1
|
Vygonskaya M, Wu Y, Price TJ, Chen Z, Smith MT, Klyne DM, Han FY. The role and treatment potential of the complement pathway in chronic pain. THE JOURNAL OF PAIN 2024:104689. [PMID: 39362355 DOI: 10.1016/j.jpain.2024.104689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/05/2024]
Abstract
The role of the complement system in pain syndromes has garnered attention on the back of preclinical and clinical evidence supporting its potential as a target for new analgesic pharmacotherapies. Of the components that make up the complement system, component 5a (C5a) and component 3a (C3a) are most strongly and consistently associated with pain. Receptors for C5a are widely found in immune resident cells (microglia, astrocytes, sensory neuron-associated macrophages (sNAMs)) in the central nervous system (CNS) as well as hematogenous immune cells (mast cells, macrophages, T-lymphocytes, etc.). When active, as is often observed in chronic pain conditions, these cells produce various inflammatory mediators including pro-inflammatory cytokines. These events can trigger nervous tissue inflammation (neuroinflammation) which coexists with and potentially maintains peripheral and central sensitization. C5a has a likely critical role in initiating this process highlighting its potential as a promising non-opioid target for treating pain. This review summarises the most up-to-date research on the role of the complement system in pain with emphasis on the C5 pathway in peripheral tissue, dorsal root ganglia (DRG) and the CNS, and explores advances in complement-targeted drug development and sex differences. A perspective on the optimal application of different C5a inhibitors for different types (e.g., neuropathic, post-surgical and chemotherapy-induced pain, osteoarthritis pain) and stages (e.g., acute, subacute, chronic) of pain is also provided to help guide future clinical trials. PERSPECTIVE: This review highlights the role and mechanisms of complement components and their receptors in physiological and pathological pain. The potential of complement-targeted therapeutics for the treatment of chronic pain is also explored with a focus on C5a inhibitors to help guide future clinical trials.
Collapse
Affiliation(s)
- Marina Vygonskaya
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Youzhi Wu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Theodore J Price
- Center for Advanced Pain Studies, Department of Neuroscience, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Zhuo Chen
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Maree T Smith
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia
| | - David M Klyne
- NHMRC Centre of Clinical Research Excellence in Spinal Pain, Injury and Health, The University of Queensland, St Lucia, Brisbane, QLD 4072, Australia
| | - Felicity Y Han
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
2
|
Schanzenbacher J, Hendrika Kähler K, Mesler E, Kleingarn M, Marcel Karsten C, Leonard Seiler D. The role of C5a receptors in autoimmunity. Immunobiology 2023; 228:152413. [PMID: 37598588 DOI: 10.1016/j.imbio.2023.152413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/04/2023] [Accepted: 06/10/2023] [Indexed: 08/22/2023]
Abstract
The complement system is an essential component of the innate immune response and plays a vital role in host defense and inflammation. Dysregulation of the complement system, particularly involving the anaphylatoxin C5a and its receptors (C5aR1 and C5aR2), has been linked to several autoimmune diseases, indicating the potential for targeted therapies. C5aR1 and C5aR2 are seven-transmembrane receptors with distinct signaling mechanisms that play both partially overlapping and opposing roles in immunity. Both receptors are expressed on a broad spectrum of immune and non-immune cells and are involved in cellular functions and physiological processes during homeostasis and inflammation. Dysregulated C5a-mediated inflammation contributes to autoimmune diseases such as rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, epidermolysis bullosa acquisita, antiphospholipid syndrome, and others. Therefore, targeting C5a or its receptors may yield therapeutic innovations in these autoimmune diseases by reducing the recruitment and activation of immune cells that lead to tissue inflammation and injury, thereby exacerbating the autoimmune response. Clinical trials focused on the inhibition of C5 cleavage or the C5a/C5aR1-axis using small molecules or monoclonal antibodies hold promise for bringing novel treatments for autoimmune diseases into practice. However, given the heterogeneous nature of (systemic) autoimmune diseases, there are still several challenges, such as patient selection, optimal dosing, and treatment duration, that require further investigation and development to realize the full therapeutic potential of C5a receptor inhibition, ideally in the context of a personalized medicine approach. Here, we aim to provide a brief overview of the current knowledge on the function of C5a receptors, the involvement of C5a receptors in autoimmune disorders, the molecular mechanisms underlying C5a receptor-mediated autoimmunity, and the potential for targeted therapies to modulate their activity.
Collapse
Affiliation(s)
- Jovan Schanzenbacher
- Institute for Systemic Inflammation Research (ISEF), University of Lübeck, Lübeck, Germany
| | - Katja Hendrika Kähler
- Institute for Systemic Inflammation Research (ISEF), University of Lübeck, Lübeck, Germany
| | - Evelyn Mesler
- Institute for Systemic Inflammation Research (ISEF), University of Lübeck, Lübeck, Germany
| | - Marie Kleingarn
- Institute for Systemic Inflammation Research (ISEF), University of Lübeck, Lübeck, Germany
| | | | - Daniel Leonard Seiler
- Institute for Systemic Inflammation Research (ISEF), University of Lübeck, Lübeck, Germany.
| |
Collapse
|
3
|
Kordowski A, Reinicke AT, Wu D, Orinska Z, Hagemann P, Huber-Lang M, Lee JB, Wang YH, Hogan SP, Köhl J. C5a receptor 1 -/- mice are protected from the development of IgE-mediated experimental food allergy. Allergy 2019; 74:767-779. [PMID: 30341777 DOI: 10.1111/all.13637] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 09/25/2018] [Accepted: 09/27/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND Food-induced anaphylaxis is a serious allergic reaction caused by Fcε-receptor activation on mast cells (MCs). The exact mechanisms breaking oral tolerance and the effector pathways driving food allergy remain elusive. As complement is activated in food-induced anaphylaxis, we aimed to assess the role of C5a in disease pathogenesis. METHODS Oral antigen-induced food-induced anaphylaxis was induced in BALB/c wild-type (wt) and C5ar1-/- mice. Readouts included diarrhea development, changes in rectal temperature, hematocrit, antigen-specific serum IgE, MCPT-1, and intestinal MC numbers, as well as FcεR1-mediated MC functions including C5a receptor 1 (C5aR1) regulation. Further, histamine-mediated hypothermia and regulation of endothelial tight junctions were determined. RESULTS Repeated oral OVA challenge resulted in diarrhea, hypothermia, increased hematocrit, high OVA-specific serum IgE, and MCPT-1 levels in wt mice. Male C5ar1-/- mice were completely whereas female C5ar1-/- mice were partially protected from anaphylaxis development. Serum MCPT-1 levels were reduced gender-independent, whereas IgE levels were reduced in male but not in female C5ar1-/- mice. Mechanistically, IgE-mediated degranulation and IL-6 production from C5ar1-/- BMMCs of both sexes were significantly reduced. Importantly, FcεR1 cross-linking strongly upregulated C5aR1 MC expression in vitro and in vivo. Finally, C5ar1-/- male mice were largely protected from histamine-induced hypovolemic shock, which was associated with protection from histamine-induced barrier dysfunction in vitro following C5aR targeting. CONCLUSIONS Our findings identify C5aR1 activation as an important driver of IgE-mediated food allergy through regulation of allergen-specific IgE production, FcεR1-mediated MC degranulation, and histamine-driven effector functions preferentially in male mice.
Collapse
Affiliation(s)
- Anna Kordowski
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Anna T Reinicke
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - David Wu
- Division of Allergy and Immunology, Cincinnati Children's Hospital and University of Cincinnati, College of Medicine, Cincinnati, Ohio, USA
| | - Zane Orinska
- Division of Experimental Pneumology, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
| | - Philipp Hagemann
- Division of Experimental Pneumology, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital of Ulm, Ulm, Germany
| | - Jee-Boong Lee
- Division of Allergy and Immunology, Cincinnati Children's Hospital and University of Cincinnati, College of Medicine, Cincinnati, Ohio, USA
| | - Yui-Hsi Wang
- Division of Allergy and Immunology, Cincinnati Children's Hospital and University of Cincinnati, College of Medicine, Cincinnati, Ohio, USA
| | - Simon P Hogan
- Division of Allergy and Immunology, Cincinnati Children's Hospital and University of Cincinnati, College of Medicine, Cincinnati, Ohio, USA
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
- Division of Immunobiology, Cincinnati Children's Hospital and University of Cincinnati, College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
4
|
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. [DOI: 10.1007/s12035-018-1428-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/13/2018] [Indexed: 02/07/2023]
|
5
|
Jackson HM, Onos KD, Pepper KW, Graham LC, Akeson EC, Byers C, Reinholdt LG, Frankel WN, Howell GR. DBA/2J genetic background exacerbates spontaneous lethal seizures but lessens amyloid deposition in a mouse model of Alzheimer's disease. PLoS One 2015; 10:e0125897. [PMID: 25933409 PMCID: PMC4416920 DOI: 10.1371/journal.pone.0125897] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 03/24/2015] [Indexed: 11/23/2022] Open
Abstract
Alzheimer’s disease (AD) is a leading cause of dementia in the elderly and is characterized by amyloid plaques, neurofibrillary tangles (NFTs) and neuronal dysfunction. Early onset AD (EOAD) is commonly caused by mutations in amyloid precursor protein (APP) or genes involved in the processing of APP including the presenilins (e.g. PSEN1 or PSEN2). In general, mouse models relevant to EOAD recapitulate amyloidosis, show only limited amounts of NFTs and neuronal cell dysfunction and low but significant levels of seizure susceptibility. To investigate the effect of genetic background on these phenotypes, we generated APPswe and PSEN1de9 transgenic mice on the seizure prone inbred strain background, DBA/2J. Previous studies show that the DBA/2J genetic background modifies plaque deposition in the presence of mutant APP but the impact of PSEN1de9 has not been tested. Our study shows that DBA/2J.APPswePSEN1de9 mice are significantly more prone to premature lethality, likely to due to lethal seizures, compared to B6.APPswePSEN1de9 mice—70% of DBA/2J.APPswePSEN1de9 mice die between 2-3 months of age. Of the DBA/2J.APPswePSEN1de9 mice that survived to 6 months of age, plaque deposition was greatly reduced compared to age-matched B6.APPswePSEN1de9 mice. The reduction in plaque deposition appears to be independent of microglia numbers, reactive astrocytosis and complement C5 activity.
Collapse
Affiliation(s)
| | - Kristen D. Onos
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | | | - Leah C. Graham
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
- Sackler School of Medicine, Tufts University, Boston, United States of America
| | - Ellen C. Akeson
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Candice Byers
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | | | - Wayne N. Frankel
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Gareth R. Howell
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
- Sackler School of Medicine, Tufts University, Boston, United States of America
- * E-mail:
| |
Collapse
|
6
|
Benson MJ, Thomas NK, Talwar S, Hodson MP, Lynch JW, Woodruff TM, Borges K. A novel anticonvulsant mechanism via inhibition of complement receptor C5ar1 in murine epilepsy models. Neurobiol Dis 2015; 76:87-97. [PMID: 25681535 DOI: 10.1016/j.nbd.2015.02.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 01/21/2015] [Accepted: 02/04/2015] [Indexed: 12/24/2022] Open
Abstract
The role of complement system-mediated inflammation is of key interest in seizure and epilepsy pathophysiology, but its therapeutic potential has not yet been explored. We observed that the pro-inflammatory C5a receptor, C5ar1, is upregulated in two mouse models after status epilepticus; the pilocarpine model and the intrahippocampal kainate model. The C5ar1 antagonist, PMX53, was used to assess potential anticonvulsant actions of blocking this receptor pathway. PMX53 was found to be anticonvulsant in several acute models (6Hz and corneal kindling) and one chronic seizure model (intrahippocampal kainate model). The effects in the 6Hz model were not found in C5ar1-deficient mice, or with an inactive PMX53 analogue suggesting that the anticonvulsant effect of PMX53 is C5ar1-specific. In the pilocarpine model, inhibition or absence of C5ar1 during status epilepticus lessened seizure power and protected hippocampal neurons from degeneration as well as halved SE-associated mortality. C5ar1-deficiency during pilocarpine-induced status epilepticus also was accompanied by attenuation of TNFα upregulation by microglia, suggesting that C5ar1 activation results in TNFα release contributing to disease. Patch clamp studies showed that C5a-induced microglial K(+) outward currents were also inhibited with PMX53 providing a potential mechanism to explain acute anticonvulsant effects. In conclusion, our data indicate that C5ar1 activation plays a role in seizure initiation and severity, as well as neuronal degeneration following status epilepticus. The widespread anticonvulsant activity of PMX53 suggests that C5ar1 represents a novel target for improved anti-epileptic drug development which may be beneficial for pharmaco-resistant patients.
Collapse
Affiliation(s)
- Melissa J Benson
- Department of Pharmacology, School of Biomedical Sciences, The University of Queensland, Skerman Building, St Lucia, Queensland 4072, Australia
| | - Nicola K Thomas
- Department of Pharmacology, School of Biomedical Sciences, The University of Queensland, Skerman Building, St Lucia, Queensland 4072, Australia
| | - Sahil Talwar
- Queensland Brain Institute, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Mark P Hodson
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Joseph W Lynch
- Queensland Brain Institute, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Trent M Woodruff
- Department of Pharmacology, School of Biomedical Sciences, The University of Queensland, Skerman Building, St Lucia, Queensland 4072, Australia
| | - Karin Borges
- Department of Pharmacology, School of Biomedical Sciences, The University of Queensland, Skerman Building, St Lucia, Queensland 4072, Australia.
| |
Collapse
|
7
|
Zimmer J, Hobkirk J, Mohamed F, Browning MJ, Stover CM. On the Functional Overlap between Complement and Anti-Microbial Peptides. Front Immunol 2015; 5:689. [PMID: 25646095 PMCID: PMC4298222 DOI: 10.3389/fimmu.2014.00689] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 12/22/2014] [Indexed: 12/19/2022] Open
Abstract
Intriguingly, activated complement and anti-microbial peptides share certain functionalities; lytic, phagocytic, and chemo-attractant activities and each may, in addition, exert cell instructive roles. Each has been shown to have distinct LPS detoxifying activity and may play a role in the development of endotoxin tolerance. In search of the origin of complement, a functional homolog of complement C3 involved in opsonization has been identified in horseshoe crabs. Horseshoe crabs possess anti-microbial peptides able to bind to acyl chains or phosphate groups/saccharides of endotoxin, LPS. Complement activity as a whole is detectable in marine invertebrates. These are also a source of anti-microbial peptides with potential pharmaceutical applicability. Investigating the locality for the production of complement pathway proteins and their role in modulating cellular immune responses are emerging fields. The significance of local synthesis of complement components is becoming clearer from in vivo studies of parenchymatous disease involving specifically generated, complement-deficient mouse lines. Complement C3 is a central component of complement activation. Its provision by cells of the myeloid lineage varies. Their effector functions in turn are increased in the presence of anti-microbial peptides. This may point to a potentiating range of activities, which should serve the maintenance of health but may also cause disease. Because of the therapeutic implications, this review will consider closely studies dealing with complement activation and anti-microbial peptide activity in acute inflammation (e.g., dialysis-related peritonitis, appendicitis, and ischemia).
Collapse
Affiliation(s)
- Jana Zimmer
- Department of Infectious Diseases - Medical Microbiology and Hygiene, Ruprecht-Karls-University of Heidelberg , Heidelberg , Germany
| | - James Hobkirk
- Department of Academic Endocrinology, Diabetes and Metabolism, Hull York Medical School, University of Hull , Hull , UK
| | - Fatima Mohamed
- Department of Infection, Immunity and Inflammation, University of Leicester , Leicester , UK
| | - Michael J Browning
- Department of Infection, Immunity and Inflammation, University of Leicester , Leicester , UK ; Department of Immunology, Leicester Royal Infirmary , Leicester , UK
| | - Cordula M Stover
- Department of Infection, Immunity and Inflammation, University of Leicester , Leicester , UK
| |
Collapse
|
8
|
Neuroprotective effect of schizandrin A on oxygen and glucose deprivation/reperfusion-induced cell injury in primary culture of rat cortical neurons. J Physiol Biochem 2014; 70:735-47. [DOI: 10.1007/s13105-014-0342-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Accepted: 06/16/2014] [Indexed: 01/13/2023]
|
9
|
Nomaru H, Sakumi K, Katogi A, Ohnishi YN, Kajitani K, Tsuchimoto D, Nestler EJ, Nakabeppu Y. Fosb gene products contribute to excitotoxic microglial activation by regulating the expression of complement C5a receptors in microglia. Glia 2014; 62:1284-98. [PMID: 24771617 DOI: 10.1002/glia.22680] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 04/05/2014] [Accepted: 04/10/2014] [Indexed: 12/18/2022]
Abstract
The Fosb gene encodes subunits of the activator protein-1 transcription factor complex. Two mature mRNAs, Fosb and ΔFosb, encoding full-length FOSB and ΔFOSB proteins respectively, are formed by alternative splicing of Fosb mRNA. Fosb products are expressed in several brain regions. Moreover, Fosb-null mice exhibit depressive-like behaviors and adult-onset spontaneous epilepsy, demonstrating important roles in neurological and psychiatric disorders. Study of Fosb products has focused almost exclusively on neurons; their function in glial cells remains to be explored. In this study, we found that microglia express equivalent levels of Fosb and ΔFosb mRNAs to hippocampal neurons and, using microarray analysis, we identified six microglial genes whose expression is dependent on Fosb products. Of these genes, we focused on C5ar1 and C5ar2, which encode receptors for complement C5a. In isolated Fosb-null microglia, chemotactic responsiveness toward the truncated form of C5a was significantly lower than that in wild-type cells. Fosb-null mice were significantly resistant to kainate-induced seizures compared with wild-type mice. C5ar1 mRNA levels and C5aR1 immunoreactivity were increased in wild-type hippocampus 24 hours after kainate administration; however, such induction was significantly reduced in Fosb-null hippocampus. Furthermore, microglial activation after kainate administration was significantly diminished in Fosb-null hippocampus, as shown by significant reductions in CD68 immunoreactivity, morphological change and reduced levels of Il6 and Tnf mRNAs, although no change in the number of Iba-1-positive cells was observed. These findings demonstrate that, under excitotoxicity, Fosb products contribute to a neuroinflammatory response in the hippocampus through regulation of microglial C5ar1 and C5ar2 expression.
Collapse
Affiliation(s)
- Hiroko Nomaru
- Division of Neurofunctional Genomics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Howell GR, Soto I, Ryan M, Graham LC, Smith RS, John SWM. Deficiency of complement component 5 ameliorates glaucoma in DBA/2J mice. J Neuroinflammation 2013; 10:76. [PMID: 23806181 PMCID: PMC3708765 DOI: 10.1186/1742-2094-10-76] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 06/12/2013] [Indexed: 01/08/2023] Open
Abstract
Background Glaucoma is an age-related neurodegenerative disorder involving the loss of retinal ganglion cells (RGCs), which results in blindness. Studies in animal models have shown that activation of inflammatory processes occurs early in the disease. In particular, the complement cascade is activated very early in DBA/2J mice, a widely used mouse model of glaucoma. A comprehensive analysis of the role of the complement cascade in DBA/2J glaucoma has not been possible because DBA/2J mice are naturally deficient in complement component 5 (C5, also known as hemolytic complement, Hc), a key mediator of the downstream processes of the complement cascade, including the formation of the membrane attack complex. Methods To assess the role of C5 in DBA/2J glaucoma, we backcrossed a functional C5 gene from strain C57BL/6J to strain DBA/2J for at least 10 generations. The prevalence and severity of glaucoma was evaluated using ocular examinations, IOP measurements, and assessments of optic nerve damage and RGC degeneration. To understand how C5 affects glaucoma, C5 expression was assessed in the retinas and optic nerves of C5-sufficient DBA/2J mice, using immunofluorescence. Results C5-sufficient DBA/2J mice developed a more severe glaucoma at an earlier age than standard DBA/2J mice, which are therefore protected by C5 deficiency. Components of the membrane attack complex were found to be deposited at sites of axonal injury in the optic nerve head and associated with RGC soma in the retina. Conclusion C5 plays an important role in glaucoma, with its deficiency lessening disease severity. These results highlight the importance of fully understanding the role of the complement cascade in neurodegenerative diseases. Inhibiting C5 may be beneficial as a therapy for human glaucoma.
Collapse
Affiliation(s)
- Gareth R Howell
- The Jackson Laboratory, 600 Main Street, Bar Harbor, Maine, USA.
| | | | | | | | | | | |
Collapse
|
11
|
Li YH, Jin H, Xu JS, Guo GQ, Chen DAL, Bo Y. Complement factor C5a and C5a receptor contribute to morphine tolerance and withdrawal-induced hyperalgesia in rats. Exp Ther Med 2012; 4:723-727. [PMID: 23170133 PMCID: PMC3501444 DOI: 10.3892/etm.2012.636] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Accepted: 06/13/2012] [Indexed: 01/03/2023] Open
Abstract
Morphine is a potent opioid analgesic. However, the repeated use of morphine causes tolerance and hyperalgesia. Neuroinflammation has been reported to be involved in morphine tolerance and withdrawal-induced hyperalgesia. The complement system is a crucial effector mechanism of immune responses. The present study investigated the roles of complement factor C5a and C5a receptor (C5aR) in the development of morphine tolerance and withdrawal-induced hyperalgesia. In the present study, the levels of C5a and C5aR were increased in the L5 lumbar spinal cords of morphine-tolerant rats. The administration of C5a promoted the development of hyperalgesia and the expression of spinal antinociceptive tolerance to intrathecal morphine in both mechanical and thermal test. However, these phenomena caused by morphine were significantly attenuated by the C5aR antagonist PMX53. These results suggest that complement activation within the spinal cord is involved in morphine tolerance and withdrawal-induced hyperalgesia. C5a and C5aR may serve as novel targets for the control of morphine tolerance and withdrawal-induced hyperalgesia.
Collapse
|
12
|
Pavlovski D, Thundyil J, Monk PN, Wetsel RA, Taylor SM, Woodruff TM. Generation of complement component C5a by ischemic neurons promotes neuronal apoptosis. FASEB J 2012; 26:3680-90. [PMID: 22651932 DOI: 10.1096/fj.11-202382] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
C5a receptors are found in the central nervous system (CNS), on both neurons and glia. However, the origin of the C5a, which activates these receptors, is unclear. In the present study, we show that primary cultured mouse cortical neurons constitutively express C5, the precursor of C5a, and express the classical receptor for C5a, CD88. With cell ischemia caused by 12 h glucose deprivation, or oxygen-glucose deprivation (OGD), neurons demonstrated increased apoptosis, up-regulation of CD88, and increased levels of C5a in the media. Exogenous murine C5a (100 nM) added to the neuronal cultures resulted in apoptosis, without affecting cell necrosis. Pretreatment of the cells with the specific CD88 receptor antagonist PMX53 (100 nM) significantly blocked ischemia-induced apoptosis (∼50%), and neurons from CD88(-/-) mice were similarly protected. In a murine model of stroke, using middle cerebral artery occlusion (MCAO), we found that C5a levels in the brain increased; this also occurred in cerebral slice cultures exposed to OGD. CD88(-/-) mice subjected to MCAO had significantly reduced infarct volumes and improved neurological scores. Taken together, our results demonstrate that neurons in the CNS have the capability to generate C5a following ischemic stress, and this has the potential to activate their C5a receptors, with deleterious consequences.
Collapse
Affiliation(s)
- Dale Pavlovski
- School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia
| | | | | | | | | | | |
Collapse
|
13
|
Geloso MC, Corvino V, Michetti F. Trimethyltin-induced hippocampal degeneration as a tool to investigate neurodegenerative processes. Neurochem Int 2011; 58:729-38. [DOI: 10.1016/j.neuint.2011.03.009] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Revised: 03/02/2011] [Accepted: 03/08/2011] [Indexed: 12/29/2022]
|
14
|
Abstract
The development of amyloid-containing neuritic plaques is an invariable characteristic of Alzheimer's diseases (AD). The conversion from monomeric amyloid β protein (Aβ) to oligomeric Aβ and finally neuritic plaques is highly dynamic. The specific Aβ species that is correlated with disease severity remains to be discovered. Oligomeric Aβ has been detected in cultured cells, rodent and human brains, as well as human cerebrospinal fluid. Synthetic, cell, and brain derived Aβ oligomers have been found to inhibit hippocampal long-term potentiation (LTP) and this effect can be suppressed by the blockage of Aβ oligomer formation. A large body of evidence suggests that Aβ oligomers inhibit N-methyl-D-aspartate receptor dependent LTP; additional receptors have also been found to elicit downstream pathways upon binding to Aβ oligomers. Amyloid antibodies and small molecular compounds that reduce brain Aβ levels and block Aβ oligomer formation are capable of reversing synaptic dysfunction and these approaches hold a promising therapeutic potential to rescue memory disruption.
Collapse
Affiliation(s)
- Weiming Xia
- Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
15
|
CNS-specific expression of C3a and C5a exacerbate demyelination severity in the cuprizone model. Mol Immunol 2010; 48:219-30. [PMID: 20813409 DOI: 10.1016/j.molimm.2010.08.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2010] [Revised: 08/03/2010] [Accepted: 08/10/2010] [Indexed: 12/18/2022]
Abstract
Demyelination in the central nervous system (CNS) is known to involve several immune effector mechanisms, including complement proteins. Local production of complement by glial cells in the brain can be both harmful and protective. To investigate the roles of C3a and C5a in demyelination and remyelination pathology we utilized the cuprizone model. Transgenic mice expressing C3a or C5a under the control of the glial fibrillary acidic protein (GFAP) promoter had exacerbated demyelination and slightly delayed remyelination in the corpus callosum compared to WT mice. C3a and C5a transgenic mice had increased cellularity in the corpus callosum due to increase activation and/or migration of microglia. Oligodendrocytes migrated to the corpus callosum in higher numbers during early remyelination events in C3a and C5a transgenic mice, thus enabling these mice to remyelinate as effectively as WT mice by the end of the 10 week study. To determine the effects of C3a and/or C5a on individual glial subsets, we created murine recombinant C3a and C5a proteins. When microglia and mixed glial cultures were stimulated with C3a and/or C5a, we observed an increase in the production of proinflammatory cytokines and chemokines. In contrast, astrocytes had decreased cytokine and chemokine production in the presence of C3a and/or C5a. We also found that the MAPK pathway proteins JNK and ERK1/2 were activated in glia upon stimulation with C3a and C5a. Overall, our findings show that although C3a and C5a production in the brain play a negative role during demyelination, these proteins may aid in remyelination.
Collapse
|
16
|
Woodruff TM, Ager RR, Tenner AJ, Noakes PG, Taylor SM. The role of the complement system and the activation fragment C5a in the central nervous system. Neuromolecular Med 2009; 12:179-92. [PMID: 19763906 DOI: 10.1007/s12017-009-8085-y] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2009] [Accepted: 08/25/2009] [Indexed: 12/28/2022]
Abstract
The complement system is a pivotal component of the innate immune system which protects the host from infection and injury. Complement proteins can be induced in all cell types within the central nervous system (CNS), where the pathway seems to play similar roles in host defense. Complement activation produces the C5 cleavage fragment C5a, a potent inflammatory mediator, which recruits and activates immune cells. The primary cellular receptor for C5a, the C5a receptor (CD88), has been reported to be on all CNS cells, including neurons and glia, suggesting a functional role for C5a in the CNS. A second receptor for C5a, the C5a-like receptor 2 (C5L2), is also expressed on these cells; however, little is currently known about its potential role in the CNS. The potent immune and inflammatory actions of complement activation are necessary for host defense. However, if over-activated, or left unchecked it promotes tissue injury and contributes to brain disease pathology. Thus, complement activation, and subsequent C5a generation, is thought to play a significant role in the progression of CNS disease. Paradoxically, complement may also exert a neuroprotective role in these diseases by aiding in the elimination of aggregated and toxic proteins and debris which are a principal hallmark of many of these diseases. This review will discuss the expression and known roles for complement in the CNS, with a particular focus on the pro-inflammatory end-product, C5a. The possible overarching role for C5a in diseases of the CNS is reviewed, and the therapeutic potential of blocking C5a/CD88 interaction is evaluated.
Collapse
Affiliation(s)
- Trent M Woodruff
- School of Biomedical Sciences, University of Queensland, St. Lucia, Brisbane, 4072, Australia.
| | | | | | | | | |
Collapse
|
17
|
Humayun S, Gohar M, Volkening K, Moisse K, Leystra-Lantz C, Mepham J, McLean J, Strong MJ. The complement factor C5a receptor is upregulated in NFL-/- mouse motor neurons. J Neuroimmunol 2009; 210:52-62. [PMID: 19286267 DOI: 10.1016/j.jneuroim.2009.01.028] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2008] [Revised: 01/28/2009] [Accepted: 01/30/2009] [Indexed: 11/28/2022]
Abstract
In NFL-/- mice, a model of motor neuron degeneration in ALS, degenerating spinal motor neurons express high levels of the receptor for the C5a anaphylatoxin (C5aR) early in the disease process. C5a is a potent in vitro neurotoxin for both Neuro2A and NGF-differentiated PC12 cells. While no interaction was observed between glutamate and C5a, both C5a and kainate upregulated the expression of activated C5aR. C5aR expression was increased in motor neurons in ALS. This data suggests that the early upregulation of C5aR may contribute to motor neuron damage that potentiates excitotoxicity in ALS.
Collapse
Affiliation(s)
- Saima Humayun
- Department of Pathology, Schulich School of Medicine, University of Western Ontario, London, Canada
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Flierl MA, Stahel PF, Rittirsch D, Huber-Lang M, Niederbichler AD, Hoesel LM, Touban BM, Morgan SJ, Smith WR, Ward PA, Ipaktchi K. Inhibition of complement C5a prevents breakdown of the blood-brain barrier and pituitary dysfunction in experimental sepsis. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2009; 13:R12. [PMID: 19196477 PMCID: PMC2688129 DOI: 10.1186/cc7710] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2008] [Revised: 01/12/2009] [Accepted: 02/06/2009] [Indexed: 12/12/2022]
Abstract
Introduction Septic encephalopathy secondary to a breakdown of the blood-brain barrier (BBB) is a known complication of sepsis. However, its pathophysiology remains unclear. The present study investigated the effect of complement C5a blockade in preventing BBB damage and pituitary dysfunction during experimental sepsis. Methods Using the standardised caecal ligation and puncture (CLP) model, Sprague-Dawley rats were treated with either neutralising anti-C5a antibody or pre-immune immunoglobulin (Ig) G as a placebo. Sham-operated animals served as internal controls. Results Placebo-treated septic rats showed severe BBB dysfunction within 24 hours, accompanied by a significant upregulation of pituitary C5a receptor and pro-inflammatory cytokine expression, although gene levels of growth hormone were significantly attenuated. The pathophysiological changes in placebo-treated septic rats were restored by administration of neutralising anti-C5a antibody to the normal levels of BBB and pituitary function seen in the sham-operated group. Conclusions Collectively, the neutralisation of C5a greatly ameliorated pathophysiological changes associated with septic encephalopathy, implying a further rationale for the concept of pharmacological C5a inhibition in sepsis.
Collapse
Affiliation(s)
- Michael A Flierl
- Department of Orthopaedic Surgery, Denver Health Medical Center, University of Colorado School of Medicine, Denver, CO 80204, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Mukherjee P, Thomas S, Pasinetti GM. Complement anaphylatoxin C5a neuroprotects through regulation of glutamate receptor subunit 2 in vitro and in vivo. J Neuroinflammation 2008; 5:5. [PMID: 18230183 PMCID: PMC2246107 DOI: 10.1186/1742-2094-5-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2007] [Accepted: 01/29/2008] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The complement system is thought to be involved in the pathogenesis of numerous neurological diseases. We previously reported that pre-treatment of murine cortico-hippocampal neuronal cultures with the complement derived anaphylatoxin C5a, protects against glutamate mediated apoptosis. Our present study with C5a receptor knock out (C5aRKO) mice corroborates that the deficiency of C5a renders C5aRKO mouse more susceptible to apoptotic injury in vivo. In this study we explored potential upstream mechanisms involved in C5a mediated neuroprotection in vivo and in vitro. METHODS Based on evidence suggesting that reduced expression of glutamate receptor subunit 2 (GluR2) may influence apoptosis in neurons, we studied the effect of human recombinant C5a on GluR2 expression in response to glutamate neurotoxicity. Glutamate analogs were injected into C5aRKO mice or used to treat in vitro neuronal culture and GluR2 expression were assessed in respect with cell death. RESULTS In C5aRKO mice we found that the neurons are more susceptible to excitotoxicity resulting in apoptotic injury in the absence of the C5a receptor compared to WT control mice. Our results suggest that C5a protects against apoptotic pathways in neurons in vitro and in vivo through regulation of GluR2 receptor expression. CONCLUSION Complement C5a neuroprotects through regulation of GluR2 receptor subunit.
Collapse
Affiliation(s)
- Piali Mukherjee
- Department of Psychiatry, Mount Sinai School of Medicine, 1 Gustav L,, Levy Place, New York, NY 10029, USA.
| | | | | |
Collapse
|
20
|
Crane JW, Buller KM. Systemic blockade of complement C5a receptors reduces lipopolysacharride-induced responses in the paraventricular nucleus and the central amygdala. Neurosci Lett 2007; 424:10-5. [PMID: 17703884 DOI: 10.1016/j.neulet.2007.07.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2007] [Revised: 06/22/2007] [Accepted: 07/07/2007] [Indexed: 11/29/2022]
Abstract
The complement anaphylatoxin C5a is a potent mediator of the innate immune response to infection. Recent evidence also reveals that C5a contributes to central nervous system effects in addition to its well-known peripheral functions. However, it is not known if C5a has a role in the activation of the hypothalamic-pituitary-adrenal (HPA) axis; a critical cascade that exemplifies neuroimmune interactions between the periphery and the brain. In the present study we examined if systemic pre-treatment with a C5a receptor antagonist, PMX53, can affect lipopolysaccharide-induced (LPS; 1 mg/kg, i.p.) activation of the HPA axis in the rat. Using Fos protein as a marker of neuronal activation, we found that systemic administration of PMX53 reduced the LPS-induced activation of paraventricular corticotropin-releasing factor (PVN CRF) and central amygdala cells. However, PMX53 did not alter LPS-induced responses in the bed nucleus of the stria terminalis, nucleus tractus solitarius and ventrolateral medulla. Our findings demonstrate that C5a may have a role in the activation of the HPA axis in response to systemic LPS.
Collapse
Affiliation(s)
- James W Crane
- Queensland Brain Institute, University of Queensland, St. Lucia, Queensland 4072, Australia
| | | |
Collapse
|
21
|
Köhl J. Self, non-self, and danger: a complementary view. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 586:71-94. [PMID: 16893066 DOI: 10.1007/0-387-34134-x_6] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Complement is a sophisticated system of molecules that is critical to the functional integrity of the body. Initially considered as a defense system to ward off infections, it becomes increasingly clear that the complement system is one of the most important humoral systems to sense danger, i.e., to recognize conserved patterns on pathogens and on altered/damaged self. In addition to this important role in danger recognition, the complement system has the ability to translate the danger information into an adequate cellular innate or adaptive immune response. This is accomplished by two distinct mechanisms: (a) danger sensors that have recognized altered cells or pathogens can directly activate cell-bound receptors (e.g., C1q/C1q receptor interaction), and/or (b) danger sensors initiate cleavage of complement factors C3 and C5, the fragments of which acquire the ability to bind to complement receptors and/or regulators. It is the specific interaction of the danger sensors and of the cleavage fragments with distinct cell-bound receptors/regulators that directs the immune response toward an innate or an adaptive phenotype. Further, the expression pattern of the complement receptors critically impacts the shape of the immune response. Complement has the ability to discriminate between physiological and pathological danger, i.e., physiological cell death and death in response to injury. In the former case, cells are merely flagged for enhanced phagocytosis (by C3 fragments) without accompanying inflammation (through CR3), whereas in the latter case inflammatory signals are accessorily triggered (e.g., by the release of ATs, which recruit and activate neutrophils, eosinophils, etc.). This function is of major importance for apoptotic cell clearance and tissue repair but plays also important roles in fibrotic tissue remodeling in response to chronic tissue injury. Further, complement cleavage fragments may prevent the development of maldaptive immune responses at the mucosal surface. Here, complement fragment C5a does not act as a danger transmitter but as a "homeostasis transmitter," as its interaction with the C5a receptor on DCs provides a signal that prevents DCs from activating CD4+ T cells. The generation of regulatory T cells in response to CD46 ligation may have a similar function, as injured cells lose CD46 expresssion, which may lead to decreased proliferation of Tregs and, consecutively, increased production of T effector cells. Although we are still at the beginning of understanding the complex interaction patterns within the complement system, recent data suggest substantial crosstalk between the signaling pathways downstream of complement receptors and other receptors of the innate immune system that function as immune sensors and/or transmitters (i.e., TLRs, FcgammaRs130,131). Given the importance of complement as a sensor and effector system of innate and adaptive immune responses, a complement-related view of the immune system might help to unravel some enigmas of autoimmunity, allergy, and transplantation.
Collapse
Affiliation(s)
- Jörg Köhl
- Division of Molecular Immunology, Cincinnati Children's Hospital Research Foundation, MLC 7021, Cincinnati, OH 45229, USA.
| |
Collapse
|
22
|
Flierl MA, Schreiber H, Huber-Lang MS. The role of complement, C5a and its receptors in sepsis and multiorgan dysfunction syndrome. J INVEST SURG 2006; 19:255-65. [PMID: 16835140 DOI: 10.1080/08941930600778263] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Sepsis continues to be a major clinical problem that is difficult to treat, as the pathophysiology of the disease is still unclear. Despite promising experimental strategies, therapeutic interventions have been largely unsuccessful. There is now increasing evidence that the disturbance of innate immunity during sepsis and multiorgan dysfunction syndrome (MODS) may be linked to uncontrolled activation of the complement system. Especially, the powerful anaphylatoxin C5a seems to play a key role in the development of immune paralysis. In this review, we describe our present understanding of the role of complement in the inflammatory response during sepsis and MODS.
Collapse
Affiliation(s)
- Michael A Flierl
- Department of Traumatology, Hand and Reconstructive Surgery, University Hospital of Ulm, Steinhoevelstrasse 9, 89075 Ulm, Germany
| | | | | |
Collapse
|
23
|
Woodruff TM, Crane JW, Proctor LM, Buller KM, Shek AB, de Vos K, Pollitt S, Williams HM, Shiels IA, Monk PN, Taylor SM. Therapeutic activity of C5a receptor antagonists in a rat model of neurodegeneration. FASEB J 2006; 20:1407-17. [PMID: 16816116 DOI: 10.1096/fj.05-5814com] [Citation(s) in RCA: 112] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The complement system is thought to be involved in the pathogenesis of numerous neurological diseases, although its precise role remains controversial. In this study we used orally active C5a receptor antagonists (PMX53 and PMX205) developed in our laboratories in a rat model of 3-nitropropionic acid (3-NP) -induced Huntington's disease. Administration of the C5a antagonists (10 mg/kg/day, oral) either 48 h pre- or 48 h post-toxin significantly reduced body weight loss, anorexia, and behavioral and motor deficits associated with 3-NP intoxication. Striatal lesion size, apoptosis, neutrophil infiltration, and hemorrhage were also significantly reduced in C5a antagonist-treated rats. Immunohistochemical analysis demonstrated marked deposition of C3 and C9, and up-regulation of C5a receptors on neuronal cells at the time of lesion formation. Inhibition of prostaglandins or TNF-alpha with ibuprofen or infliximab had no effect in this model. The C5a antagonists did not affect 3-NP-induced cell death when added directly to rat striatal neuronal cultures, indicating a secondary mechanism of action in vivo. Our findings demonstrate for the first time that complement activation in the brain, particularly C5a, is a key event in the pathogenesis of this disease model, and suggest a future role for inhibitors of C5a in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Trent M Woodruff
- Promics Ltd., The University of Queensland, Brisbane, QLD 4072, Australia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Köhl J. The role of complement in danger sensing and transmission. Immunol Res 2006; 34:157-76. [PMID: 16760575 DOI: 10.1385/ir:34:2:157] [Citation(s) in RCA: 121] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 01/08/2023]
Abstract
Self-non-self discrimination has long been considered the main function of the immune system. Increasing evidence supports the view of the immune system as a network of complex danger sensors and transmitters in which self-non-self discrimination is only one facet. To meet the challenge of danger sensing, the immune system carries a large stock of germline-encoded, highly conserved molecules that can recognize microbial as well as modified host structures. Among those are the Toll-like receptors (TLR), which comprise a dozen membrane-bound pattern-recognition receptors that directly link danger recognition to danger transmission through activation of several distinct cellular signaling pathways. Here, I discuss the function and biology of a complex, evolutionary ancient system, the complement system, which has long been considered critical to host defense. In contrast to TLRs, the complement system senses danger by a panel of soluble molecules that can directly bind to specific complement receptors and/or initiate a complex cascade of proteolytic events that lead to the generation of soluble complement fragments able to bind to another, distinct set of specific complement receptors. As I will outline in this review, complement- mediated danger sensing and the complex transition of this information into distinct cellular activation profiles is critical for tissue homeostasis under steady-state conditions and in response to infection and cell injury. Furthermore, I will discuss recent findings that support a concept of intense cross-talk between the complement system and TLRs, which defines the quality and the magnitude of immune responses in vivo.
Collapse
Affiliation(s)
- Jörg Köhl
- Division of Molecular Immunology, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, Ohio 45229, USA.
| |
Collapse
|
25
|
Blatteis CM. Endotoxic fever: New concepts of its regulation suggest new approaches to its management. Pharmacol Ther 2006; 111:194-223. [PMID: 16460809 DOI: 10.1016/j.pharmthera.2005.10.013] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2005] [Accepted: 10/07/2005] [Indexed: 01/09/2023]
Abstract
Endotoxic fever is regulated by endogenous factors that provide pro- and anti-pyretic signals at different points along the febrigenic pathway, from the periphery to the brain. Current evidence indicates that the febrile response to invading Gram-negative bacteria and their products is initiated upon their arrival in the liver via the circulation and their uptake by Kupffer cells (Kc). These pathogens activate the complement cascade on contact, hence generating complement component 5a. It, in turn, very rapidly stimulates Kc to release prostaglandin (PG)E2. Pyrogenic cytokines (TNF-alpha, etc.) are produced later and are no longer considered to be the immediate triggers of fever. The Kc-generated PGE2 either (1) may be transported by the bloodstream to the ventromedial preoptic-anterior hypothalamus (POA, the locus of the temperature-regulating center), presumptively diffusing into it and acting on thermoregulatory neurons; PGE2 is thus taken to be the final, central fever mediator. Or (2) it may activate hepatic vagal afferents projecting to the medulla oblongata, thence to the POA via the ventral noradrenergic bundle. Norepinephrine consequently secreted stimulates alpha1-adrenoceptors on thermoregulatory neurons, rapidly evoking an initial rise in core temperature (Tc) not associated with any change in POA PGE2; this neural, PGE2-independent signaling pathway is quicker than the blood-borne route. Elevated POA PGE2 and a secondary Tc rise occur later, consequent to alpha2 stimulation. Endogenous counter-regulatory factors are also elaborated peripherally and centrally at different points during the course of the febrile response; they are, therefore, anti-pyretic. These multiple interacting pathways are the subject of this review.
Collapse
Affiliation(s)
- Clark M Blatteis
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, 894 Union Avenue, Memphis, 38163, USA.
| |
Collapse
|
26
|
Wang Q, Yu S, Simonyi A, Sun GY, Sun AY. Kainic acid-mediated excitotoxicity as a model for neurodegeneration. Mol Neurobiol 2006; 31:3-16. [PMID: 15953808 DOI: 10.1385/mn:31:1-3:003] [Citation(s) in RCA: 268] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2004] [Accepted: 11/15/2004] [Indexed: 02/06/2023]
Abstract
Neuronal excitation involving the excitatory glutamate receptors is recognized as an important underlying mechanism in neurodegenerative disorders. Excitation resulting from stimulation of the ionotropic glutamate receptors is known to cause the increase in intracellular calcium and trigger calcium-dependent pathways that lead to neuronal apoptosis. Kainic acid (KA) is an agonist for a subtype of ionotropic glutamate receptor, and administration of KA has been shown to increase production of reactive oxygen species, mitochondrial dysfunction, and apoptosis in neurons in many regions of the brain, particularly in the hippocampal subregions of CA1 and CA3, and in the hilus of dentate gyrus (DG). Systemic injection of KA to rats also results in activation of glial cells and inflammatory responses typically found in neurodegenerative diseases. KA-induced selective vulnerability in the hippocampal neurons is related to the distribution and selective susceptibility of the AMPA/kainate receptors in the brain. Recent studies have demonstrated ability of KA to alter a number of intracellular activities, including accumulation of lipofuscin-like substances, induction of complement proteins, processing of amyloid precursor protein, and alteration of tau protein expression. These studies suggest that KA-induced excitotoxicity can be used as a model for elucidating mechanisms underlying oxidative stress and inflammation in neurodegenerative diseases. The focus of this review is to summarize studies demonstrating KA-induced excitotoxicity in the central nervous system and possible intervention by anti-oxidants.
Collapse
Affiliation(s)
- Qun Wang
- Department of Medical Pharmacology, University of Missouri School of Medicine, Columbia, MO, USA
| | | | | | | | | |
Collapse
|
27
|
Twining CM, Sloane EM, Schoeniger DK, Milligan ED, Martin D, Marsh H, Maier SF, Watkins LR. Activation of the spinal cord complement cascade might contribute to mechanical allodynia induced by three animal models of spinal sensitization. THE JOURNAL OF PAIN 2005; 6:174-83. [PMID: 15772911 DOI: 10.1016/j.jpain.2004.11.011] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The present series of experiments examined whether the complement cascade might play a key role in the expression of mechanical allodynia. Soluble complement receptor 1 (sCR1) was used to block the activation of the membrane attack pathway of the complement cascade. In doing so, sCR1 prevents the formation of the biologically active end products C3a, C5a, and membrane attack complexes (MACs). Intrathecal sCR1 had no effect on the behavioral responses of control groups. In contrast, blockade of this pathway abolished the expression of mechanical allodynia induced by peripheral nerve inflammation (sciatic inflammatory neuropathy model), partial sciatic nerve injury (chronic constriction injury model), and intrathecal injection of human immunodeficiency virus type 1 gp120, a viral envelope protein that activates glia. The fact that enhanced nociception was prevented or reversed in all 3 paradigms suggests that complement might be broadly involved in spinally mediated pain enhancement. The mechanisms whereby complement activation might potentially affect the functioning of microglia, astrocytes, and neurons are discussed. The complement cascade has not been previously implicated in spinal sensitization. These data suggest that complement activation within the spinal cord might contribute to enhanced pain states and provide additional evidence for immune regulation of pain transmission.
Collapse
Affiliation(s)
- Carin M Twining
- Department of Psychology and the Center of Neuroscience, University of Colorado at Boulder, 80309, USA
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Li S, Boackle SA, Holers VM, Lambris JD, Blatteis CM. Complement component c5a is integral to the febrile response of mice to lipopolysaccharide. Neuroimmunomodulation 2005; 12:67-80. [PMID: 15785108 DOI: 10.1159/000083578] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2004] [Accepted: 03/23/2004] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES The complement system is critical to the febrile response of mice to intraperitoneally administered lipopolysaccharide (LPS). We previously identified C3 and C5 as two components potentially involved in this response. This study was designed to examine whether the complement system is also pivotal in the response of mice to intravenously or intracerebroventricularly injected LPS, to distinguish between C3 and C5 and their cognate derivatives as the essential mediator(s), and to determine whether the failure of complement-deficient mice to develop a fever could be due to their possible inability to secrete pyrogenic cytokines. METHODS Wild-type (WT; C57BL/6J) mice, hypocomplemented or not by intravenously injected cobra venom factor (10 U/mouse), and C3-, CR3- and C5-sufficient and -deficient mice were intravenously challenged with LPS (0.25 mug/mouse); WT and C3-/- mice pretreated with a C5a receptor antagonist (C5aRa) were similarly challenged. In addition, the serum levels of interleukin (IL)-1beta, tumor necrosis factor (TNF)-alpha and IL-6 were compared in LPS-treated C5+/+ and C5-/- mice. RESULTS LPS induced a 1 degrees C rise in core temperature in all the mice, except C5-/- mice and those pretreated with C5aRa. C5+/+ and C5-/- mice challenged intracerebroventricularly with LPS exhibited identical febrile responses. LPS induced similar increases in the serum levels of IL-1beta, TNFalpha and IL-6 in C5+/+ and C5-/- mice. CONCLUSIONS C5a is crucial for the development of febrile responses to LPS in mice; its site of action is peripheral, not central. The possibility that an inability to produce cytokines could account for the failure of C5-/- mice to develop a fever is not supported.
Collapse
Affiliation(s)
- S Li
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | | | | | | | | |
Collapse
|
29
|
Mullick A, Elias M, Picard S, Bourget L, Jovcevski O, Gauthier S, Tuite A, Harakidas P, Bihun C, Massie B, Gros P. Dysregulated inflammatory response to Candida albicans in a C5-deficient mouse strain. Infect Immun 2004; 72:5868-76. [PMID: 15385488 PMCID: PMC517586 DOI: 10.1128/iai.72.10.5868-5876.2004] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2004] [Revised: 06/16/2004] [Accepted: 07/12/2004] [Indexed: 11/20/2022] Open
Abstract
Experimental infection of inbred mouse strains with Candida albicans provides a good model system to identify host genetic determinants that regulate onset of, response to, and ultimate outcome of disseminated candidiasis. The A/J mouse strain is exquisitely sensitive to infection with C. albicans, while the C57BL/6J strain is relatively resistant, as measured by survival following intravenous injection of Candida blastospores. This differential susceptibility is caused by an A/J-specific loss-of-function mutation in the C5 component of the complement pathway. C5 plays several critical roles in host response to infection, including target lysis and phagocyte recruitment. Therefore, to determine which of its functions were required for host resistance to candidiasis, a detailed comparative analysis of pathophysiology and host response to acute C. albicans infection was conducted in A/J and C57BL/6J mice. C5-sufficient C57BL/6J mice were found to succumb late in infection due to severe kidney pathology, typified by fungal replication and robust neutrophil-based inflammatory response associated with extensive tissue damage. In contrast, A/J mice were moribund within 24 h postinfection but displayed little if any kidney damage despite an inability to mobilize granulocytes and a high fungal load in the kidney. Rather, C5 deficiency in A/J mice was associated with higher levels of circulating cytokines tumor necrosis factor alpha, interleukin-6, monocyte chemotactic protein 1 (MCP-1), MCP-5, and eotaxin in response to C. albicans. Transfer of the C5-defective allele from A/J onto a C57BL/6J genetic background in recombinant congenic strain BcA17 recapitulated the phenotypic aspects of the susceptibility of A/J mice to C. albicans, confirming the causative role of C5 deficiency in the dysregulated cytokine response.
Collapse
Affiliation(s)
- Alaka Mullick
- Biotechnology Research Institute, National Research Council, Montréal, Québec, Canada.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Speth C, Williams K, Hagleitner M, Westmoreland S, Rambach G, Mohsenipour I, Schmitz J, Würzner R, Lass-Flörl C, Stoiber H, Dierich MP, Maier H. Complement synthesis and activation in the brain of SIV-infected monkeys. J Neuroimmunol 2004; 151:45-54. [PMID: 15145603 DOI: 10.1016/j.jneuroim.2004.02.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2003] [Revised: 01/12/2004] [Accepted: 02/19/2004] [Indexed: 11/25/2022]
Abstract
Complement is one of the most critical defence tools against cerebral infections, but uncontrolled complement biosynthesis and activation can induce profound brain tissue damage. To clarify the role of complement in the pathogenesis of AIDS-associated neurological disorders, we analysed the synthesis of complement in the brains of SIV-infected rhesus macaques. Using immunohistochemical staining we could show that the cerebral synthesis of complement factors C1q and C3 was strongly upregulated in SIV-infected monkeys compared to the spontaneous synthesis in uninfected control monkeys. Astrocytes, neurons, microglia, infiltrating macrophages and multinuclear giant cells all contribute to the high amounts of C1q and C3 in the brain. Secreted C1q and C3 are also deposited on the membrane of neurons, a prerequisite for formation of the membrane-driven lytic membrane attack complex. The membrane deposition thus might suggest complement-induced lysis of bystander neurons as a potential mechanism for cell damage during viral infection of the brain.
Collapse
Affiliation(s)
- Cornelia Speth
- Institute of Hygiene and Social Medicine, Innsbruck Medical University and Ludwig-Bolthmann-Institute for AIDS Research, Fritz-Pregl-Str. 3, Innsbruck A-6020, Austria.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
van Beek J. Complement activation: beneficial and detrimental effects in the CNS. ERNST SCHERING RESEARCH FOUNDATION WORKSHOP 2004:67-85. [PMID: 15032054 DOI: 10.1007/978-3-662-05426-0_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
32
|
Abstract
The chromosomal protein HMGB1 is now regarded as a proinflammatory cytokine. Importantly, HMGB1 has chemotactic activity suggesting its involvement in the early and late events of the inflammatory reaction. Therefore, HMGB1 has all the hallmarks of a chemokine (chemotactic cytokine). We propose to classify HMGB1 into a new group of proteins unrelated structurally to chemokines but having chemokine-like functions, and to name this class CLF (chemokine-like functions). The CLF class should include other unrelated molecules such as urokinase and its receptor, cytokines macrophage migration inhibitory factor (MIF) and interleukin (IL)-6, anaphylatoxin C5a, ribosomal protein S19, and thioredoxin that have similar chemokine-like activities. This innovative concept may lead to the identification of new therapeutic targets.
Collapse
Affiliation(s)
- Bernard Degryse
- Department of Cell Biology, Division of Vascular Biology/VB5, The Scripps Research Institute, La Jolla, CA 92130, USA
| | | |
Collapse
|
33
|
Huber-Lang MS, Sarma JV, McGuire SR, Lu KT, Padgaonkar VA, Younkin EM, Guo RF, Weber CH, Zuiderweg ER, Zetoune FS, Ward PA. Structure-function relationships of human C5a and C5aR. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:6115-24. [PMID: 12794141 DOI: 10.4049/jimmunol.170.12.6115] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Using peptides that represent linear regions of the powerful complement activation product, C5a, or loops that connect the four alpha helices of C5a, we have defined the ability of these peptides to reduce binding of (125)I-C5a to human neutrophils, inhibit chemotactic responses of neutrophils to C5a, and reduce H(2)O(2) production in neutrophils stimulated with PMA. The data have defined likely sites of interaction of C5a with C5aR. The peptides had no functional activity per se on neutrophils and did not interfere with neutrophil responses to the unrelated chemotactic peptide, N-formyl-Met-Leu-Phe. Although previous data have suggested that there are two separate sites on C5a reactive with C5aR, the current data suggest that C5a interacts with C5aR in a manner that engages three discontinuous regions of C5a.
Collapse
MESH Headings
- Amino Acid Sequence
- Antigens, CD/chemistry
- Antigens, CD/metabolism
- Antigens, CD/physiology
- Binding, Competitive/immunology
- Cell Migration Inhibition
- Chemotaxis, Leukocyte
- Complement C5a/antagonists & inhibitors
- Complement C5a/chemistry
- Complement C5a/metabolism
- Complement C5a/physiology
- Dose-Response Relationship, Immunologic
- Humans
- Hydrogen Peroxide/antagonists & inhibitors
- Hydrogen Peroxide/metabolism
- Iodine Radioisotopes/metabolism
- Molecular Sequence Data
- N-Formylmethionine Leucyl-Phenylalanine/pharmacology
- Neutrophils/drug effects
- Neutrophils/immunology
- Neutrophils/metabolism
- Peptide Fragments/antagonists & inhibitors
- Peptide Fragments/chemical synthesis
- Peptide Fragments/metabolism
- Peptide Fragments/physiology
- Receptor, Anaphylatoxin C5a
- Receptors, Complement/chemistry
- Receptors, Complement/metabolism
- Receptors, Complement/physiology
- Structure-Activity Relationship
- Tetradecanoylphorbol Acetate/antagonists & inhibitors
- Tetradecanoylphorbol Acetate/pharmacology
Collapse
Affiliation(s)
- Markus S Huber-Lang
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
van Beek J, Elward K, Gasque P. Activation of complement in the central nervous system: roles in neurodegeneration and neuroprotection. Ann N Y Acad Sci 2003; 992:56-71. [PMID: 12794047 DOI: 10.1111/j.1749-6632.2003.tb03138.x] [Citation(s) in RCA: 155] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The complement system is an essential effector of the humoral and cellular immunity involved in cytolysis and immune/inflammatory responses. Complement participates in host defense against pathogens by triggering the formation of the membrane attack complex. Complement opsonins (C1q, C3b, and iC3b) interact with surface complement receptors to promote phagocytosis, whereas complement anaphylatoxins C3a and C5a initiate local inflammatory responses that ultimately contribute to the protection and healing of the host. However, activation of complement to an inappropriate extent has been proposed to promote tissue injury. There is now compelling evidence that complement activation in the brain is a double-edged sword in that it can exert beneficial or detrimental effects depending on the pathophysiological context. This review focuses on the roles of the complement system in the pathogenesis of acute brain injury (cerebral ischemia and trauma) and chronic neurodegeneration (Alzheimer's disease). Because many effects of the complement appear to promote neuronal survival and tissue remodeling, directing activation of the complement system in the brain may provide a better therapeutic rationale than inhibiting it.
Collapse
Affiliation(s)
- Johan van Beek
- Brain Inflammation and Immunity Group, Department of Medical Biochemistry and Immunology, University of Wales College of Medicine, Cardiff, UK.
| | | | | |
Collapse
|
35
|
Sayah S, Jauneau AC, Patte C, Tonon MC, Vaudry H, Fontaine M. Two different transduction pathways are activated by C3a and C5a anaphylatoxins on astrocytes. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2003; 112:53-60. [PMID: 12670702 DOI: 10.1016/s0169-328x(03)00046-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
C3a and C5a anaphylatoxins are two proinflammatory peptides generated during complement system activation. C3a and C5a exert several biological activities through binding to their specific receptors, named C3aR and C5aR, respectively. We have previously shown that C3aR and C5aR are constitutively expressed by astrocytes, a cell type that actively participates in inflammatory events in the central nervous system. In this article, we focus on the transduction signal pathways activated by these two receptors on astrocytes. We show that the stimulation of C3aR or C5aR results in the activation of the mitogen activated protein kinase pathway by phosphorylation of the p44 and p42 kinases. On the contrary, the binding of C3a or C5a to their receptors on astrocytes decreases the production of cAMP, revealing an inhibition of the adenylyl cyclase pathway. Stimulation of C3aR and C5aR induces an increase in intracellular calcium concentration, arising from the opening of intracellular calcium channels. The observed calcium wave results from the activation of the phospholipase C pathway. Taken together, our results suggest that the binding of C3a or C5a to their receptors on astrocytes would be of functional importance since it induces the activation of two important transduction pathways leading to several cellular events such as neurotrophin and cytokine production.
Collapse
Affiliation(s)
- S Sayah
- INSERM U519, Faculté Mixte de Médecine et Pharmacie, 22 Bld. Gambetta, 76183, Cedex, Rouen, France
| | | | | | | | | | | |
Collapse
|
36
|
Huber-Lang MS, Riedeman NC, Sarma JV, Younkin EM, McGuire SR, Laudes IJ, Lu KT, Guo RF, Neff TA, Padgaonkar VA, Lambris JD, Spruce L, Mastellos D, Zetoune FS, Ward PA. Protection of innate immunity by C5aR antagonist in septic mice. FASEB J 2002; 16:1567-74. [PMID: 12374779 DOI: 10.1096/fj.02-0209com] [Citation(s) in RCA: 118] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Innate immune functions are known to be compromised during sepsis, often with lethal consequences. There is also evidence in rats that sepsis is associated with excessive complement activation and generation of the potent anaphylatoxin C5a. In the presence of a cyclic peptide antagonist (C5aRa) to the C5a receptor (C5aR), the binding of murine 125I-C5a to murine neutrophils was reduced, the in vitro chemotactic responses of mouse neutrophils to mouse C5a were markedly diminished, the acquired defect in hydrogen peroxide (H2O2) production of C5a-exposed neutrophils was reversed, and the lung permeability index (extravascular leakage of albumin) in mice after intrapulmonary deposition of IgG immune complexes was markedly diminished. Mice that developed sepsis after cecal ligation/puncture (CLP) and were treated with C5aRa had greatly improved survival rates. These data suggest that C5aRa interferes with neutrophil responses to C5a, preventing C5a-induced compromise of innate immunity during sepsis, with greatly improved survival rates after CLP.
Collapse
Affiliation(s)
- Markus S Huber-Lang
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109-0602, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Huber-Lang MS, Younkin EM, Sarma JV, McGuire SR, Lu KT, Guo RF, Padgaonkar VA, Curnutte JT, Erickson R, Ward PA. Complement-induced impairment of innate immunity during sepsis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:3223-31. [PMID: 12218141 DOI: 10.4049/jimmunol.169.6.3223] [Citation(s) in RCA: 139] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
This study defines the molecular basis for defects in innate immunity involving neutrophils during cecal ligation/puncture (CLP)-induced sepsis in rats. Blood neutrophils from CLP rats demonstrated defective phagocytosis and defective assembly of NADPH oxidase, the latter being due to the inability of p47(phox) to translocate from the cytosol to the cell membrane of neutrophils after cell stimulation by phorbol ester (PMA). The appearance of these defects was prevented by in vivo blockade of C5a in CLP rats. In vitro exposure of neutrophils to C5a led to reduced surface expression of C5aR and defective assembly of NADPH oxidase, as defined by failure in phosphorylation of p47(phox) and its translocation to the cell membrane, together with failure in phosphorylation of p42/p44 mitogen-activated protein kinases. These data identify a molecular basis for defective innate immunity involving neutrophils during sepsis.
Collapse
Affiliation(s)
- Markus S Huber-Lang
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
O'Hanlon GM, Bullens RWM, Plomp JJ, Willison HJ. Complex gangliosides as autoantibody targets at the neuromuscular junction in Miller Fisher syndrome: a current perspective. Neurochem Res 2002; 27:697-709. [PMID: 12374204 DOI: 10.1023/a:1020284302718] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Glycosphingolipid biology has increasingly interfaced with the field of human autoimmune neuropathy over the last two decades. There are currently over 20 distinct glycolipids that have been identified as autoantibody targets in a wide range of clinical neuropathy syndromes. This review sets out the clinical and experimental background to one interesting example of anti-glycolipid antibody-associated neuropathy termed Miller Fisher syndrome. This syndrome, comprising the triad of ataxia, areflexia, and ophthalmoplegia, correlates highly with the presence of serum anti-GQ1b antibodies, arising through molecular mimicry with microbial oligosaccharides. Anti-GQ1b antibodies mediate neural injury through binding to GQ1b-enriched sites in the peripheral nervous system, including extraocular nerves. Animal experimental evidence, along with a hypothetical background, indicates the motor nerve terminal may be a key site for anti-GQ1b antibody binding with consequent defects in synaptic transmission, as occurs in botulism and other toxinopathies. Our work in recent years on this hypothesis is summarized.
Collapse
Affiliation(s)
- Graham M O'Hanlon
- University Department of Neurology, Institute of Neurological Sciences, Southern General Hospital, Glasgow, Scotland, United Kingdom
| | | | | | | |
Collapse
|
39
|
Riedemann NC, Guo RF, Neff TA, Laudes IJ, Keller KA, Sarma VJ, Markiewski MM, Mastellos D, Strey CW, Pierson CL, Lambris JD, Zetoune FS, Ward PA. Increased C5a receptor expression in sepsis. J Clin Invest 2002; 110:101-8. [PMID: 12093893 PMCID: PMC151030 DOI: 10.1172/jci15409] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Excessive production of the complement activation product C5a appears to be harmful during the development of sepsis in rodents. Little is known about the role of the C5a receptor (C5aR) and its presence in different organs during sepsis. Using the cecal ligation/puncture (CLP) model in mice, we show here that C5aR immunoreactivity was strikingly increased in lung, liver, kidney, and heart early in sepsis in both control and neutrophil-depleted mice. C5aR mRNA expression in these organs was also significantly increased during sepsis. Immunohistochemical analysis revealed patterns of increased C5aR expression in parenchymal cells in all four organs following CLP. Mice injected at the start of CLP with a blocking IgG to C5aR (alphaC5aR) showed dramatically improved survival when compared with animals receiving nonspecific IgG, as did mice injected with alphaC5a. In alphaC5aR-treated mice, serum levels of IL-6 and TNF-alpha and bacterial counts in various organs were significantly reduced during CLP when compared with control CLP animals. These studies demonstrate for the first time that C5aR is upregulated in lung, liver, kidney, and heart during the early phases of sepsis and that blockade of C5aR is highly protective from the lethal outcome of sepsis.
Collapse
MESH Headings
- Animals
- Antibodies/administration & dosage
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Disease Models, Animal
- Immunohistochemistry
- Kidney/immunology
- Liver/immunology
- Lung/immunology
- Lymphocyte Depletion
- Male
- Mice
- Mice, Inbred BALB C
- Myocardium/immunology
- Neutrophils/immunology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptor, Anaphylatoxin C5a
- Receptors, Complement/antagonists & inhibitors
- Receptors, Complement/genetics
- Receptors, Complement/metabolism
- Sepsis/genetics
- Sepsis/immunology
- Sepsis/prevention & control
- Tissue Distribution
- Up-Regulation
Collapse
Affiliation(s)
- Niels C Riedemann
- Department of Pathology, University of Michigan Medical School, 1301 Catherine Road, Ann Arbor, MI 48109, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Riedemann NC, Guo RF, Neff TA, Laudes IJ, Keller KA, Sarma VJ, Markiewski MM, Mastellos D, Strey CW, Pierson CL, Lambris JD, Zetoune FS, Ward PA. Increased C5a receptor expression in sepsis. J Clin Invest 2002. [DOI: 10.1172/jci0215409] [Citation(s) in RCA: 127] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
41
|
Reiss CS, Chesler DA, Hodges J, Ireland DDC, Chen N. Innate immune responses in viral encephalitis. Curr Top Microbiol Immunol 2002; 265:63-94. [PMID: 12014196 DOI: 10.1007/978-3-662-09525-6_4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The innate immune system is multifaceted, comprised of preformed factors, cells, and many proteins and lipid mediators produced by those cells. In the CNS these are critical in initiation and amplification of the inflammatory response and in the subsequent elicitation of the specific T cell response to viral encephalitis. Cells that are resident in brain parenchyma and peripheral cells that are recruited both play key roles in the hosts's responses. Unlike the peripheral compartments, in the CNS, non-cytolytic means of eliminating viral infections have been critical, since, in contrast to columnar epithelial cells, neurons are non-renewing. When the innate immune responses are inefficient or absent in viral encephalitis, pathology is more likely. Much more work remains to elucidate all of the critical cells and their mediators, as well as to develop new therapies for infections of the CNS.
Collapse
Affiliation(s)
- C S Reiss
- Biology Department, New York University, 100 Washington Square East, New York, NY 10003-6688, USA
| | | | | | | | | |
Collapse
|
42
|
Speth C, Schabetsberger T, Mohsenipour I, Stöckl G, Würzner R, Stoiber H, Lass-Flörl C, Dierich MP. Mechanism of human immunodeficiency virus-induced complement expression in astrocytes and neurons. J Virol 2002; 76:3179-88. [PMID: 11884542 PMCID: PMC136041 DOI: 10.1128/jvi.76.7.3179-3188.2002] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The cerebral complement system is hypothesized to contribute to neurodegeneration in the pathogenesis of AIDS-associated neurological disorders. Our former results have shown that the human immunodeficiency virus (HIV) strongly induces the synthesis of complement factor C3 in astrocytes. This upregulation explains in vivo data showing elevated complement levels in the cerebrospinal fluid of patients with AIDS-associated neurological symptoms. Since inhibition of complement synthesis and activation in the brain may represent a putative therapeutic goal to prevent virus-induced damage, we analyzed in detail the mechanisms of HIV-induced modulation of C3 expression. HIV-1 increased the C3 levels in astrocyte culture supernatants from 30 to up to 400 ng/ml; signal transduction studies revealed that adenylate cyclase activation with upregulation of cyclic AMP is the central signaling pathway to mediate that increase. Furthermore, activity of protein kinase C is necessary for HIV induction of C3, since inhibition of protein kinase C by prolonged exposure to the phorbol ester tetradecanoyl phorbol acetate partly abolished the HIV effect. The cytokines tumor necrosis factor alpha and gamma interferon were not involved in mediating the HIV-induced C3 upregulation, since neutralizing antibodies had no effect. Besides whole HIV virions, the purified viral proteins Nef and gp41 are biologically active in upregulating C3, whereas Tat, gp120, and gp160 were not able to modulate C3 synthesis. Further experiments revealed that neurons were also able to respond on incubation with HIV with increased C3 synthesis, although the precise pattern was slightly different from that in astrocytes. This strengthens the hypothesis that HIV-induced complement synthesis represents an important mechanism for the pathogenesis of AIDS in the brain.
Collapse
Affiliation(s)
- Cornelia Speth
- Institute of Hygiene and Social Medicine, University of Innsbruck Ludwig Boltzmann Institute for AIDS Research, Innsbruck, Austria.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Alzheimer's disease (AD) is a worldwide problem that affects 5 million people in the United States alone. Until the approval of tacrine in the mid-1990s, there was no effective therapy for the cognitive symptoms of AD. Although cholinergic therapy provides modest but significant symptomatic relief, the development of effective disease-modifying therapy is essential. It has been demonstrated that a number of inflammatory processes are active in the brain of patients with AD, and therefore it is believed that an anti-inflammatory regimen may offer some degree of neuroprotection. Several studies have indicated that use of nonsteroidal anti-inflammatory drugs (NSAIDs) is associated with delayed onset and/or slowed cognitive decline in AD. Although not currently approved for this condition, recent findings have demonstrated that cyclooxygenase (COX)-2 is of primary importance in the inflammatory response and may have a role in neurodegeneration. Therefore, selective COX-2 inhibitors (coxibs) may have an advantage over traditional NSAIDs as potential therapeutic agents in AD. The Alzheimer's Disease Cooperative Study (ADCS) is conducting an ongoing multicenter, double-blind, placebo-controlled trial to determine whether rofecoxib, a coxib, or naproxen, a nonselective NSAID, will slow the rate of cognitive and clinical decline in AD. This study, along with other clinical studies currently under way, will determine the utility of selective and nonselective COX inhibitors for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Paul S Aisen
- Departments of Neurology and Medicine, Georgetown University Medical Center, Washington, DC 20007, USA
| |
Collapse
|
44
|
Reiman R, Gerard C, Campbell IL, Barnum SR. Disruption of the C5a receptor gene fails to protect against experimental allergic encephalomyelitis. Eur J Immunol 2002; 32:1157-63. [PMID: 11932923 DOI: 10.1002/1521-4141(200204)32:4<1157::aid-immu1157>3.0.co;2-m] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Activation of the complement system generates the anaphylatoxic peptide C5a, which elicits a broad range of inflammatory activities. The biological activities of C5a are mediated through its binding to the widely expressed C5a receptor (C5aR), a G-protein-coupled seven transmembrane domain receptor. In experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis, the C5aR is expressed on monocytes/macrophages, reactive astrocytes and T cells infiltrating the central nervous system (CNS). To investigate the role of the C5aR in this T cell-driven autoimmune model, we induced EAE in C5aR-deficient mice (C5aR(-/-)) and wild-type mice using a myelin oligodendrocyte glycoprotein (MOG) peptide as the immunogen. We found that C5aR(-/-) mice were fully susceptible to MOG-induced EAE with no difference in disease onset or severity in C5aR(-/-) mice compared to control mice. Cellular infiltrates (macrophages and T cells) were similar in the spinal cords of both animal groups and splenic T cells from C5aR(-/-) mice and control mice responded identically to MOG in T cell proliferation assays. Ribonuclease protection assays demonstrated no significant differences in pro-inflammatory gene expression between receptor-deficient and sufficient mice. These results indicate that the C5aR is not an essential mediator in the induction and progression of EAE.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/physiology
- CD3 Complex/biosynthesis
- CD3 Complex/genetics
- Cells, Cultured/immunology
- Chemokines/biosynthesis
- Chemokines/genetics
- Complement Activation
- Complement C5a/physiology
- Cytokines/biosynthesis
- Cytokines/genetics
- Disease Models, Animal
- Disease Progression
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/prevention & control
- Gene Expression Regulation
- Genetic Predisposition to Disease
- Lymphocyte Activation
- Macrophage-1 Antigen/biosynthesis
- Macrophage-1 Antigen/genetics
- Macrophages/pathology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Microglia/pathology
- RNA, Messenger/biosynthesis
- Receptor, Anaphylatoxin C5a
- Receptors, Complement/deficiency
- Receptors, Complement/genetics
- Receptors, Complement/physiology
- Spinal Cord/pathology
- T-Lymphocyte Subsets/immunology
Collapse
Affiliation(s)
- Rachael Reiman
- Department of Microbiology, University of Alabama at Birmingham, Birmingham 35294, USA
| | | | | | | |
Collapse
|
45
|
Koleva M, Schlaf G, Landmann R, Götze O, Jungermann K, Schieferdecker HL. Induction of anaphylatoxin C5a receptors in rat hepatocytes by lipopolysaccharide in vivo: mediation by interleukin-6 from Kupffer cells. Gastroenterology 2002; 122:697-708. [PMID: 11875003 DOI: 10.1053/gast.2002.31883] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND & AIMS In normal rat liver, anaphylatoxin C5a induces glucose output from hepatocytes indirectly via prostanoids released from Kupffer cells. Correspondingly, it was found that hepatocytes, in contrast to Kupffer cells, did not express C5a receptors. Lipopolysaccharide (LPS) has been reported to enhance C5a receptor expression in murine livers. This might be the result of de novo expression in hepatocytes. METHODS C5a receptor expression was investigated in hepatocytes after in vivo treatment of rats with LPS and in vitro stimulation of isolated cells with LPS and proinflammatory cytokines on messenger RNA (mRNA) and protein level, and functionally in isolated hepatocytes and perfused liver. RESULTS In vivo treatment of rats with LPS induced C5a receptor mRNA and protein in hepatocytes with a maximum after 8-10 hours. At this time-point, C5a directly activated glycogen phosphorylase in isolated hepatocytes and enhanced glucose output in perfused livers without the involvement of prostanoids. LPS failed to induce C5a receptors in cultured hepatocytes in vitro, whereas interleukin (IL) 6 and IL-1beta, which are known to be released from Kupffer cells on stimulation with LPS, did so. In cocultures of hepatocytes with Kupffer cells, LPS induced C5a receptors in hepatocytes in an IL-6-dependent manner. CONCLUSIONS Thus, IL-6 from Kupffer cells appears to be the main mediator of LPS-induced de novo expression of C5a receptors in hepatocytes.
Collapse
Affiliation(s)
- Milena Koleva
- Institut für Biochemie und Molekulare Zellbiologie, Georg-August-Universität Göttingen, Germany
| | | | | | | | | | | |
Collapse
|
46
|
Abstract
Complement (C) is one of the most critical defence mechanisms of the innate immunity against cerebral infection by viruses, bacteria and fungi, with different molecular pathways contributing to the clearance of the invading pathogens. There is now compelling evidence that C proteins can be synthesized by brain cells in response to the infectious challenge and leading to cytotoxic and cytolytic activities against the harmful intruders. However, since there is also emerging evidence that uncontrolled C biosynthesis/activation can lead to brain inflammation with loss of neurons and oligodendrocytes, it is important to highlight that C may have adverse effects in infectious diseases of the CNS and induce profound tissue damage. The role of C in brain infection may even be more versatile. Many invading pathogens are not helpless against C attack and can use the membrane-bound C molecules to invade the host, either by binding directly or after decoration with C fragments. During budding viruses can acquire complement inhibitors from the host cell membrane and thus behave like 'Trojan horses' that are sheltered from the local innate immune response. Moreover, pathogens have evolved means of molecular mimicry with the expression of C inhibitor-like molecules to escape recognition and clearance by the C system. We herein provide a comprehensive and insightful review of the expression and the role of the C system in the brain. The three main focuses are: (i) C activation and lysis of pathogens in the brain; (ii) C-dependent neuroinvasion mechanisms (iii) uncontrolled C activation in inflamed CNS contributing to tissue damage.
Collapse
Affiliation(s)
- Cornelia Speth
- Institute of Hygiene and Social Medicine, University of Innsbruck and Ludwig Boltzmann-Institute for AIDS Research, Fritz-Pergl-Str.3, A-6020 Innsbruck, Austria.
| | | | | |
Collapse
|
47
|
Yu JX, Bradt BM, Cooper NR. Constitutive expression of proinflammatory complement components by subsets of neurons in the central nervous system. J Neuroimmunol 2002; 123:91-101. [PMID: 11880154 DOI: 10.1016/s0165-5728(01)00483-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The brain is largely protected from damage due to infection, trauma, and aberrant processes by the innate immune system. These studies were undertaken to determine whether neurons in normal brains constitutively express complement components. In situ hybridization and immunohistochemical studies with specific riboprobes and antibodies, respectively, revealed that most hippocampal neurons, many pyramidal cortical neurons and cerebellar Purkinje neurons in normal murine brains constitutively express C3, C5 and C6. The constitutive expression by neuronal subsets of components of the complement activation and membrane attack pathways suggests that the complement system represents a "first line" of host defense in the brain.
Collapse
Affiliation(s)
- Jack X Yu
- Department of Immunology, The Scripps Research Institute, IMM-19, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
48
|
Abstract
Antigen-induced airway hyperresponsiveness and airway inflammation are features of both human asthma and animal models of this disease. The genesis of these key asthma phenotypes represents the summation of a complex cascade of immune responses. It is hypothesized that multiple cell types are involved in the induction, propagation, and maintenance of these immune processes. Several molecules have been reported to be essential for cell-cell interactions, inflammatory cell recruitment, and effector functions leading to the overall expression of the asthmatic phenotype. This review summarizes the genetic evidence supporting a role for these molecules in antigen-driven airway hyperresponsiveness and inflammation.
Collapse
Affiliation(s)
- A Daser
- Institute of Laboratory Medicine and Pathobiochemistry, Charite, Berlin, Germany
| | | | | |
Collapse
|
49
|
Schieferdecker HL, Schlaf G, Jungermann K, Götze O. Functions of anaphylatoxin C5a in rat liver: direct and indirect actions on nonparenchymal and parenchymal cells. Int Immunopharmacol 2001; 1:469-81. [PMID: 11367531 DOI: 10.1016/s1567-5769(00)00038-2] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Growing evidence obtained in recent years indicates that anaphylatoxin C5a receptors (C5aR) are not restricted to myeloid cells but are also expressed on nonmyeloid cells in different tissues such as brain, lung, skin and liver. In contrast to its well-defined systemic functions, the actions of anaphylatoxins in these organs are poorly characterized. The liver can be a primary target organ for the C5a anaphylatoxin since the liver is directly connected to the gut, via the mesenteric veins and portal vein which is a main source of complement activating lipopolysaccharides (LPS). In the normal rat liver, the C5aR is only expressed by nonparenchymal cells, i.e. strongly by Kupffer cells (KC) and hepatic stellate cells (HSC) and weakly by sinusoidal endothelial cells (SEC), but not expressed by the parenchymal hepatocytes (HC). Accordingly, direct effects of C5a were only found in the C5aR-expressing KC and HSC: C5a induced the release of prostanoids from KC and HSC and enhanced the LPS-dependent release of interleukin-6 from KC. These soluble mediators indirectly influenced effector functions of the C5aR-free HC. C5a enhanced the glycogen phosphorylase activity and thus the glucose output from HC indirectly via prostanoids released from KC and HSC. Glucose can serve as an energy substrate as well as an electron donor for the synthesis of reactive oxygen intermediates by KC. Moreover, C5a also enhanced transcription of the gene for the type-2 acute phase protein alpha 2-macroglobulin in HC indirectly by increasing LPS-dependent IL-6 release from KC. Under pathological conditions, C5aR was found to be upregulated in various organs including the liver. Simulation of inflammatory conditions by treatment of rats with IL-6, a main inflammatory mediator in the liver, caused a de novo expression of functional C5aR in HC. In livers of IL-6-treated rats, C5a initiated glucose output from HC and perhaps other HC-specific defense reactions directly without the intervention of soluble mediators from nonparenchymal cells.
Collapse
Affiliation(s)
- H L Schieferdecker
- Institut für Biochemie und Molekulare Zellbiologie, Georg-August-Universität Göttingen, Humboldtallee 23, D-37073 Göttingen, Germany.
| | | | | | | |
Collapse
|
50
|
Mukherjee P, Pasinetti GM. Complement anaphylatoxin C5a neuroprotects through mitogen-activated protein kinase-dependent inhibition of caspase 3. J Neurochem 2001; 77:43-9. [PMID: 11279260 DOI: 10.1046/j.1471-4159.2001.00167.x] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
We previously reported that pretreatment of murine cortico-hippocampal neuronal cultures with the complement-derived anaphylatoxin C5a, protects against glutamate neurotoxicity. In this study we explored the potential mechanisms involved in C5a-mediated neuroprotection. We found that C5a neuroprotects in vitro through inhibition of apoptotic death because pretreatment with human recombinant (hr)C5a prevented nuclear DNA fragmentation coincidental to inhibition of the pro-apoptotic caspase 3 activity mediated by glutamate treatment. Also, hrC5a-mediated responses appeared to be receptor-mediated because pretreatment of cultures with the specific C5a receptor antagonist C177, prevented hrC5a-mediated neuroprotection. Based on this evidence, we further explored possible signaling pathways involved in hrC5a inhibition of caspase 3 activation and apoptotic neuronal death. We found that treatment of cultures with the mitogen-activated protein kinase (MAPK) pathway inhibitor PD98059 prevented hrC5a-mediated inhibition of caspase 3 and apoptotic neuron death. MAPK pathways, whose activation by hrC5a is inhibited by PD98059 and C177, include the extracellular signal-regulated kinase (ERK)2 and, to a lesser extent, ERK1. The study suggests that C5a may protect against glutamate-induced apoptosis in neurons through MAPK-mediated regulation of caspase cascades.
Collapse
Affiliation(s)
- P Mukherjee
- Neuroinflammation Research Center, Department of Psychiatry, Mount Sinai School of Medicine, New York 10029, USA
| | | |
Collapse
|