1
|
Steggerda JA, Heeger PS. The Promise of Complement Therapeutics in Solid Organ Transplantation. Transplantation 2024; 108:1882-1894. [PMID: 38361233 DOI: 10.1097/tp.0000000000004927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Transplantation is the ideal therapy for end-stage organ failure, but outcomes for all transplant organs are suboptimal, underscoring the need to develop novel approaches to improve graft survival and function. The complement system, traditionally considered a component of innate immunity, is now known to broadly control inflammation and crucially contribute to induction and function of adaptive T-cell and B-cell immune responses, including those induced by alloantigens. Interest of pharmaceutical industries in complement therapeutics for nontransplant indications and the understanding that the complement system contributes to solid organ transplantation injury through multiple mechanisms raise the possibility that targeting specific complement components could improve transplant outcomes and patient health. Here, we provide an overview of complement biology and review the roles and mechanisms through which the complement system is pathogenically linked to solid organ transplant injury. We then discuss how this knowledge has been translated into novel therapeutic strategies to improve organ transplant outcomes and identify areas for future investigation. Although the clinical application of complement-targeted therapies in transplantation remains in its infancy, the increasing availability of new agents in this arena provides a rich environment for potentially transformative translational transplant research.
Collapse
Affiliation(s)
- Justin A Steggerda
- Division of Abdominal Transplant Surgery, Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Peter S Heeger
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA
- Division of Nephrology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA
| |
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
|
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
|
4
|
Heja D, Zhao D, Cody E, Cumpelik A, Lim PC, Prado-Acosta M, Palma L, Dellepiane S, Chun N, Ferrara J, Heeger PS. Mannan-Binding Lectin Promotes Murine Graft-versus-Host Disease by Amplifying Lipopolysaccharide-Initiated Inflammation. Transplant Cell Ther 2022; 28:472.e1-472.e11. [PMID: 35643350 PMCID: PMC9357100 DOI: 10.1016/j.jtct.2022.05.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 11/27/2022]
Abstract
Conditioning regimens used for hematopoietic stem cell transplantation (HCT) can escalate the severity of acute T cell-mediated graft-versus-host disease (GVHD) by disrupting gastrointestinal integrity and initiating lipopolysaccharide (LPS)-dependent innate immune cell activation. Activation of the complement cascade has been associated with murine GVHD, and previous work has shown that alternative pathway complement activation can amplify T cell immunity. Whether and how mannan-binding lectin (MBL), a component of the complement system that binds mannose as well as oligosaccharide components of LPS and lipoteichoic acid, affects GVHD is unknown. In this study, we tested the hypothesis that MBL modulates murine GVHD and examined the mechanisms by which it does so. We adoptively transferred C3.SW bone marrow (BM) cells ± T cells into irradiated wild type (WT) or MBL-deficient C57Bl/6 (B6) recipients with or without inhibiting MBL-initiated complement activation using C1-esterase inhibitor (C1-INH). We analyzed the clinical severity of disease expression and analyzed intestinal gene and cell infiltration. In vitro studies assessed MBL expression on antigen-presenting cells (APCs) and compared LPS-induced responses of WT and MBL-deficient APCs. MBL-deficient recipients of donor BM ± T cells exhibited significantly less weight loss over the first 2 weeks post-transplantation weeks compared with B6 controls (P < .05), with similar donor engraftment in the 2 groups. In recipients of C3.SW BM + T cells, the clinical expression of GVHD was less severe (P < .05) and overall survival was better (P < .05) in MBL-deficient mice compared with WT mice. On day-7 post-transplantation, analyses showed that the MBL-deficient recipients exhibited less intestinal IL1b, IL17, and IL12 p40 gene expression (P < .05 for each) and fewer infiltrating intestinal CD11c+, CD11b+, and F4/80+ cells and TCRβ+, CD4+, CD4+IL17+, and CD8+ T cells (P < .05 for each). Ovalbumin or allogeneic cell immunizations induced equivalent T cell responses in MBL-deficient and WT mice, demonstrating that MBL-deficiency does not directly impact T cell immunity in the absence of irradiation conditioning. Administration of C1-INH did not alter the clinical expression of GVHD in preconditioned WT B6 recipients, suggesting that MBL amplifies clinical expression of GVHD via a complement-independent mechanism. WT, but not MBL-deficient, APCs express MBL on their surfaces. LPS-stimulated APCs from MBL-deficient mice produced less proinflammatory cytokines (P < .05) and induced weaker alloreactive T cell responses (P < .05) compared with WT APCs. Together, our data show that MBL modulates murine GVHD, likely by amplifying complement-independent, LPS-initiated gastrointestinal inflammation. The results suggest that devising strategies to block LPS/MBL ligation on APCs has the potential to reduce the clinical expression of GVHD.
Collapse
Affiliation(s)
- David Heja
- Translational Transplant Research Center, Tisch Cancer Institute, Precision Immunology Institute, Tisch Cancer Institute and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Dongchang Zhao
- Translational Transplant Research Center, Tisch Cancer Institute, Precision Immunology Institute, Tisch Cancer Institute and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Evan Cody
- Translational Transplant Research Center, Tisch Cancer Institute, Precision Immunology Institute, Tisch Cancer Institute and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Arun Cumpelik
- Translational Transplant Research Center, Tisch Cancer Institute, Precision Immunology Institute, Tisch Cancer Institute and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Pik Chin Lim
- Translational Transplant Research Center, Tisch Cancer Institute, Precision Immunology Institute, Tisch Cancer Institute and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Mariano Prado-Acosta
- Translational Transplant Research Center, Tisch Cancer Institute, Precision Immunology Institute, Tisch Cancer Institute and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Liv Palma
- Translational Transplant Research Center, Tisch Cancer Institute, Precision Immunology Institute, Tisch Cancer Institute and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sergio Dellepiane
- Translational Transplant Research Center, Tisch Cancer Institute, Precision Immunology Institute, Tisch Cancer Institute and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Nicholas Chun
- Translational Transplant Research Center, Tisch Cancer Institute, Precision Immunology Institute, Tisch Cancer Institute and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - James Ferrara
- Translational Transplant Research Center, Tisch Cancer Institute, Precision Immunology Institute, Tisch Cancer Institute and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Peter S Heeger
- Translational Transplant Research Center, Tisch Cancer Institute, Precision Immunology Institute, Tisch Cancer Institute and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
5
|
Wu L, Chung JY, Cao T, Jin G, Edmiston WJ, Hickman S, Levy ES, Whalen JA, Abrams ESL, Degterev A, Lo EH, Tozzi L, Kaplan DL, El Khoury J, Whalen MJ. Genetic inhibition of RIPK3 ameliorates functional outcome in controlled cortical impact independent of necroptosis. Cell Death Dis 2021; 12:1064. [PMID: 34753914 PMCID: PMC8578385 DOI: 10.1038/s41419-021-04333-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 09/20/2021] [Accepted: 10/04/2021] [Indexed: 02/05/2023]
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability with no specific effective therapy, in part because disease driving mechanisms remain to be elucidated. Receptor interacting protein kinases (RIPKs) are serine/threonine kinases that assemble multi-molecular complexes that induce apoptosis, necroptosis, inflammasome and nuclear factor kappa B activation. Prior studies using pharmacological inhibitors implicated necroptosis in the pathogenesis of TBI and stroke, but these studies cannot be used to conclusively demonstrate a role for necroptosis because of the possibility of off target effects. Using a model of cerebral contusion and RIPK3 and mixed lineage kinase like knockout (MLKL-/-) mice, we found evidence for activation of RIPK3 and MLKL and assembly of a RIPK1-RIPK3-MLKL necrosome complex in pericontusional brain tissue. Phosphorylated forms of RIPK3 and MLKL were detected in endothelium, CD11b + immune cells, and neurons, and RIPK3 was upregulated and activated in three-dimensional human endothelial cell cultures subjected to CCI. RIPK3-/- and MLKL-/- mice had reduced blood-brain barrier damage at 24 h (p < 0.05), but no differences in neuronal death (6 h, p = ns in CA1, CA3 and DG), brain edema (24 h, p = ns), or lesion size (4 weeks, p = ns) after CCI. RIPK3-/-, but not MLKL-/- mice, were protected against postinjury motor and cognitive deficits at 1-4 weeks (RIPK3-/- vs WT: p < 0.05 for group in wire grip, Morris water maze hidden platform trials, p < 0.05 for novel object recognition test, p < 0.01 for rotarod test). RIPK3-/- mice had reduced infiltrating leukocytes (p < 0.05 vs WT in CD11b + cells, microglia and macrophages), HMGB1 release and interleukin-1 beta activation at 24-48 h (p < 0.01) after CCI. Our data indicate that RIPK3 contributes to functional outcome after cerebral contusion by mechanisms involving inflammation but independent of necroptosis.
Collapse
Affiliation(s)
- Limin Wu
- grid.38142.3c000000041936754XDepartment of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA
| | - Joon Yong Chung
- grid.38142.3c000000041936754XDepartment of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA
| | - Tian Cao
- grid.38142.3c000000041936754XDepartment of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA ,grid.13291.380000 0001 0807 1581Department of Neurology, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan China
| | - Gina Jin
- grid.38142.3c000000041936754XDepartment of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA
| | - William J. Edmiston
- grid.38142.3c000000041936754XDepartment of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA
| | - Suzanne Hickman
- grid.32224.350000 0004 0386 9924Department of Medicine, Center for Immunology and Inflammatory Disease, Massachusetts General Hospital, Boston, USA
| | - Emily S. Levy
- grid.38142.3c000000041936754XDepartment of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA
| | - Jordyn A. Whalen
- grid.38142.3c000000041936754XDepartment of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA
| | - Eliza Sophie LaRovere Abrams
- grid.38142.3c000000041936754XDepartment of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA
| | - Alexei Degterev
- grid.67033.310000 0000 8934 4045Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA USA
| | - Eng H. Lo
- grid.32224.350000 0004 0386 9924Department of Radiology, Massachusetts General Hospital, Boston, MA 02115 USA ,grid.32224.350000 0004 0386 9924Department of Neurology, Massachusetts General Hospital, Boston, MA 02115 USA
| | - Lorenzo Tozzi
- grid.429997.80000 0004 1936 7531Department of Biomedical Engineering, Tufts University, Medford, MA 02155 USA
| | - David L. Kaplan
- grid.429997.80000 0004 1936 7531Department of Biomedical Engineering, Tufts University, Medford, MA 02155 USA
| | - Joseph El Khoury
- grid.32224.350000 0004 0386 9924Department of Medicine, Center for Immunology and Inflammatory Disease, Massachusetts General Hospital, Boston, USA
| | - Michael J. Whalen
- grid.38142.3c000000041936754XDepartment of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA
| |
Collapse
|
6
|
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: 10] [Impact Index Per Article: 2.5] [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
|
7
|
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: 29] [Impact Index Per Article: 7.3] [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
|
8
|
Auriti C, Prencipe G, Inglese R, Moriondo M, Nieddu F, Mondì V, Longo D, Bucci S, Del Pinto T, Timelli L, Di Ciommo VM. Mannose Binding Lectin, S100 B Protein, and Brain Injuries in Neonates With Perinatal Asphyxia. Front Pediatr 2020; 8:527. [PMID: 33042903 PMCID: PMC7527601 DOI: 10.3389/fped.2020.00527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 07/24/2020] [Indexed: 11/13/2022] Open
Abstract
Perinatal asphyxia triggers an acute inflammatory response in the injured brain. Complement activation and neuroinflammation worsen brain damage after a systemic ischemia/reperfusion insult. The increase of mannose binding lectin (MBL) during asphyxia may contribute to the brain damage, via activation of the complement lectin pathway. The possible role of MBL2 gene variants in influencing the severity of post-asphyxia brain injuries is still unexplored. This retrospective study included 53 asphyxiated neonates: 42 underwent therapeutic hypothermia (TH) and 11 did not because they were admitted to the NICU later than 6 h after the hypoxic insult. Blood samples from TH-treated and untreated patients were genotyped for MBL2 gene variants, and biomarker plasma levels (MBL and S100 B protein) were measured at different time points: during hypothermia, during rewarming, and at 7-10 days of life. The timing of blood sampling, except for the T1 sample, was the same in untreated infants. Highest (peak) levels of MBL and MBL2 genotypes were correlated to neuroimaging brain damage or death and long-term neurodevelopmental delay. MBL2 wild-type genotype was associated with the highest MBL levels and worst brain damage on MRI (p = 0.046) at 7-10 days after hypoxia. MBL increased in both groups and S100B decreased, slightly more in treated than in untreated neonates. The progressive increase of MBL (p = 0.08) and to be untreated with TH (p = 0.08) increased the risk of brain damage or death at 7-10 days of life, without affecting neurodevelopmental outcomes at 1 year. The effect of TH on MBL plasma profiles is uncertain.
Collapse
Affiliation(s)
- Cinzia Auriti
- Neonatal Intensive Care Unit, Department of Medical and Surgical Neonatology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Giusi Prencipe
- Laboratory of Rheumatology Department of Laboratories, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Rita Inglese
- Laboratory of Chemical Chemistry, Department of Laboratories, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Maria Moriondo
- Laboratory of Immunology, Department of Pediatrics, Anna Meyer Children's University Hospital, Florence, Italy
| | - Francesco Nieddu
- Laboratory of Immunology, Department of Pediatrics, Anna Meyer Children's University Hospital, Florence, Italy
| | - Vito Mondì
- Neonatal Intensive Care Unit, Department of Medical and Surgical Neonatology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.,Neonatology and Neonatal Intensive Care Unit, Policlinico Casilino Hospital, Rome, Italy
| | - Daniela Longo
- Neuroimaging Unit, Imaging Department, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Silvia Bucci
- Clinical Psychology Unit, Department of Neurosciences, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Tamara Del Pinto
- Neonatal Intensive Care Unit, Department of Medical and Surgical Neonatology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Laura Timelli
- Neonatal Intensive Care Unit, Department of Medical and Surgical Neonatology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | |
Collapse
|
9
|
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: 85] [Impact Index Per Article: 14.2] [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
|
10
|
Izzy S, Liu Q, Fang Z, Lule S, Wu L, Chung JY, Sarro-Schwartz A, Brown-Whalen A, Perner C, Hickman SE, Kaplan DL, Patsopoulos NA, El Khoury J, Whalen MJ. Time-Dependent Changes in Microglia Transcriptional Networks Following Traumatic Brain Injury. Front Cell Neurosci 2019; 13:307. [PMID: 31440141 PMCID: PMC6694299 DOI: 10.3389/fncel.2019.00307] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 06/24/2019] [Indexed: 12/21/2022] Open
Abstract
The neuroinflammatory response to traumatic brain injury (TBI) is critical to both neurotoxicity and neuroprotection, and has been proposed as a potentially modifiable driver of secondary injury in animal and human studies. Attempts to broadly target immune activation have been unsuccessful in improving outcomes, in part because the precise cellular and molecular mechanisms driving injury and outcome at acute, subacute, and chronic time points after TBI remain poorly defined. Microglia play a critical role in neuroinflammation and their persistent activation may contribute to long-term functional deficits. Activated microglia are characterized by morphological transformation and transcriptomic changes associated with specific inflammatory states. We analyzed the temporal course of changes in inflammatory genes of microglia isolated from injured brains at 2, 14, and 60 days after controlled cortical impact (CCI) in mice, a well-established model of focal cerebral contusion. We identified a time dependent, injury-associated change in the microglial gene expression profile toward a reduced ability to sense tissue damage, perform housekeeping, and maintain homeostasis in the early stages following CCI, with recovery and transition to a specialized inflammatory state over time. This later state starts at 14 days post-injury and is characterized by a biphasic pattern of IFNγ, IL-4, and IL-10 gene expression changes, with concurrent proinflammatory and anti-inflammatory gene changes. Our transcriptomic data sets are an important step to understand microglial role in TBI pathogenesis at the molecular level and identify common pathways that affect outcome. More studies to evaluate gene expression at the single cell level and focusing on subacute and chronic timepoint are warranted.
Collapse
Affiliation(s)
- Saef Izzy
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.,Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States.,Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States.,Harvard Medical School, Boston, MA, United States
| | - Qiong Liu
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention, Shanghai, China
| | - Zhou Fang
- Harvard Medical School, Boston, MA, United States.,Systems Biology and Computer Science Program, Ann Romney Center for Neurological Diseases, Department of Neurology, Brigham and Women's Hospital, Boston, MA, United States.,Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.,Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Sevda Lule
- Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States.,Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Limin Wu
- Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States.,Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Joon Yong Chung
- Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States.,Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Aliyah Sarro-Schwartz
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States
| | - Alexander Brown-Whalen
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States.,Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Caroline Perner
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States.,Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States.,Harvard Medical School, Boston, MA, United States
| | - Suzanne E Hickman
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States.,Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States.,Harvard Medical School, Boston, MA, United States
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Nikolaos A Patsopoulos
- Harvard Medical School, Boston, MA, United States.,Systems Biology and Computer Science Program, Ann Romney Center for Neurological Diseases, Department of Neurology, Brigham and Women's Hospital, Boston, MA, United States.,Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.,Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Joseph El Khoury
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States.,Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States.,Harvard Medical School, Boston, MA, United States
| | - Michael J Whalen
- Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States.,Harvard Medical School, Boston, MA, United States.,Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
11
|
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: 142] [Impact Index Per Article: 23.7] [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
|
12
|
Younger D, Murugan M, Rama Rao KV, Wu LJ, Chandra N. Microglia Receptors in Animal Models of Traumatic Brain Injury. Mol Neurobiol 2018; 56:5202-5228. [PMID: 30554385 DOI: 10.1007/s12035-018-1428-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/13/2018] [Indexed: 02/07/2023]
|
13
|
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: 105] [Impact Index Per Article: 15.0] [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
|
14
|
Kim DS, Li YK, Kim JH, Bergquist CS, Gerdes M, Bernbaum JC, Burnham N, McDonald-McGinn DM, Zackai EH, Nicolson SC, Spray TL, Nickerson DA, Hakonarson H, Jarvik GP, Gaynor JW. Autosomal dominant mannose-binding lectin deficiency is associated with worse neurodevelopmental outcomes after cardiac surgery in infants. J Thorac Cardiovasc Surg 2017; 155:1139-1147.e2. [PMID: 29452463 DOI: 10.1016/j.jtcvs.2017.08.035] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 07/01/2017] [Accepted: 08/03/2017] [Indexed: 11/19/2022]
Abstract
OBJECTIVES The MBL2 gene is the major genetic determinant of mannose-binding lectin (MBL)-an acute phase reactant. Low MBL levels have been associated with adverse outcomes in preterm infants. The MBL2Gly54Asp missense variant causes autosomal dominant MBL deficiency. We tested the hypothesis that MBL2Gly54Asp is associated with worse neurodevelopmental outcomes after cardiac surgery in neonates. METHODS This is an analysis of a previously described cohort of patients with nonsyndromic congenital heart disease who underwent cardiac surgery with cardiopulmonary bypass before age 6 months (n = 295). Four-year neurodevelopment was assessed in 3 domains: Full-Scale Intellectual Quotient, the Visual Motor Integration development test, and the Child Behavior Checklist to assess behavior problems. The Child Behavior Checklist measured total behavior problems, pervasive developmental problems, and internalizing/externalizing problems. A multivariable linear regression model, adjusting for confounders, was fit. RESULTS MBL2Gly54Asp was associated with a significantly increased covariate-adjusted pervasive developmental problem score (β = 3.98; P = .0025). Sensitivity analyses of the interaction between age at first surgery and MBL genotype suggested effect modification for the patients with MBL2Gly54Asp (Pinteraction = .039), with the poorest neurodevelopment outcomes occurring in children who had surgery earlier in life. CONCLUSIONS We report the novel finding that carriers of MBL2Gly54Asp causing autosomal dominant MBL deficiency have increased childhood pervasive developmental problems after cardiac surgery, independent of other covariates. Sensitivity analyses suggest that this effect may be larger in children who underwent surgery at earlier ages. These data support the role of nonsyndromic genetic variation in determining postsurgical neurodevelopment-related outcomes in children with congenital heart disease.
Collapse
MESH Headings
- Age Factors
- Cardiac Surgical Procedures/adverse effects
- Checklist
- Child Behavior
- Child Development
- Child Development Disorders, Pervasive/diagnosis
- Child Development Disorders, Pervasive/etiology
- Child Development Disorders, Pervasive/physiopathology
- Child Development Disorders, Pervasive/psychology
- Child, Preschool
- Female
- Gene-Environment Interaction
- Genetic Predisposition to Disease
- Heart Defects, Congenital/complications
- Heart Defects, Congenital/diagnostic imaging
- Heart Defects, Congenital/surgery
- Humans
- Infant
- Infant, Newborn
- Male
- Mannose-Binding Lectin/deficiency
- Mannose-Binding Lectin/genetics
- Metabolism, Inborn Errors/complications
- Metabolism, Inborn Errors/diagnosis
- Metabolism, Inborn Errors/genetics
- Metabolism, Inborn Errors/physiopathology
- Motor Skills
- Mutation, Missense
- Nervous System/growth & development
- Neurologic Examination
- Phenotype
- Risk Assessment
- Risk Factors
- Time Factors
- Treatment Outcome
Collapse
Affiliation(s)
- Daniel Seung Kim
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Wash; Department of Genome Sciences, University of Washington, Seattle, Wash; Department of Biostatistics, University of Michigan, Ann Arbor, Mich
| | - Yatong K Li
- Department of Biostatistics, University of Michigan, Ann Arbor, Mich
| | - Jerry H Kim
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Wash
| | - Curtis S Bergquist
- Section of Thoracic Surgery, Department of Surgery, University of Michigan, Ann Arbor, Mich
| | - Marsha Gerdes
- Department of Pediatrics, The Children's Hospital of Philadelphia and the Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | - Judy C Bernbaum
- Department of Pediatrics, The Children's Hospital of Philadelphia and the Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | - Nancy Burnham
- Division of Cardiothoracic Surgery, The Children's Hospital of Philadelphia and the Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | - Donna M McDonald-McGinn
- Division of Genetics, The Children's Hospital of Philadelphia and the Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | - Elaine H Zackai
- Division of Genetics, The Children's Hospital of Philadelphia and the Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | - Susan C Nicolson
- Division of Cardiothoracic Anesthesiology, The Children's Hospital of Philadelphia and the Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | - Thomas L Spray
- Division of Cardiothoracic Surgery, The Children's Hospital of Philadelphia and the Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | | | - Hakon Hakonarson
- Center for Applied Genomics, The Children's Hospital of Philadelphia and the Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | - Gail P Jarvik
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Wash; Department of Genome Sciences, University of Washington, Seattle, Wash
| | - J William Gaynor
- Division of Cardiothoracic Surgery, The Children's Hospital of Philadelphia and the Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa.
| |
Collapse
|
15
|
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
|
16
|
Hammel JM. Assembling the puzzle of polymorphism. J Thorac Cardiovasc Surg 2017; 155:1138. [PMID: 29042103 DOI: 10.1016/j.jtcvs.2017.09.075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 09/16/2017] [Indexed: 10/18/2022]
Affiliation(s)
- James M Hammel
- Department of Cardiothoracic Surgery, Children's Hospital and Medical Center, Omaha, Neb.
| |
Collapse
|
17
|
Gordon EE, Jaquiss RDB. Yet another risk factor appears. J Thorac Cardiovasc Surg 2017; 155:1148-1149. [PMID: 28986043 DOI: 10.1016/j.jtcvs.2017.08.105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Accepted: 08/30/2017] [Indexed: 10/18/2022]
Affiliation(s)
- Erin E Gordon
- Department of Pediatrics, UT Southwestern Medical Center and Children's Medical Center, Dallas, Tex
| | - R D B Jaquiss
- Department of Thoracic and Cardiovascular Surgery, UT Southwestern Medical Center and Children's Medical Center, Dallas, Tex.
| |
Collapse
|
18
|
Cheng C, Yu Z, Zhao S, Liao Z, Xing C, Jiang Y, Yang YG, Whalen MJ, Lo EH, Sun X, Wang X. Thrombospondin-1 Gene Deficiency Worsens the Neurological Outcomes of Traumatic Brain Injury in Mice. Int J Med Sci 2017; 14:927-936. [PMID: 28924363 PMCID: PMC5599915 DOI: 10.7150/ijms.18812] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Accepted: 03/14/2017] [Indexed: 12/13/2022] Open
Abstract
Background: Thrombospondin-1 (TSP-1) is an extracellular matrix protein that plays multiple physiological and pathophysiological roles in the brain. Experimental reports suggest that TSP-1 may have an adverse role in neuronal function recovery under certain injury conditions. However, the roles of TSP-1 in traumatic brain injury (TBI) have not been elucidated. In this study we for the first time investigated the roles of TSP-1 in a controlled cortical impact (CCI) model of TBI in TSP-1 knockout (TSP-1 KO) and wild type (WT) mice. Methods: We examined blood brain-barrier (BBB) damage using at 1 day post-TBI by measuring Evans Blue leakage, and neurological functional recovery at 3 weeks post-TBI by measuring neurological severity score (NSS), wire gripping, corner test and Morris Water Maze (MWM). Mechanistically, we quantified pro-angiogenic biomarkers including cerebral vessel density, vascular endothelial growth factors (VEGF) and angiopoietin-1 (Ang-1) protein expression, synaptic biomarker synaptophysin, and synaptogenesis marker brain-derived neurotrophic factor (BDNF) protein expression in contralateral and ipsilateral (peri-lesion) cortex at 21 days after TBI using immunohistochemistry and Western Blot. Results: TSP-1 is upregulated at early phase of TBI in WT mice. Compared to WT mice, TSP-1 KO (1) significantly worsened TBI-induced BBB leakage at 1 day after TBI; (2) had similar lesion size as WT mice at 3 weeks after TBI; (3) exhibited a significantly worse neurological deficits in motor and cognitive functions; (4) had no significant difference in cerebral vessel density, but significant increase of VEGF and Ang-1 protein expressions in peri-lesion cortex; (5) significantly increased BDNF but not synaptophysin protein level in peri-lesion cortex compared to sham, but both synaptophysin and BDNF expressions were significantly decreased in contralateral cortex compared to WT. Conclusion: Our results suggest that TSP-1 may be beneficial for maintaining BBB integrity in the early phase and functional recovery in late phase after TBI. The molecular mechanisms of TSP-1 in early BBB pathophysiology, and long-term neurological function recovery after TBI need to be further investigated.
Collapse
Affiliation(s)
- Chongjie Cheng
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Neuroprotection Research Laboratory, Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Zhanyang Yu
- Neuroprotection Research Laboratory, Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Song Zhao
- Departments of Orthopedic and Neurosurgery, The First Bethune Hospital of Jilin University, Changchun, Jilin, China
| | - Zhengbu Liao
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Neuroprotection Research Laboratory, Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Changhong Xing
- Neuroprotection Research Laboratory, Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Yinghua Jiang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Neuroprotection Research Laboratory, Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Yong-Guang Yang
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Michael J. Whalen
- Department of Pediatrics, Pediatric Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Eng H. Lo
- Neuroprotection Research Laboratory, Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Xiaochuan Sun
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoying Wang
- Neuroprotection Research Laboratory, Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
19
|
Osthoff M, Walder B, Delhumeau C, Trendelenburg M, Turck N. Association of Lectin Pathway Protein Levels and Genetic Variants Early after Injury with Outcomes after Severe Traumatic Brain Injury: A Prospective Cohort Study. J Neurotrauma 2017; 34:2560-2566. [PMID: 28482760 DOI: 10.1089/neu.2016.4941] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The lectin pathway of the complement system has been implicated in secondary ischemic/inflammatory injury after traumatic brain injury (TBI). However, previous experimental studies have yielded conflicting results, and human studies are scarce. In this exploratory study, we investigated associations of several lectin pathway proteins early after injury and single-nucleotide polymorphisms (SNP) with outcomes after severe TBI (mortality at 14 days [primary outcome] and consciousness assessed with the Glasgow Coma Scale [GCS] at 14 days, disability assessed with the Glasgow Outcome Scale Extended [GOSE] at 90 days). Forty-four patients with severe TBI were included. Plasma levels of lectin pathway proteins were sampled at 6, 12, 24, and 48 h after injury and eight mannose-binding lectin (MBL) and ficolin (FCN)2 SNPs were analyzed by enzyme-linked immunosorbent assay (ELISA) and genotyping, respectively. Plasma protein levels were stable with only a slight increase in mannose-binding protein-associated serine protease (MASP)-2 and FCN2 levels after 48 h (p < 0.05), respectively. Neither lectin protein plasma levels (6 h or mean levels) nor MBL2 genotypes or FCN2 variant alleles were associated with 14 day mortality or 14 day consciousness. However, FCN2, FCN3, and MASP-2 levels were higher in patients with an unfavorable outcome (GOSE 1-4) at 90 days (p < 0.05), whereas there was no difference in MBL2 genotypes or FCN2 variant alleles. In particular, higher mean MASP-2 levels over 48 h were independently associated with a GOSE score < 4 at 90 days after adjustment (odds ratio 3.46 [95% confidence interval 1.12-10.68] per 100 ng/mL increase, p = 0.03). No association was observed between the lectin pathway of the complement system and 14 day mortality or 14 day consciousness. However, higher plasma FCN2, FCN3, and, in particular, MASP-2 levels early after injury were associated with an unfavorable outcome at 90 days (death, vegetative state, and severe disability) which may be related to an increased activation of the lectin pathway.
Collapse
Affiliation(s)
- Michael Osthoff
- 1 Division of Internal Medicine, University Hospital Basel , Basel, Switzerland .,2 Department of Biomedicine, University Hospital Basel , Basel, Switzerland
| | - Bernhard Walder
- 3 Division of Anaesthesiology, Department of Anaesthesiology, Intensive Care and Clinical Pharmacology, University Hospitals of Geneva , Geneva, Switzerland
| | - Cécile Delhumeau
- 3 Division of Anaesthesiology, Department of Anaesthesiology, Intensive Care and Clinical Pharmacology, University Hospitals of Geneva , Geneva, Switzerland
| | - Marten Trendelenburg
- 1 Division of Internal Medicine, University Hospital Basel , Basel, Switzerland .,2 Department of Biomedicine, University Hospital Basel , Basel, Switzerland
| | - Natacha Turck
- 4 OPTICS Group, Department of Human Protein Sciences, University of Geneva , Geneva, Switzerland
| |
Collapse
|
20
|
De Blasio D, Fumagalli S, Longhi L, Orsini F, Palmioli A, Stravalaci M, Vegliante G, Zanier ER, Bernardi A, Gobbi M, De Simoni MG. Pharmacological inhibition of mannose-binding lectin ameliorates neurobehavioral dysfunction following experimental traumatic brain injury. J Cereb Blood Flow Metab 2017; 37:938-950. [PMID: 27165013 PMCID: PMC5363468 DOI: 10.1177/0271678x16647397] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Mannose-binding lectin is present in the contusion area of traumatic brain-injured patients and in that of traumatic brain-injured mice, where mannose-binding lectin-C exceeds mannose-binding lectin-A. The reduced susceptibility to traumatic brain injury of mannose-binding lectin double knock-out mice (mannose-binding lectin-/-) when compared to wild type mice suggests that mannose-binding lectin may be a therapeutic target following traumatic brain injury. Here, we evaluated the effects of a multivalent glycomimetic mannose-binding lectin ligand, Polyman9, following traumatic brain injury in mice. In vitro surface plasmon resonance assay indicated that Polyman9 dose-dependently inhibits the binding to immobilized mannose residues of plasma mannose-binding lectin-C selectively over that of mannose-binding lectin-A. Male C57Bl/6 mice underwent sham/controlled cortical impact traumatic brain injury and intravenous treatment with Polyman9/saline. Ex-vivo surface plasmon resonance studies confirmed that Polyman9 effectively reduces the binding of plasma mannose-binding lectin-C to immobilized mannose residues. In vivo studies up to four weeks post injury, showed that Polyman9 induces significant improvement in sensorimotor deficits (by neuroscore and beam walk), promotes neurogenesis (73% increase in doublecortin immunoreactivity), and astrogliosis (28% increase in glial fibrillary acid protein). Polyman9 administration in brain-injured mannose-binding lectin-/- mice had no effect on post-traumatic brain-injured functional deficits, suggestive of the specificity of its neuroprotective effects. The neurobehavioral efficacy of Polyman9 implicates mannose-binding lectin-C as a novel therapeutic target for traumatic brain injury.
Collapse
Affiliation(s)
- Daiana De Blasio
- 1 IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milano, Italy.,2 Department of Anesthesia and Critical Care Medicine, Fondazione IRCCS Ca'Granda - Ospedale Maggiore Policlinico, Milano, Italy
| | - Stefano Fumagalli
- 1 IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milano, Italy.,2 Department of Anesthesia and Critical Care Medicine, Fondazione IRCCS Ca'Granda - Ospedale Maggiore Policlinico, Milano, Italy
| | - Luca Longhi
- 3 Department of Anesthesia and Critical Care Medicine, Neurosurgical Intensive Care Unit, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Franca Orsini
- 1 IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milano, Italy
| | | | - Matteo Stravalaci
- 1 IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milano, Italy
| | - Gloria Vegliante
- 1 IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milano, Italy
| | - Elisa R Zanier
- 1 IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milano, Italy
| | - Anna Bernardi
- 4 Department of Chemistry, Università degli Studi di Milano, Milano, Italy
| | - Marco Gobbi
- 1 IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milano, Italy
| | | |
Collapse
|
21
|
Mannose-Binding Lectin: Biologic Characteristics and Role in the Susceptibility to Infections and Ischemia-Reperfusion Related Injury in Critically Ill Neonates. J Immunol Res 2017; 2017:7045630. [PMID: 28246614 PMCID: PMC5299167 DOI: 10.1155/2017/7045630] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Revised: 11/08/2016] [Accepted: 12/28/2016] [Indexed: 01/14/2023] Open
Abstract
The mannose-binding lectin (MBL) is a member of the collectin family, belonging to the innate immunity system. Genetic, biologic, and clinical properties of MBL have been widely investigated throughout the last decades, although some interesting aspects of its potential clinical relevance are still poorly understood. Low circulating concentrations of MBL have been associated with increased risk of infection and poor neurologic outcome in neonates. On the other hand, an excessive and uncontrolled inflammatory response by the neonatal intestine after the exposure to luminal bacteria, leading to an increased production of MBL, may be involved in the onset of necrotizing enterocolitis. The purpose of the present review is to summarize the current knowledge about genetic and biologic characteristics of MBL and its role in the susceptibility to infections and to ischemia-reperfusion related tissue injuries to better explore its clinical relevance during the perinatal period and the possible future therapeutic applications.
Collapse
|
22
|
Shultz SR, McDonald SJ, Vonder Haar C, Meconi A, Vink R, van Donkelaar P, Taneja C, Iverson GL, Christie BR. The potential for animal models to provide insight into mild traumatic brain injury: Translational challenges and strategies. Neurosci Biobehav Rev 2016; 76:396-414. [PMID: 27659125 DOI: 10.1016/j.neubiorev.2016.09.014] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 09/07/2016] [Accepted: 09/16/2016] [Indexed: 12/14/2022]
Abstract
Mild traumatic brain injury (mTBI) is a common health problem. There is tremendous variability and heterogeneity in human mTBI, including mechanisms of injury, biomechanical forces, injury severity, spatial and temporal pathophysiology, genetic factors, pre-injury vulnerability and resilience factors, and clinical outcomes. Animal models greatly reduce this variability and heterogeneity, and provide a means to study mTBI in a rigorous, controlled, and efficient manner. Rodent models, in particular, are time- and cost-efficient, and they allow researchers to measure morphological, cellular, molecular, and behavioral variables in a single study. However, inter-species differences in anatomy, morphology, metabolism, neurobiology, and lifespan create translational challenges. Although the term "mild" TBI is used often in the pre-clinical literature, clearly defined criteria for mild, moderate, and severe TBI in animal models have not been agreed upon. In this review, we introduce current issues facing the mTBI field, summarize the available research methodologies and previous studies in mTBI animal models, and discuss how a translational research approach may be useful in advancing our understanding and management of mTBI.
Collapse
Affiliation(s)
- Sandy R Shultz
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia.
| | - Stuart J McDonald
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, VIC, Australia
| | - Cole Vonder Haar
- Department of Psychology, The University of British Columbia, Vancouver, BC, Canada
| | - Alicia Meconi
- Division of Medical Sciences, The University of Victoria, Victoria, BC, Canada
| | - Robert Vink
- Division of Health Sciences, The University of South Australia, Adelaide, SA, Australia
| | - Paul van Donkelaar
- School of Health and Exercise Sciences, The University of British Columbia Okanagan, Kelowna, BC, Canada
| | - Chand Taneja
- Division of Medical Sciences, The University of Victoria, Victoria, BC, Canada
| | - Grant L Iverson
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Spaulding Rehabilitation Hospital, Home Base, A Red Sox Foundation and Massachusetts General Hospital Program, and MassGeneral Hospital for Children™ Sports Concussion Program, Boston, MA, USA
| | - Brian R Christie
- Division of Medical Sciences, The University of Victoria, Victoria, BC, Canada
| |
Collapse
|
23
|
Mannose-binding lectin is expressed after clinical and experimental traumatic brain injury and its deletion is protective. Crit Care Med 2016; 42:1910-8. [PMID: 24810526 DOI: 10.1097/ccm.0000000000000399] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Mannose-binding lectin protein is the activator of the lectin complement pathway. Goals were (1) to investigate mannose-binding lectin expression after human and experimental traumatic brain injury induced by controlled cortical impact and (2) to evaluate whether mannose-binding lectin deletion is associated with reduced sequelae after controlled cortical impact. DESIGN Translational research, combining a human/experimental observational study and a prospective experimental study. SETTING University hospital/research laboratory. PATIENTS AND SUBJECTS Brain-injured patients, C57Bl/6 mice, and mannose-binding lectin-A and mannose-binding lectin-C double-knockout (-/-) mice. INTERVENTIONS Using anti-human mannose-binding lectin antibody, we evaluated mannose-binding lectin expression in tissue samples from six patients who underwent surgery for a cerebral contusion. Immunohistochemistry was also performed on tissues obtained from mice at 30 minutes; 6, 12, 24, 48, and 96 hours; and 1 week after controlled cortical impact using anti-mouse mannose-binding lectin-A and mannose-binding lectin-C antibodies. We evaluated the effects of mannose-binding lectin deletion in wild-type and mannose-binding lectin-A and mannose-binding lectin-C double-knockout mice. Functional outcome was evaluated using the neuroscore and beam walk tests for 4 weeks postinjury (n = 11). Histological injury was evaluated by comparing neuronal cell counts in the cortex adjacent to the contusion (n = 11). MEASUREMENTS AND MAIN RESULTS Following human traumatic brain injury, we observed mannose-binding lectin-positive immunostaining in the injured cortex as early as few hours and up to 5 days postinjury. Similarly in mice, we observed mannose-binding lectin-C-positive immunoreactivity in the injured cortex beginning 30 minutes and persisting up to 1 week postinjury. The extent of mannose-binding lectin-A expression was lower when compared with that of mannose-binding lectin-C. We observed attenuated sensorimotor deficits in mannose-binding lectin (-/-) mice compared with wild-type mice at 2-4 weeks postinjury. Furthermore, we observed reduced cortical cell loss at 5 weeks postinjury in mannose-binding lectin (-/-) mice compared with wild-type mice. CONCLUSIONS Mannose-binding lectin expression was documented after traumatic brain injury. The reduced sequelae associated with mannose-binding lectin absence suggest that mannose-binding lectin modulation might be a potential target after traumatic brain injury.
Collapse
|
24
|
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
|
25
|
Huang Z, Cheng C, Jiang L, Yu Z, Cao F, Zhong J, Guo Z, Sun X. Intraventricular apolipoprotein ApoJ infusion acts protectively in Traumatic Brain Injury. J Neurochem 2016; 136:1017-25. [PMID: 26670094 DOI: 10.1111/jnc.13491] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 11/24/2015] [Accepted: 12/04/2015] [Indexed: 11/29/2022]
Affiliation(s)
- Zhijian Huang
- Department of Neurosurgery; the First Affiliated Hospital of Chongqing Medical University; Chongqing China
| | - Chongjie Cheng
- Department of Neurosurgery; the First Affiliated Hospital of Chongqing Medical University; Chongqing China
| | - Li Jiang
- Department of Neurosurgery; the First Affiliated Hospital of Chongqing Medical University; Chongqing China
| | - Zhanyang Yu
- Department of Neurology and Radiology; Neuroprotection Research Laboratory; Harvard Medical School; Cambridge MA USA
| | - Fang Cao
- Department of Cerebrovascular; the Affiliated Hospital of Zunyi Medical College; Guizhou China
| | - Jianjun Zhong
- Department of Neurosurgery; the First Affiliated Hospital of Chongqing Medical University; Chongqing China
| | - Zongduo Guo
- Department of Neurosurgery; the First Affiliated Hospital of Chongqing Medical University; Chongqing China
| | - Xiaochuan Sun
- Department of Neurosurgery; the First Affiliated Hospital of Chongqing Medical University; Chongqing China
| |
Collapse
|
26
|
Yu W, Le HW, Lu YG, Hu JA, Yu JB, Wang M, Shen W. High levels of serum mannose-binding lectins are associated with the severity and clinical outcomes of severe traumatic brain injury. Clin Chim Acta 2015; 451:111-6. [PMID: 26525964 DOI: 10.1016/j.cca.2015.10.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 10/16/2015] [Accepted: 10/16/2015] [Indexed: 11/28/2022]
Abstract
BACKGROUND Mannose-binding lectin (MBL) is a key component of innate immunity. The expression of cortical MBL is up-regulated after clinical and experimental head trauma. This study aimed to assess the association of serum MBL levels with injury severity and long-term clinical outcomes after severe traumatic brain injury (STBI). METHODS Serum MBL levels were measured in 122 patients and 100 healthy controls. Multivariate analyses were used to analyze the relationship between serum MBL levels and trauma severity reflected by Glasgow Coma Scale scores as well as between serum MBL levels and 6-month mortality and unfavorable outcome (Glasgow Outcome Scale score: 1-3). A receiver operating characteristic (ROC) curve was structured to evaluate the prognostic predictive performance of serum MBL levels. RESULTS Compared with healthy controls, serum MBL levels of patients were markedly elevated. Using multivariate analyses, serum MBL levels were found to be associated closely with Glasgow Coma Scale (GCS) scores and MBL emerged as an independent predictor for 6-month mortality and unfavorable outcome. Under ROC curve, serum MBL levels and GCS scores possessed similar prognostic predictive values. CONCLUSION Increased serum level of MBL was independently associated with head trauma severity and long-term clinical outcomes of STBI.
Collapse
Affiliation(s)
- Wei Yu
- Department of Neurosurgery, The People's Hospital of Beilun District, Beilun Branch Hospital of The First Affiliated Hospital of Medical School of Zhejiang University, 1288 Lushan East Road, Beilun District, Ningbo 315800, China
| | - Hai-Wei Le
- Department of Neurosurgery, The People's Hospital of Beilun District, Beilun Branch Hospital of The First Affiliated Hospital of Medical School of Zhejiang University, 1288 Lushan East Road, Beilun District, Ningbo 315800, China
| | - Yi-Gao Lu
- Department of Neurosurgery, The People's Hospital of Beilun District, Beilun Branch Hospital of The First Affiliated Hospital of Medical School of Zhejiang University, 1288 Lushan East Road, Beilun District, Ningbo 315800, China
| | - Jun-An Hu
- Department of Neurosurgery, The People's Hospital of Beilun District, Beilun Branch Hospital of The First Affiliated Hospital of Medical School of Zhejiang University, 1288 Lushan East Road, Beilun District, Ningbo 315800, China
| | - Jian-Bo Yu
- Department o f Neurosurgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou 310003, China
| | - Ming Wang
- Department o f Neurosurgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou 310003, China
| | - Wei Shen
- Department of Neurosurgery, The People's Hospital of Beilun District, Beilun Branch Hospital of The First Affiliated Hospital of Medical School of Zhejiang University, 1288 Lushan East Road, Beilun District, Ningbo 315800, China.
| |
Collapse
|
27
|
Xue J, Liu AH, Zhao B, Si M, Li YQ. Low levels of mannose-binding lectin at admission increase the risk of adverse neurological outcome in preterm infants: a 1-year follow-up study. J Matern Fetal Neonatal Med 2015; 29:1425-9. [DOI: 10.3109/14767058.2015.1050372] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
28
|
Orsini F, De Blasio D, Zangari R, Zanier ER, De Simoni MG. Versatility of the complement system in neuroinflammation, neurodegeneration and brain homeostasis. Front Cell Neurosci 2014; 8:380. [PMID: 25426028 PMCID: PMC4224073 DOI: 10.3389/fncel.2014.00380] [Citation(s) in RCA: 153] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 10/22/2014] [Indexed: 01/30/2023] Open
Abstract
The immune response after brain injury is highly complex and involves both local and systemic events at the cellular and molecular level. It is associated to a dramatic over-activation of enzyme systems, the expression of proinflammatory genes and the activation/recruitment of immune cells. The complement system represents a powerful component of the innate immunity and is highly involved in the inflammatory response. Complement components are synthesized predominantly by the liver and circulate in the bloodstream primed for activation. Moreover, brain cells can produce complement proteins and receptors. After acute brain injury, the rapid and uncontrolled activation of the complement leads to massive release of inflammatory anaphylatoxins, recruitment of cells to the injury site, phagocytosis and induction of blood brain barrier (BBB) damage. Brain endothelial cells are particularly susceptible to complement-mediated effects, since they are exposed to both circulating and locally synthesized complement proteins. Conversely, during neurodegenerative disorders, complement factors play distinct roles depending on the stage and degree of neuropathology. In addition to the deleterious role of the complement, increasing evidence suggest that it may also play a role in normal nervous system development (wiring the brain) and adulthood (either maintaining brain homeostasis or supporting regeneration after brain injury). This article represents a compendium of the current knowledge on the complement role in the brain, prompting a novel view that complement activation can result in either protective or detrimental effects in brain conditions that depend exquisitely on the nature, the timing and the degree of the stimuli that induce its activation. A deeper understanding of the acute, subacute and chronic consequences of complement activation is needed and may lead to new therapeutic strategies, including the ability of targeting selective step in the complement cascade.
Collapse
Affiliation(s)
- Franca Orsini
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri Milan, Italy
| | - Daiana De Blasio
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri Milan, Italy ; Department of Experimental and Clinical Sciences, University of Chieti Pescara, Italy
| | - Rosalia Zangari
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri Milan, Italy ; Department of Anesthesia and Critical Care Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, University of Milan Milan, Italy
| | - Elisa R Zanier
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri Milan, Italy
| | - Maria-Grazia De Simoni
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri Milan, Italy
| |
Collapse
|
29
|
MBL2 gene polymorphisms increase the risk of adverse neurological outcome in preterm infants: a preliminary prospective study. Pediatr Res 2014; 76:464-9. [PMID: 25119337 DOI: 10.1038/pr.2014.118] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Accepted: 05/19/2014] [Indexed: 02/02/2023]
Abstract
BACKGROUND As described in animal models, the lectin-complement pathway is central to the propagation of ischemia-reperfusion injuries in many tissues, including the brain. Similarly, it might affect the genesis of brain damage in preterm infants. MBL2 gene single-nucleotide polymorphisms (SNPs), regulating mannose-binding lectin (MBL) serum levels, could predict the risk of adverse neurological outcome in these infants. METHODS To evaluate the association between SNPs of the MBL2 gene and long-term neurological outcomes in preterm infants, 75 infants (gestational age (GA) ≤ 32 wk) were observed in a prospective longitudinal study and assessed by clinical and instrumental exams at 12 and 24 mo of corrected age (CA). They were genotyped for the promoter polymorphism -221 and for the exon-1 variant alleles (at codons 52, 54, and 57) of the MBL2 gene. RESULTS The MBL2 exon-1 OO genotype was more frequent in children with an adverse neurological outcome (5/35; 7%) than in controls (0/40; 0%), P = 0.045. The risk of intraventricular hemorrhage in carriers of the genotype OO was marked, without reaching statistical significance (odds ratio: 8.67; 95% confidence interval: 0.87-86.06; P = 0.07). CONCLUSION Preterm infants who are carriers of MBL2 exon-1 OO genotype are exposed to an increased risk of adverse neurological outcomes.
Collapse
|
30
|
The pharmacokinetics and pharmacodynamics of Kollidon VA64 dissociate its protective effects from membrane resealing after controlled cortical impact in mice. J Cereb Blood Flow Metab 2014; 34:1347-53. [PMID: 24824916 PMCID: PMC4126095 DOI: 10.1038/jcbfm.2014.89] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 03/24/2014] [Accepted: 04/27/2014] [Indexed: 11/08/2022]
Abstract
Membrane-resealing agents such as poloxamer P188 improve the outcome in experimental brain injury paradigms; however, whether membrane resealing is a key mechanism for protection has not been shown in vivo. We previously reported that Kollidon VA64, a polymeric membrane-resealing agent, reduces cell membrane permeability and improves brain edema, brain tissue damage, and functional outcome after controlled cortical impact in mice, without rescuing resealed cells from death. To reconcile these disparate findings, we used a dual-pulse labeling protocol to determine membrane-resealing kinetics by VA64/P188 in vivo. Membrane resealing after controlled cortical impact in mice by intravenous or intracerebroventricular VA64 and poloxamer P188 was transient, with most cells becoming repermeabilized within 2 hours, even with multiple-dose paradigms that maintained high VA64 blood levels. Moreover, VA64 reduced cytotoxic brain edema in a water intoxication model devoid of plasmalemma permeability (P<0.05 versus P188, VA30, mannitol, and vehicle). We conclude that VA64 reduces cytotoxic and traumatic brain edema independent of membrane resealing. The results suggest that classic membrane-resealing agents such as poloxamer P188, and the newly discovered VA64, exert protective effects in central nervous system injury paradigms by mechanisms other than or in addition to maintaining permeable cell membranes sealed.
Collapse
|
31
|
Nature has no principle-inflammation following brain injury is neither good nor evil*. Crit Care Med 2014; 42:1958-9. [PMID: 25029142 DOI: 10.1097/ccm.0000000000000479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
32
|
Dapul HR, Park J, Zhang J, Lee C, DanEshmand A, Lok J, Ayata C, Gray T, Scalzo A, Qiu J, Lo EH, Whalen MJ. Concussive injury before or after controlled cortical impact exacerbates histopathology and functional outcome in a mixed traumatic brain injury model in mice. J Neurotrauma 2013; 30:382-91. [PMID: 23153355 DOI: 10.1089/neu.2012.2536] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Traumatic brain injury (TBI) may involve diverse injury mechanisms (e.g., focal impact vs. diffuse impact loading). Putative therapies developed in TBI models featuring a single injury mechanism may fail in clinical trials if the model does not fully replicate multiple injury subtypes, which may occur concomitantly in a given patient. We report development and characterization of a mixed contusion/concussion TBI model in mice using controlled cortical impact (CCI; 0.6 mm depth, 6 m/sec) and a closed head injury (CHI) model at one of two levels of injury (53 vs. 83 g weight drop from 66 in). Compared with CCI or CHI alone, sequential CCI-CHI produced additive effects on loss of consciousness (p<0.001), acute cell death (p<0.05), and 12-day lesion size (p<0.05) but not brain edema or 48-h contusion volume. Additive effects of CHI and CCI on post-injury motor (p<0.05) and cognitive (p<0.005) impairment were observed with sequential CCI-CHI (83 g). The data suggest that concussive forces, which in isolation do not induce histopathological damage, exacerbate histopathology and functional outcome after cerebral contusion. Sequential CHI-CCI may model complex injury mechanisms that occur in some patients with TBI and may prove useful for testing putative therapies.
Collapse
Affiliation(s)
- Heda R Dapul
- Neuroscience Center , Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Takahashi K. Mannose-binding lectin and the balance between immune protection and complication. Expert Rev Anti Infect Ther 2012; 9:1179-90. [PMID: 22114968 DOI: 10.1586/eri.11.136] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The innate immune system is evolutionarily ancient and biologically primitive. Historically, it was first identified as an element of the immune system that provides the first-line response to pathogens, and increasingly it is recognized for its central housekeeping role and its essential functions in tissue homeostasis, including coagulation and inflammation, among others. A pivotal link between the innate immune system and other functions is mannose-binding lectin (MBL), a pattern recognition molecule. Multiple studies have demonstrated that MBL deficiency increases susceptibility to infection, and the mechanisms associated with this susceptibility to infection include reduced opsonophagocytic killing and reduced activation of the lectin complement pathway. Results from our laboratory have demonstrated that MBL and MBL-associated serine protease (MASP)-1/3 together mediate coagulation factor-like activities, including thrombin-like activity. MBL and/or MASP-1/3-deficient hosts demonstrate in vivo evidence that MBL and MASP-1/3 are involved with hemostasis following injury. Staphylococcus aureus-infected MBL null mice developed disseminated intravascular coagulation, which was associated with elevated blood IL-6 levels (but not TNF-α) and systemic inflammatory responses. Infected MBL null mice also develop liver injury. These findings suggest that MBL deficiency may manifest as disseminated intravascular coagulation and organ failure with infection. Beginning from these observations, this review focuses on the interaction of innate immunity and other homeostatic systems, the derangement of which may lead to complications in infection and other inflammatory states.
Collapse
Affiliation(s)
- Kazue Takahashi
- Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, GRJ1402, Boston, MA 02114, USA.
| |
Collapse
|
34
|
Mannose-binding lectin binds to amyloid β protein and modulates inflammation. J Biomed Biotechnol 2012; 2012:929803. [PMID: 22536027 PMCID: PMC3322523 DOI: 10.1155/2012/929803] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Revised: 11/26/2011] [Accepted: 12/04/2011] [Indexed: 01/20/2023] Open
Abstract
Mannose-binding lectin (MBL), a soluble factor of the innate immune system, is a pattern recognition molecule with a number of known ligands, including viruses, bacteria, and molecules from abnormal self tissues. In addition to its role in immunity, MBL also functions in the maintenance of tissue homeostasis. We present evidence here that MBL binds to amyloid β peptides. MBL binding to other known carbohydrate ligands is calcium-dependent and has been attributed to the carbohydrate-recognition domain, a common feature of other C-type lectins. In contrast, we find that the features of MBL binding to Aβ are more similar to the reported binding characteristics of the cysteine-rich domain of the unrelated mannose receptor and therefore may involve the MBL cysteine-rich domain. Differences in MBL ligand binding may contribute to modulation of inflammatory response and may correlate with the function of MBL in processes such as coagulation and tissue homeostasis.
Collapse
|
35
|
Yip PK, Malaspina A. Spinal cord trauma and the molecular point of no return. Mol Neurodegener 2012; 7:6. [PMID: 22315999 PMCID: PMC3299607 DOI: 10.1186/1750-1326-7-6] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Accepted: 02/08/2012] [Indexed: 12/13/2022] Open
Abstract
A mechanical trauma to the spinal cord can be followed by the development of irreversible and progressive neurodegeneration, as opposed to a temporary or partially reversible neurological damage. An increasing body of experimental and clinical evidence from humans and animal models indicates that spinal cord injury may set in motion the development of disabling and at times fatal neuromuscular disorders, whose occurrence is not normally associated with any major environmental event. This outcome appears to be dependent on the co-occurrence of a particular form of mechanical stress and of a genetically-determined vulnerability. This increased vulnerability to spinal cord injury may depend on a change of the nature and of the timing of activation of a number of neuroprotective and neurodestructive molecular signals in the injured cord. Among the main determinants, we could mention an altered homeostasis of lipids and neurofilaments, an earlier inflammatory response and the failure of the damaged tissue to rein in oxidative damage and apoptotic cell death. These changes could force injured tissue beyond a point of no return and precipitate an irreversible neurodegenerative process. A better knowledge of the molecular signals activated in a state of increased vulnerability to trauma can inform future treatment strategies and the prediction of the neurological outcome after spinal cord injury.
Collapse
Affiliation(s)
- Ping K Yip
- Centre for Neuroscience and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, UK.
| | | |
Collapse
|
36
|
Morrison H, Frye J, Davis-Gorman G, Funk J, McDonagh P, Stahl G, Ritter L. The contribution of mannose binding lectin to reperfusion injury after ischemic stroke. Curr Neurovasc Res 2012; 8:52-63. [PMID: 21208161 DOI: 10.2174/156720211794520260] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Accepted: 09/19/2010] [Indexed: 12/23/2022]
Abstract
After complement system (CS) activation, the sequential production of complement products increases cell injury and death through opsonophagocytosis, cytolysis, adaptive, and inflammatory cell responses. These responses potentiate cerebral ischemia-reperfusion (IR) injury after ischemic stroke and reperfusion. Activation of the CS via mannose binding lectin (MBL)-initiated lectin pathway is known to increase tissue damage in response to IR in muscle, myocardium and intestine tissue. In contrast, the contribution of this pathway to cerebral IR injury, a neutrophil-mediated event, is less clear. Therefore, we investigated the potential protective role of MBL deficiency in neutrophil-mediated cerebral injury after IR. Using an intraluminal filament method, neutrophil activation and cerebral injury were compared between MBL-deficient and wild type C57Bl/6 mice subjected to 60 minutes of MCA ischemia and reperfusion. Systemic neutrophil activation was not decreased in MBL-deficient animals after IR. In MBL-deficient animals, cerebral injury was significantly decreased only in the striatum (p < 0.05). Despite MBL deficiency, C3 depositions were evident in the injured hemisphere during reperfusion. These results indicate that while MBL deficiency results in a modest protection of a sub-cortical brain region during IR, redundant complement pathway activation may overwhelm further beneficial effects of MBL deficiency during reperfusion.
Collapse
Affiliation(s)
- Helena Morrison
- College of Nursing, University of Arizona, Tucson Arizona, USA.
| | | | | | | | | | | | | |
Collapse
|
37
|
Iadecola C, Anrather J. Reply to: Mannose-binding lectin—the forgotten molecule? Nat Med 2011. [DOI: 10.1038/nm.2589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
38
|
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
|
39
|
Helmy A, De Simoni MG, Guilfoyle MR, Carpenter KLH, Hutchinson PJ. Cytokines and innate inflammation in the pathogenesis of human traumatic brain injury. Prog Neurobiol 2011; 95:352-72. [PMID: 21939729 DOI: 10.1016/j.pneurobio.2011.09.003] [Citation(s) in RCA: 158] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2011] [Revised: 09/04/2011] [Accepted: 09/06/2011] [Indexed: 01/31/2023]
Abstract
There is an increasing recognition that following traumatic brain injury, a cascade of inflammatory mediators is produced, and contributes to the pathological consequences of central nervous system injury. This review summarises the key literature from pre-clinical models that underlies our understanding of innate inflammation following traumatic brain injury before focussing on the growing evidence from human studies. In addition, the underlying molecular mediators responsible for blood brain barrier dysfunction have been discussed. In particular, we have highlighted the different sampling methodologies available and the difficulties in interpreting human data of this sort. Ultimately, understanding the innate inflammatory response to traumatic brain injury may provide a therapeutic avenue in the treatment of central nervous system disease.
Collapse
Affiliation(s)
- Adel Helmy
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Box 167, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK.
| | | | | | | | | |
Collapse
|
40
|
Khuman J, Zhang J, Park J, Carroll JD, Donahue C, Whalen MJ. Low-level laser light therapy improves cognitive deficits and inhibits microglial activation after controlled cortical impact in mice. J Neurotrauma 2011; 29:408-17. [PMID: 21851183 DOI: 10.1089/neu.2010.1745] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Low-level laser light therapy (LLLT) exerts beneficial effects on motor and histopathological outcomes after experimental traumatic brain injury (TBI), and coherent near-infrared light has been reported to improve cognitive function in patients with chronic TBI. However, the effects of LLLT on cognitive recovery in experimental TBI are unknown. We hypothesized that LLLT administered after controlled cortical impact (CCI) would improve post-injury Morris water maze (MWM) performance. Low-level laser light (800 nm) was applied directly to the contused parenchyma or transcranially in mice beginning 60-80 min after CCI. Injured mice treated with 60 J/cm² (500 mW/cm²×2 min) either transcranially or via an open craniotomy had modestly improved latency to the hidden platform (p<0.05 for group), and probe trial performance (p<0.01) compared to non-treated controls. The beneficial effects of LLLT in open craniotomy mice were associated with reduced microgliosis at 48 h (21.8±2.3 versus 39.2±4.2 IbA-1+ cells/200×field, p<0.05). Little or no effect of LLLT on post-injury cognitive function was observed using the other doses, a 4-h administration time point and 7-day administration of 60 J/cm². No effect of LLLT (60 J/cm² open craniotomy) was observed on post-injury motor function (days 1-7), brain edema (24 h), nitrosative stress (24 h), or lesion volume (14 days). Although further dose optimization and mechanism studies are needed, the data suggest that LLLT might be a therapeutic option to improve cognitive recovery and limit inflammation after TBI.
Collapse
Affiliation(s)
- Jugta Khuman
- Neuroscience Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | | | | | | | | | | |
Collapse
|
41
|
Khuman J, Meehan WP, Zhu X, Qiu J, Hoffmann U, Zhang J, Giovannone E, Lo EH, Whalen MJ. Tumor necrosis factor alpha and Fas receptor contribute to cognitive deficits independent of cell death after concussive traumatic brain injury in mice. J Cereb Blood Flow Metab 2011; 31:778-89. [PMID: 20940727 PMCID: PMC3049532 DOI: 10.1038/jcbfm.2010.172] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2010] [Revised: 08/21/2010] [Accepted: 09/01/2010] [Indexed: 11/09/2022]
Abstract
Tumor necrosis factor alpha (TNFα) and Fas receptor contribute to cell death and cognitive dysfunction after focal traumatic brain injury (TBI). We examined the role of TNFα/Fas in postinjury functional outcome independent of cell death in a novel closed head injury (CHI) model produced with weight drop and free rotational head movement in the anterior-posterior plane. The CHI produced no cerebral edema or blood-brain barrier damage at 24 to 48 hours, no detectable cell death, occasional axonal injury (24 hours), and no brain atrophy or hippocampal cell loss (day 60). Microglia and astrocytes were activated (48 to 72 hours). Tumor necrosis factor-α mRNA, Fas mRNA, and TNFα protein were increased in the brain at 3 to 6 hours after injury (P<0.001 versus sham injured). In wild-type (WT) mice, CHI produced hidden platform (P=0.009) and probe deficits (P=0.001) in the Morris water maze versus sham. Surprisingly, injured TNFα/Fas knockout (KO) mice performed worse in hidden platform trials (P=0.036) but better in probe trials than did WT mice (P=0.0001). Administration of recombinant TNFα to injured TNFα/Fas KO mice reduced probe trial performance to that of WT. Thus, TNFα/Fas influence cognitive deficits independent of cell death after CHI. Therapies targeting TNFα/Fas together may be inappropriate for patients with concussive TBI.
Collapse
Affiliation(s)
- Jugta Khuman
- Neuroscience Center, Harvard Medical School, Massachusetts General Hospital, Charlestown, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - William P Meehan
- Neuroscience Center, Harvard Medical School, Massachusetts General Hospital, Charlestown, Massachusetts, USA
- Division of Emergency Medicine, Department of Medicine, Harvard Medical School, Children's Hospital Boston, Boston, Massachusetts, USA
- Division of Sports Medicine, Department of Orthopedics, Sports Concussion Clinic, Children's Hospital Boston, Harvard Medical School, Boston, Massachusetts, USA
| | - Xiaoxia Zhu
- Neuroscience Center, Harvard Medical School, Massachusetts General Hospital, Charlestown, Massachusetts, USA
- Department of Rheumatology Huashan Hospital, Fudan University, Shanghai, China
| | - Jianhua Qiu
- Neuroscience Center, Harvard Medical School, Massachusetts General Hospital, Charlestown, Massachusetts, USA
- Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Ulrike Hoffmann
- Neuroscience Center, Harvard Medical School, Massachusetts General Hospital, Charlestown, Massachusetts, USA
- Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Jimmy Zhang
- Neuroscience Center, Harvard Medical School, Massachusetts General Hospital, Charlestown, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Eric Giovannone
- Neuroscience Center, Harvard Medical School, Massachusetts General Hospital, Charlestown, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Eng H Lo
- Neuroscience Center, Harvard Medical School, Massachusetts General Hospital, Charlestown, Massachusetts, USA
- Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Michael J Whalen
- Neuroscience Center, Harvard Medical School, Massachusetts General Hospital, Charlestown, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| |
Collapse
|
42
|
Yang J, You Z, Kim HH, Hwang SK, Khuman J, Guo S, Lo EH, Whalen MJ. Genetic analysis of the role of tumor necrosis factor receptors in functional outcome after traumatic brain injury in mice. J Neurotrauma 2010; 27:1037-46. [PMID: 20205514 DOI: 10.1089/neu.2009.1229] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
We previously reported that tumor necrosis factor-alpha (TNF-alpha) and Fas receptor induce acute cellular injury, tissue damage, and motor and cognitive deficits after controlled cortical impact (CCI) in mice (Bermpohl et al. 2007 ); however, the TNF receptors (TNFR) involved are unknown. Using a CCI model and novel mutant mice deficient in TNFR1/Fas, TNFR2/Fas, or TNFR1/TNFR2/Fas, we tested the hypothesis that the combination of TNFR2/Fas is protective, whereas TNFR1/Fas is detrimental after CCI. Uninjured knockout (KO) mice showed no differences in baseline physiological variables or motor or cognitive function. Following CCI, mice deficient in TNFR2/Fas had worse post-injury motor and Morris water maze (MWM) performance than wild-type (WT) mice (p < 0.05 group effect for wire grip score and MWM performance by repeated measures ANOVA). No differences in motor or cognitive outcome were observed in TNFR1/Fas KO, or in TNFR2 or TNFR1 single KO mice, versus WT mice. Additionally, no differences in propidium iodide (PI)-positive cells (at 6 h) or lesion size (at 14 days) were observed between WT and TNFR1/Fas or TNFR2/Fas KO mice. Somewhat surprisingly, mice deficient in TNFR1/TNFR2/Fas also had PI-positive cells, lesion size, and motor and MWM deficits similar to those of WT mice. These data suggest a protective role for TNFR2/Fas in the pathogenesis of TBI. Further studies are needed to determine whether direct or indirect effects of TNFR1 deletion in TNFR2/Fas KO mice mediate improved functional outcome in TNFR1/TNFR2/Fas KO mice after CCI.
Collapse
Affiliation(s)
- Jinsheng Yang
- Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Mannose-binding lectin pathway is not involved in myasthenia gravis pathogenesis. J Neuroimmunol 2009; 208:40-5. [PMID: 19193448 DOI: 10.1016/j.jneuroim.2008.12.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2008] [Revised: 12/29/2008] [Accepted: 12/30/2008] [Indexed: 12/18/2022]
Abstract
Classical complement pathway factor, C4 is required for experimental autoimmune myasthenia gravis (EAMG) pathogenesis. C4 is also a central component of the mannose binding lectin (MBL) pathway suggesting that this pathway might also be involved in MG pathogenesis. However, MBL gene deficient mice displayed intact anti-acetylcholine receptor (AChR)-immune response and neuromuscular junction (NMJ) IgG and complement accumulation following AChR-immunization. Moreover, no significant difference was observed between the serum MBL levels of 77 anti-AChR antibody positive generalized MG patients and 105 healthy controls. Therefore, MBL pathway does not play a role in EAMG/MG pathogenesis.
Collapse
|
44
|
Stahel PF, Flierl MA, Morgan BP, Persigehl I, Stoll C, Conrad C, Touban BM, Smith WR, Beauchamp K, Schmidt OI, Ertel W, Leinhase I. Absence of the complement regulatory molecule CD59a leads to exacerbated neuropathology after traumatic brain injury in mice. J Neuroinflammation 2009; 6:2. [PMID: 19133139 PMCID: PMC2631471 DOI: 10.1186/1742-2094-6-2] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2008] [Accepted: 01/08/2009] [Indexed: 12/02/2022] Open
Abstract
Background Complement represents a crucial mediator of neuroinflammation and neurodegeneration after traumatic brain injury. The role of the terminal complement activation pathway, leading to generation of the membrane attack complex (MAC), has not been thoroughly investigated. CD59 is the major regulator of MAC formation and represents an essential protector from homologous cell injury after complement activation in the injured brain. Methods Mice deleted in the Cd59a gene (CD59a-/-) and wild-type littermates (n = 60) were subjected to focal closed head injury. Sham-operated (n = 60) and normal untreated mice (n = 14) served as negative controls. The posttraumatic neurological impairment was assessed for up to one week after trauma, using a standardized Neurological Severity Score (NSS). The extent of neuronal cell death was determined by serum levels of neuron-specific enolase (NSE) and by staining of brain tissue sections in TUNEL technique. The expression profiles of pro-apoptotic (Fas, FasL, Bax) and anti-apoptotic (Bcl-2) mediators were determined at the gene and protein level by real-time RT-PCR and Western blot, respectively. Results Clinically, the brain-injured CD59a-/- mice showed a significantly impaired neurological outcome within 7 days, as determined by a higher NSS, compared to wild-type controls. The NSE serum levels, an indirect marker of neuronal cell death, were significantly elevated in CD59a-/- mice at 4 h and 24 h after trauma, compared to wild-type littermates. At the tissue level, increased neuronal cell death and brain tissue destruction was detected by TUNEL histochemistry in CD59a-/- mice within 24 hours to 7 days after head trauma. The analysis of brain homogenates for potential mediators and regulators of cell death other than the complement MAC (Fas, FasL, Bax, Bcl-2) revealed no difference in gene expression and protein levels between CD59a-/- and wild-type mice. Conclusion These data emphasize an important role of CD59 in mediating protection from secondary neuronal cell death and further underscore the key role of the terminal complement pathway in the pathophysiology of traumatic brain injury. The exact mechanisms of complement MAC-induced secondary neuronal cell death after head injury require further investigation.
Collapse
Affiliation(s)
- Philip F Stahel
- Department of Orthopedic Surgery, Denver Health Medical Center, University of Colorado School of Medicine, Denver, CO 80204, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|