Elsevier

Molecular Immunology

Volume 48, Issue 14, August 2011, Pages 1592-1603
Molecular Immunology

Review
Complement in the brain

https://doi.org/10.1016/j.molimm.2011.04.003Get rights and content

Abstract

The brain is considered to be an immune privileged site, because the blood–brain barrier limits entry of blood borne cells and proteins into the central nervous system (CNS). As a result, the detection and clearance of invading microorganisms and senescent cells as well as surplus neurotransmitters, aged and glycated proteins, in order to maintain a healthy environment for neuronal and glial cells, is largely confined to the innate immune system. In recent years it has become clear that many factors of innate immunity are expressed throughout the brain. Neuronal and glial cells express Toll like receptors as well as complement receptors, and virtually all complement components can be locally produced in the brain, often in response to injury or developmental cues. However, as inflammatory reactions could interfere with proper functioning of the brain, tight and fine tuned regulatory mechanisms are warranted. In age related diseases, such as Alzheimer's disease (AD), accumulating amyloid proteins elicit complement activation and a local, chronic inflammatory response that leads to attraction and activation of glial cells that, under such activation conditions, can produce neurotoxic substances, including pro-inflammatory cytokines and oxygen radicals. This process may be exacerbated by a disturbed balance between complement activators and complement regulatory proteins such as occurs in AD, as the local synthesis of these proteins is differentially regulated by pro-inflammatory cytokines. Much knowledge about the role of complement in neurodegenerative diseases has been derived from animal studies with transgenic overexpressing or knockout mice for specific complement factors or receptors. These studies have provided insight into the potential therapeutic use of complement regulators and complement receptor antagonists in chronic neurodegenerative diseases as well as in acute conditions, such as stroke. Interestingly, recent animal studies have also indicated that complement activation products are involved in brain development and synapse formation. Not only are these findings important for the understanding of how brain development and neural network formation is organized, it may also give insights into the role of complement in processes of neurodegeneration and neuroprotection in the injured or aged and diseased adult central nervous system, and thus aid in identifying novel and specific targets for therapeutic intervention.

Highlights

► Complement (C), a powerful tool of innate immunity, is important for defense of the brain. ► Most C proteins can be synthesized in the brain, mainly by glia but also by neurons. ► The C system is involved in both acute and chronic disorders of the brain. ► A delicate balance between C activators and regulators directs C to act as friend or foe. ► Recent findings suggests novel functions of C in brain development and synapse formation.

Introduction

Complement (C) is a major component of innate immunity, recognizing danger, as well as discriminating self from non-self (Ricklin et al., 2010). The C system is best known for its role in the recognition and killing of pathogenic microbes. Activation of the C system, which consists of over 30 soluble and cell-associated factors, can occur through three pathways, each triggered by different types of agents. All three pathways lead to the assembly of C3 convertases that, in turn, can cleave C3 resulting in formation of C3b and C3a activation products. The larger C3b fragment, a major effector molecule of the C system, acts as an opsonin and, in addition, together with other factors can assemble the C5 convertase, which enables further activation of the C cascade ultimately leading to generation of the chemotactic C5a fragment and the formation of the terminal complement complex C5b-9, also called membrane attack complex (Fig. 1). Thus, when the cascade is fully activated, C leads to assembly of the membrane attack complex (C5b-9; MAC) and lysis of invading microorganisms. However, if C5b-9 production is excessive or targeted to host cells, C5b-9 can induce host cell death. On the other hand, some C activation products also facilitate the generation of adaptive immune responses (Carroll, 2004, Erdei et al., 2009), while other components contribute to the control of autoimmunity during the clearance of apoptotic cells (Sjoberg et al., 2009, Fraser et al., 2009, van Kooten et al., 2008). Interestingly, sublytic amounts of C5b-9 on host cells may cause an influx of extracellular calcium that leads to activation and/or proliferation of the cells and resistance to induction of apoptosis (Cole and Morgan, 2003), further illustrating the diverse functions of this ancient pathway.

Binding of C1, a Ca2+-dependent complex of the recognition unit C1q and a tetramer of the proenzymes C1r and C1s, to an activator is the initial event in classical pathway (CP) activation of C. Activators can be immune complexes, certain microbes, apoptotic cells, and other specific protein motifs, such as amyloid in a fibrillar beta sheet structure found in the plaques in brain from Alzheimer's disease (AD) patients.

Mannose-binding lectin (MBL) and ficolins (Ficolin-1, -2 and -3) bind to mannan and other carbohydrate moieties or acetylated moieties on microorganisms or dying cells initiating C activation, through the lectin pathway (LP). MBL and ficolins share homology with C1q and, like C1q, are associated with proenzymes, MBL-associated serine proteases (MASPs).

The alternative pathway (AP) is initiated by spontaneous hydrolysis of the internal thioester within C3, resulting in C3b-like C3 (“tick-over”) or by recruitment of C3 by properdin bound to specific targets (Kemper and Hourcade, 2008). A range of microbial and also eukaryotic cell surfaces with a low sialic acid content allow AP C activation, whereas the inactivation of C3b by the C regulatory proteins (Creg) factor H (fH) and factor I (fI) is more efficient on surfaces rich in sialic acid (Austen and Fearon, 1979). C3b generated by the CP or MP activation pathways can enlist the alternative pathway components, thereby amplifying the amount of downstream complement cascade events (Fig. 1).

The brains of organisms with a well developed central nervous system (CNS) are shielded by the blood–brain-barrier (BBB) with tight junction formations at three principal barrier sites (i) the BBB formed by endothelial cells in the cerebral capillaries, (ii) the arachnoid barrier formed by the arachnoid multi-layered epithelium and (iii) the blood-CSF barrier formed by the CSF-secreting choroid plexus epithelium. The integrity properties are further defined by BBB-associated cells including pericytes and astrocytes. Together these brain barriers efficiently prevent infiltration of circulating immune cells, such as B- and T lymphocytes, and minimize influx of plasma proteins as well as neuroexcitatory and neurotoxic substances from the blood (reviewed in Abbott et al., 2010). The functions of immune surveillance and differentiation between “self” and “nonself” in non-CNS tissue, provided by neutrophils, dendritic cells, macrophages and natural killer cells in the periphery, are in the CNS attributed to resident glial cells including astrocytes, microglia, oligodendrocytes, and NG2 chondroitin sulphate (NG2) and platelet-derived growth factor-α receptor (PDGFα) positive oligodendrocyte precursor cells (NG2+PDGFα+ OPCs) (Butt et al., 2005 and reviewed in Dong and Benveniste, 2001, Griffiths et al., 2009). Much of our understanding of the occurrence and role of complement in the CNS derives from studies into the pathogenic mechanisms involved in various diseases affecting the brain. In a variety of CNS diseases, including bacterial meningitis, transmissible spongiform encephalopathies (TSE; prion disease), stroke and in more chronic conditions such as multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), Huntington's (HD) and Parkinson's disease (PD), AD, as well as age-related macular degeneration (AMD), C contributes to the inflammatory process (Woodruff et al., 2008, Bonifati and Kishore, 2007). In this review we discuss the findings that have led to insight on the role of C in acute and chronic brain disorders and, importantly, its role in the normal homeostasis and brain functioning, as more recent studies (Stevens et al., 2007) indicate that C is also involved in brain development and synapse pruning.

Section snippets

Local synthesis of complement and complement regulators in the brain

The liver is the main source of C proteins, but although the BBB is not absolute and some macromolecules can by-pass the barriers by use of extracellular routes (Broadwell and Sofroniew, 1993) most C proteins are unlikely to penetrate the brain parenchyma unless the BBB integrity is disrupted. Therefore, local synthesis of C components by resident cells in the brain is crucial to appropriate functions of the local defense system. Local synthesis of C proteins in the brain was suggested after

The role of complement in normal CNS

Similar to other proteins that are part of the immune system, such as proinflammatory cytokines (e.g., TNFα, IL-6) and proteins of the adaptive immune system (e.g. major histocompatibility complex class I [MHCI] molecules and MHCI-binding immunoreceptors and their components (e.g., PIRB, Ly49, DAP12, CD3ζ) (for a review see Boulanger, 2009), C factors are now thought to also have nonimmune functions in the brain. Complement proteins were found to promote proliferation and regeneration in

Complement during acute brain injury

Acute brain injuries including infections, brain trauma, ischemic and hemorrhagic stroke and subsequent reperfusion injuries are to date associated with a limited repertoire of effective treatments and high morbidity. Neurodegeneration and death in these acute conditions can be via necrosis or via apoptosis. For example, apoptosis was recently shown to be dominant in the peri-infarct area after ischemic stroke in humans (Sairanen et al., 2006). Most likely neuronal death following acute

Complement during chronic conditions of brain injury

Substantial advances in understanding the effects of C in the brain comes from research in neurodegenerative diseases (ND) and subsequent studies with animal models for ND including C knock outs or genetically manipulated animals over expressing certain C factors bred to AD mouse models. Here we will focus on only a few diseases that demonstrate some of the mechanisms of disease acceleration and begin to provide insight on potential therapeutic targets. Recent reviews of the pathogenesis of age

Summary and future directions

In summary, various cell types in the CNS were shown to synthesize C factors, and the synthesis rates of many factors increase during development and within the injured brain. Data from human immunohistochemistry, animal models of diseases, and in vitro studies suggest that the role of C in AD is complex, with evidence for both detrimental and beneficial functions, presumably dependent on location, timing, and environmental signals. The potential disease associated polymorphisms of C factors

Acknowledgements

The work in the authors’ lab reported here was supported by NIH grants NS35144 and AG 00538 (AT), Stchting Dioraphte, Hersenstichting Nederland and Internationale Stichting Alzheimer Onderzoek (06-517) (RV) and Demensfonden and Alzheimerfonden (HN). The authors thank Dr. Maria I. Fonseca (University of California, Irvine) for Fig. 2.

References (181)

  • R.D. Broadwell et al.

    Serum proteins bypass the blood-brain fluid barriers for extracellular entry to the central nervous system

    Exp. Neurol.

    (1993)
  • E.D. Cadman et al.

    Beta-amyloid peptides initiate the complement cascade without producing a comparable effect on the terminal pathway in vitro

    Exp. Neurol.

    (1997)
  • B.H. Choi et al.

    Decreases in protease nexins in Alzheimer's disease brain

    Neurobiol. Aging

    (1995)
  • P. Eikelenboom et al.

    The role of complement and activated microglia in the pathogenesis of Alzheimer's disease

    Neurobiol. Aging

    (1996)
  • A. Erdei et al.

    Expression and role of CR1 and CR2 on B and T lymphocytes under physiological and autoimmune conditions

    Mol. Immunol.

    (2009)
  • A. Familian et al.

    Minocycline does not affect amyloid beta phagocytosis by human microglial cells

    Neurosci. Lett.

    (2007)
  • M.I. Fonseca et al.

    Neuronal localization of C1q in preclinical Alzheimer's disease

    Neurobiol. Dis.

    (2004)
  • L. Gasparini et al.

    Activity of flurbiprofen and chemically related anti-inflammatory drugs in models of Alzheimer's disease

    Brain Res. Brain Res. Rev.

    (2005)
  • P. Gasque et al.

    Expression of the complement classical pathway by human glioma in culture, A model for complement expression by nerve cells

    J. Biol. Chem.

    (1993)
  • P. Gasque et al.

    Complement activation on human neuroblastoma cell lines in vitro: route of activation and expression of functional complement regulatory proteins

    J. Neuroimmunol.

    (1996)
  • E. Head et al.

    Complement association with neurons and beta-amyloid deposition in the brains of aged individuals with Down Syndrome

    Neurobiol. Dis.

    (2001)
  • W. Hu et al.

    Development of a novel therapeutic suppressor of brain proinflammatory cytokine up-regulation that attenuates synaptic dysfunction and behavioral deficits

    Bioorg. Med. Chem. Lett.

    (2007)
  • S. Itagaki et al.

    Ultrastructural localization of complement membrane attack complex (MAC)-like immunoreactivity in brains of patients with Alzheimer's disease

    Brain Res.

    (1994)
  • C. Kemper et al.

    Properdin: New roles in pattern recognition and target clearance

    Mol. Immunol.

    (2008)
  • A. Klos et al.

    The role of the anaphylatoxins in health and disease

    Mol. Immunol.

    (2009)
  • K.M. Lucin et al.

    Immune activation in brain aging and neurodegeneration: too much or too little?

    Neuron

    (2009)
  • C.E. Lumsden

    The immunogenesis of the multiple sclerosis plaque

    Brain Res.

    (1971)
  • P.L. McGeer et al.

    Activation of the classical complement pathway in brain tissue of Alzheimer patients

    Neurosci. Lett.

    (1989)
  • P.L. McGeer et al.

    Distribution of clusterin in Alzheimer brain tissue

    Brain Res.

    (1992)
  • P. Mukherjee et al.

    The role of complement anaphylatoxin C5a in neurodegeneration: implications in Alzheimer's disease

    J. Neuroimmunol.

    (2000)
  • T. Osterwalder et al.

    The axonally secreted serine proteinase inhibitor, neuroserpin, inhibits plasminogen activators and plasmin but not thrombin

    J. Biol. Chem.

    (1998)
  • J.J. Alexander et al.

    The complement cascade: Yin-Yang in neuroinflammation–neuro-protection and -degeneration

    J. Neurochem.

    (2008)
  • K.F. Austen et al.

    A molecular basis of activation of the alternative pathway of human complement

    Adv. Exp. Med. Biol.

    (1979)
  • M.H. Barnett et al.

    Immunoglobulins and complement in postmortem multiple sclerosis tissue

    Ann. Neurol.

    (2009)
  • M.H. Barnett et al.

    Relapsing and remitting multiple sclerosis: pathology of the newly forming lesion

    Ann. Neurol.

    (2004)
  • B.M. Bellander et al.

    Complement activation in the human brain after traumatic head injury

    J. Neurotrauma

    (2001)
  • M.E. Benoit et al.

    Complement protein C1q-mediated neuroprotection is correlated with regulation of neuronal gene and microRNA expression

    J. Neurosci.

    (2011)
  • L. Bergamaschini et al.

    Activation of the contact system in cerebrospinal fluid of patients with Alzheimer disease

    Alzheimer Dis. Assoc. Disord.

    (1998)
  • L. Bergamaschini et al.

    Peripheral treatment with enoxaparin, a low molecular weight heparin, reduces plaques and beta-amyloid accumulation in a mouse model of Alzheimer's disease

    J. Neurosci.

    (2004)
  • L. Bo et al.

    Subpial demyelination in the cerebral cortex of multiple sclerosis patients

    J. Neuropathol. Exp. Neurol.

    (2003)
  • Y.R. Bogestal et al.

    Signaling through C5aR is not involved in basal neurogenesis

    J. Neurosci. Res.

    (2007)
  • M.F. Bolliger et al.

    The cell-adhesion G protein-coupled receptor BAI3 is a high-affinity receptor for C1q-like proteins

    Proc. Natl. Acad. Sci. U. S. A.

    (2011)
  • B.M. Bradt et al.

    Complement-dependent proinflammatory properties of the Alzheimer's disease beta-peptide

    J. Exp. Med.

    (1998)
  • E.C. Breij et al.

    Homogeneity of active demyelinating lesions in established multiple sclerosis

    Ann. Neurol.

    (2008)
  • B.P. Brink et al.

    The pathology of multiple sclerosis is location-dependent: no significant complement activation is detected in purely cortical lesions

    J. Neuropathol. Exp. Neurol.

    (2005)
  • A.M. Butt et al.

    Synantocytes: the fifth element

    J. Anat.

    (2005)
  • C. Canova et al.

    Expression of innate immune complement regulators on brain epithelial cells during human bacterial meningitis

    J. Neuroinflamm.

    (2006)
  • J.C. Carel et al.

    Analysis of epitope expression and the functional repertoire of recombinant complement receptor 2 (CR2/CD21) in mouse and human cells

    J. Immunol.

    (1989)
  • M.C Carroll

    The complement system in regulation of adaptive immunity

    Nat. Immunol.

    (2004)
  • P. Chakrabarty et al.

    IFN-gamma promotes complement expression and attenuates amyloid plaque deposition in amyloid beta precursor protein transgenic mice

    J. Immunol.

    (2010)
  • Cited by (310)

    View all citing articles on Scopus
    View full text