1
|
Sialylation-dependent pharmacokinetics and differential complement pathway inhibition are hallmarks of CR1 activity in vivo. Biochem J 2022; 479:1007-1030. [PMID: 35470373 DOI: 10.1042/bcj20220054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/21/2022] [Accepted: 04/26/2022] [Indexed: 11/17/2022]
Abstract
Human Complement Receptor 1 (HuCR1) is a potent membrane-bound regulator of complement both in vitro and in vivo, acting via interaction with its ligands C3b and C4b. Soluble versions of HuCR1 have been described such as TP10, the recombinant full-length extracellular domain, and more recently CSL040, a truncated version lacking the C-terminal long homologous repeat domain D (LHR-D). However, the role of N-linked glycosylation in determining its pharmacokinetic (PK) and pharmacodynamic (PD) properties is only partly understood. We demonstrated a relationship between the asialo-N-glycan levels of CSL040 and its PK/PD properties in rats and non-human primates (NHPs), using recombinant CSL040 preparations with varying asialo-N-glycan levels. The clearance mechanism likely involves the asialoglycoprotein receptor (ASGR), as clearance of CSL040 with a high proportion of asialo-N-glycans was attenuated in vivo by co-administration of rats with asialofetuin, which saturates the ASGR. Biodistribution studies also showed CSL040 localisation to the liver following systemic administration. Our studies uncovered differential PD effects by CSL040 on complement pathways, with extended inhibition in both rats and NHPs of the alternative pathway compared to the classical and lectin pathways that were not correlated with its PK profile. Further studies showed that this effect was dose dependent and observed with both CSL040 and the full-length extracellular domain of HuCR1. Taken together, our data suggests that sialylation optimization is an important consideration for developing HuCR1-based therapeutic candidates such as CSL040 with improved PK properties and shows that CSL040 has superior PK/PD responses compared to full-length soluble HuCR1.
Collapse
|
2
|
The role of complement in brain injury following intracerebral hemorrhage: A review. Exp Neurol 2021; 340:113654. [PMID: 33617886 DOI: 10.1016/j.expneurol.2021.113654] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 02/05/2021] [Accepted: 02/17/2021] [Indexed: 02/08/2023]
Abstract
Intracerebral hemorrhage (ICH) is a significant cause of death and disability and current treatment is limited to supportive measures to reduce brain edema and secondary hematoma expansion. Current evidence suggests that the complement cascade is activated early after hemorrhage and contributes to brain edema/injury in multiple ways. The aim of this review is to summarize the most recent literature about the role of the complement cascade after ICH. Primary literature demonstrating complement mediated brain edema and neurologic injury through the membrane attack complex (MAC) as well as C3a and C5a are reviewed. Further, attenuation of brain edema and improved functional outcomes are demonstrated after inhibition of specific components of the complement cascade. Conversely, complement also plays a significant role in neurologic recovery after ICH and in other neurologic disorders. We conclude that the role of complement after ICH is complex. Understanding the role of complement after ICH is essential and may elucidate possible interventions to reduce brain edema and injury.
Collapse
|
3
|
Clarke AR, Christophe BR, Khahera A, Sim JL, Connolly ES. Therapeutic Modulation of the Complement Cascade in Stroke. Front Immunol 2019; 10:1723. [PMID: 31417544 PMCID: PMC6682670 DOI: 10.3389/fimmu.2019.01723] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 07/09/2019] [Indexed: 01/22/2023] Open
Abstract
Stroke is a leading cause of death and disability worldwide and an increasing number of ischemic stroke patients are undergoing pharmacological and mechanical reperfusion. Both human and experimental models of reperfused ischemic stroke have implicated the complement cascade in secondary tissue injury. Most data point to the lectin and alternative pathways as key to activation, and C3a and C5a binding of their receptors as critical effectors of injury. During periods of thrombolysis use to treat stroke, acute experimental complement cascade blockade has been found to rescue tissue and improves functional outcome. Blockade of the complement cascade during the period of tissue reorganization, repair, and recovery is by contrast not helpful and in fact is likely to be deleterious with emerging data suggesting downstream upregulation of the cascade might even facilitate recovery. Successful clinical translation will require the right clinical setting and pharmacologic strategies that are capable of targeting the key effectors early while not inhibiting delayed repair. Early reports in a variety of disease states suggest that such pharmacologic strategies appear to have a favorable risk profile and offer substantial hope for patients.
Collapse
Affiliation(s)
- Alison R Clarke
- Cerebrovascular Research Laboratory, Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, United States
| | - Brandon R Christophe
- Cerebrovascular Research Laboratory, Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, United States
| | - Anadjeet Khahera
- Cerebrovascular Research Laboratory, Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, United States
| | - Justin L Sim
- Cerebrovascular Research Laboratory, Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, United States
| | - E Sander Connolly
- Cerebrovascular Research Laboratory, Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, United States
| |
Collapse
|
4
|
Malone K, Amu S, Moore AC, Waeber C. Immunomodulatory Therapeutic Strategies in Stroke. Front Pharmacol 2019; 10:630. [PMID: 31281252 PMCID: PMC6595144 DOI: 10.3389/fphar.2019.00630] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 05/16/2019] [Indexed: 12/14/2022] Open
Abstract
The role of immunity in all stages of stroke is increasingly being recognized, from the pathogenesis of risk factors to tissue repair, leading to the investigation of a range of immunomodulatory therapies. In the acute phase of stroke, proposed therapies include drugs targeting pro-inflammatory cytokines, matrix metalloproteinases, and leukocyte infiltration, with a key objective to reduce initial brain cell toxicity. Systemically, the early stages of stroke are also characterized by stroke-induced immunosuppression, where downregulation of host defences predisposes patients to infection. Therefore, strategies to modulate innate immunity post-stroke have garnered greater attention. A complementary objective is to reduce longer-term sequelae by focusing on adaptive immunity. Following stroke onset, the integrity of the blood–brain barrier is compromised, exposing central nervous system (CNS) antigens to systemic adaptive immune recognition, potentially inducing autoimmunity. Some pre-clinical efforts have been made to tolerize the immune system to CNS antigens pre-stroke. Separately, immune cell populations that exhibit a regulatory phenotype (T- and B- regulatory cells) have been shown to ameliorate post-stroke inflammation and contribute to tissue repair. Cell-based therapies, established in oncology and transplantation, could become a strategy to treat the acute and chronic stages of stroke. Furthermore, a role for the gut microbiota in ischaemic injury has received attention. Finally, the immune system may play a role in remote ischaemic preconditioning-mediated neuroprotection against stroke. The development of stroke therapies involving organs distant to the infarct site, therefore, should not be overlooked. This review will discuss the immune mechanisms of various therapeutic strategies, surveying published data and discussing more theoretical mechanisms of action that have yet to be exploited.
Collapse
Affiliation(s)
- Kyle Malone
- Department of Pharmacology and Therapeutics, School of Pharmacy, University College Cork, Cork, Ireland
| | - Sylvie Amu
- Cancer Research @UCC, University College Cork, Cork, Ireland
| | - Anne C Moore
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Christian Waeber
- Department of Pharmacology and Therapeutics, School of Pharmacy, University College Cork, Cork, Ireland
| |
Collapse
|
5
|
Ma Y, Liu Y, Zhang Z, Yang GY. Significance of Complement System in Ischemic Stroke: A Comprehensive Review. Aging Dis 2019; 10:429-462. [PMID: 31011487 PMCID: PMC6457046 DOI: 10.14336/ad.2019.0119] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 01/19/2019] [Indexed: 12/14/2022] Open
Abstract
The complement system is an essential part of innate immunity, typically conferring protection via eliminating pathogens and accumulating debris. However, the defensive function of the complement system can exacerbate immune, inflammatory, and degenerative responses in various pathological conditions. Cumulative evidence indicates that the complement system plays a critical role in the pathogenesis of ischemic brain injury, as the depletion of certain complement components or the inhibition of complement activation could reduce ischemic brain injury. Although multiple candidates modulating or inhibiting complement activation show massive potential for the treatment of ischemic stroke, the clinical availability of complement inhibitors remains limited. The complement system is also involved in neural plasticity and neurogenesis during cerebral ischemia. Thus, unexpected side effects could be induced if the systemic complement system is inhibited. In this review, we highlighted the recent concepts and discoveries of the roles of different kinds of complement components, such as C3a, C5a, and their receptors, in both normal brain physiology and the pathophysiology of brain ischemia. In addition, we comprehensively reviewed the current development of complement-targeted therapy for ischemic stroke and discussed the challenges of bringing these therapies into the clinic. The design of future experiments was also discussed to better characterize the role of complement in both tissue injury and recovery after cerebral ischemia. More studies are needed to elucidate the molecular and cellular mechanisms of how complement components exert their functions in different stages of ischemic stroke to optimize the intervention of targeting the complement system.
Collapse
Affiliation(s)
- Yuanyuan Ma
- 1Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,2Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yanqun Liu
- 3Department of Neurology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Zhijun Zhang
- 2Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Guo-Yuan Yang
- 1Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,2Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
6
|
Malone K, Amu S, Moore AC, Waeber C. The immune system and stroke: from current targets to future therapy. Immunol Cell Biol 2018; 97:5-16. [DOI: 10.1111/imcb.12191] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 07/11/2018] [Accepted: 07/16/2018] [Indexed: 01/15/2023]
Affiliation(s)
- Kyle Malone
- Department of Pharmacology and Therapeutics; School of Pharmacy; University College Cork; Cork Ireland
| | - Sylvie Amu
- Department of Pharmacology and Therapeutics; School of Pharmacy; University College Cork; Cork Ireland
| | - Anne C Moore
- Department of Pharmacology and Therapeutics; School of Pharmacy; University College Cork; Cork Ireland
| | - Christian Waeber
- Department of Pharmacology and Therapeutics; School of Pharmacy; University College Cork; Cork Ireland
| |
Collapse
|
7
|
Tissue-targeted complement therapeutics. Mol Immunol 2018; 102:120-128. [PMID: 30220307 DOI: 10.1016/j.molimm.2018.06.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 06/04/2018] [Accepted: 06/05/2018] [Indexed: 02/07/2023]
Abstract
Complement activation contributes to the pathogenesis of numerous inflammatory and autoimmune diseases. Therapeutic complement inhibitors have proven effective in several of these diseases and have now entered clinical use. Complement activation has multiple different biologic effects, however, and the currently available drugs can have undesirable side-effects, such as an increased risk of infection. Several different complement inhibitors have been developed that bind to target molecules, thereby concentrating the drug at a specific anatomic site. This approach appears to be both more effective than untargeted drugs and to have fewer side effects. In this article we review different targeting strategies that have been developed and the evidence supporting the use and benefits of targeted drugs.
Collapse
|
8
|
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: 7.4] [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
|
9
|
Chen D, Song MQ, Liu YJ, Xue YK, Cheng P, Zheng H, Chen LB. Inhibition of complement C3 might rescue vascular hyporeactivity in a conscious hemorrhagic shock rat model. Microvasc Res 2015; 105:23-9. [PMID: 26687560 DOI: 10.1016/j.mvr.2015.12.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 12/08/2015] [Accepted: 12/08/2015] [Indexed: 11/26/2022]
Abstract
BACKGROUND Vascular hyporeactivity in severe hemorrhagic shock could induce refractory hypotension and is an important cause of death. The global acute inflammatory response induced in shock triggers the over-expression of reactive oxygen species, NO, ET1 and TNF-α, which play essential roles in the pathology of vascular hyporeactivity. This leads to a hypothesis that inhibition of the complement system, the mediator of the inflammatory cascade, might be a promising therapeutic exploration for vascular hyporeactivity. METHODS We use cobra venom factor (CVF) and the soluble form of CR1 (sCR1) which deplete or inhibit complement C3 respectively to examine its role in vascular hyporeactivity in a conscious hemorrhagic shock rat model. RESULTS We first confirmed the over-activation of C3 during shock and the down-regulation effects of CVF and sCR1 on C3. Then, both CVF and sCR1 could significantly mitigate the over-expression of serum NO, ET-1, TNF-α and reactive oxygen species. Finally, the vascular reactivity of superior mesenteric arteries (SMA) was examined in vitro, which confirmed the massive reduction of vascular reactivity in shock, which was significantly rescued by both CVF and sCR1. CONCLUSIONS Inhibition of C3 might improve the reactivity of SMA to norepinephrine during hemorrhagic shock possibly through the downregulation of NO, ET1, TNF-α and reactive oxygen radicals.
Collapse
Affiliation(s)
- Ding Chen
- Department of Emergency, Wuhan Union Hospital affiliated with Tongji Medical College, Huazhong University of Science and Technology, PR China.
| | - Meng-Qi Song
- Department of Emergency, Wuhan Union Hospital affiliated with Tongji Medical College, Huazhong University of Science and Technology, PR China.
| | - Yan-Jun Liu
- Department of Emergency, Wuhan Union Hospital affiliated with Tongji Medical College, Huazhong University of Science and Technology, PR China.
| | - Yin-Kai Xue
- Department of Emergency, Wuhan Union Hospital affiliated with Tongji Medical College, Huazhong University of Science and Technology, PR China.
| | - Ping Cheng
- Department of Emergency, Wuhan Union Hospital affiliated with Tongji Medical College, Huazhong University of Science and Technology, PR China.
| | - Hai Zheng
- Department of Emergency, Wuhan Union Hospital affiliated with Tongji Medical College, Huazhong University of Science and Technology, PR China.
| | - Li-Bo Chen
- Department of Emergency, Wuhan Union Hospital affiliated with Tongji Medical College, Huazhong University of Science and Technology, PR China.
| |
Collapse
|
10
|
Alawieh A, Elvington A, Tomlinson S. Complement in the Homeostatic and Ischemic Brain. Front Immunol 2015; 6:417. [PMID: 26322048 PMCID: PMC4533015 DOI: 10.3389/fimmu.2015.00417] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 07/30/2015] [Indexed: 11/29/2022] Open
Abstract
The complement system is a component of the immune system involved in both recognition and response to pathogens, and it is implicated in an increasing number of homeostatic and disease processes. It is well documented that reperfusion of ischemic tissue results in complement activation and an inflammatory response that causes post-reperfusion injury. This occurs following cerebral ischemia and reperfusion and triggers secondary damage that extends beyond the initial infarcted area, an outcome that has rationalized the use of complement inhibitors as candidate therapeutics after stroke. In the central nervous system, however, recent studies have revealed that complement also has essential roles in synaptic pruning, neurogenesis, and neuronal migration. In the context of recovery after stroke, these apparent divergent functions of complement may account for findings that the protective effect of complement inhibition in the acute phase after stroke is not always maintained in the subacute and chronic phases. The development of effective stroke therapies based on modulation of the complement system will require a detailed understanding of complement-dependent processes in both early neurodegenerative events and delayed neuro-reparatory processes. Here, we review the role of complement in normal brain physiology, the events initiating complement activation after cerebral ischemia-reperfusion injury, and the contribution of complement to both injury and recovery. We also discuss how the design of future experiments may better characterize the dual role of complement in recovery after ischemic stroke.
Collapse
Affiliation(s)
- Ali Alawieh
- Neuroscience Institute, Department of Neurosciences, Medical University of South Carolina , Charleston, SC , USA
| | - Andrew Elvington
- Department of Pathology and Immunology, Washington University School of Medicine , St. Louis, MO , USA
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Ralph H. Johnson Veteran Affairs Medical Center, Medical University of South Carolina , Charleston, SC , USA
| |
Collapse
|
11
|
Li S, Xian J, He L, Luo X, Tan B, Yang Y, Liu G, Wang Z. The protective effect of SCR15-18on cerebral ischemia-reperfusion injury. Neurol Res 2013; 33:866-74. [DOI: 10.1179/1743132811y.0000000016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
12
|
Yang S, Wang X, Zhang X, Lu Y, Wang Z. Neuroprotective effects of the SCR1-3 functional domain of CR1 on acute cerebral ischemia and reperfusion injury in rats. Neurol Res 2013; 35:976-83. [PMID: 23815859 DOI: 10.1179/1743132813y.0000000238] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVE Complement receptor type 1 (CR1), one of the most potent inhibitors in complement activation, shows a protective effect on cerebral ischemia/reperfusion (CI/R) injury due to its ability to bind C3b and C4b and to inactivate C3/C5 convertases. So far, no study assessed the effect of the first three short consensus repeats (SCR1-3) with low molecular weight, one of the most active functional domains of CR1, binding C4b with a powerful decay-acceleration effect on classical and alternative C3/C5 convertases pathways. Therefore, we aim to assess this effect on CI/R injury in the present study. METHODS Seventy-five adult male Sprague-Dawley rats were randomly divided into three groups: sham operation group (n = 15), CI/R group (n = 30), and CI/R group treated with CR1-SCR1-3 protein (n = 30). After middle cerebral artery occlusion (MCAO) for 1 hour and reperfusion for 24 hours, neurological motor deficits, cerebral infarct size, and biochemical parameters including myeloperoxidase (MPO), malondialdehyde (MDA), and superoxide dismutase (SOD) were assessed. Meanwhile, tissues in cerebral cortex were collected and processed for western blotting, immunohistochemistry, and HE staining. RESULTS CR1-SCR1-3 could improve neurological functions in brain with a 26.8% decrease in neurological motor deficit score and could lead to a 63.8% reduction in cerebral infarct size. Besides, pretreatment using CR1-SCR1-3 could prevent neutrophil infiltration and alleviate inflammation severity and subsequent tissue damage. Decreased C4b expression and action, as well as improved morphological changes, were also observed in cerebral tissues of CI/R+CR1-SCR1-3 rats. CONCLUSION CR1-SCR1-3 protein could possess a neuroprotective effect on acute CI/R injury.
Collapse
Affiliation(s)
- Shaojun Yang
- Third Military Medical University, Chongqing, China
| | | | | | | | | |
Collapse
|
13
|
Brennan FH, Anderson AJ, Taylor SM, Woodruff TM, Ruitenberg MJ. Complement activation in the injured central nervous system: another dual-edged sword? J Neuroinflammation 2012; 9:137. [PMID: 22721265 PMCID: PMC3464784 DOI: 10.1186/1742-2094-9-137] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 06/21/2012] [Indexed: 11/28/2022] Open
Abstract
The complement system, a major component of the innate immune system, is becoming increasingly recognised as a key participant in physiology and disease. The awareness that immunological mediators support various aspects of both normal central nervous system (CNS) function and pathology has led to a renaissance of complement research in neuroscience. Various studies have revealed particularly novel findings on the wide-ranging involvement of complement in neural development, synapse elimination and maturation of neural networks, as well as the progression of pathology in a range of chronic neurodegenerative disorders, and more recently, neurotraumatic events, where rapid disruption of neuronal homeostasis potently triggers complement activation. The purpose of this review is to summarise recent findings on complement activation and acquired brain or spinal cord injury, i.e. ischaemic-reperfusion injury or stroke, traumatic brain injury (TBI) and spinal cord injury (SCI), highlighting the potential for complement-targeted therapeutics to alleviate the devastating consequences of these neurological conditions.
Collapse
Affiliation(s)
- Faith H Brennan
- The University of Queensland, School of Biomedical Sciences, St Lucia, Brisbane, QLD 4072, Australia
| | | | | | | | | |
Collapse
|
14
|
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]
|
15
|
Cook DJ, Tymianski M. Translating promising preclinical neuroprotective therapies to human stroke trials. Expert Rev Cardiovasc Ther 2011; 9:433-49. [PMID: 21517728 DOI: 10.1586/erc.11.34] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Stroke is the third leading cause of mortality and carries the greatest socioeconomic burden of disease in North America. Despite several promising therapies discovered in the preclinical setting, there have been no positive results in human stroke clinical trials to date. In this article, we review the potential causes for failure and discuss strategies that have been proposed to overcome the barrier to translation of stroke therapies. To improve the chance of success in future human stroke trials, we propose that therapies be tested in stroke models that closely resemble the human condition with molecular, imaging and functional outcomes that relate to outcomes utilized in clinical trials. These strategies include higher-order, old-world, nonhuman primate models of stroke with clinically relevant outcome measures. Although stroke neuroprotection has been looked upon pessimistically given the many failures in clinical trials to date, we propose that neuroprotection in humans is feasible and will be realized with rigorous translational science.
Collapse
Affiliation(s)
- Douglas James Cook
- University of Toronto, Department of Surgery, Division of Neurosurgery, Toronto Western Research Institute Neuroprotection Laboratory, 11-414 MCl 399 Bathurst St, Toronto, ON, M5T 2S8, Canada
| | | |
Collapse
|
16
|
Preclinical evaluation of postischemic dehydroascorbic Acid administration in a large-animal stroke model. Transl Stroke Res 2011; 2:399-403. [PMID: 24323656 DOI: 10.1007/s12975-011-0084-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Revised: 04/01/2011] [Accepted: 04/26/2011] [Indexed: 10/18/2022]
Abstract
Dehydroascorbic acid (DHA), a blood-brain barrier transportable form of ascorbic acid, confers robust neuroprotection following murine stroke. In an effort to translate this promising neuroprotective strategy into human clinical trial, we evaluated postischemic DHA administration in a large-animal stroke model. Thirty-six adult male baboons were initially randomized to undergo transorbital craniectomy to induce transient cerebral artery occlusion and to receive postischemic dosing of either 500 mg/kg of DHA or vehicle. Primary outcomes included infarct volume, determined by magnetic resonance imaging, as well as neurological function evaluated on the day of sacrifice. The midpoint interim analysis (n = 9 per cohort) revealed that DHA administration did not significantly improve either infarct volume or neurological function. The study was terminated after a determination of statistical futility. We were unable to confirm a neuroprotective effect for postischemic DHA administration in our large-animal model using a dosing scheme that was previously successful in rodents. Further analysis of the efficacy of DHA administration must thus be undertaken prior to clinical translation.
Collapse
|
17
|
Abstract
The complement system is an essential component of innate immunity that has been more recently recognized as an unexpected player in various pathological states. These include age-related macular degeneration, atypical haemolytic uraemic syndrome, allergy, foetal loss, and axonal and myelin degradation after trauma. Its importance has also been recognized in physiological processes including haematopoietic stem cell homing to the bone marrow, liver regeneration and modulation of adaptive immune responses. Although the complement system has long been known to be involved in autoimmune and inflammatory diseases, few agents that target the complement system are currently approved for clinical use. However, renewed interest in modulating this system in various pathological conditions has emerged, and several agents are now in development.
Collapse
|
18
|
Sughrue ME, Mocco J, Mack WJ, Ducruet AF, Komotar RJ, Fischbach RL, Martin TE, Connolly ES. Bioethical considerations in translational research: primate stroke. THE AMERICAN JOURNAL OF BIOETHICS : AJOB 2009; 9:3-12. [PMID: 19396671 DOI: 10.1080/15265160902788652] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Controversy and activism have long been linked to the subject of primate research. Even in the midst of raging ethical debates surrounding fertility treatments, genetically modified foods and stem-cell research, there has been no reduction in the campaigns of activists worldwide. Playing their trade of intimidation aimed at ending biomedical experimentation in all animals, they have succeeded in creating an environment where research institutions, often painted as guilty until proven innocent, have avoided addressing the issue for fear of becoming targets. One area of intense debate is the use of primates in stroke research. Despite the fact that stroke kills more people each year than AIDS and malaria, and less than 5% of patients are candidates for current therapies, there is significant opposition to primate stroke research. A balanced examination of the ethics of primate stroke research is thus of broad interest to all areas of biomedical research.
Collapse
Affiliation(s)
- Michael E Sughrue
- Department of Neurological Surgery, Columbia University, College of Physicians & Surgeons, New York, NY 10032, USA
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Arumugam TV, Woodruff TM, Lathia JD, Selvaraj PK, Mattson MP, Taylor SM. Neuroprotection in stroke by complement inhibition and immunoglobulin therapy. Neuroscience 2009; 158:1074-89. [PMID: 18691639 PMCID: PMC2639633 DOI: 10.1016/j.neuroscience.2008.07.015] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2008] [Revised: 07/08/2008] [Accepted: 07/08/2008] [Indexed: 12/18/2022]
Abstract
Activation of the complement system occurs in a variety of neuroinflammatory diseases and neurodegenerative processes of the CNS. Studies in the last decade have demonstrated that essentially all of the activation components and receptors of the complement system are produced by astrocytes, microglia, and neurons. There is also rapidly growing evidence to indicate an active role of the complement system in cerebral ischemic injury. In addition to direct cell damage, regional cerebral ischemia and reperfusion (I/R) induces an inflammatory response involving complement activation and generation of active fragments, such as C3a and C5a anaphylatoxins, C3b, C4b, and iC3b. The use of specific inhibitors to block complement activation or their mediators such as C5a, can reduce local tissue injury after I/R. Consistent with therapeutic approaches that have been successful in models of autoimmune disorders, many of the same complement inhibition strategies are proving effective in animal models of cerebral I/R injury. One new form of therapy, which is less specific in its targeting of complement than monodrug administration, is the use of immunoglobulins. Intravenous immunoglobulin (IVIG) has the potential to inhibit multiple components of inflammation, including complement fragments, pro-inflammatory cytokine production and leukocyte cell adhesion. Thus, IVIG may directly protect neurons, reduce activation of intrinsic inflammatory cells (microglia) and inhibit transendothelial infiltration of leukocytes into the brain parenchyma following an ischemic stroke. The striking neuroprotective actions of IVIG in animal models of ischemic stroke suggest a potential therapeutic potential that merits consideration for clinical trials in stroke patients.
Collapse
Affiliation(s)
- T V Arumugam
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, 1300 Coulter Drive, Amarillo, TX 79106, USA.
| | | | | | | | | | | |
Collapse
|
20
|
Molecular mechanisms of thrombus formation in ischemic stroke: novel insights and targets for treatment. Blood 2008; 112:3555-62. [DOI: 10.1182/blood-2008-04-144758] [Citation(s) in RCA: 171] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In ischemic stroke, treatment options are limited. Therapeutic thrombolysis is restricted to the first few hours after stroke, and the utility of current platelet aggregation inhibitors, including GPIIb/IIIa receptor antagonists, and anticoagulants is counterbalanced by the risk of intracerebral bleeding complications. Numerous attempts to establish neuroprotection in ischemic stroke have been unfruitful. Thus, there is strong demand for novel treatment strategies. Major advances have been made in understanding the molecular functions of platelet receptors such as glycoprotein Ib (GPIb) and GPVI and their downstream signaling pathways that allow interference with their function. Inhibition of these receptors in the mouse stroke model of transient middle cerebral artery occlusion prevented infarctions without increasing the risk of intracerebral bleeding. Similarly, it is now clear that the intrinsic coagulation factor XII (FXII) and FXI play a functional role in thrombus formation and stabilization during stroke: their deficiency or blockade protects from cerebral ischemia without overtly affecting hemostasis. Based on the accumulating evidence that thrombus formation and hemostasis are not inevitably linked, new concepts for prevention and treatment of ischemic stroke may eventually emerge without the hazard of severe bleeding complications. This review discusses recent advances related to antithrombotic strategies in experimental stroke research.
Collapse
|
21
|
Strong complement activation after acute ischemic stroke is associated with unfavorable outcomes. Atherosclerosis 2008; 204:315-20. [PMID: 18804761 DOI: 10.1016/j.atherosclerosis.2008.07.044] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2008] [Revised: 07/03/2008] [Accepted: 07/20/2008] [Indexed: 10/21/2022]
Abstract
OBJECTIVE According to data from animal models, complement activation plays a major role in the brain injury after acute ischemic stroke. Scarce findings are, however, available on the detection of complement activation products in stroke patients. METHODS We have measured plasma levels of the five complement activation products (C1rC1sC1inh, C4d, C3a, C5a and SC5b-9) in samples of 26 patients with ischemic stroke upon admission. Twenty-six patients with severe carotid atherosclerosis served as patient controls. RESULTS Levels of two activation products (SC5b-9 and C4d)) were significantly elevated in the plasma of stroke patients, SC5b-9 levels, exhibited significant positive correlation with the clinical severity of stroke, the severity of neurological deficit, as well as with the level of functional disability. CONCLUSION These findings suggest that complement activation plays an active role in the development of brain infarct. The measurement of complement activation products might help to determine the clinical prognosis after acute ischemic stroke. Furthermore, there is potential usefulness of complement modulating therapy in ischemic stroke.
Collapse
|
22
|
|
23
|
Arumugam TV, Selvaraj PK, Woodruff TM, Mattson MP. Targeting ischemic brain injury with intravenous immunoglobulin. Expert Opin Ther Targets 2007; 12:19-29. [DOI: 10.1517/14728222.12.1.19] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
24
|
Ducruet AF, Mocco J, Mack WJ, Coon AL, Marsh HC, Pinsky DJ, Hickman ZL, Kim GH, Connolly ES. Pre-clinical evaluation of an sLex-glycosylated complement inhibitory protein in a non-human primate model of reperfused stroke. J Med Primatol 2007; 36:375-80. [DOI: 10.1111/j.1600-0684.2007.00213.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
25
|
Abstract
Age-related macular degeneration (AMD) is the leading cause of blindness in developed countries. There is no effective treatment for the most prevalent atrophic (dry) form of AMD. Atrophic AMD is triggered by abnormalities in the retinal pigment epithelium (RPE) that lies beneath the photoreceptor cells and normally provides critical metabolic support to these light-sensing cells. Secondary to RPE dysfunction, macular rods and cones degenerate leading to the irreversible loss of vision. Oxidative stress, formation of drusen, accumulation of lipofuscin, local inflammation and reactive gliosis represent the pathologic processes implicated in pathogenesis of atrophic AMD. This review discusses potential target areas for small-molecule and biologic intervention, which may lead to development of new therapeutic treatments for atrophic AMD.
Collapse
Affiliation(s)
- Konstantin Petrukhin
- Columbia University, Department of Ophthalmology, Eye Institute Annex, New York, NY 10032, USA.
| |
Collapse
|