition of the MAC should be considered to reduce posttraumatic neurologic damage. This work identifies a novel therapeutic target for TBI and will guide the development of new therapy for patients.
Collapse
Affiliation(s)
- Kees Fluiter
- Department of Genome Analysis, Academic Medical Center, 1105 AZ Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
40
|
Li Y, Chavko M, Slack JL, Liu B, McCarron RM, Ross JD, Dalle Lucca JJ. Protective effects of decay-accelerating factor on blast-induced neurotrauma in rats. Acta Neuropathol Commun 2013; 1:52. [PMID: 24252631 PMCID: PMC3893442 DOI: 10.1186/2051-5960-1-52] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 08/05/2013] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Blast-induced neurotrauma (BINT) is the signature life threatening injury of current military casualties. Neuroinflammation is a key pathological occurrence of secondary injury contributing to brain damage after blast injury. We have recently demonstrated that blast-triggered complement activation and cytokine release are associated with BINT. Here, we evaluated if administration of the complement inhibitor recombinant human decay-accelerating factor (rhDAF) is beneficial on neuroinflammation and neurodegeneration in a rat model of moderate BINT. Administration of rhDAF after exposure to moderate blast overpressure (BOP, 120 kPa) mitigated brain injury characterized by neuronal degeneration. rhDAF treatment reduced complement hemolytic activity at 3 hours and tissue complement deposition at 3, 24, and 48 hours as well as systemic and local cytokine release at 24 hours post BOP. Furthermore, rhDAF protected blood-brain barrier (BBB) integrity and reduced cytotoxic edema. Interaction between complement cleavage component, C3a and C3a receptor and tau phosphorylation were also attenuated in rhDAF treated animals at 3 and 24 hours after BOP. These novel findings suggest early complement targeted inhibition as a new therapeutic strategy to decrease neuroinflammation and neurodegeneration after blast TBI. RESULT Administration of rhDAF after exposure to moderate blast overpressure (BOP, 120 kPa) mitigated brain injury characterized by neuronal degeneration. rhDAF treatment reduced complement hemolytic activity at 3 hours and tissue complement deposition at 3, 24, and 48 hours as well as systemic and local cytokine release at 24 hours post BOP. Furthermore, rhDAF protected blood-brain barrier (BBB) integrity and reduced cytotoxic edema. Interaction between complement cleavage component, C3a and C3a receptor and tau phosphorylation were also attenuated in rhDAF treated animals at 3 and 24 hours after BOP. CONCLUSION These novel findings suggest early complement targeted inhibition as a new therapeutic strategy to decrease neuroinflammation and neurodegeneration after blast TBI.
Collapse
|
41
|
Hu X, Holers VM, Thurman JM, Schoeb TR, Ramos TN, Barnum SR. Therapeutic inhibition of the alternative complement pathway attenuates chronic EAE. Mol Immunol 2013; 54:302-8. [PMID: 23337717 PMCID: PMC3602149 DOI: 10.1016/j.molimm.2012.12.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2012] [Accepted: 12/14/2012] [Indexed: 12/13/2022]
Abstract
Previous studies from our laboratory using complement-mutant mice demonstrated that the alternative pathway is the dominant activation pathway responsible for complement-mediated pathology in demyelinating disease. Using a well-characterized inhibitory monoclonal antibody (mAb 1379) directed against mouse factor B, we assessed the therapeutic value of inhibiting the alternative complement pathway in experimental autoimmune encephalomyelitis (EAE), the animal model for multiple sclerosis. Administration of anti-factor B antibody to mice prior to the onset of clinical signs of active EAE had no affect on the onset or acute phase of disease, but significantly attenuated the chronic phase of disease resulting in reduced cellular infiltration, inflammation and demyelination in antibody-treated mice. Attenuation of the chronic phase of disease was long lasting even though antibody administration was terminated shortly after disease onset. Chronic disease was also attenuated in transferred EAE when anti-factor B antibody was administered before or after disease onset. Similar levels of disease attenuation were observed in transferred EAE using MOG-specific encephalitogenic T cells. These studies demonstrate the therapeutic potential for inhibition of factor B in the chronic phase of demyelinating disease, where treatment options are limited.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- Complement Factor B/classification
- Complement Pathway, Alternative/drug effects
- Complement Pathway, Alternative/immunology
- Complement System Proteins/immunology
- Demyelinating Diseases/immunology
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Inflammation/immunology
- Mice
- Mice, Inbred C57BL
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Xianzhen Hu
- Department of Microbiology, University of Alabama at Birmingham, 845 19th St. S., Birmingham, AL 35294, USA
| | | | | | | | | | | |
Collapse
|
42
|
Abstract
Multiorgan failure (MOF) represents the leading cause of death in patients with sepsis and systemic inflammatory response syndrome (SIRS) following severe trauma. The underlying immune response is highly complex and involves activation of the complement system as a crucial entity of innate immunity. Uncontrolled activation of the complement system during sepsis and SIRS with in excessive generation of complement activation products contributes to an ensuing dysfunction of various organ systems. In the present review, mechanisms of the inflammatory response in the development of MOF in sepsis and SIRS with particular focus on the complement system are discussed.
Collapse
|
43
|
Stephan AH, Barres BA, Stevens B. The complement system: an unexpected role in synaptic pruning during development and disease. Annu Rev Neurosci 2012; 35:369-89. [PMID: 22715882 DOI: 10.1146/annurev-neuro-061010-113810] [Citation(s) in RCA: 752] [Impact Index Per Article: 57.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
An unexpected role for the classical complement cascade in the elimination of central nervous system (CNS) synapses has recently been discovered. Complement proteins are localized to developing CNS synapses during periods of active synapse elimination and are required for normal brain wiring. The function of complement proteins in the brain appears analogous to their function in the immune system: clearance of cellular material that has been tagged for elimination. Similarly, synapses tagged with complement proteins may be eliminated by microglial cells expressing complement receptors. In addition, developing astrocytes release signals that induce the expression of complement components in the CNS. In the mature brain, early synapse loss is a hallmark of several neurodegenerative diseases. Complement proteins are profoundly upregulated in many CNS diseases prior to signs of neuron loss, suggesting a reactivation of similar developmental mechanisms of complement-mediated synapse elimination potentially driving disease progression.
Collapse
Affiliation(s)
- Alexander H Stephan
- Department of Neurobiology, Stanford University School of Medicine, Stanford, California 94305-5125, USA.
| | | | | |
Collapse
|
44
|
Elvington A, Atkinson C, Zhu H, Yu J, Takahashi K, Stahl GL, Kindy MS, Tomlinson S. The alternative complement pathway propagates inflammation and injury in murine ischemic stroke. THE JOURNAL OF IMMUNOLOGY 2012; 189:4640-7. [PMID: 23028050 DOI: 10.4049/jimmunol.1201904] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
There is mounting evidence indicating an important role for complement in the pathogenesis of cerebral ischemia-reperfusion injury, or ischemic stroke. The role of the alternative complement pathway in ischemic stroke has not been investigated, and there is conflicting data on the role of the terminal pathway. In this study, we show that compared with wild-type mice, mice deficient in the alternative pathway protein factor B or mice treated with the alternative pathway inhibitor CR2-fH have improved outcomes after 60-min middle cerebral artery occlusion and 24-h reperfusion. Factor B-deficient or CR2-fH-treated mice were protected in terms of improved neurologic function and reduced cerebral infarct, demyelination, P-selectin expression, neutrophil infiltration, and microthrombi formation. Mice deficient in both the classical and lectin pathways (C1q/MBL deficient) were also protected from cerebral ischemia-reperfusion injury, and there was no detectable C3d deposition in the ipsilateral brain of these mice. These data demonstrate that the alternative pathway is not alone sufficient to initiate complement activation and indicate that the alternative pathway propagates cerebral injury via amplification of the cascade. Deficiency of C6, a component of the terminal cytolytic membrane attack complex, had no effect on outcome after ischemic stroke, indicating that the membrane attack complex is not involved in mediating injury in this model. We additionally show that the protective effect of factor B deficiency and CR2-fH treatment is sustained in the subacute stage of infarct development, adding to the clinical relevance of these findings.
Collapse
Affiliation(s)
- Andrew Elvington
- Department of Microbiology and Immunology, Children's Research Institute, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Ehrnthaller C, Amara U, Weckbach S, Kalbitz M, Huber-Lang M, Bahrami S. Alteration of complement hemolytic activity in different trauma and sepsis models. J Inflamm Res 2012; 5:59-66. [PMID: 22879778 PMCID: PMC3413207 DOI: 10.2147/jir.s31787] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Indexed: 11/23/2022] Open
Abstract
Complement activation is involved in various diseases in which innate immunity plays a crucial role. However, its pathophysiological relevance is not clearly understood. Experimental models have been widely used to characterize the role of complement activation under different pathological conditions, such as hypoxemia, ischemia and reperfusion, tissue damage, and polymicrobial invasion. Screening of the complement status and function is, however, strongly dependent on the laboratory-specific techniques being used to sample and measure complement, making it difficult to compare the results found in different laboratories. Therefore, we evaluated complement function by measuring complement hemolytic activity (CH50) in various animal models of isolated ischemia reperfusion (I/R: kidney, liver, gut), hemorrhagic traumatic shock (HTS), endotoxic shock (LPS), and sepsis (CLP). Complement activation was less pronounced in isolated models of ischemia and reperfusion, whereas a strong complement response was observed early after HTS, CLP, and LPS. In summary, CH50 is a well-established, quick, and cost-effective screening method of complement function. However, because we obtained different results in clinically relevant animal models, further differentiation using specific complement factor analysis is necessary.
Collapse
Affiliation(s)
- Christian Ehrnthaller
- Department of Traumatology, Hand, Plastic, and Reconstructive Surgery, Center of Surgery, University of Ulm, Germany
| | | | | | | | | | | |
Collapse
|
46
|
NMDA preconditioning prevents object recognition memory impairment and increases brain viability in mice exposed to traumatic brain injury. Brain Res 2012; 1466:82-90. [DOI: 10.1016/j.brainres.2012.05.041] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Revised: 05/09/2012] [Accepted: 05/21/2012] [Indexed: 12/22/2022]
|
47
|
Brennan FH, Anderson AJ, Taylor SM, Woodruff TM, Ruitenberg MJ. Complement activation in the injured central nervous system: another dual-edged sword? J Neuroinflammation 2012; 9:137. [PMID: 22721265 PMCID: PMC3464784 DOI: 10.1186/1742-2094-9-137] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 06/21/2012] [Indexed: 11/28/2022] Open
Abstract
The complement system, a major component of the innate immune system, is becoming increasingly recognised as a key participant in physiology and disease. The awareness that immunological mediators support various aspects of both normal central nervous system (CNS) function and pathology has led to a renaissance of complement research in neuroscience. Various studies have revealed particularly novel findings on the wide-ranging involvement of complement in neural development, synapse elimination and maturation of neural networks, as well as the progression of pathology in a range of chronic neurodegenerative disorders, and more recently, neurotraumatic events, where rapid disruption of neuronal homeostasis potently triggers complement activation. The purpose of this review is to summarise recent findings on complement activation and acquired brain or spinal cord injury, i.e. ischaemic-reperfusion injury or stroke, traumatic brain injury (TBI) and spinal cord injury (SCI), highlighting the potential for complement-targeted therapeutics to alleviate the devastating consequences of these neurological conditions.
Collapse
Affiliation(s)
- Faith H Brennan
- The University of Queensland, School of Biomedical Sciences, St Lucia, Brisbane, QLD 4072, Australia
| | | | | | | | | |
Collapse
|
48
|
Guilty molecules, guilty minds? The conflicting roles of the innate immune response to traumatic brain injury. Mediators Inflamm 2012; 2012:356494. [PMID: 22701273 PMCID: PMC3373171 DOI: 10.1155/2012/356494] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Accepted: 03/26/2012] [Indexed: 12/11/2022] Open
Abstract
Traumatic brain injury (TBI) is a complex disease in the most complex organ of the body, whose victims endure lifelong debilitating physical, emotional, and psychosocial consequences. Despite advances in clinical care, there is no effective neuroprotective therapy for TBI, with almost every compound showing promise experimentally having disappointing results in the clinic. The complex and highly interrelated innate immune responses govern both the beneficial and deleterious molecular consequences of TBI and are present as an attractive therapeutic target. This paper discusses the positive, negative, and often conflicting roles of the innate immune response to TBI in both an experimental and clinical settings and highlights recent advances in the search for therapeutic candidates for the treatment of TBI.
Collapse
|
49
|
Weckbach S, Neher M, Losacco JT, Bolden AL, Kulik L, Flierl MA, Bell SE, Holers VM, Stahel PF. Challenging the role of adaptive immunity in neurotrauma: Rag1(-/-) mice lacking mature B and T cells do not show neuroprotection after closed head injury. J Neurotrauma 2012; 29:1233-42. [PMID: 22335783 DOI: 10.1089/neu.2011.2169] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The role of adaptive immunity in contributing to post-traumatic neuroinflammation and neuropathology after head injury remains largely unexplored. The present study was designed to investigate the pathophysiological sequelae of closed head injury in Rag1(-/-) mice devoid of mature B and T lymphocytes. C57BL/6 wild-type and Rag1(-/-) mice were subjected to experimental closed head injury, using a standardized weight-drop device. Outcome parameters consisted of neurological scoring, quantification of blood-brain barrier (BBB) function, measurement of inflammatory markers and mediators of apoptosis in serum and brain tissue, and assessment of neuronal cell death, astrogliosis, and tissue destruction. There was no difference between wild-type and Rag1(-/-) mice with regard to injury severity and neurological impairment for up to 7 days after head injury. The extent of BBB dysfunction was in a similar range for both groups. Quantification of complement activation fragments in serum revealed significantly attenuated C3a levels in Rag1(-/-) mice compared to wild-type animals. In contrast, the levels of pro- and anti-inflammatory cytokines and pro-apoptotic and anti-apoptotic mediators remained in a similar range for both groups, and the histological analysis of brain sections did not reveal a difference in reactive astrogliosis, tissue destruction, and neuronal cell death in Rag1(-/-) compared to wild-type mice. These findings suggest that adaptive immunity is not of crucial importance for initiating and sustaining the inflammatory neuropathology after closed head injury. The attenuated extent of post-traumatic complement activation seen in Rag1(-/-) mice implies a cross-talk between innate and adaptive immune responses, which requires further investigation in future studies.
Collapse
Affiliation(s)
- Sebastian Weckbach
- Department of Orthopaedics, Denver Health Medical Center, University of Colorado School of Medicine, Denver, Colorado 80204, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Neher MD, Weckbach S, Flierl MA, Huber-Lang MS, Stahel PF. Molecular mechanisms of inflammation and tissue injury after major trauma--is complement the "bad guy"? J Biomed Sci 2011; 18:90. [PMID: 22129197 PMCID: PMC3247859 DOI: 10.1186/1423-0127-18-90] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2011] [Accepted: 11/30/2011] [Indexed: 02/07/2023] Open
Abstract
Trauma represents the leading cause of death among young people in industrialized countries. Recent clinical and experimental studies have brought increasing evidence for activation of the innate immune system in contributing to the pathogenesis of trauma-induced sequelae and adverse outcome. As the "first line of defense", the complement system represents a potent effector arm of innate immunity, and has been implicated in mediating the early posttraumatic inflammatory response. Despite its generic beneficial functions, including pathogen elimination and immediate response to danger signals, complement activation may exert detrimental effects after trauma, in terms of mounting an "innocent bystander" attack on host tissue. Posttraumatic ischemia/reperfusion injuries represent the classic entity of complement-mediated tissue damage, adding to the "antigenic load" by exacerbation of local and systemic inflammation and release of toxic mediators. These pathophysiological sequelae have been shown to sustain the systemic inflammatory response syndrome after major trauma, and can ultimately contribute to remote organ injury and death. Numerous experimental models have been designed in recent years with the aim of mimicking the inflammatory reaction after trauma and to allow the testing of new pharmacological approaches, including the emergent concept of site-targeted complement inhibition. The present review provides an overview on the current understanding of the cellular and molecular mechanisms of complement activation after major trauma, with an emphasis of emerging therapeutic concepts which may provide the rationale for a "bench-to-bedside" approach in the design of future pharmacological strategies.
Collapse
Affiliation(s)
- Miriam D Neher
- Department of Orthopaedic Surgery, University of Colorado Denver, School of Medicine, Denver Health Medical Center, 777 Bannock Street, Denver, CO 80204, USA
| | | | | | | | | |
Collapse
|