Complement is an important component of innate immune defence against pathogens and crucial for efficient immune complex disposal. These core protective activities are dependent in large part on properly regulated complement-mediated inflammation. Dysregulated complement activation, often driven by persistence of activating triggers, is a cause of pathological inflammation in numerous diseases, including neurological diseases. Increasingly, this has become apparent not only in well-recognized neuroinflammatory diseases like multiple sclerosis but also in neurodegenerative and neuropsychiatric diseases where inflammation was previously either ignored or dismissed as a secondary event. There is now a large and rapidly growing body of evidence implicating complement in neurological diseases that cannot be comprehensively addressed in a brief review. Here, we will focus on neurodegenerative diseases, including not only the ‘classical’ neurodegenerative diseases such as Alzheimer’s disease and Parkinson’s disease, but also two other neurological diseases where neurodegeneration is a neglected feature and complement is implicated, namely, schizophrenia, a neurodevelopmental disorder with many mechanistic features of neurodegeneration, and multiple sclerosis, a demyelinating disorder where neurodegeneration is a major cause of progressive decline. We will discuss the evidence implicating complement as a driver of pathology in these diverse diseases and address briefly the potential and pitfalls of anti-complement drug therapy for neurodegenerative diseases.

In this brief review we will discuss how complement, essential for immune defence and homeostasis in the healthy brain, can also contribute to pathology in neuroinflammation and neurodegeneration. The basics of the system are summarized in Figure 1. Complement activation in the periphery is a ‘double-edged sword’, functioning to fight infection and resolve inflammation in health but, when dysregulated in disease, causing or exacerbating pathology – and precisely the same applies to complement in the brain. Before considering what complement does in the brain it is first necessary to address some key aspects of brain complement: Are complement proteins present in the brain made locally or acquired from the periphery? How and when is complement activated in brain? How do brain cells respond to complement activation? How is complement regulated in the brain?

The complement cascade

Figure 1
The complement cascade

The complement system is activated via the classical, lectin, or alternative pathways. The classical pathway is initiated by C1 binding to antigen-antibody complexes or in brain directly to amyloid, tau or α-Syn aggregates. C1s in the C1 complex cleaves C4 and C2 to form the C3 convertase C4b2a. The lectin pathway is activated by MBL or other lectins binding surface carbohydrates; attached MASPs are activated to cleave C4 and C2 to generate C4b2a. The C3 convertase cleaves C3 to C3b and C3a, the former is further processed by Factor I (FI) to form the opsonin iC3b, a ligand for phagocyte complement receptors CR3 and CR4 to initiate phagocytosis. C3b also feeds back to generate more C3 convertase via the amplification loop; factor B (FB) binds C3b and is then cleaved by factor D (FD) to create the alternative pathway C3 convertase C3bBb. Binding of a further C3b to either C3 convertase creates a C5 convertase which cleaves C5 to initiate the terminal pathway culminating in formation of the membrane attack complex (MAC). The small fragments generated from the C3 and C5 convertases, C3a and C5a, are anaphylatoxins and signal via their receptors, C3aR and C5aR, to recruit immune cells. Numerous cell surface and fluid-phase proteins regulate complement activation and terminal pathway, including C1-inhibitor (C1INH), which inhibits C1s and MASPs, decay accelerating factor (DAF), complement receptor 1 (CR1) and Factor H (FH) which regulate the C3 convertases clusterin, vitronectin and CD59 which control MAC assembly. Complement drugs in clinical/pre-clinical trials are shown in red. GWAS hits in specific diseases are shown in blue. Figure created with BioRender (BioRender.com).

Figure 1
The complement cascade

The complement system is activated via the classical, lectin, or alternative pathways. The classical pathway is initiated by C1 binding to antigen-antibody complexes or in brain directly to amyloid, tau or α-Syn aggregates. C1s in the C1 complex cleaves C4 and C2 to form the C3 convertase C4b2a. The lectin pathway is activated by MBL or other lectins binding surface carbohydrates; attached MASPs are activated to cleave C4 and C2 to generate C4b2a. The C3 convertase cleaves C3 to C3b and C3a, the former is further processed by Factor I (FI) to form the opsonin iC3b, a ligand for phagocyte complement receptors CR3 and CR4 to initiate phagocytosis. C3b also feeds back to generate more C3 convertase via the amplification loop; factor B (FB) binds C3b and is then cleaved by factor D (FD) to create the alternative pathway C3 convertase C3bBb. Binding of a further C3b to either C3 convertase creates a C5 convertase which cleaves C5 to initiate the terminal pathway culminating in formation of the membrane attack complex (MAC). The small fragments generated from the C3 and C5 convertases, C3a and C5a, are anaphylatoxins and signal via their receptors, C3aR and C5aR, to recruit immune cells. Numerous cell surface and fluid-phase proteins regulate complement activation and terminal pathway, including C1-inhibitor (C1INH), which inhibits C1s and MASPs, decay accelerating factor (DAF), complement receptor 1 (CR1) and Factor H (FH) which regulate the C3 convertases clusterin, vitronectin and CD59 which control MAC assembly. Complement drugs in clinical/pre-clinical trials are shown in red. GWAS hits in specific diseases are shown in blue. Figure created with BioRender (BioRender.com).

Close modal

Most complement proteins are produced in the liver, secreted into plasma and pass from there into extra-hepatic tissues and organs; local synthesis of complement proteins does occur but is of limited importance in most organs. The brain is different from other organs because it is enclosed in the blood–brain barrier (BBB), restricting the influx of complement proteins and other macromolecules from plasma. The brain, like other organs, must be defended against infection, but the role of complement in this defence was unclear. Early studies demonstrated the presence of complement proteins in cerebrospinal fluid (CSF) and in brain extracts, but the sources of these were uncertain – were they made locally or infiltrated from the periphery [1–3]? Evidence regarding local production of complement in the brain is patchy and often derived from studies in immortalized cell lines of tenuous relevance to primary brain cells. For example, ex vivo studies using human brain cell lines or primary cells isolated from postmortem brain demonstrated that almost all complement components, regulators and receptors can be produced by brain cells with neurons, astrocytes, oligodendrocytes and microglia all contributing (reviewed in [4]) (Table 1). Astrocytes are the most abundant non-neuronal cell type in the brain [5], primary astrocytes expressed the regulators CD59, CD55 and CD46, the complement receptors CR1, C3aR and C5aR [6–8], the terminal pathway components C6, C7 and C9, and terminal pathway regulators vitronectin and clusterin [9]. More recently, activated astrocytes have been shown to abundantly express and secrete C3 ex vivo and in situ in brain; indeed, C3 is now used as a marker for activated astrocytes [10]. Microglia, the principal brain phagocytes, are the predominant source of C1q in the brain [11]; cultured human primary microglia also expressed C3 [12] and induced pluripotent stem cell (iPSC)-derived microglia expressed C1q, C2, C3, complement receptor 3 (CR3) and complement receptor 1 (CR1/CD35) and several other complement proteins (Table 1) [13–15]. Neurons also express complement proteins; human neuronal cell lines expressed the components C1q, C2, C3, factor B (FB), C6 and C9, and the regulators factor H (FH) and C1 inhibitor (C1-INH) [16–18]. Human fetal neurons expressed C4, C3 and the regulators CD59 and CD46 and were positive for C3 and terminal pathway activation products, suggesting that complement was activated on them in situ [9]. Neurons derived from iPSC expressed and secreted C1, C4, FD, FI and FB as well as the regulators C1-INH, C1qBP and DAF [15,19]. Complement expression by the myelin-producing oligodendrocytes has been little studied; one report using a human oligodendrocyte cell line (HOG) showed expression of the membrane regulators CD55, CD46 and CD59 and the fluid-phase regulators C1-INH, vitronectin and clusterin [20,21]. RNA-sequencing analyses of individual brain cells have confirmed that most but not all complement genes are expressed in the human brain [22]. However, the majority of these studies were performed on non-inflamed brain; the release of transcriptomic data from the Seattle Alzheimer’s Disease Brain Cell Atlas should clarify brain cell expression of complement proteins in disease [23].

Table 1
Evidence of complement expression in different brain cell types
ProteinNeuronMicrogliaAstrocyteOligodendrocyteReferences
C1q Cell lines iPSC, primary cells Cell lines Primary cells [12,15,17,18,20,247,248
C1r iPSC iPSC Cell lines Primary cells [15,20,247–249
C1s iPSC iPSC Cell lines Primary cells [15,20,247–249
C2 Cell lines iPSC Cell lines Primary cells [15,18,20,247–249
C3 Cell lines, primary cells Primary cells, iPSC Cell lines, primary cells Cell lines, primary cells [9,12,15,17,18,20,247,250–253
C4 Cell lines, iPSC, primary cells Primary cells, iPSC Cell lines, primary cells Primary cells [9,12,15,17,18,20,247–251,253–255
C5 Cell lines, iPSC iPSC Cell lines Primary cells [15,17–20,247–249
C6 Cell lines iPSC Cell lines, primary cells Primary cells [7,17,18,20,247,249
C7 Cell lines iPSC Primary cells Primary cells [7,17,18,20,247
C8 Cell lines iPSC  Primary cells [18,20,247
C9 Cell lines, primary cells iPSC Cell lines, primary cells Primary cells [7,9,15,17,18,20,247,250,255
C1-INH Cell lines, iPSC, tissue iPSC, primary cells Cell lines, primary cells, tissue Cell lines [7,15,17,21,247,248,251,256,257
C3aR Cell lines iPSC, primary cells Cell lines, primary cells  [8,247,258–259
C5aR  iPSC Cell lines, primary cells  [6,15,247
C4BP  iPSC  Cell lines [21,247
C1qBP iPSC iPSC   [15,19,247
MCP/CD46 Cell lines, primary cells iPSC Cell lines, primary cells Cell lines [7,9,21,247,260–262
DAF/CD55 iPSC iPSC Cell lines, primary cells Cell lines [9,19,21,247,259,261
CD59 Cell lines, primary cells iPSC Cell lines, primary cells Cell lines [7,9,21,247,252,260,261
CFB Cell lines, iPSC iPSC Cell lines  [15,17,247,252
CFD iPSC iPSC   [15,247
CFH Cell lines iPSC Cell lines, primary cells Cell lines [17,21,247,252,260,263
CFI iPSC  Cell lines  [15,252
Properdin  iPSC Primary cells  [15,247,263
CLU Cell lines, iPSC iPSC Cell lines, primary cells Cell lines [7,15,19,21,247,260
CR1  Cell lines, iPSC Cell lines, primary cells  [14,21,247
CR1L  iPSC   [247
CR2  iPSC   [247
CR4/CD11c  iPSC   [15
CR3/CD11b/ITAM  iPSC   [15
CSMD1 Tissue    [204
FCN1  iPSC   [247
FCN3  iPSC   [247
Itgax  iPSC   [247
MASP1 iPSC iPSC Cell lines  [15,247,249
MASP2 iPSC iPSC Cell lines  [15,247,249
MASP3   Cell lines  [249
MBL2  iPSC   [249
VTN Cell lines, iPSC iPSC Cell lines, primary cells Cell lines [7,15,21,247,260
ProteinNeuronMicrogliaAstrocyteOligodendrocyteReferences
C1q Cell lines iPSC, primary cells Cell lines Primary cells [12,15,17,18,20,247,248
C1r iPSC iPSC Cell lines Primary cells [15,20,247–249
C1s iPSC iPSC Cell lines Primary cells [15,20,247–249
C2 Cell lines iPSC Cell lines Primary cells [15,18,20,247–249
C3 Cell lines, primary cells Primary cells, iPSC Cell lines, primary cells Cell lines, primary cells [9,12,15,17,18,20,247,250–253
C4 Cell lines, iPSC, primary cells Primary cells, iPSC Cell lines, primary cells Primary cells [9,12,15,17,18,20,247–251,253–255
C5 Cell lines, iPSC iPSC Cell lines Primary cells [15,17–20,247–249
C6 Cell lines iPSC Cell lines, primary cells Primary cells [7,17,18,20,247,249
C7 Cell lines iPSC Primary cells Primary cells [7,17,18,20,247
C8 Cell lines iPSC  Primary cells [18,20,247
C9 Cell lines, primary cells iPSC Cell lines, primary cells Primary cells [7,9,15,17,18,20,247,250,255
C1-INH Cell lines, iPSC, tissue iPSC, primary cells Cell lines, primary cells, tissue Cell lines [7,15,17,21,247,248,251,256,257
C3aR Cell lines iPSC, primary cells Cell lines, primary cells  [8,247,258–259
C5aR  iPSC Cell lines, primary cells  [6,15,247
C4BP  iPSC  Cell lines [21,247
C1qBP iPSC iPSC   [15,19,247
MCP/CD46 Cell lines, primary cells iPSC Cell lines, primary cells Cell lines [7,9,21,247,260–262
DAF/CD55 iPSC iPSC Cell lines, primary cells Cell lines [9,19,21,247,259,261
CD59 Cell lines, primary cells iPSC Cell lines, primary cells Cell lines [7,9,21,247,252,260,261
CFB Cell lines, iPSC iPSC Cell lines  [15,17,247,252
CFD iPSC iPSC   [15,247
CFH Cell lines iPSC Cell lines, primary cells Cell lines [17,21,247,252,260,263
CFI iPSC  Cell lines  [15,252
Properdin  iPSC Primary cells  [15,247,263
CLU Cell lines, iPSC iPSC Cell lines, primary cells Cell lines [7,15,19,21,247,260
CR1  Cell lines, iPSC Cell lines, primary cells  [14,21,247
CR1L  iPSC   [247
CR2  iPSC   [247
CR4/CD11c  iPSC   [15
CR3/CD11b/ITAM  iPSC   [15
CSMD1 Tissue    [204
FCN1  iPSC   [247
FCN3  iPSC   [247
Itgax  iPSC   [247
MASP1 iPSC iPSC Cell lines  [15,247,249
MASP2 iPSC iPSC Cell lines  [15,247,249
MASP3   Cell lines  [249
MBL2  iPSC   [249
VTN Cell lines, iPSC iPSC Cell lines, primary cells Cell lines [7,15,21,247,260

Abbreviation: iPSC, induced pluripotent stem cell.

Together, the data show that brain cells can make all the key complement proteins and regulators and express membrane complement regulators and receptors that enable them to resist and respond to complement activation (Table 1). Most complement proteins are acute phase reactants; peripheral inflammation drives increased hepatic synthesis with resultant increased plasma levels [24]. In brain, a few studies in primary cells, cell lines and mouse models have demonstrated that complement gene expression is increased by inflammatory triggers. Repeated systemic injection of lipopolysaccharide (LPS) in mice caused increased C1q expression in the hippocampus associated with increased microglial phagocytosis [25], while LPS treatment of neuron/glial co-cultures increased C3 expression and secretion [26]. The impact of inflammation on complement gene and protein expression in brain pathology will be discussed in later sections.

CNS is an immune-privileged site isolated behind the BBB; complement proteins are locally synthesized to maintain immunosurveillance [27,28]. Protective roles include the clearance of pathogens and cellular debris, synaptic pruning and neural circuit refinement, and maintaining BBB integrity. C1q has anti-inflammatory and neuroprotective properties, accomplished by enhancing the removal of apoptotic cells, suppressing inflammatory cytokines, encouraging the quiet elimination of cell debris, and reducing the accompanying inflammation [29,30]. Additionally, throughout retinal development, C1q with C3 contribute to pruning and remodeling of synapses [31]. The local activation of C3 to C3b, which can subsequently function as an opsonin, occurs at synapses that are C1q tagged. This enables microglia expressing the C3b receptor, CR3, to specifically target and eliminate these neurons. C1q and C3 have also been linked to memory regulation, specifically in the process of forgetting, where they direct microglial engulfment of specific engram cells during restructuring of the brain [32]. Downstream complement pathway activation leads to neurotoxicity [28,33]. Studies utilizing animal models of AD, stroke and NMO demonstrate the significance of complement in maintaining the integrity of the BBB and the healthy balance of the brain, although the precise protective mechanisms are uncertain [34]. Together, these data show that complement is critical for immunosurveillance and essential for normal brain development. However, chronic activation of the cascade contributes to the breakdown of the BBB and brain injury.

The demonstration that complement is required for synaptic pruning during development was a game-changer, focussing attention on roles of complement in brain health and disease [31,35]. These elegant studies in the developing rodent visual system showed that synapses destined for elimination were tagged with C1q [31]. Later studies showed that synaptic C1q is part of the C1 complex, and that binding of C1 initiated activation of the classical complement pathway on the targeted synapse, leading to the deposition of C3 activation fragments (C3b and iC3b) and synaptic engulfment by microglia expressing C3 fragment receptors (CR3 and CR4) [36,37]. Mice deficient in C1q, C3, C4 or the receptor CR3, all showed defects in developmental synaptic pruning [35–38]. Together, these studies demonstrated a critical homeostatic role for complement in synapse elimination and confirmed that complement was present and active in the healthy brain, provoking further research activity.

In the following sections, we will discuss the evidence implicating complement in the various neurodegenerative diseases (NDDs) and strategies to modulate complement activation in brain. Likely roles of inflammation and complement dysregulation in NDDs are summarized in Figure 2.

Complement in healthy and neurodegenerative brain

Figure 2
Complement in healthy and neurodegenerative brain

In the healthy brain, as in other organs, complement plays a crucial role in defending against infections and maintaining tissue homeostasis. These homeostatic roles include the core complement functions of immune surveillance to detect and eliminate pathogens and to remove dead cells and debris (in collaboration with microglia), and the brain-specific role of synaptic pruning. In the NDD brain, complement is dysregulated, generating an excess of pro-inflammatory products that trigger hyper-activation of microglia (and other glia), production of diverse proinflammatory mediators and increased phagocytosis. Terminal pathway activation and MAC formation directly damages neurones and synapses, precipitating neurodegeneration. Figure created with BioRender (BioRender.com).

Figure 2
Complement in healthy and neurodegenerative brain

In the healthy brain, as in other organs, complement plays a crucial role in defending against infections and maintaining tissue homeostasis. These homeostatic roles include the core complement functions of immune surveillance to detect and eliminate pathogens and to remove dead cells and debris (in collaboration with microglia), and the brain-specific role of synaptic pruning. In the NDD brain, complement is dysregulated, generating an excess of pro-inflammatory products that trigger hyper-activation of microglia (and other glia), production of diverse proinflammatory mediators and increased phagocytosis. Terminal pathway activation and MAC formation directly damages neurones and synapses, precipitating neurodegeneration. Figure created with BioRender (BioRender.com).

Close modal

Alzheimer’s disease (AD) is the commonest cause of dementia, a NDD affecting more than 55 million individuals worldwide and projected to increase dramatically in the next decade as the population ages [39,40]. AD causes a progressive deterioration in cognitive abilities, memory, and ability to perform everyday tasks, ultimately resulting in a loss of independence. The pathological hallmarks of AD are the accumulation in brain of protein deposits, amyloid-β (Aβ) plaques and hyperphosphorylated tau tangles. Cognitive impairment is caused by loss of neurons and synapses in specific brain regions. Neurodegeneration is accompanied by inflammation; brain-resident immune cells, astrocytes and microglia, adopt a neurotoxic, phagocytic, and proinflammatory phenotype and interact with plaques, tangles and damaged neurons [41,42]. This neuroinflammation, once dismissed as a secondary event, is now recognized as a significant driver of AD pathology. The most compelling evidence came from genetic studies; several of the genes most strongly associated with AD risk are related to inflammation and immunity [43–45]. The realization that neuroinflammation is causative in AD has refocussed research to gain a deeper understanding of the timing, location and mechanisms of inflammation in the AD brain, identify the inflammatory pathways, develop improved diagnostic tests and design innovative therapeutic interventions.

Complement in AD: immunohistochemistry (IHC) evidence

While restricted to postmortem AD brain and thus to late-stage disease, IHC analysis has nevertheless convincingly demonstrated the presence of complement proteins and activation products in and around areas of pathology. Classical pathway proteins, including C1q, C3 and C4, co-localized specifically with amyloid plaques but not tangles in temporal cortex, amygdala, and hippocampus, the brain regions most affected in AD [2,46,47]. Complement activation products, including C3 fragments and the terminal complement complex (TCC), were abundant in AD cortex, associating with plaques, tangles, and neuropil threads [48]. Microglia proximal to areas of pathology in the hippocampus and frontal cortex showed up-regulation of C5a receptors, C5aR1, and C5aR2 [49], while microglial CR1 expression was increased 10-fold in AD cases compared with controls [14].

Complement in AD: genetic evidence

Multiple genome-wide association studies (GWAS) have identified single-nucleotide polymorphisms in complement genes as contributing factors to late onset AD (LOAD) risk [45,50–52]. Two complement genes are reproducibly identified in the top five genetic hits: CR1 encoding CR1, an important receptor and regulator of complement activation, and CLU encoding clusterin, a chaperone protein involved in several homeostatic processes, including complement regulation [50,51,53]. The demonstration by microarray analysis that clusterin expression was upregulated in AD brain supported the association [54]. A recent GWAS additionally identified C1S, encoding the key C1-associated enzyme C1s, as a potential genetic factor in LOAD, firmly implicating the classical pathway in AD pathogenesis [50]. Other AD GWAS hits include CD33 and TREM2, both encoding myeloid cell receptors that bind C1q and/or C3 [55,56]. Expression studies have reported elevated expression of the complement receptors C3aR1 and C5aR1 and complement proteins C4A, C4B and CFHR1 in AD compared with age-matched controls, while C1q binding protein (C1qBP) expression was decreased [54]. In a separate study, the macular degeneration-associated risk polymorphism in FH (1277C) was associated with higher risk of AD [57]. While these genetic associations provide strong evidence of the involvement of complement in AD pathology, the precise mechanisms by which complement gene variants influence AD risk remain largely unknown. Research in this area is ongoing, aiming to unravel the complex interplay between genetic factors and complement dysregulation in AD development, essential to inform future therapeutic strategies and interventions.

Complement in AD: biomarker evidence

Numerous studies have identified distinct alterations in complement proteins and activation products in blood and/or CSF from AD patients compared with controls, or as predictors of progression to AD in individuals with mild cognitive impairment (MCI) (Table 2) [58–62]. These have been comprehensively reviewed elsewhere [63] and will be summarized briefly here. In CSF, several studies have reported increased levels of C3 and FH in AD, correlating with cognitive decline [64–66]; however, another study reported that lower levels of C3 and FH were associated with faster cognitive decline and increased brain atrophy [67]. Others reported increased C4 and soluble CR1 in AD CSF [68]. In plasma, low C3 levels were associated with risk of progression to AD, particularly in those with apolipoprotein E4 (APOE4) genotype [62]. Individuals with Down Syndrome (DS) and dementia had significantly lower plasma levels of C3 and factor I compared with those without dementia [69]. A recent study linking genetics and plasma complement levels demonstrated that AD-associated SNPs in CR1, C1S and FH impacted plasma levels of the encoded proteins, suggesting a mechanism for the observed impact on disease risk [70]. Although there is a lack of consistency between these many studies, likely influenced by different populations and very different assays, collectively they support altered complement activity and regulation in AD patients.

Table 2
Complement biomarkers in neurodegenerative diseases
DiseaseSample typeComplement componentChangeReference
Alzheimer’s disease Plasma C3 Decrease [62
 CSF C3, FH Increased [64–66
 CSF C3, FH Decrease [67
 CSF C4, CR1 Increased [68
Down syndrome Plasma C3, FI Decrease [69
Frontotemporal dementia Plasma C2, C3 Increased in C9orf72 and MAPT carriers [112
 CSF C1q, C3b Increased [112
 CSF C2, C3, FD Increased [112
Multiple systems atrophy Plasma C3 Decrease [65,66
 CSF C3 Decreased [65,66
Parkinson’s disease Plasma C3 Decrease [65
 Plasma C3 No change [120
 Plasma C3, C4 Increased baseline levels [120,121
 CSF FH, C9 Increased [123
Huntington’s disease Plasma C7, C9, Clusterin Increased [141
 CSF C1q, C4B Increased [141
Multiple sclerosis Plasma MASP-2, C3, C4, C4a, FH, C1inh Increased [150,160–162,171
 Plasma iC3b, Bb, C1inh, C1s, C9, TCC Decreased [160,163
 Plasma C4, FB, Bb, C1s, Clusterin, FI No difference [150,160,162
 CSF C1q, C3, C3a, C4, C4a, TCC Increased [150,165,166,168,169,171
 CSF C3, C9, FB Decreased [160,166,167
 CSF C3, C4 No difference [162
Schizophrenia Plasma C4a Increased [218
DiseaseSample typeComplement componentChangeReference
Alzheimer’s disease Plasma C3 Decrease [62
 CSF C3, FH Increased [64–66
 CSF C3, FH Decrease [67
 CSF C4, CR1 Increased [68
Down syndrome Plasma C3, FI Decrease [69
Frontotemporal dementia Plasma C2, C3 Increased in C9orf72 and MAPT carriers [112
 CSF C1q, C3b Increased [112
 CSF C2, C3, FD Increased [112
Multiple systems atrophy Plasma C3 Decrease [65,66
 CSF C3 Decreased [65,66
Parkinson’s disease Plasma C3 Decrease [65
 Plasma C3 No change [120
 Plasma C3, C4 Increased baseline levels [120,121
 CSF FH, C9 Increased [123
Huntington’s disease Plasma C7, C9, Clusterin Increased [141
 CSF C1q, C4B Increased [141
Multiple sclerosis Plasma MASP-2, C3, C4, C4a, FH, C1inh Increased [150,160–162,171
 Plasma iC3b, Bb, C1inh, C1s, C9, TCC Decreased [160,163
 Plasma C4, FB, Bb, C1s, Clusterin, FI No difference [150,160,162
 CSF C1q, C3, C3a, C4, C4a, TCC Increased [150,165,166,168,169,171
 CSF C3, C9, FB Decreased [160,166,167
 CSF C3, C4 No difference [162
Schizophrenia Plasma C4a Increased [218

Complement in AD: evidence from animal models

There have been numerous studies of the impact of complement component knockout or complement inhibition on disease in mouse models of amyloid and tau pathology with rather mixed results. The demonstration that Aβ oligomers directly injected into mouse brain triggered complement activation and synapse loss, prevented by administration of C1q blocking antibody, implicated complement in amyloid-induced pathology [71]. In multiple mouse AD models, knockdown of complement proteins protects synapses [72]. For example, C1q knockout reduced synaptic pruning by both astrocytes and microglia and rescued synaptic density [73], and anti-C1q treatment rescued synapse loss in the P301S mouse model [74]. Knockout of the pivotal complement protein C3 protected synapses and neurons in the APP/PS1 mouse AD model despite increased plaque load, demonstrating a disconnect between amyloid plaque accumulation and neurodegeneration [75]. Nevertheless, the absence of C3 in the APP-C3−/− mice resulted in an accelerated deposition of amyloid beta plaques and increased neurodegeneration [76]. Moreover, 1-year-old APP mice expressing the C3 inhibitor soluble complement receptor-related protein y (sCrry) exhibited a 2- to 3-fold higher amyloid beta deposition accompanied by accumulation of degenerating neurons when compared with age-matched APP mice [77]. This dichotomy highlights the intricate interplay between complement and amyloid beta pathology, underscoring the need for a nuanced understanding of their relationship in the context of neurodegenerative processes. The terminal pathway, responsible for MAC formation, has also been implicated in models; knocking out C6 in 3xTg AD mice reduced synapse loss and pharmacological blockade of the terminal pathway using an anti-C7 blocking antibody was neuroprotective in APPNLGF mice, together indicating that MAC formation plays a significant role in driving synapse loss in AD models [78]. The C3a/C3aR pathway has been implicated in numerous animal studies. Helicobacter pylori-derived outer membrane vesicles caused accelerated disease development in a mouse AD model and this was reversed by disrupting C3a/C3aR signaling [79]. Knockout of C3aR reduced microglial activation, synapse loss and cognitive decline in both tauopathy [80] and amyloidopathy [81,82] models. The C5a/C5aR pathway has also been implicated; both C5aR1-KO and inhibition of the C5a/C5aR pathway restored microglial homeostasis, reduced neuronal loss and improved cognition in AD models [83,84].

Complement in AD: summary

These combined findings in human AD samples and AD models highlight the involvement of diverse components of the complement system in pathogenesis and provide valuable insights into the likely roles of complement in the AD brain. A better understanding of complement dysregulation in early disease stages and progression would aid development of effective diagnostic and therapeutic strategies for AD.

Vascular dementia (VaD) is the second most common cause of dementia and a frequent comorbidity in AD and other neurogenerative diseases, present in >50% of dementia cases [85]. In VaD, blocked or reduced blood flow to the brain leads to reduced brain oxygen and glucose, essential for neuronal survival. Underlying pathologies include atherosclerosis, small vessel disease (SVD) and cerebral amyloid angiopathy (CAA); all can lead to cerebrovascular infarcts, intracerebral haemorrhage (ICH) and chronic cerebral hypoperfusion. Neuroinflammation is an integral part of VaD and increasing evidence demonstrates the importance of complement in its pathology.

Complement in VaD: contributions to risk factors

Risk factors for VaD include stroke, diabetes and hypertension, all of which are associated with atherosclerosis [85]. Complement dysregulation is strongly implicated in atherosclerosis as a driver of inflammation and so likely plays a role in SVD and VaD [86]. Early studies reported deposition of complement proteins, including the activation product iC3b and regulator FH, in atherosclerotic plaques [87]. In two large GWAS, increased risk of ICH was strongly associated with genetic variation in the complement genes CR1 and Clu, both also AD GWAS significant [88], implying a causal role of complement in VaD pathogenesis. In a mouse model of ICH, neurodegeneration and edema were reduced in C3-KO mice [89], while pharmacological inhibition of complement with N-acetylheparin or tricarboxylic acid reduced brain injury in a rat ICH model [90]. These findings implicate complement in the pathogenesis of VaD induced by microinfarcts and suggest that complement inhibition might ameliorate injury.

Complement in VaD: roles in CAA

CAA, an accumulation of amyloid in the walls of small cerebral arteries, is present to varying degrees in ∼90% of AD cases, particularly those with underlying SVD [91]. Impaired clearance of amyloid along intramural periarterial drainage (IPAD) pathways is implicated in the development and progression of CAA [92]. Vascular amyloid deposits are associated with damage to the endothelium and perivascular inflammation resulting in vasogenic oedema observed as white matter hyperintensities (WMHI) in MRI [92–94]. The recent demonstration that plasma C3 levels are elevated and a potential blood biomarker of CAA in MCI patients suggests a role for complement [95]; indeed, an IHC study of CAA in AD brain showed deposition of C3 fragments and C9 in the amyloid-laden vessels that correlated with severity of CAA [96], while others report staining for C1q, C3d, C6 and C5b-9 in amyloid-positive cerebral vessels from CAA cases [91]. Brain vascular smooth muscle cells expressed all classical and terminal pathway complement proteins, increased on exposure to Aβ, IFN-γ or IL1β, suggesting that local production might contribute to complement dysregulation in the vessel wall [97]. Proteomic studies in white matter or isolated amyloid-laden blood vessels from CAA donors showed enrichment in the terminal pathway regulator clusterin, suggesting a compensatory mechanism to limit MAC formation [98,99]; in contrast, genetic ablation of clusterin in AD mice reduced amyloid load and brain inflammation by causing redistribution of amyloid from plaques to the perivascular compartment [100]. Of note, immune complex formation and complement activation within IPAD pathways is a frequent complication in trials of anti-amyloid immunotherapy and a major driver of toxic side effects including microhaemorrhages [101]; inhibiting complement activation could alleviate the toxicity.

Complement in VaD: roles of C3a/C3aR

The C3a/C3aR pathway is an important contributor to vascular inflammation. Human brain microvascular endothelial cells stained in situ or in culture, expressed C3aR; exposure to C3a increased BBB permeability through disruption of tight junctions and enhanced perivascular inflammation via VCAM up-regulation [102]. Hypoperfusion, a common characteristic of VaD, caused microglial activation in mice with up-regulation of C3 and C3 fragment receptors, C3aR and CR3 and resultant demyelination with complement activation and C3 fragment deposition on myelin [103]. Genetic ablation or pharmacological inhibition of C3aR reduced hypoperfusion-induced microglial activation and prevented myelin loss, confirming the primary role of the C3a/C3aR axis in hypoperfusion injury and its potential as a target for therapeutic intervention in VaD [103].

Complement in VaD: summary

Studies in man and models convincingly demonstrate the relevance of complement dysregulation in the conditions that predispose to VaD, notably vascular diseases and stroke. Dysregulation of complement in the vessel walls, particularly in the context of CAA, is likely an important exacerbating factor and trigger for vascular inflammation in VaD. Targeted complement therapies might benefit patients by impacting complement dysregulation.

Frontotemporal dementia (FTD) describes a group of diseases characterized by degeneration of neuronal circuits in frontal and temporal lobes. FTDs are pathologically diverse, presenting with proteinaceous inclusions of tau in FTD-tau, transactive response DNA-binding protein (TDP-43) aggregates in FTD-TDP, or fused in sarcoma (FUS) aggregates in FTD-FUS [104]. FTD is a largely heritable disorder with 10–20% of cases reporting autosomal dominant mutations within microtubule associated protein tau (MAPT), progranulin (GRN) or chromosome 9 open reading frame 72 (C9orf72) [104]. The most common form of sporadic FTD is FTD-tau; this term includes progressive supranuclear palsy (PSP), corticobasal degeneration (CBD), Pick’s disease (PiD), argyrophilic grain disease (AGD), globular glial tauopathy (GGT), age related tau astrogliopathy (ARTAG) and primary age-related tauopathy (PART) [105].

Although GWAS have revealed strong associations with microglia function and inflammatory processes in FTD-tau, a direct genetic association with complement has not been identified [105–107]. The demonstration that Tau aggregates, when tested in vitro, potently and directly activate classical complement pathway in an antibody independent manner [108], together with mounting evidence implicating complement from postmortem and animal model studies, suggests that complement dysregulation contributes to the underlying pathology.

Complement in FTD-tau: IHC evidence

Direct evidence for complement dysregulation in the brain in FTD is scarce. Early IHC analyses in postmortem brain tissue identified complement components and activation products, predominantly from the classical and terminal pathways, localized to reactive astrocytes and neurons containing Pick bodies in cases of PiD [109], and on neurofibrillary tangles in cases of AD with tau pathology [110] and Guam Parkinsonism-dementia [111]. More recent evidence for complement association with tau pathology comes from AD, which is considered a secondary tauopathy, although complement is mainly localized to plaques, not tangles [2], likely because tau accumulation is mainly intracellular only released after cell death. Nevertheless, these findings suggest that tau aggregates can activate complement and downstream events in the brain.

Complement in FTD-tau: biomarker evidence

In contrast to AD where complement biomarkers are well studied, there is a paucity of published studies of complement levels in FTD (Table 2). A recent report from the Genetic Frontotemporal Dementia Initiative (GENFI) used multiplexed assays to measure CSF and plasma levels of complement components and activation products in FTD cases with known genetic mutations in MAPT, C9ORF or GRN at different stages of the disease [112]. C1q and C3b were elevated in CSF during the symptomatic stage compared with presymptomatic carriers and non-carriers. The increased C1q and C3b levels correlated with CSF Nfl levels and decreased brain volume, indicative of cerebral atrophy and neurodegeneration [112]. In the symptomatic stage, C2, C3 and FD also correlated with reduced whole brain volume, further implicating complement in disease progression [112]. In plasma, C2 and C3 were elevated in FTD linked to C9orf72 and MAPT, but not GRN [112].

Complement in FTD-tau: evidence from animal models

Studies in mice harboring the P301S/L tau mutation have informed understanding of complement dysregulation in response to tau pathology [113]. Neuronal accumulation of mutant tau triggered classical complement gene expression by microglia and astrocytes and consequent neurodegeneration [74]; C1q was enriched at the synapse correlating with tau pathology, and provoking the suggestion that C1q-initiated synaptic loss causes neurodegeneration in this model. Indeed, knockout of C1q protected the P301S mice from synapse and neuronal loss and brain atrophy [73,113]. Knockout of C3 or C3 inhibition in brain by overexpression of the C3 inhibitor sCrry conferred similar protection in the mouse model, further implicating complement dysregulation [114]. sCrry over-expression also reduced tau pathology in P301L mice, suggesting that complement dysregulation contributes to tau aggregation [114]. In marked contrast, lentiviral silencing of Crry in P301S mice reduced tau phosphorylation and neuronal loss with improvement in behavioral outcomes [115]; these authors suggested that increased C3 turnover and C3b generation in the absence of Crry enhanced clearance of neurotoxic tau, illustrating the complex, double-edged nature of complement.

Involvement of the terminal pathway in tau pathology was also explored in P301L mice. Knockout of the cell-bound MAC inhibitor CD59 caused increased tau phosphorylation in the mice, likely due to MAC-induced cell activation, accompanied by increased synapse and neuronal loss [114]. C3aR expression on microglia was markedly increased in PS19 mice compared with controls, and C3aR KO reversed vascular abnormalities, reduced gliosis, tau hyperphosphorylation and synapse loss, and protected hippocampal dependent memory and LTP [80,102]. Pharmacologic blockade of C3aR in P301S mice impacted p35/CDK5 signaling in microglia, reduced synapse loss and tau hyperphosphorylation, and improved spatial memory [116]. Contributions of the C5a/C5aR axis were tested using the C5aR antagonist PMX205 in the 3xTG AD model (includes the P301L tau mutation); treatment for 3 months reduced tau hyperphosphorylation and rescued cognitive function compared with controls [117]. These latter studies suggest that drugs targeting C3a/C3aR and/or C5a/C5aR pathways may ameliorate disease in FTD-tau.

Complement in FTD-tau: summary

Despite the scarcity of research on complement in human FTD cases, the studies reported here demonstrate a clear involvement of complement dysregulation. The role of complement in tauopathies resembles that in AD, particularly roles in synapse loss. The impact of complement inhibition in mouse models of FTD-tau provides a strong rationale for further studies to inform future development of complement therapeutics in FTD.

Synucleinopathies comprise a group of NDDs characterized by accumulation of α-synuclein (αSyn), predominantly in the form of Lewy bodies (LB) that accumulate in dopaminergic (DA) neurons and/or glial cells. Synucleinopathies are broadly divided into two disease groups, the LB diseases Parkinson’s disease (PD) and Dementia with LBs (DLB), and the Multiple systems atrophy (MSA) group, itself divided into MSA with predominant cerebellar ataxia and MSA with predominant parkinsonism [118]. Why and how defects in αSyn processing cause different types of aggregates and different pathologies remains unclear. Regardless, αSyn accumulation leads to the progressive loss of neurons in the basal ganglia of patients [119]. Reactive microglia are abundant in areas of neuronal loss and GWAS have implicated inflammation and immunity pathways, implying that neuroinflammation is a core feature [119].

Complement in synucleinopathies: biomarker evidence

A few studies have investigated CSF or blood complement levels in synucleinopathies with varying results (Table 2). C3 levels were consistently decreased compared with controls in both CSF and serum from MSA but not PD [65,66]. Others reported decreased plasma C3 levels [65], or no difference in PD compared with controls [120], while high baseline levels of C3 and C4 predicted worse outcome in a longitudinal biomarker study in PD [120,121]. A proteomic analysis of PD CSF identified the alternative pathway regulator FH as one of the most significant differentially altered proteins [122]. A recent large proteomic study in over 1000 serum samples from PD and healthy controls identified enrichment of complement proteins; C9 was strongly associated with disease status, cognitive decline and motor symptoms implicating the terminal pathway [123]. Analysis of complement activation products is still largely lacking in these studies and would clarify the extent of complement dysregulation in synucleinopathies.

Complement in synucleinopathies: IHC evidence

Several IHC studies of postmortem brain from PD and DLB cases have demonstrated association of complement proteins and activation products with the hallmark lesions. C1q and the activation markers iC3b, C3d and C4d all localized to LBs in dopaminergic neurons in the substantia nigra, but AP-specific markers were absent, implicating classical pathway activation [124–128]. Terminal pathway components C7 and C9 were also localized to the lesions, implying activation through to MAC. Activated microglia were closely opposed to C1q/C4d-positive LB-bearing dopaminergic neurons, suggesting that phagocytic clearance of damaged neurons was underway [124,125]. In support of these findings, the microglial receptors for C3 activation products, CR3 and CR4, were markedly up-regulated in RNAseq analysis of cortex from 29 PD cases [129,130].

Complement in synucleinopathies: evidence from in vitro studies and animal models

αSyn is a 140 amino acid protein with an amphipathic lysine-rich amino terminus involved in interactions with membranes and a carboxy-terminal region implicated in nuclear localization. Direct activation of complement by αSyn, either expressed on a neuronal cell line or as isolated protein, has been demonstrated in vitro. Fibrillar αSyn incubated with serum bound C1 via the carboxy-terminus activated the classical pathway and became coated with C4b [131–133]; microglia selectively bound this opsonized fibrillar αSyn through the phagocytic receptors CR3 and CR4 to mediate clearance.

A few studies have investigated roles of complement in PD caused by exposure to pesticides and toxins that cause dopaminergic neuron loss. In mice, repeated dosing of rotenone caused neuronal loss accompanied by up-regulated microglial CR3 expression; knockout of CR3 prevented the neurodegeneration, suggesting a primary role for complement [134]; however, prevention of complement activation by knocking out C1q or C3 did not reduce neurodegeneration in another toxin-induced PD model, again demonstrating the inconsistencies between studies and models [135,136].

Complement in synucleinopathies: summary

Taken together, these studies implicate complement dysregulation in some types of synucleinopathies; complement is activated by αSyn aggregates via the classical pathway, may facilitate removal of aggregates via opsonization, but also enhances inflammation, a double-edged sword. To date, no trials of anti-complement drugs in this group of diseases have been reported.

Huntington’s disease (HD) is an inherited NDD characterized by a choreatic movement disorder with psychiatric symptoms, progressive neurodegeneration and dementia. The underlying cause is an expansion of a CAG triplet repeat in exon 1 of the HTT gene encoding the ∼350 kDa protein huntingtin. This generates an elongated polyglutamine tract that causes mis-folding, aggregation and accumulation of protein in neurons and other cells in the striatum and cortex leading to neuronal loss, gliosis and brain atrophy [137]. Neuroinflammation is a feature of the disease but has been considered a secondary event and attracted limited attention [138].

Complement in Huntington’s disease: evidence from IHC studies

IHC in postmortem brain tissue from HD cases has demonstrated complement dysregulation specifically in areas of pathology. Neurons, astrocytes and myelin sheaths in the caudate and striatum of HD patients showed staining for the complement proteins C1q, C4 and C3, and the activation products iC3b and TCC, strongly indicating complement dysregulation [139]. Astrocytes and microglia up-regulated C5aR expression in the caudate nucleus of HD patients [140].

Complement in Huntington’s disease: evidence from biomarker studies

There have been very few studies measuring complement proteins in HD blood and/or CSF; plasma proteomic profiling identified increased levels of C7, C9 and clusterin, the latter also increased in CSF [141], although another study failed to replicate these changes in HD plasma, perhaps due to differences in assays or patient cohorts [142]. CSF levels of C1q and C4b were strongly associated with disease severity and predictive of rate of progression in premanifest HD mutation carriers [143]. No studies of complement activation products in HD are reported.

Complement in Huntington’s disease: evidence from in vitro studies and animal models

Studies of complement in mouse HD models have predominantly focused on the R6/2 model, expressing human HTT exon 1 with 150 CAG repeats under the human huntingtin promoter [144]. R6/2 transgenic mice display progressive neurological and behavioral features resembling HD. Complement proteins were not up-regulated in the mice and crossing to C3 deficiency had no impact on HD disease progression [145]. In a chemically induced rat model of HD (3-nitropropionic acid, 3-NP) C3, C9 and C5a receptor (C5aR1 and C5aR2) expression was increased in striatal neurons in regions of pathology [146]. Administration of C5aR antagonists (PMX53 and PMX205) either pre- or post-3-NP induction reduced HD phenotypes; including weight loss and motor deficits such as ataxia and dystonia [146], suggesting a role for complement-driven inflammation in the disease process.

Complement in Huntington’s disease: summary

There is limited but growing evidence that complement is dysregulated in HD while animal models implicate inflammation and complement in pathogenesis. The limited data have provoked testing of anti-complement therapeutics in AD. Annexon has completed a Phase 2 study of their anti-C1q mAb in HD with promising results in a subset of patients displaying evidence of complement dysregulation [147]; expansion to Phase 3 is planned in 2024.

Multiple sclerosis (MS) is a complex neuroinflammatory disease characterized by demyelination and neurodegeneration. While the primary cause of MS remains unknown, neuroinflammation plays key roles in demyelination, neurodegeneration, and disease progression [148–159]. Originally considered a disease of white matter (WM), it is now clear that demyelinating lesions are found globally in the MS brain. Current therapies target adaptive immunity and are effective in many, but not all cases.

Complement in MS: biomarker evidence

There have been numerous studies measuring complement biomarkers in MS plasma and/or CSF (Table 2); however, reported results are variable and contradictory, likely because of a lack of standardized assays. Significant differences have been reported for plasma levels of components (C1s, MASP-2, C3, C4 and C9), regulators (FH and C1inh) and activation products (iC3b, Bb, C4a, C4b and TCC) between MS patients and controls and between MS patients in remission, relapse or with progressive disease [150,160–164]. A recent study reported increased CSF levels of C1q and C3a in newly diagnosed patients with clinically isolated syndrome and RRMS compared with healthy controls, which correlated with several inflammatory chemokines, implicating complement dysregulation in early disease [164–171].

Complement in MS: IHC evidence

MS is pathologically heterogeneous. Lassmann and co-workers classified cases based on pathology into four groups (I–IV) and showed that complement deposition in demyelinating lesions was a defining feature in many cases, including most of those with primary progressive MS (ppMS) [172,173]. Complement components, regulators and activation products all localize to lesions; the presence of the latter, C3 fragments and terminal pathway complexes, confirm complement dysregulation in the lesions [148,158]. In myelin oligodendrocyte glycoprotein (MOG) antibody associated disorders (MOGAD), an MS-related condition defined by the presence of anti-MOG antibodies, all active demyelinating plaques were strongly positive for complement activation products [174].

Complement activation products have also been demonstrated in the leptomeninges associated with underlying cortical demyelination, inflammation, and microglial activation in MS brain [159]. Recently, complement activation products were found in abundance in thalamic grey matter lesion in ppMS, implicating complement as a driver of grey matter pathology [156]. Taken together, the IHC data suggest an ‘outside-in’ gradient of complement dysregulation that causes further barrier disruption and increased pathology, reviewed elsewhere [175]. Overall, studies in human postmortem brain demonstrate that complement is dysregulated in a large proportion of MS cases, including most of those with ppMS.

Complement in MS: evidence from in vitro and animal models

Experimental autoimmune encephalomyelitis (EAE) is the benchmark mouse model of MS; roles of complement have been explored in the model either by testing the effect of complement gene knockouts or using anti-complement drugs. Systemic delivery of the C3 blocker sCR1 inhibited demyelination and ameliorated paralysis and weight loss in acute EAE [176]; however, intraparenchymal delivery of another C3 inhibitor, Crry, decreased synaptic loss but did not impact demyelination [177]. In knockout mice, C3 KO was protective against microglial activation and synapse loss in the hippocampus and reduced the severity of motor deficits; in contrast, C1qa KO showed no significant effect [178]. The terminal pathway was implicated in demyelination in EAE in two studies; deficiency of C6 in rats protected against demyelination and paralysis whereas knockout of the terminal pathway inhibitor CD59 in mice exacerbated demyelination and symptoms [179,180].

Complement in MS: summary

There is overwhelming evidence from EAE and human studies demonstrating the utility of complement as a biomarker of disease severity, to distinguish between the different MS subtypes, and as a marker of pathology. Evidence of complement involvement in the pathogenesis of MS, at least in some subtypes of the disease, continues to emerge as more studies implicate locally generated and circulating complement and demonstrate complement dysregulation centrally and peripherally. The evidence from EAE models supports a critical role for complement in demyelination, likely involving both opsonization of myelin to facilitate phagocytic removal and terminal pathway-mediated damage impacting myelin and axons. Although more research is needed to understand the role of complement dysregulation in MS, its impact at different stages of the disease and precisely which complement effectors are responsible for myelin loss, axonal injury, synapse loss and neuronal damage, trials of anti-complement drugs in those with proven complement dysregulation are long overdue.

Schizophrenia is a chronic and debilitating neuropsychiatric disorder that affects approximately 0.7% of the population [181]. Symptoms include delusions, hallucinations, disorganized speech and negative symptoms (e.g., anhedonia); the disorder is associated with an increased rate of mortality especially due to suicide and smoking-related diseases [182,183]. Postmortem studies reveal reduced cortical synaptic density, cortical thickness, and grey matter volume, consistent with neurodegeneration [184–187], supporting suggestions that defective cortical synaptic pruning underlies the condition [188]. In the normal developing brain, an excess of synaptic connections are generated that are removed during maturation of neural circuits; in schizophrenia this latter process is impaired [189,190]. Complement is established as a driver of developmental synaptic pruning [31,35] and evidence implicating complement dysregulation in the pathogenesis of schizophrenia is accumulating.

Complement in schizophrenia: genetic evidence

GWAS have shown that schizophrenia is highly polygenic; indeed, twin studies show that ∼80% of variance in schizophrenia susceptibility is inherited [191–193]. The first evidence of a link to complement came from the demonstration that the major GWAS association in the major histocompatibility complex (MHC) on chromosome 6 was in the C4 locus [38,193–195]. This locus comprises two alleles, C4A and C4B, encoding C4A and C4B respectively; these isoforms differ in just a few amino acids but are functionally distinct, the C4A thioester preferentially binding amino groups while C4B thioester preferentially binds hydroxyl groups [196,197]. C4 copy number varies and schizophrenia risk is increased in proportion to both the number of C4A alleles present and the expression level of C4A in the brain, leading to the suggestion that C4A more efficiently labels synapses to drive increased complement-mediated synaptic pruning [38]. More recent GWAS have identified additional risk-associated loci including CUB and sushi multiple domains 1 (CSMD1) [198–200]. CSMD1 encodes a large transmembrane receptor predominately expressed in brain and reproductive tissues, reported to be a complement inhibitor [201–204]. Precisely how CSMD1 variants predispose to schizophrenia is unknown. Reduced PBMC expression of CSMD1 in people with schizophrenia was reported in a small Han Chinese cohort [205]. Another study replicated the finding of reduced CSMD1 expression in PBMC and correlated this with reduced CSMD1 levels in plasma [206]; however, as CSMD1 has no reported secreted form, the source of CSMD1 in plasma is unclear. Recently, reduced CSMD1 expression was also reported in first episode psychosis PBMCs [207].

SNPs at the CSMD1 locus have been linked to several other neurological disorders, including Parkinson’s disease [208], bipolar disorder [209], multiple sclerosis [210,211], autism and learning disorders [212,213], implying that CSMD1 has a broad role in brain development and function, perhaps linked to synaptic loss, a common feature of these disorders. CSMD1 expressed at the synapse may regulate complement activation and synaptic opsonization locally, restricting microglial elimination [31,35,38].

Complement in schizophrenia: evidence from animal models

Several mouse studies have explored the impact of C4 on schizophrenia, although they are complicated by the fact that mice only have one C4 gene; mice humanised to express C4A and C4B have been made and used to show that C4A expression caused excessive synaptic pruning in the developing and adult mouse brain compared with C4B [214]. Others reported that C4A overexpression in mice induced enhanced microglial synaptic removal and schizophrenia-like behaviors [214,215].

Complement in schizophrenia: evidence from biomarker studies

Complement component levels and mRNA expression in plasma and brain have been repeatedly investigated as candidate biomarkers of schizophrenia and the results thoroughly reviewed elsewhere [216]. Results are inconsistent although multivariate models including multiple complement analytes measured in plasma show some promise [217]. A recent proteomic study reported elevated plasma C4A levels in schizophrenia cases, linking with the genetics and implying that measurement of the C4A isotype may provide a more suitable biomarker than measurement of total C4 [218].

Complement in schizophrenia: summary

The critical role of complement in developmental synaptic pruning and the strong genetic evidence implicating complement proteins and regulators together provide significant evidence implicating complement dysregulation in schizophrenia. Further work to better define roles of complement in synaptic pruning and to select the best markers of complement dysregulation, particularly in the early stages of the disease, are needed to identify whether and when complement dysregulation occurs in schizophrenia.

The sections above implicate complement dysregulation in diverse NDDs. Numerous studies using anti-complement drugs in animal models of NDDs have provided proof of concept for complement inhibition, summarized in Table 3. Here we will briefly discuss the evolution of anti-complement drug therapy of NDDs. Several of the existing toolbox of anti-complement drugs are already in the clinic or in clinical trials in a handful of NDDs. New agents in development will broaden the utility of this approach to other NDDs.

Table 3
Summary of complement drugs used in NDDs including preclinical and clinical studies
DiseaseDrug nameTypeTargetStageCompany
NMO Eculizumab mAb C5 cleavage In clinic 2019 Alexion/AZ 
 Ravulizumab mAb C5 cleavage Approved in EU; awaiting final FDA approval Alexion/AZ 
 Zilucoplan Cyclic peptide C5 cleavage Pre-trial UCB 
ALS Pegcetacoplan Linear peptide C3 cleavage Phase II trial discontinued (NCT04579666Annexon 
 ANX-005 mAb C1q blocker Phase II trial (NCT04569435Apellis 
 Ravulizumab mAb C5 cleavage Phase III failed 2021 (NCT04248465Alexion/AZ 
 Zilucoplan Cyclic peptide C5 cleavage Phase II/III discontinued (NCT04436497UCB 
 CP010 (anti-C6) mAb C6 blocker Preclinical Complement Pharma 
HD ANX-005 mAb C1q blocker Phase II trial (NCT04514367Annexon 
MS None; No tested anti-complement drugs Many tested in models n/a All preclinical n/a 
AD None; No tested anti-complement drugs Some data from models n/a All preclinical n/a 
PD None; No tested anti-complement drugs Minimal model data n/a All preclinical n/a 
Schz None; No tested anti-complement drugs Minimal model data n/a All preclinical n/a 
Tauopathies None; No tested anti-complement drugs Minimal model data n/a All preclinical n/a 
DiseaseDrug nameTypeTargetStageCompany
NMO Eculizumab mAb C5 cleavage In clinic 2019 Alexion/AZ 
 Ravulizumab mAb C5 cleavage Approved in EU; awaiting final FDA approval Alexion/AZ 
 Zilucoplan Cyclic peptide C5 cleavage Pre-trial UCB 
ALS Pegcetacoplan Linear peptide C3 cleavage Phase II trial discontinued (NCT04579666Annexon 
 ANX-005 mAb C1q blocker Phase II trial (NCT04569435Apellis 
 Ravulizumab mAb C5 cleavage Phase III failed 2021 (NCT04248465Alexion/AZ 
 Zilucoplan Cyclic peptide C5 cleavage Phase II/III discontinued (NCT04436497UCB 
 CP010 (anti-C6) mAb C6 blocker Preclinical Complement Pharma 
HD ANX-005 mAb C1q blocker Phase II trial (NCT04514367Annexon 
MS None; No tested anti-complement drugs Many tested in models n/a All preclinical n/a 
AD None; No tested anti-complement drugs Some data from models n/a All preclinical n/a 
PD None; No tested anti-complement drugs Minimal model data n/a All preclinical n/a 
Schz None; No tested anti-complement drugs Minimal model data n/a All preclinical n/a 
Tauopathies None; No tested anti-complement drugs Minimal model data n/a All preclinical n/a 

Abbreviations: AD, Alzheimer’s disease; ALS, Amyotrophic lateral sclerosis; HD, Huntington’s disease; mAb, monoclonal antibodies; MS, multiple sclerosis; NMO, neuromyelitis optica; PD, Parkinson’s disease; Schz, Schizophrenia.

Complement therapeutics: current anti-complement drugs for brain diseases

Several of the current crop of complement inhibitors, developed to treat complement dysregulation in peripheral diseases, are being evaluated for treatment of NDDs (Table 3). The anti-C5 (mAb) eculizumab, already in the clinic for more than 16 years for the treatment of rare systemic complement dysregulation diseases, was FDA approved for the treatment of the neuromuscular conduction disorder myasthenia gravis (MG) in 2017 and for the demyelinating disease NMO spectrum disorder (NMOSD) in 2019 [219,220]. These successes raise the prospect of using complement inhibitors to treat NDDs currently lacking effective treatments.

Amyotrophic lateral sclerosis (ALS) is a progressive NDD characterized by loss of motor neurones in the brain and spinal cord. Blood–spinal cord barrier (BSCB) and BBB degradation occurs early in ALS, provoking the testing of non-brain penetrant anti-complement drugs, including the C3-blocking linear peptide pegcetacoplan (Apellis) [221], C5-blocking peptide zilucoplan and the anti-C5 mAb ravulizumab [222,223]. Disappointingly, none of these continued beyond Phase II and trials were discontinued [221]. The anti-C1q mAb ANX-005 (Annexon) is currently in Phase IIb for ALS, was well tolerated and the trial should report in 2024 [147].

NMOSD is a demyelinating disease specifically targeting optic tracts and spinal cord; here too the BBB is disrupted early in the disease process permitting access of drugs from the periphery. The C5-blocking mAb eculizumab was FDA approved for NMOSD in 2019 and has become the standard for therapy [162,221,224]. The next-generation, extended half-life version of eculizumab, ravulizumab is also now in the clinic for NMOSD.

Complement therapeutics in AD

To date, only a handful of drugs are approved for treatment of AD; most of these provide only symptomatic relief and have no impact on disease course [225–229]. In the quest for disease-modifying drugs, most attention has focussed on removing amyloid, culminating in the recent FDA approval of the anti-Aβ antibodies aducanumab and lecanemab despite limited efficacy [230–233]. Lecanemab effectively eliminated amyloid (68% clearance when compared with placebo) but its impact on cognitive decline was modest and side effects were common [234]. The modest effect on cognition despite efficient amyloid clearance reveals a disconnect between amyloid and disease, suggesting the need for alternative treatment strategies. Inhibiting neuroinflammation provides an alternative approach to therapy of AD and complement is an obvious target. Complement inhibition has proved effective in several mouse AD models: inhibition of MAC assembly reduced synapse loss [31,78,108,235]; blocking C5aR1 with the antagonist PMX53 prevented C5a-mediated neuronal death [236,237] and reduced amyloid, microglial activation and inflammation in Tg2576 and 3xTg AD mice [84,117]; C5aR1-antagonist EP67 reduced amyloid load and synapse loss in 5xFAD mice [238].

Despite the above evidence, it has been difficult to convince Pharma that anti-complement drugs should be trialled in AD. Anti-complement drugs have been in the clinic for systemic diseases for more than 15 years; the anti-C5 monoclonal antibody (mAb) Eculizumab is approved for therapy of two neurological conditions, MG and NMOSD [224]. Modifying and repurposing current anti-complement drugs for therapy of AD should be prioritized.

Complement therapeutics in MS

Current MS treatments use mAbs to target B cells expressing the cell-specific marker CD20; these are effective in most MS subtypes, with the exception of ppMS which responds poorly or not at all to these biological therapies [175]. Despite the evidence above implicating complement in MS and the demonstration that systemic complement blockade is effective in animal models [219,239], no attempts have been made to use currently available complement-blocking drugs for treatment of MS [221]. This is particularly surprising given that anti-complement therapy has proven highly effective in NMOSD, a disease that, although localized to optic nerve and spinal cord, resembles MS in many ways. Eculizumab, the C5 blocking mAb was FDA-approved for NMOSD in 2019 and has become the gold standard [162,221,224]. Trials in MS sub-groups selected based on failure to respond to existing drugs and evidence of complement dysregulation, are overdue.

Complement in therapeutics schizophrenia

Schizophrenia symptoms are managed with antipsychotics but responses are variable and often poor. Whether targeting complement and inflammation can be beneficial remains to be tested. Evidence from mouse models is sparse, in part due to the lack of good models, and precisely how complement contributes to the disease is unclear. Nevertheless, the above evidence implicating complement as a pathological factor raises the possibility that complement-blocking drugs may ameliorate the course of the illness in some patients.

Complement therapeutics for NDDs: optimizing the drugs and selecting the patients

In most NDDs the BBB is intact, at least in early disease, preventing access of drugs from the periphery; for example, access for systemically delivered mAb or other large drugs is minimal (<0.1% of peripheral levels). Efficacy of systemically delivered drugs in ALS and NMOSD are the exceptions to this rule because the BBB is impaired early. In ALS patients, BBB dysfunction is evidenced by infiltration of antibodies and other large proteins (e.g., thrombin, plasminogen and hemoglobin), leading to pericyte degeneration and tight junction dysfunction. Albumin (∼67 kDa), a protein typically excluded by the intact BBB, has been observed in the spinal cord and brain tissues of MND patients and is used as a marker of BBB leakage [240–242]. CSF biomarkers for MND include immunoglobulins and albumin, as well as specific markers like GFAP, NSE, and S100 beta proteins, demonstrating an impairment of BBB integrity [243]. For other NDDs, the BBB represents a considerable hurdle for drugs and none of the complement blocking drugs in use today cross the BBB. Current efforts to design and develop BBB-penetrant anti-complement drugs for the early treatment of diverse NDDs are extensively reviewed elsewhere [221].

The second issue to be resolved is patient selection. The different NDDs are all heterogenous conditions and selecting those diseases and cases where complement dysregulation is a driver is key to success in clinical trials and subsequent therapeutic use. Patient stratification and selection is thus a core consideration when designing a clinical study, choosing those with evidence of complement dysregulation for inclusion. For instance, in the trial of the anti-C1q mAb ANX-005 in HD, retrospective analysis showed that a subset of patients with high C4/C4a ratios (indicative of classical pathway activation) responded best to the drug (75% vs. 36% response) [244]. It is thus essential that patients are selected and monitored using tests to measure markers of complement dysregulation in blood and brain, something that has been lacking in trials to date.

In the recent clinical trials (NCT04569435), ANX-005 reduced neurotoxicity and preserve synapses, was well-tolerated in both healthy individuals and the patient populations, with some patients undergoing treatment for up to a year, and robust target engagement was demonstrated in blood, brain and eye [245,246]. To date a handful of other anti-complement drugs, including C3 blockers and anti-C5 agents, have been tested in ALS with disappointing outcomes. Carefully designed clinical trials are required to identify responder subgroups with pronounced complement dysregulation. Measurement of plasma and CSF complement activation products (e.g., TCC, Ba, C3 or C4 fragments, C5a) show promise and may improve the efficiency of future clinical trials. The ongoing clinical trials of anti-complement drugs in NDDs, most in the early phases, are summarized in Table 3. Preliminary results from some trials have shown encouraging outcomes, supporting the potential therapeutic value of complement-targeted drugs. A major challenge that needs to be addressed for the effective targeting of complement in NDDs is the lack of understanding of underpinning mechanisms: When and how is complement dysregulation initiated? Which pathways are involved? Where is complement coming from? What are the main targets of complement attack? Addressing these questions will enable proper targeting of the relevant pathways at the right time in the disease process as new brain-penetrant complement-blocking drugs emerge for therapy of NDDs.

This is a review article with no primary data so not applicable.

The authors confirm that there are no competing interests associated with the manuscript.

This work is supported by the UK Dementia Research Institute through UK DRI Ltd, principally funded by the Medical Research Council.

Open access for this article was enabled by the participation of Cardiff University in an all-inclusive Read & Publish agreement with Portland Press and the Biochemical Society under a transformative agreement with JISC.

Jacqui Nimmo: Writing—original draft, Project administration, Writing—review & editing. Robert Byrne: Writing—original draft, Writing—review & editing. Nikoleta Daskoulidou: Writing—original draft, Writing—review & editing. Lewis Watkins: Writing—original draft, Writing—review & editing. Sarah Carpanini: Supervision, Writing—original draft, Project administration, Writing—review & editing. Wioleta Zelek: Supervision, Writing—original draft, Project administration, Writing—review & editing. Bryan Paul Morgan: Supervision, Writing—original draft, Project administration, Writing—review & editing.

αSyn

Alpha synuclein

AD

Alzheimer’s disease

AGD

argyrophilic grain disease

ALS

amyotrophic lateral sclerosis

APOE

apolipoprotein E

APP

amyloid precursor protein

BBB

blood–brain barrier

BSCB

blood–spinal cord barrier

C1-INH

C1 inhibitor

C1qBP

C1q-binding protein

C9orf72

chromosome 9 open reading frame 72

CAA

cerebral amyloid angiopathy

CBD

corticobasal degeneration

CR1

complement receptor 1

CR3

complement receptor 3

CSF

cerebrospinal fluid

CSMD1

CUB and sushi multiple domains 1

DA

dopaminergic

DAF

decay accelerating factor

DLB

dementia with Lewy bodies

DS

Down’s syndrome

FB

factor B

Fcn

ficolin

FH

factor H

FTD

frontotemporal dementia

FUS

fused in sarcoma

GGT

globular glial tauopathy

GRN

progranulin

GWAS

Genome Wide Association Study

HD

Huntington’s disease

ICH

intracerebral haemorrhage

IHC

immunohistochemistry

IPAD

intramural periarterial drainage

iPSC

induced pluripotent stem cell

KO

knockout

LOAD

late onset AD

LPS

lipopolysaccharide

mAb

monoclonal antibody

MAC

membrane attack complex

MAPT

microtubule-associated protein tau

MASP

Mannan-binding lectin-associated serine protease

MBL

Mannan-binding lectin

MCI

mild cognitive impairment

MCP

membrane cofactor protein

MG

Myasthenia Gravis

MHC

major histocompatibility complex

MOG

myelin oligodendrocyte glycoprotein

MOGAD

MOG-associated disorder

MS

multiple sclerosis

MSA

multiple system atrophy

NDD

neurodegenerative disease

NFL

neurofilament light chain

NMOSD

neuromyelitis optica spectrum disorder

NMO

neuromyelitis optica

PBMC

peripgeral blood mononuclear cell

PD

Parkinson’s disease

PiD

Pick’s disease

ppMS

primary progressive MS

PSP

progressive supranuclear palsy

SVD

small vessel disease

TCC

terminal complement complex

TDP-43

transactive response DNA-binding protein

VaD

vascular dementia

VTR

vitronectin

WM

white matter

1.
Heidelberger
M.
and
Muller
R.H.
(
1949
)
Complement and its components in human cerebrospinal fluid
.
J. Clin. Invest.
28
,
282
285
[PubMed]
2.
Ishii
T.
and
Haga
S.
(
1984
)
Immuno-electron-microscopic localization of complements in amyloid fibrils of senile plaques
.
Acta Neuropathol.
63
,
296
300
[PubMed]
3.
Eikelenboom
P.
and
Stam
F.C.
(
1982
)
Immunoglobulins and complement factors in senile plaques. An immunoperoxidase study
.
Acta Neuropathol.
57
,
239
242
[PubMed]
4.
Fatoba
O.
,
Itokazu
T.
and
Yamashita
T.
(
2022
)
Complement cascade functions during brain development and neurodegeneration
.
FEBS J.
289
,
2085
2109
[PubMed]
5.
Miller
S.J.
(
2018
)
Astrocyte heterogeneity in the adult central nervous system
.
Front Cell Neurosci.
12
,
401
[PubMed]
6.
Gasque
P.
,
Chan
P.
,
Fontaine
M.
,
Ischenko
A.
,
Lamacz
M.
,
Gotze
O.
et al.
(
1995
)
Identification and characterization of the complement C5a anaphylatoxin receptor on human astrocytes
.
J. Immunol.
155
,
4882
4889
[PubMed]
7.
Gasque
P.
,
Fontaine
M.
and
Morgan
B.P.
(
1995
)
Complement expression in human brain. Biosynthesis of terminal pathway components and regulators in human glial cells and cell lines
.
J. Immunol.
154
,
4726
4733
[PubMed]
8.
Ischenko
A.
,
Sayah
S.
,
Patte
C.
,
Andreev
S.
,
Gasque
P.
,
Schouft
M.T.
et al.
(
1998
)
Expression of a functional anaphylatoxin C3a receptor by astrocytes
.
J. Neurochem.
71
,
2487
2496
[PubMed]
9.
Singhrao
S.K.
,
Neal
J.W.
,
Rushmere
N.K.
,
Morgan
B.P.
and
Gasque
P.
(
2000
)
Spontaneous classical pathway activation and deficiency of membrane regulators render human neurons susceptible to complement lysis
.
Am. J. Pathol.
157
,
905
918
[PubMed]
10.
Pekna
M.
and
Pekny
M.
(
2021
)
The complement system: a powerful modulator and effector of astrocyte function in the healthy and diseased central nervous system
.
Cells
10
,
1812
11.
Fonseca
M.I.
,
Chu
S.H.
,
Hernandez
M.X.
,
Fang
M.J.
,
Modarresi
L.
,
Selvan
P.
et al.
(
2017
)
Cell-specific deletion of C1qa identifies microglia as the dominant source of C1q in mouse brain
.
J. Neuroinflammation
14
,
48
[PubMed]
12.
Walker
D.G.
,
Kim
S.U.
and
McGeer
P.L.
(
1995
)
Complement and cytokine gene expression in cultured microglial derived from postmortem human brains
.
J. Neurosci. Res.
40
,
478
493
[PubMed]
13.
Abud
E.M.
,
Ramirez
R.N.
,
Martinez
E.S.
,
Healy
L.M.
,
Nguyen
C.H.H.
,
Newman
S.A.
et al.
(
2017
)
iPSC-derived human microglia-like cells to study neurological diseases
.
Neuron
94
,
278e9
293e9
[PubMed]
14.
Daskoulidou
N.
,
Shaw
B.
,
Torvell
M.
,
Watkins
L.
,
Cope
E.L.
,
Carpanini
S.M.
et al.
(
2023
)
Complement receptor 1 is expressed on brain cells and in the human brain
.
Glia
71
,
1522
1535
[PubMed]
15.
Marques
E.T.A.
,
Demers
M.
,
D'Aiuto
L.
,
Castanha
P.M.S.
,
Yeung
J.
,
Wood
J.A.
et al.
(
2023
)
Herpesvirus infections in the human brain: a neural cell model of the complement system derived from induced pluripotent stem cells
.
Curr. Top. Behav. Neurosci.
61
,
243
264
[PubMed]
16.
Agoropoulou
C.
,
Wing
M.G.
and
Wood
A.
(
1996
)
CD59 expression and complement susceptibility of human neuronal cell line (NTera2)
.
Neuroreport
7
,
997
1004
[PubMed]
17.
Thomas
A.
,
Gasque
P.
,
Vaudry
D.
,
Gonzalez
B.
and
Fontaine
M.
(
2000
)
Expression of a complete and functional complement system by human neuronal cells in vitro
.
Int. Immunol.
12
,
1015
1023
[PubMed]
18.
Shen
Y.
,
Sullivan
T.
,
Lee
C.
,
Meri
S.
,
Shiosaki
K.
and
Lin
C.
(
1998
)
Induced expression of neuronal membrane attack complex and cell death by Alzheimer's beta-amyloid peptide
.
Brain Res.
796
,
187
197
19.
Lin
M.
,
Pedrosa
E.
,
Shah
A.
,
Hrabovsky
A.
,
Maqbool
S.
,
Zheng
D.
et al.
(
2011
)
RNA-Seq of human neurons derived from iPS cells reveals candidate long non-coding RNAs involved in neurogenesis and neuropsychiatric disorders
.
PLoS ONE
6
,
20.
Hosokawa
M.
,
Klegeris
A.
,
Maguire
J.
and
McGeer
P.L.
(
2003
)
Expression of complement messenger RNAs and proteins by human oligodendroglial cells
.
Glia
42
,
417
423
[PubMed]
21.
Gasque
P.
and
Morgan
B.P.
(
1996
)
Complement regulatory protein expression by a human oligodendrocyte cell line: cytokine regulation and comparison with astrocytes
.
Immunology
89
,
338
347
[PubMed]
22.
(
2023
)
Science AIfB: Seattle Alzheimer's Disease Brain Cell Atlas
.
23.
(
2023
)
Science AIfB: Accelerating progress toward understanding the brain
.
https://portal.brain-map.org/ Accessed 21/09/2023
24.
Gabay
C.
and
Kushner
I.
(
1999
)
Acute-phase proteins and other systemic responses to inflammation
.
N. Engl. J. Med.
340
,
448
454
[PubMed]
25.
Wu
X.
,
Gao
Y.
,
Shi
C.
,
Tong
J.
,
Ma
D.
,
Shen
J.
et al.
(
2023
)
Complement C1q drives microglia-dependent synaptic loss and cognitive impairments in a mouse model of lipopolysaccharide-induced neuroinflammation
.
Neuropharmacology
237
,
109646
[PubMed]
26.
Zhou
R.
,
Chen
S.H.
,
Zhao
Z.
,
Tu
D.
,
Song
S.
,
Wang
Y.
et al.
(
2023
)
Complement C3 Enhances LPS-Elicited Neuroinflammation and Neurodegeneration Via the Mac1/NOX2 Pathway
.
Mol. Neurobiol.
,
60
5167
5183
27.
Veerhuis
R.
,
Nielsen
H.
and
Tenner
A.
(
2011
)
Complement in the brain
.
Mol. Immunol.
48
,
1592
1603
[PubMed]
28.
Schartz
N.
and
Tenner
A.
(
2020
)
The good, the bad, and the opportunities of the complement system in neurodegenerative disease
.
J. Neuroinflammation
17
,
354
[PubMed]
29.
Thielens
N.
,
Tedesco
F.
,
Bohlson
S.
,
Gaboriaud
C.
and
Tenner
A.
(
2017
)
C1q: A fresh look upon an old molecule
.
Mol. Immunol.
89
,
73
83
[PubMed]
30.
Lu
J.
,
Teh
B.
,
Wang
L.
,
Wang
Y.
,
Tan
Y.
,
Lai
M.
et al.
(
2008
)
The classical and regulatory functions of C1q in immunity and autoimmunity
.
Cell. Mol. Immunol.
5
,
9
21
31.
Stevens
B.
,
Allen
N.
,
Vazquez
L.
,
Howell
G.
,
Christopherson
K.
,
Nouri
N.
et al.
(
2007
)
The classical complement cascade mediates CNS synapse elimination
.
Cell
131
,
1164
1178
[PubMed]
32.
Wang
C.
,
Yue
H.
,
Hu
Z.
,
Shen
Y.
,
Ma
J.
,
Li
J.
et al.
(
2020
)
Microglia mediate forgetting via complement-dependent synaptic elimination
.
Science (New York, NY)
367
,
688
694
33.
Bohlson
S.
and
Tenner
A.
(
2023
)
Complement in the brain: contributions to neuroprotection, neuronal plasticity, and neuroinflammation
.
Annu. Rev. Immunol.
41
,
431
452
[PubMed]
34.
Alexander
J.
(
2018
)
Blood-brain barrier (BBB) and the complement landscape
.
Mol. Immunol.
102
,
26
31
[PubMed]
35.
Schafer
D.P.
,
Lehrman
E.K.
,
Kautzman
A.G.
,
Koyama
R.
,
Mardinly
A.R.
,
Yamasaki
R.
et al.
(
2012
)
Microglia sculpt postnatal neural circuits in an activity and complement-dependent manner
.
Neuron
74
,
691
705
[PubMed]
36.
Chu
Y.
,
Jin
X.
,
Parada
I.
,
Pesic
A.
,
Stevens
B.
,
Barres
B.
et al.
(
2010
)
Enhanced synaptic connectivity and epilepsy in C1q knockout mice
.
Proc. Natl. Acad. Sci. U.S.A.
107
,
7975
7980
[PubMed]
37.
Shi
Q.
,
Colodner
K.J.
,
Matousek
S.B.
,
Merry
K.
,
Hong
S.
,
Kenison
J.E.
et al.
(
2015
)
Complement C3-deficient mice fail to display age-related hippocampal decline
.
J. Neurosci.
35
,
13029
13042
[PubMed]
38.
Sekar
A.
,
Bialas
A.R.
,
de Rivera
H.
,
Davis
A.
,
Hammond
T.R.
,
Kamitaki
N.
et al.
(
2016
)
Schizophrenia risk from complex variation of complement component 4
.
Nature
530
,
177
183
[PubMed]
39.
Gauthier
S.
,
Webster
C.
,
Servaes
S.
,
Morais
J.A.
and
Rosa-Neto
P.
(
2022
)
World Alzheimer Report 2022: Life after diagnosis: Navigating treatment, care and support
,
Alzheimer's Disease International
,
London, England
40.
Li
X.
,
Feng
X.
,
Sun
X.
,
Hou
N.
,
Han
F.
and
Liu
Y.
(
2022
)
Global, regional, and national burden of Alzheimer’s disease and other dementias, 1990-2019
.
Front. Aging Neurosci.
14
, ​​​​​​
937486
41.
Kinney
J.W.
,
Bemiller
S.M.
,
Murtishaw
A.S.
,
Leisgang
A.M.
,
Salazar
A.M.
and
Lamb
B.T.
(
2018
)
Inflammation as a central mechanism in Alzheimer’s disease
.
Alzheimers Dement (N Y)
4
,
575
590
[PubMed]
42.
Morgan
B.P.
(
2018
)
Complement in the pathogenesis of Alzheimer’'s disease
.
Seminars Immunopathol.
40
,
113
124
[PubMed]
43.
Jones
L.
,
Holmans
P.A.
,
Hamshere
M.L.
,
Harold
D.
,
Moskvina
V.
,
Ivanov
D.
et al.
(
2010
)
Genetic evidence implicates the immune system and cholesterol metabolism in the aetiology of Alzheimer’s disease
.
PLoS ONE
5
,
e13950
[PubMed]
44.
Kunkle
B.W.
,
Grenier-Boley
B.
,
Sims
R.
,
Bis
J.C.
,
Damotte
V.
,
Naj
A.C.
et al.
(
2019
)
Genetic meta-analysis of diagnosed Alzheimer’s disease identifies new risk loci and implicates Aβ, tau, immunity and lipid processing
.
Nat. Genet.
51
,
414
430
[PubMed]
45.
Torvell
M.
,
Carpanini
S.M.
,
Daskoulidou
N.
,
Byrne
R.A.J.
,
Sims
R.
and
Morgan
B.P.
(
2021
)
Genetic insights into the impact of complement in Alzheimer's disease
.
Genes
12
,
1990
46.
Rogers
J.
,
Cooper
N.R.
,
Webster
S.
,
Schultz
J.
,
McGeer
P.L.
,
Styren
S.D.
et al.
(
1992
)
Complement activation by beta-amyloid in Alzheimer disease
.
Proc. Natl. Acad. Sci. U.S.A.
89
,
10016
10020
[PubMed]
47.
Veerhuis
R.
,
van der Valk
P.
,
Janssen
I.
,
Zhan
S.S.
,
Van Nostrand
W.E.
and
Eikelenboom
P.
(
1995
)
Complement activation in amyloid plaques in Alzheimer's disease brains does not proceed further than C3
.
Virchows Archiv: Int. J. Pathol.
426
,
603
610
48.
Webster
S.
,
Lue
L.F.
,
Brachova
L.
,
Tenner
A.J.
,
McGeer
P.L.
,
Terai
K.
et al.
(
1997
)
Molecular and cellular characterization of the membrane attack complex, C5b-9, in Alzheimer's disease
.
Neurobiol. Aging
18
,
415
421
[PubMed]
49.
Fonseca
M.I.
,
McGuire
S.O.
,
Counts
S.E.
and
Tenner
A.J.
(
2013
)
Complement activation fragment C5a receptors, CD88 and C5L2, are associated with neurofibrillary pathology
.
J Neuroinflammation
10
,
25
[PubMed]
50.
Bellenguez
C.
,
Küçükali
F.
,
Jansen
I.E.
,
Kleineidam
L.
,
Moreno-Grau
S.
,
Amin
N.
et al.
(
2022
)
New insights into the genetic etiology of Alzheimer's disease and related dementias
.
Nat. Genet.
54
,
412
436
[PubMed]
51.
Lambert
J.C.
,
Heath
S.
,
Even
G.
,
Campion
D.
,
Sleegers
K.
,
Hiltunen
M.
et al.
(
2009
)
Genome-wide association study identifies variants at CLU and CR1 associated with Alzheimer's disease
.
Nat. Genet.
41
,
1094
1099
[PubMed]
52.
Carpanini
S.M.
,
Harwood
J.C.
,
Baker
E.
,
Torvell
M.
,
The Gerad C, Sims R
et al.
(
2021
)
The impact of complement genes on the risk of late-onset Alzheimer's disease
.
Genes
12
,
443
53.
Harold
D.
,
Abraham
R.
,
Hollingworth
P.
,
Sims
R.
,
Gerrish
A.
,
Hamshere
M.L.
et al.
(
2009
)
Genome-wide association study identifies variants at CLU and PICALM associated with Alzheimer's disease
.
Nat. Genet.
41
,
1088
1093
[PubMed]
54.
Cribbs
D.H.
,
Berchtold
N.C.
,
Perreau
V.
,
Coleman
P.D.
,
Rogers
J.
,
Tenner
A.J.
et al.
(
2012
)
Extensive innate immune gene activation accompanies brain aging, increasing vulnerability to cognitive decline and neurodegeneration: a microarray study
.
J Neuroinflammation
9
,
179
[PubMed]
55.
Guerreiro
R.
,
Wojtas
A.
,
Bras
J.
,
Carrasquillo
M.
,
Rogaeva
E.
,
Majounie
E.
et al.
(
2013
)
TREM2 variants in Alzheimer's disease
.
N. Engl. J. Med.
368
,
117
127
[PubMed]
56.
Naj
A.C.
,
Jun
G.
,
Beecham
G.W.
,
Wang
L.S.
,
Vardarajan
B.N.
,
Buros
J.
et al.
(
2011
)
Common variants at MS4A4/MS4A6E, CD2AP, CD33 and EPHA1 are associated with late-onset Alzheimer's disease
.
Nat. Genet.
43
,
436
441
[PubMed]
57.
Zetterberg
M.
,
Landgren
S.
,
Andersson
M.
,
Palmér
M.
,
Gustafson
D.
,
Skoog
I.
et al.
(
2008
)
Association of complement factor H Y402H gene polymorphism with Alzheimer's disease
.
Am. J. Med. Genetics Part B, Neuropsychiatric Genetics: Off. Publ. Int. Soc. Psychiatric Genet.
147B
,
720
726
58.
Hakobyan
S.
,
Harding
K.
,
Aiyaz
M.
,
Hye
A.
,
Dobson
R.
,
Baird
A.
et al.
(
2016
)
Complement biomarkers as predictors of disease progression in Alzheimer's disease
.
J. Alzheimer's Dis.: JAD
54
,
707
716
[PubMed]
59.
Hu
W.T.
,
Watts
K.D.
,
Tailor
P.
,
Nguyen
T.P.
,
Howell
J.C.
,
Lee
R.C.
et al.
(
2016
)
CSF complement 3 and factor H are staging biomarkers in Alzheimer's disease
.
Acta Neuropathol. Commun.
4
,
14
[PubMed]
60.
Morgan
A.R.
,
Touchard
S.
,
Leckey
C.
,
O'Hagan
C.
,
Nevado-Holgado
A.J.
,
Barkhof
F.
et al.
(
2019
)
Inflammatory biomarkers in Alzheimer's disease plasma
.
Alzheimers Dement
15
,
776
787
[PubMed]
61.
Morgan
A.R.
,
Touchard
S.
,
O'Hagan
C.
,
Sims
R.
,
Majounie
E.
,
Escott-Price
V.
et al.
(
2017
)
The correlation between inflammatory biomarkers and polygenic risk score in Alzheimer's disease
.
J. Alzheimers Dis.
56
,
25
36
[PubMed]
62.
Rasmussen
K.L.
,
Nordestgaard
B.G.
,
Frikke-Schmidt
R.
and
Nielsen
S.F.
(
2018
)
An updated Alzheimer hypothesis: Complement C3 and risk of Alzheimer's disease-A cohort study of 95,442 individuals
.
Alzheimers Dement.
14
,
1589
1601
[PubMed]
63.
Krance
S.
,
Wu
C.
,
Zou
Y.
,
Mao
H.
,
Toufighi
S.
,
He
X.
et al.
(
2021
)
The complement cascade in Alzheimer's disease: a systematic review and meta-analysis
.
Mol. Psychiatry
26
,
5532
5541
[PubMed]
64.
Daborg
J.
,
Andreasson
U.
,
Pekna
M.
,
Lautner
R.
,
Hanse
E.
,
Minthon
L.
et al.
(
2012
)
Cerebrospinal fluid levels of complement proteins C3, C4 and CR1 in Alzheimer's disease
.
J. Neural. Transm. (Vienna)
119
,
789
797
[PubMed]
65.
Wang
Y.
,
Hancock
A.M.
,
Bradner
J.
,
Chung
K.A.
,
Quinn
J.F.
,
Peskind
E.R.
et al.
(
2011
)
Complement 3 and factor h in human cerebrospinal fluid in Parkinson's disease, Alzheimer's disease, and multiple-system atrophy
.
Am. J. Pathol.
178
,
1509
1516
[PubMed]
66.
Ayton
S.
,
Janelidze
S.
,
Roberts
B.
,
Palmqvist
S.
,
Kalinowski
P.
,
Diouf
I.
et al.
(
2021
)
Acute phase markers in CSF reveal inflammatory changes in Alzheimer's disease that intersect with pathology, APOE ε4, sex and age
.
Prog. Neurobiol.
198
,
[PubMed]
67.
Toledo
J.B.
,
Korff
A.
,
Shaw
L.M.
,
Trojanowski
J.Q.
and
Zhang
J.
(
2014
)
Low levels of cerebrospinal fluid complement 3 and factor H predict faster cognitive decline in mild cognitive impairment
.
Alzheimer's Res. Therapy
6
,
36
68.
Mulder
C.
,
Schoonenboom
S.
,
Wahlund
L.
,
Scheltens
P.
,
van Kamp
G.
,
Veerhuis
R.
et al.
(
2002
)
CSF markers related to pathogenetic mechanisms in Alzheimer's disease
.
J. Neural Transmission (Vienna, Austria: 1996)
109
1491
1498
69.
Veteleanu
A.
,
Pape
S.
,
Davies
K.
,
Kodosaki
E.
,
Hye
A.
,
Zelek
W.M.
et al.
(
2023
)
Complement dysregulation and Alzheimer's disease in Down syndrome
.
Alzheimers Dement
19
,
1383
1392
[PubMed]
70.
Veteleanu
A.
,
Stevenson-Hoare
J.
,
Keat
S.
,
Daskoulidou
N.
,
Zetterberg
H.
,
Heslegrave
A.
et al.
(
2023
)
Alzheimer's disease-associated complement gene variants influence plasma complement protein levels
.
J. Neuroinflammation
20
,
169
[PubMed]
71.
Hong
S.
,
Beja-Glasser
V.F.
,
Nfonoyim
B.M.
,
Frouin
A.
,
Li
S.
,
Ramakrishnan
S.
et al.
(
2016
)
Complement and microglia mediate early synapse loss in Alzheimer mouse models
.
Science (New York, NY)
352
,
712
716
72.
Hansen
D.V.
,
Hanson
J.E.
and
Sheng
M.
(
2018
)
Microglia in Alzheimer's disease
.
J. Cell Biol.
217
,
459
472
[PubMed]
73.
Dejanovic
B.
,
Wu
T.
,
Tsai
M.C.
,
Graykowski
D.
,
Gandham
V.D.
,
Rose
C.M.
et al.
(
2022
)
Complement C1q-dependent excitatory and inhibitory synapse elimination by astrocytes and microglia in Alzheimer's disease mouse models
.
Nat Aging
2
,
837
850
[PubMed]
74.
Dejanovic
B.
,
Huntley
M.A.
,
De Mazière
A.
,
Meilandt
W.J.
,
Wu
T.
,
Srinivasan
K.
et al.
(
2018
)
Changes in the synaptic proteome in tauopathy and rescue of tau-induced synapse loss by C1q antibodies
.
Neuron
100
,
1322.e7
1336.e7
[PubMed]
75.
Shi
Q.
,
Chowdhury
S.
,
Ma
R.
,
Le
K.X.
,
Hong
S.
,
Caldarone
B.J.
et al.
(
2017
)
Complement C3 deficiency protects against neurodegeneration in aged plaque-rich APP/PS1 mice
.
Sci. Transl. Med.
9
,
392
eaaf6295
76.
Maier
M.
,
Peng
Y.
,
Jiang
L.
,
Seabrook
T.J.
,
Carroll
M.C.
and
Lemere
C.A.
(
2008
)
Complement C3 deficiency leads to accelerated amyloid beta plaque deposition and neurodegeneration and modulation of the microglia/macrophage phenotype in amyloid precursor protein transgenic mice
.
J. Neurosci.
28
,
6333
6341
[PubMed]
77.
Wyss-Coray
T.
,
Yan
F.
,
Lin
A.H.
,
Lambris
J.D.
,
Alexander
J.J.
,
Quigg
R.J.
et al.
(
2002
)
Prominent neurodegeneration and increased plaque formation in complement-inhibited Alzheimer's mice
.
Proc. Natl. Acad. Sci. U.S.A.
99
,
10837
10842
[PubMed]
78.
Carpanini
S.M.
,
Torvell
M.
,
Bevan
R.J.
,
Byrne
R.A.J.
,
Daskoulidou
N.
,
Saito
T.
et al.
(
2022
)
Terminal complement pathway activation drives synaptic loss in Alzheimer's disease models
.
Acta Neuropathologica Commun.
10
,
99
[PubMed]
79.
Xie
J.
,
Cools
L.
,
Van Imschoot
G.
,
Van Wonterghem
E.
,
Pauwels
M.J.
,
Vlaeminck
I.
et al.
(
2023
)
Helicobacter pylori-derived outer membrane vesicles contribute to Alzheimer's disease pathogenesis via C3-C3aR signalling
.
J. Extracellular Vesicles
12
,
e12306
[PubMed]
80.
Litvinchuk
A.
,
Wan
Y.W.
,
Swartzlander
D.B.
,
Chen
F.
,
Cole
A.
,
Propson
N.E.
et al.
(
2018
)
Complement C3aR inactivation attenuates tau pathology and reverses an immune network deregulated in tauopathy models and Alzheimer's disease
.
Neuron
100
,
1337.e5
1353.e5
[PubMed]
81.
Gedam
M.
,
Comerota
M.M.
,
Propson
N.E.
,
Chen
T.
,
Jin
F.
,
Wang
M.C.
et al.
(
2023
)
Complement C3aR depletion reverses HIF-1α-induced metabolic impairment and enhances microglial response to Aβ pathology
.
J. Clin. Invest.
133
,
e167501
[PubMed]
82.
Hernandez
M.X.
,
Jiang
S.
,
Cole
T.A.
,
Chu
S.H.
,
Fonseca
M.I.
,
Fang
M.J.
et al.
(
2017
)
Prevention of C5aR1 signaling delays microglial inflammatory polarization, favors clearance pathways and suppresses cognitive loss
.
Mol. Neurodegener
12
,
66
[PubMed]
83.
Carvalho
K.
,
Schartz
N.D.
,
Balderrama-Gutierrez
G.
,
Liang
H.Y.
,
Chu
S.H.
,
Selvan
P.
et al.
(
2022
)
Modulation of C5a-C5aR1 signaling alters the dynamics of AD progression
.
J. Neuroinflammation
19
,
178
[PubMed]
84.
Gomez-Arboledas
A.
,
Carvalho
K.
,
Balderrama-Gutierrez
G.
,
Chu
S.H.
,
Liang
H.Y.
,
Schartz
N.D.
et al.
(
2022
)
C5aR1 antagonism alters microglial polarization and mitigates disease progression in a mouse model of Alzheimer's disease
.
Acta Neuropathol. Commun.
10
,
116
[PubMed]
85.
Vijayan
M.
and
Reddy
P.
(
2016
)
Stroke, vascular dementia, and Alzheimer's disease: molecular links
.
J. Alzheimer's Dis.: JAD
54
,
427
443
[PubMed]
86.
Kiss
M.
and
Binder
C.
(
2022
)
The multifaceted impact of complement on atherosclerosis
.
Atherosclerosis
351
,
29
40
[PubMed]
87.
Gamble
C.
(
1986
)
The pathogenesis of hyaline arteriolosclerosis
.
Am. J. Pathol.
122
,
410
420
[PubMed]
88.
Sawyer
R.
,
Demel
S.
,
Comeau
M.
,
Marion
M.
,
Rosand
J.
,
Langefeld
C.
et al.
(
2022
)
Alzheimer's disease related single nucleotide polymorphisms and correlation with intracerebral hemorrhage incidence
.
Medicine (Baltimore).
101
,
89.
Wang
M.
,
Xia
F.
,
Wan
S.
,
Hua
Y.
,
Keep
R.
and
Xi
G.
(
2021
)
Role of complement component 3 in early erythrolysis in the hematoma after experimental intracerebral hemorrhage
.
Stroke
52
,
2649
2660
90.
Wang
M.
,
Hua
Y.
,
Keep
R.
,
Wan
S.
,
Novakovic
N.
and
Xi
G.
(
2019
)
Complement inhibition attenuates early erythrolysis in the hematoma and brain injury in aged rats
.
Stroke
50
,
1859
1868
91.
Matsuo
K.
,
Shindo
A.
,
Niwa
A.
,
Tabei
K.
,
Akatsu
H.
,
Hashizume
Y.
et al.
(
2017
)
Complement activation in capillary cerebral amyloid angiopathy
.
Dement. Geriatr. Cogn. Disord.
44
,
343
353
[PubMed]
92.
Weidauer
S.
,
Neuhaus
E.
and
Hattingen
E.
(
2022
)
Cerebral superficial siderosis
.
Clin. Neuroradiol.
33
,
293
306
[PubMed]
93.
Weller
R.
,
Hawkes
C.
,
Kalaria
R.
,
Werring
D.
and
Carare
R.
(
2015
)
White matter changes in dementia: role of impaired drainage of interstitial fluid
.
Brain Pathol.
25
,
63
78
[PubMed]
94.
Kim
S.
,
Ahn
J.
,
Yang
H.
,
Lee
P.
,
Koh
G.
and
Jeong
Y.
(
2020
)
Cerebral amyloid angiopathy aggravates perivascular clearance impairment in an Alzheimer's disease mouse model
.
Acta Neuropathologica Commun.
8
,
95.
Saito
S.
,
Yamashiro
T.
,
Yamauchi
M.
,
Yamamoto
Y.
,
Noguchi
M.
,
Tomita
T.
et al.
(
2022
)
Complement 3 is a potential biomarker for cerebral amyloid angiopathy
.
J. Alzheimer's Dis.:JAD
89
,
381
387
[PubMed]
96.
Tanskanen
M.
,
Lindsberg
P.
,
Tienari
P.
,
Polvikoski
T.
,
Sulkava
R.
,
Verkkoniemi
A.
et al.
(
2005
)
Cerebral amyloid angiopathy in a 95+ cohort: complement activation and apolipoprotein E (ApoE) genotype
.
Neuropathol. Appl. Neurobiol.
31
,
589
599
[PubMed]
97.
Walker
D.
,
Dalsing-Hernandez
J.
and
Lue
L.
(
2008
)
Human postmortem brain-derived cerebrovascular smooth muscle cells express all genes of the classical complement pathway: a potential mechanism for vascular damage in cerebral amyloid angiopathy and Alzheimer's disease
.
Microvasc. Res.
75
,
411
419
[PubMed]
98.
Hondius
D.
,
Eigenhuis
K.
,
Morrema
T.
,
van der Schors
R.
,
van Nierop
P.
,
Bugiani
M.
et al.
(
2018
)
Proteomics analysis identifies new markers associated with capillary cerebral amyloid angiopathy in Alzheimer's disease
.
Acta Neuropathologica Commun.
6
,
[PubMed]
99.
Manousopoulou
A.
,
Yuen
H.
,
Sharp
M.
,
Saito
S.
and
Aldea
R.
(
2020
)
Maze rN, et al. Quantitative proteomic profiling of white matter in cases of cerebral amyloid angiopathy reveals upregulation of extracellular matrix proteins and clusterin
.
Free Neuropathol.
1
,
1
28
[PubMed]
100.
Wojtas
A.
,
Kang
S.
,
Olley
B.
,
Gatherer
M.
,
Shinohara
M.
,
Lozano
P.
et al.
(
2017
)
Loss of clusterin shifts amyloid deposition to the cerebrovasculature via disruption of perivascular drainage pathways
.
PNAS
114
,
6962
6971
[PubMed]
101.
Hettmann
T.
,
Gillies
S.
,
Kleinschmidt
M.
,
Piechotta
A.
,
Makioka
K.
,
Lemere
C.
et al.
(
2020
)
Development of the clinical candidate PBD-C06, a humanized pGlu3-Aβ-specific antibody against Alzheimer's disease with reduced complement activation
.
Sci. Rep.
10
,
3294
[PubMed]
102.
Propson
N.
,
Roy
E.
,
Litvinchuk
A.
,
Köhl
J.
and
Zheng
H.
(
2021
)
Endothelial C3a receptor mediates vascular inflammation and blood-brain barrier permeability during aging
.
J. Clin. Invest.
131
,
[PubMed]
103.
Zhang
L.
,
Pan
J.
,
Mamtilahun
M.
,
Zhu
Y.
,
Wang
L.
,
Venkatesh
A.
et al.
(
2020
)
Microglia exacerbate white matter injury via complement C3/C3aR pathway after hypoperfusion
.
Theranostics
10
,
104.
Convery
R.
,
Mead
S.
and
Rohrer
J.
(
2019
)
Review: Clinical, genetic and neuroimaging features of frontotemporal dementia
.
Neuropathol. Appl. Neurobiol.
45
,
6
18
[PubMed]
105.
Hartnell
I.J.
,
Blum
D.
,
Nicoll
J.A.R.
,
Dorothee
G.
and
Boche
D.
(
2023
)
Glial cells and adaptive immunity in frontotemporal dementia with tau pathology
.
Brain
144
,
724
745
106.
Rexach
J.
,
Polioudakis
D.
,
Yin
A.
,
Swarup
V.
,
Chang
T.
,
Nguyen
T.
et al.
(
2020
)
Tau pathology drives dementia risk-associated gene networks toward chronic inflammatory states and immunosuppression
.
Cell Rep.
33
,
[PubMed]
107.
Bright
F.
,
Werry
E.L.
,
Dobson-Stone
C.
,
Piguet
O.
,
Ittner
L.M.
,
Halliday
G.M.
et al.
(
2019
)
Neuroinflammation in frontotemporal dementia
.
Nat. Rev. Neurol.
15
,
540
555
[PubMed]
108.
Shen
Y.
,
Lue
L.
,
Yang
L.
,
Roher
A.
,
Kuo
Y.
,
Strohmeyer
R.
et al.
(
2001
)
Complement activation by neurofibrillary tangles in Alzheimer's disease
.
Neurosci. Lett.
305
,
165
8
[PubMed]
109.
Singhrao
S.
,
Neal
J.
,
Gasque
P.
,
Morgan
B.
and
Newman
G.
(
1996
)
Role of complement in the aetiology of Pick's disease?
J. Neuropathol. Exp. Neurol.
55
,
578
593
[PubMed]
110.
McGeer
P.L.
,
Akiyama
H.
,
Itagaki
S.
and
McGeer
E.G.
(
1998
)
Immune System Response in Alzheimer's Disease
.
Can. J. Neurol. Sci.
16
516
527
111.
Schwab
C.
,
Steele
J.
and
McGeer
P.
(
1996
)
Neurofibrillary tangles of Guam parkinson-dementia are associated with reactive microglia and complement proteins
.
Brain Res.
707
,
196
205
[PubMed]
112.
van der Ende
E.
,
Heller
C.
,
Sogorb-Esteve
A.
,
Swift
I.
,
McFall
D.
,
Peakman
G.
et al.
(
2022
)
Elevated CSF and plasma complement proteins in genetic frontotemporal dementia: results from the GENFI study
.
J. Neuroinflammation
19
,
217
[PubMed]
113.
Wu
T.
,
Dejanovic
B.
,
Gandham
V.
,
Gogineni
A.
,
Edmonds
R.
,
Schauer
S.
et al.
(
2019
)
Complement C3 Is Activated in Human AD Brain and Is Required for Neurodegeneration in Mouse Models of Amyloidosis and Tauopathy
.
Cell Reports
28
,
2111
2123
114.
Britschgi
M.
,
Takeda-Uchimura
Y.
,
Rockenstein
E.
,
Johns
H.
,
Masliah
E.
and
Wyss-Coray
T.
(
2012
)
Deficiency of terminal complement pathway inhibitor promotes neuronal tau pathology and degeneration in mice
.
J. Neuroinflammation
9
,
220
[PubMed]
115.
Zhu
X.
,
Liu
L.
,
Dai
W.
and
Ma
T.
(
2022
)
Crry silencing alleviates Alzheimer's disease injury by regulating neuroinflammatory cytokines and the complement system
.
Neural Regeneration Res.
17
,
1841
1849
116.
Yao
Y.
,
Chang
Y.
,
Li
S.
,
Zhu
J.
,
Wu
Y.
,
Jiang
X.
et al.
(
2023
)
Complement C3a receptor antagonist alleviates tau pathology and ameliorates cognitive deficits in P301S mice
.
Brain Res. Bull.
200
,
[PubMed]
117.
Fonseca
M.
,
Ager
R.
,
Chu
S.
,
Yazan
O.
,
Sanderson
S.
,
LaFerla
F.
et al.
(
2009
)
Treatment with a C5aR antagonist decreases pathology and enhances behavioral performance in murine models of Alzheimer's disease
.
J. Immunol.(Baltimore, Md: 1950)
183
,
1375
1383
118.
Outeiro
T.
,
Koss
D.
,
Erskine
D.
,
Walker
L.
,
Kurzawa-Akanbi
M.
,
Burn
D.
et al.
(
2019
)
Dementia with Lewy bodies: an update and outlook
.
Mol. Neurodegeneration
14
,
5
[PubMed]
119.
Tansey
M.G.
,
Wallings
R.L.
,
Houser
M.C.
,
Herrick
M.K.
,
Keating
C.E.
and
Joers
V.
(
2022
)
Inflammation and immune dysfunction in Parkinson disease
.
Nat. Rev. Immunol.
22
,
657
673
[PubMed]
120.
Khosousi
S.
,
Hye
A.
,
Velayudhan
L.
,
Bloth
B.
,
Tsitsi
P.
,
Markaki
I.
et al.
(
2023
)
Complement system changes in blood in Parkinson's disease and progressive Supranuclear Palsy/Corticobasal Syndrome
.
Parkinsonism Related Disorders
108
,
[PubMed]
121.
Veselý
B.
,
Dufek
M.
,
Thon
V.
,
Brozman
M.
,
Királová
S.
,
Halászová
T.
et al.
(
2018
)
Interleukin 6 and complement serum level study in Parkinson's disease
.
J. Neural. Transmission (Vienna, Austria: 1996)
125
,
875
881
[PubMed]
122.
Naskar
A.
,
Stezin
A.
,
Dharmappa
A.
,
Hegde
S.
,
Philip
M.
,
Kamble
N.
et al.
(
2022
)
Fibrinogen and Complement Factor H Are Promising CSF Protein Biomarkers for Parkinson's Disease with Cognitive Impairment─A Proteomics-ELISA-Based Study
.
ACS Chem. Neurosci.
13
,
1030
1045
[PubMed]
123.
Winchester
L.
,
Barber
I.
,
Lawton
M.
,
Ash
J.
,
Liu
B.
,
Evetts
S.
et al.
(
2022
)
Identification of a possible proteomic biomarker in Parkinson's disease: discovery and replication in blood, brain and cerebrospinal fluid
.
Brain Commun.
5
,
fcac343
[PubMed]
124.
Togo
T.
,
Iseki
E.
,
Marui
W.
,
Akiyama
H.
,
Uéda
K.
and
Kosaka
K.
(
2001
)
Glial involvement in the degeneration process of Lewy body-bearing neurons and the degradation process of Lewy bodies in brains of dementia with Lewy bodies
.
J. Neurol. Sci.
184
,
71
75
[PubMed]
125.
Depboylu
C.
,
Schäfer
M.
,
Arias-Carrión
O.
,
Oertel
W.
,
Weihe
E.
and
Höglinger
G.
(
2011
)
Possible involvement of complement factor C1q in the clearance of extracellular neuromelanin from the substantia nigra in Parkinson disease
.
J. Neuropathol. Exp. Neurol.
70
,
125
132
[PubMed]
126.
Loeffler
D.
,
Camp
D.
and
Conant
S.
(
2006
)
Complement activation in the Parkinson's disease substantia nigra: an immunocytochemical study
.
J. Neuroinflammation
3
,
[PubMed]
127.
Yamada
T.
,
McGeer
P.
and
McGeer
E.
(
1992
)
Lewy bodies in Parkinson's disease are recognized by antibodies to complement proteins
.
Acta Neuropathol. (Berl)
84
,
100
104
128.
Iseki
E.
,
Marui
W.
,
Akiyama
H.
,
Uéda
K.
and
Kosaka
K.
(
2000
)
Degeneration process of Lewy bodies in the brains of patients with dementia with Lewy bodies using alpha-synuclein-immunohistochemistry
.
Neurosci. Lett.
286
,
69
73
[PubMed]
129.
Dumitriu
A.
,
Golji
J.
,
Labadorf
A.
,
Gao
B.
,
Beach
T.
,
Myers
R.
et al.
(
2016
)
Integrative analyses of proteomics and RNA transcriptomics implicate mitochondrial processes, protein folding pathways and GWAS loci in Parkinson disease
.
BMC Med. Genet.
9
130.
Juul-Madsen
K.
,
Qvist
P.
,
Bendtsen
K.L.
,
Vestergaard
B.
,
Howard
K.A.
,
Paludan
S.R.
et al.
(
2020
)
Size-Selective Phagocytic Clearance of Fibrillar α-Synuclein through Conformational Activation of Complement Receptor 4
.
J. Immunol.
204
,
1345
1361
[PubMed]
131.
Klegeris
A.
and
McGeer
P.
(
2007
)
Complement activation by islet amyloid polypeptide (IAPP) and alpha-synuclein 112
.
Biochem. Biophys. Res. Commun.
357
,
1096
9
[PubMed]
132.
Gregersen
E.
,
Betzer
C.
,
Kim
W.
,
Kovacs
G.
,
Reimer
L.
,
Halliday
G.
et al.
(
2021
)
Alpha-synuclein activates the classical complement pathway and mediates complement-dependent cell toxicity
.
J. Neuroinflammation
18
, 177
[PubMed]
133.
Juul-Madsen
K.
,
Qvist
P.
,
Bendtsen
K.
,
Langkilde
A.
,
Vestergaard
B.
,
Howard
K.
et al.
(
2020
)
Size-selective phagocytic clearance of fibrillar α-synuclein through conformational activation of complement receptor 4
.
J. Immunol. (Baltimore, Md: 1950)
204
,
1345
1361
134.
Jing
L.
,
Hou
L.
,
Zhang
D.
,
Li
S.
,
Ruan
Z.
,
Zhang
X.
et al.
(
2021
)
Microglial activation mediates noradrenergic locus coeruleus neurodegeneration via complement receptor 3 in a rotenone-induced Parkinson's disease mouse model
.
J. Inflammation Res.
14
,
1341
1356
135.
Liang
Y.
,
Li
S.
,
Guo
Q.
,
Zhang
Y.
,
Wen
C.
,
Zou
Q.
et al.
(
2007
)
Complement 3-deficient mice are not protected against MPTP-induced dopaminergic neurotoxicity
.
Brain Res.
1178
,
132
40
[PubMed]
136.
Depboylu
C.
,
Schorlemmer
K.
,
Klietz
M.
,
Oertel
W.
,
Weihe
E.
,
Höglinger
G.
et al.
(
2011
)
Upregulation of microglial C1q expression has no effects on nigrostriatal dopaminergic injury in the MPTP mouse model of Parkinson disease
.
J. Neuroimmunol.
236
,
39
46
[PubMed]
137.
Bates
G.P.
,
Dorsey
R.
,
Gusella
J.F.
,
Hayden
M.R.
,
Kay
C.
,
Leavitt
B.R.
et al.
(
2015
)
Huntington disease
.
Nat. Rev. Dis. Primers
1
,
15005
[PubMed]
138.
Moller
T.
(
2010
)
Neuroinflammation in Huntington's disease
.
J. Neural. Transm. (Vienna)
117
,
1001
1008
[PubMed]
139.
Singhrao
S.K.
,
Neal
J.W.
,
Morgan
B.P.
and
Gasque
P.
(
1999
)
Increased complement biosynthesis by microglia and complement activation on neurons in Huntington's disease
.
Exp. Neurol.
159
,
362
376
[PubMed]
140.
Gasque
P.
,
Singhrao
S.
,
Neal
J.
,
Götze
O.
and
Morgan
B.
(
1997
)
Expression of the receptor for complement C5a (CD88) is up-regulated on reactive astrocytes, microglia, and endothelial cells in the inflamed human central nervous system
.
Am. J. Pathol.
150
,
31
41
[PubMed]
141.
Dalrymple
A.
,
Wild
E.J.
,
Joubert
R.
,
Sathasivam
K.
,
Bjorkqvist
M.
,
Petersen
A.
et al.
(
2007
)
Proteomic profiling of plasma in Huntington's disease reveals neuroinflammatory activation and biomarker candidates
.
J. Proteome Res.
6
,
2833
2840
[PubMed]
142.
Silajdzic
E.
,
Rezeli
M.
,
Vegvari
A.
,
Lahiri
N.
,
Andre
R.
,
Magnusson-Lind
A.
et al.
(
2013
)
A critical evaluation of inflammatory markers in Huntington's Disease plasma
.
J. Huntingtons Dis.
2
,
125
134
[PubMed]
143.
Caron
N.S.
,
Haqqani
A.S.
,
Sandhu
A.
,
Aly
A.E.
,
Findlay Black
H.
,
Bone
J.N.
et al.
(
2022
)
Cerebrospinal fluid biomarkers for assessing Huntington disease onset and severity
.
Brain Commun.
4
,
309
[PubMed]
144.
Mangiarini
L.
,
Sathasivam
K.
,
Seller
M.
,
Cozens
B.
,
Harper
A.
,
Hetherington
C.
et al.
(
1996
)
Exon 1 of the HD gene with an expanded CAG repeat is sufficient to cause a progressive neurological phenotype in transgenic mice
.
Cell
87
,
493
506
[PubMed]
145.
Larkin
P.B.
and
Muchowski
P.J.
(
2012
)
Genetic deficiency of complement component 3 does not alter disease progression in a mouse model of Huntington's disease
.
J. Huntingtons Dis.
1
,
107
118
[PubMed]
146.
Woodruff
T.M.
,
Crane
J.W.
,
Proctor
L.M.
,
Buller
K.M.
,
Shek
A.B.
,
de Vos
K.
et al.
(
2006
)
Therapeutic activity of C5a receptor antagonists in a rat model of neurodegeneration
.
FASEB J.
20
,
1407
1417
[PubMed]
147.
(
2023
)
Annexon: Pipeline - Annexon Biosciences
.
148.
Compston
D.
,
Morgan
B.
,
Campbell
A.
,
Wilkins
P.
,
Cole
G.
,
Thomas
N.
et al.
(
1989
)
Immunocytochemical localization of the terminal complement complex in multiple sclerosis
.
Neuropathol. Appl. Neurobiol.
15
,
307
316
[PubMed]
149.
Peterson
J.
,
L.
,
Mörk
S.
,
Chang
A.
and
Trapp
B.
(
2001
)
Transected neurites, apoptotic neurons, and reduced inflammation in cortical multiple sclerosis lesions
.
Ann. Neurol.
50
,
389
400
[PubMed]
150.
Ingram
G.
,
Hakobyan
S.
,
Robertson
N.
and
Morgan
B.P.
(
2010
)
Elevated plasma C4a levels in multiple sclerosis correlate with disease activity
.
J. Neuroimmunol.
223
,
124
127
[PubMed]
151.
Lucchinetti
C.
,
Popescu
B.
,
Bunyan
R.
,
Moll
N.
,
Roemer
S.
,
Lassmann
H.
et al.
(
2011
)
Inflammatory cortical demyelination in early multiple sclerosis
.
N. Engl. J. Med.
365
,
2188
2197
[PubMed]
152.
Popescu
B.
and
Lucchinetti
C.
(
2012
)
Meningeal and cortical grey matter pathology in multiple sclerosis
.
BMC Neurol.
12
, 11
[PubMed]
153.
Ingram
G.
,
Loveless
S.
,
Howell
O.W.
,
Hakobyan
S.
,
Dancey
B.
,
Harris
C.L.
et al.
(
2014
)
Complement activation in multiple sclerosis plaques: an immunohistochemical analysis
.
Acta Neuropathologica Commun.
2
,
1
15
[PubMed]
154.
Michailidou
I.
,
Willems
J.
,
Kooi
E.
,
van Eden
C.
,
Gold
S.
,
Geurts
J.
et al.
(
2015
)
Complement C1q-C3-associated synaptic changes in multiple sclerosis hippocampus
.
Ann. Neurol.
77
,
1007
1026
[PubMed]
155.
Bevan
R.
,
Evans
R.
,
Griffiths
L.
,
Watkins
L.
,
Rees
M.
,
Magliozzi
R.
et al.
(
2018
)
Meningeal inflammation and cortical demyelination in acute multiple sclerosis
.
Ann. Neurol.
84
,
829
842
[PubMed]
156.
Cooze
B.
,
Dickerson
M.
,
Loganathan
R.
,
Watkins
L.
,
Grounds
E.
,
Pearson
B.
et al.
(
2022
)
The association between neurodegeneration and local complement activation in the thalamus to progressive multiple sclerosis outcome
.
Brain Pathol.
32
,
[PubMed]
157.
Watkins
L.
,
Neal
J.
,
Loveless
S.
,
Michailidou
I.
,
Ramaglia
V.
,
Rees
M.
et al.
(
2016
)
Complement is activated in progressive multiple sclerosis cortical grey matter lesions
.
J. Neuroinflammation
13
,
161
[PubMed]
158.
Loveless
S.
,
Neal
J.
,
Howell
O.
,
Harding
K.
,
Sarkies
P.
,
Evans
R.
et al.
(
2018
)
Tissue microarray methodology identifies complement pathway activation and dysregulation in progressive multiple sclerosis
.
Brain Pathol.
28
,
507
520
[PubMed]
159.
Evans
R.
,
Watkins
L.
,
Hawkins
K.
,
Santiago
G.
,
Demetriou
C.
,
Naughton
M.
et al.
(
2023
)
Complement activation and increased anaphylatoxin receptor expression are associated with cortical grey matter lesions and the compartmentalised inflammatory response of multiple sclerosis
.
Front. Cell. Neurosci.
17
,
1094106
[PubMed]
160.
Ingram
G.
,
Hakobyan
S.
,
Hirst
C.
,
Harris
C.
,
Loveless
S.
,
Mitchell
J.
et al.
(
2012
)
Systemic complement profiling in multiple sclerosis as a biomarker of disease state
.
Mult. Scler.
18
,
1401
1411
161.
Ingram
G.
,
Hakobyan
S.
,
Hirst
C.
,
Harris
C.
,
Pickersgill
T.
,
Cossburn
M.
et al.
(
2010
)
Complement regulator factor H as a serum biomarker of multiple sclerosis disease state
.
Brain: J. Neurol.
133
,
1602
1611
162.
Ingram
G.
,
Hakobyan
S.
,
Robertson
N.P.
and
Morgan
B.P.
(
2009
)
Complement in multiple sclerosis: its role in disease and potential as a biomarker
.
Clin. Exp. Immunol.
155
,
128
139
[PubMed]
163.
Hakobyan
S.
,
Luppe
S.
,
Evans
D.
,
Harding
K.
,
Loveless
S.
,
Robertson
N.
et al.
(
2017
)
Plasma complement biomarkers distinguish multiple sclerosis and neuromyelitis optica spectrum disorder
.
Mult. Scler.
23
,
946
955
[PubMed]
164.
Tatomir
A.
,
Talpos-Caia
A.
,
Anselmo
F.
,
Kruszewski
A.
,
Boodhoo
D.
,
Rus
V.
et al.
(
2017
)
The complement system as a biomarker of disease activity and response to treatment in multiple sclerosis
.
Immunol. Res.
65
,
1103
1109
[PubMed]
165.
Sanders
M.
,
Koski
C.
,
Robbins
D.
,
Shin
M.
,
Frank
M.
and
Joiner
K.
(
1986
)
Activated terminal complement in cerebrospinal fluid in Guillain-Barré syndrome and multiple sclerosis
.
J. Immunol.(Baltimore, Md: 1950)
136
,
4456
4459
166.
Jongen
P.
,
Doesburg
W.
,
Ibrahim-Stappers
J.
,
Lemmens
W.
,
Hommes
O.
and
Lamers
K.
(
2000
)
Cerebrospinal fluid C3 and C4 indexes in immunological disorders of the central nervous system
.
Acta Neurol. Scand.
101
,
[PubMed]
167.
Finehout
E.
,
Franck
Z.
and
Lee
K.
(
2005
)
Complement protein isoforms in CSF as possible biomarkers for neurodegenerative disease
.
Dis. Markers
21
,
93
101
[PubMed]
168.
Aeinehband
S.
,
Lindblom
R.
,
Al Nimer
F.
,
Vijayaraghavan
S.
,
Sandholm
K.
,
Khademi
M.
et al.
(
2015
)
Complement component C3 and butyrylcholinesterase activity are associated with neurodegeneration and clinical disability in multiple sclerosis
.
PloS ONE
10
,
[PubMed]
169.
Lindblom
R.
,
Aeinehband
S.
,
Ström
M.
,
Al Nimer
F.
,
Sandholm
K.
,
Khademi
M.
et al.
(
2016
)
Complement Receptor 2 is increased in cerebrospinal fluid of multiple sclerosis patients and regulates C3 function
.
Clin. Immunol.
166-167
89
95
170.
Magliozzi
R.
,
Hametner
S.
,
Facchiano
F.
,
Marastoni
D.
,
Rossi
S.
,
Castellaro
M.
et al.
(
2019
)
Iron homeostasis, complement, and coagulation cascade as CSF signature of cortical lesions in early multiple sclerosis
.
Ann. Clin. Transl. Neurol.
6
2150
2163
171.
Håkansson
I.
,
Ernerudh
J.
,
Vrethem
M.
,
Dahle
C.
and
Ekdahl
K.
(
2020
)
Complement activation in cerebrospinal fluid in clinically isolated syndrome and early stages of relapsing remitting multiple sclerosis
.
J. Neuroimmunol.
340
,
[PubMed]
172.
Lucchinetti
C.
,
Brück
W.
,
Parisi
J.
,
Scheithauer
B.
,
Rodriguez
M.
and
Lassmann
H.
(
2000
)
Heterogeneity of multiple sclerosis lesions: implications for the pathogenesis of demyelination
.
Ann. Neurol.
47
,
707
17
[PubMed]
173.
Brück
W.
,
Lucchinetti
C.
and
Lassmann
H.
(
2002
)
The pathology of primary progressive multiple sclerosis
.
Mult. Scler.
8
,
93
7
[PubMed]
174.
Höftberger
R.
,
Guo
Y.
,
Flanagan
E.
,
Lopez-Chiriboga
A.
,
Endmayr
V.
,
Hochmeister
S.
et al.
(
2020
)
The pathology of central nervous system inflammatory demyelinating disease accompanying myelin oligodendrocyte glycoprotein autoantibody
.
Acta Neuropathol. (Berl)
139
,
875
892
175.
Morgan
B.
,
Gommerman
J.
and
Ramaglia
V.
(
2021
)
An “Outside-In” and “Inside-Out” Consideration of Complement in the Multiple Sclerosis Brain: Lessons From Development and Neurodegenerative Diseases
.
Front. Cell. Neurosci.
14
,
[PubMed]
176.
Piddlesden
S.
,
Storch
M.
,
Hibbs
M.
,
Freeman
A.
,
Lassmann
H.
and
Morgan
B.
(
1994
)
Soluble recombinant complement receptor 1 inhibits inflammation and demyelination in antibody-mediated demyelinating experimental allergic encephalomyelitis
.
J. Immunol.(Baltimore, Md: 1950)
152
,
5477
84
177.
Werneburg
S.
,
Jung
J.
,
Kunjamma
R.
,
Ha
S.
,
Luciano
N.
,
Willis
C.
et al.
(
2020
)
Targeted Complement Inhibition at Synapses Prevents Microglial Synaptic Engulfment and Synapse Loss in Demyelinating Disease
.
Immunity
52
,
167
182
[PubMed]
178.
Hammond
J.
,
Bellizzi
M.
,
Ware
C.
,
Qiu
W.
,
Saminathan
P.
,
Li
H.
et al.
(
2020
)
Complement-dependent synapse loss and microgliosis in a mouse model of multiple sclerosis
.
Brain Behav. Immun.
87
,
739
750
[PubMed]
179.
Mead
R.
,
Singhrao
S.
,
Neal
J.
,
Lassmann
H.
and
Morgan
B.
(
2002
)
The membrane attack complex of complement causes severe demyelination associated with acute axonal injury
.
J. Immunol. (Baltimore, Md: 1950)
168
,
458
65
180.
Mead
R.
,
Neal
J.
,
Griffiths
M.
,
Linington
C.
,
Botto
M.
,
Lassmann
H.
et al.
(
2004
)
Deficiency of the complement regulator CD59a enhances disease severity, demyelination and axonal injury in murine acute experimental allergic encephalomyelitis
.
Lab. Invest.
84
,
21
8
[PubMed]
181.
Saha
S.
,
Chant
D.
,
Welham
J.
and
McGrath
J.
(
2005
)
A systematic review of the prevalence of schizophrenia
.
PLoS Med.
2
,
[PubMed]
182.
Brown
S.
(
1997
)
Excess mortality of schizophrenia. A meta-analysis
.
Br. J. Psychiatry: J. Mental Sci.
171
502
508
183.
Brown
S.
,
Inskip
H.
and
Barraclough
B.
(
2000
)
Causes of the excess mortality of schizophrenia
.
Br. J. Psychiatry: J. Mental Sci.
177
,
212
7
184.
Cannon
T.D.
,
Thompson
P.M.
,
van Erp
T.G.M.
,
Toga
A.W.
,
Poutanen
V.
,
Huttunen
M.
et al.
(
2002
)
Cortex mapping reveals regionally specific patterns of genetic and disease-specific gray-matter deficits in twins discordant for schizophrenia
.
Proc. Natl. Acad. Sci. U.S.A.
99
5
3228
33
185.
Cannon
T.
,
Chung
Y.
,
He
G.
,
Sun
D.
,
Jacobson
A.
,
van Erp
T.
et al.
(
2015
)
Progressive reduction in cortical thickness as psychosis develops: a multisite longitudinal neuroimaging study of youth at elevated clinical risk
.
Biol. Psychiatry
77
,
147
57
186.
Garey
L.J.
,
Ong
W.Y.
,
Patel
T.S.
,
Kanani
M.
,
Davis
A.
,
Mortimer
A.M.
et al.
(
1998
)
Reduced dendritic spine density on cerebral cortical pyramidal neurons in schizophrenia
.
Journal of Neurology, Neurosurgery & Psychiatry
446
453
65
187.
Glantz
L.A.
and
Lewis
D.A.
(
2002
)
Decreased dendritic spine density on prefrontal cortical pyramidal neurons in schizophrenia
.
Arch. Gen. Psychiatry
57
,
65
73
188.
Feinberg
I.
(
1982
)
Schizophrenia: caused by a fault in programmed synaptic elimination during adolescence?
J. Psychiatr. Res.
17
,
319
34
[PubMed]
189.
Huttenlocher
P.
(
1979
)
Synaptic density in human frontal cortex - developmental changes and effects of aging
.
Brain Res.
163
,
195
205
[PubMed]
190.
Katz
L.
and
Shatz
C.
(
1996
)
Synaptic activity and the construction of cortical circuits
.
Science (New York, NY)
274
,
1133
8
191.
Cardno
A.
and
Gottesman
I.
(
2000
)
Twin studies of schizophrenia: from bow-and-arrow concordances to star wars Mx and functional genomics
.
Am. J. Med. Genet.
97
,
12
7
[PubMed]
192.
Sullivan
P.
,
Kendler
K.
and
Neale
M.
(
2003
)
Schizophrenia as a complex trait: evidence from a meta-analysis of twin studies
.
Arch. Gen. Psychiatry
60
,
1187
92
193.
Purcell
S.M.
,
Wray
N.R.
,
Stone
J.L.
,
Visscher
P.M.
,
O'Donovan
M.C.
,
Sullivan
P.F.
et al.
(
2009
)
Common polygenic variation contributes to risk of schizophrenia and bipolar disorder
.
Nature
460
,
748
752
[PubMed]
194.
Stefansson
H.
,
Ophoff
R.A.
,
Steinberg
S.
,
Andreassen
O.A.
,
Cichon
S.
,
Rujescu
D.
et al.
(
2009
)
Common variants conferring risk of schizophrenia
.
Nature
460
,
744
747
[PubMed]
195.
Shi
J.
,
Levinson
D.F.
,
Duan
J.
,
Sanders
A.R.
,
Zheng
Y.
,
Pe'er
I.
et al.
(
2009
)
Common variants on chromosome 6p22.1 are associated with schizophrenia
.
Nature
460
,
753
757
[PubMed]
196.
Isenman
D.
and
Young
J.
(
1984
)
The molecular basis for the difference in immune hemolysis activity of the Chido and Rodgers isotypes of human complement component C4
.
J. Immunol.
132
3019
3027
[PubMed]
197.
Law
S.
,
Dodds
A.
and
Porter
R.
(
1984
)
A comparison of the properties of two classes, C4A and C4B, of the human complement component C4
.
EMBO J.
3
,
1819
23
[PubMed]
198.
Consortium
S.W.GotP.G.
(
2014
)
Biological insights from 108 schizophrenia-associated genetic loci
.
Nature
511
,
421
427
[PubMed]
199.
Study
T.S.P.G.-W.A.
(
2011
)
Genome-wide association study identifies five new schizophrenia loci
.
Nat. Genet.
43
,
969
976
[PubMed]
200.
Trubetskoy
V.
,
Pardiñas
A.F.
,
Qi
T.
,
Panagiotaropoulou
G.
,
Awasthi
S.
,
Bigdeli
T.B.
et al.
(
2022
)
Mapping genomic loci implicates genes and synaptic biology in schizophrenia
.
Nature
604
,
502
508
[PubMed]
201.
Escudero-Esparza
A.
,
Kalchishkova
N.
,
Kurbasic
E.
,
Jiang
W.
and
Blom
A.
(
2013
)
The novel complement inhibitor human CUB and Sushi multiple domains 1 (CSMD1) protein promotes factor I-mediated degradation of C4b and C3b and inhibits the membrane attack complex assembly
.
FASEB J.: Off. Publ. Federation Am. Societies Exp. Biol.
27
,
5083
93
[PubMed]
202.
Kraus
D.
,
Elliott
G.
,
Chute
H.
,
Horan
T.
,
Pfenninger
K.
,
Sanford
S.
et al.
(
2006
)
CSMD1 is a novel multiple domain complement-regulatory protein highly expressed in the central nervous system and epithelial tissues
.
J. Immunol. (Baltimore, Md: 1950)
176
,
4419
30
203.
Lee
A.S.
,
Rusch
J.
,
Lima
A.C.
,
Usmani
A.
,
Huang
N.
,
Lepamets
M.
et al.
(
2019
)
Rare mutations in the complement regulatory gene CSMD1 are associated with male and female infertility
.
Nat. Commun.
10
,
1
16
[PubMed]
204.
Baum
M.L.
,
Wilton
D.K.
,
Muthukumar
A.
,
Fox
R.G.
,
Carey
A.
,
Crotty
W.
et al.
(
2020
)
CUB and Sushi Multiple Domains 1 (CSMD1) opposes the complement cascade in neural tissues
.
bioRxiv
205.
Liu
Y.
,
Fu
X.
,
Tang
Z.
,
Li
C.
,
Xu
Y.
,
Zhang
F.
et al.
(
2019
)
Altered expression of the CSMD1 gene in the peripheral blood of schizophrenia patients
.
BMC Psychiatry
19
,
206.
Abd El Gayed
E.
,
Rizk
M.
,
Ramadan
A.
and
Bayomy
N.
(
2021
)
mRNA expression of the CUB and sushi multiple domains 1 (CSMD1) and its serum protein level as predictors for psychosis in the familial high-risk children and young adults
.
ACS Omega
6
,
[PubMed]
207.
Hatzimanolis
A.
,
Foteli
S.
,
Stefanatou
P.
,
Ntigrintaki
A.-A.
,
Ralli
I.
,
Kollias
K.
et al.
(
2022
)
Deregulation of complement components C4A and CSMD1 peripheral expression in first-episode psychosis and links to cognitive ability
.
Eur. Arch. Psychiatry Clin. Neurosci.
272
,
1219
1228
[PubMed]
208.
Ruiz-Martínez
J.
,
Azcona
L.
,
Bergareche
A.
,
Martí-Massó
J.
and
Paisán-Ruiz
C.
(
2017
)
Whole-exome sequencing associates novel CSMD1 gene mutations with familial Parkinson disease
.
Neurol. Genet.
3
,
[PubMed]
209.
Xu
W.
,
Cohen-Woods
S.
,
Chen
Q.
,
Noor
A.
,
Knight
J.
,
Hosang
G.
et al.
(
2014
)
Genome-wide association study of bipolar disorder in Canadian and UK populations corroborates disease loci including SYNE1 and CSMD1
.
BMC Med. Genet.
15
,
1
13
[PubMed]
210.
Baranzini
S.
,
Wang
J.
,
Gibson
R.
,
Galwey
N.
,
Naegelin
Y.
,
Barkhof
F.
et al.
(
2009
)
Genome-wide association analysis of susceptibility and clinical phenotype in multiple sclerosis
.
Hum. Mol. Genet.
18
,
767
78
211.
Cavanillas
M.L.
,
Fernández
O.
,
Comabella
M.
,
Alcina
A.
,
Fedetz
M.
,
Izquierdo
G.
et al.
(
2010
)
Replication of top markers of a genome-wide association study in multiple sclerosis in Spain
.
Genes Immunity
12
,
110
115
[PubMed]
212.
Cukier
H.N.
,
Dueker
N.D.
,
Slifer
S.H.
,
Lee
J.M.
,
Whitehead
P.L.
,
Lalanne
E.
et al.
(
2014
)
Exome sequencing of extended families with autism reveals genes shared across neurodevelopmental and neuropsychiatric disorders
.
Mol. Autism
5
,
1
10
[PubMed]
213.
Glancy
M.
,
Barnicoat
A.
,
Vijeratnam
R.
,
de Souza
S.
,
Gilmore
J.
,
Huang
S.
et al.
(
2008
)
Transmitted duplication of 8p23.1–8p23.2 associated with speech delay, autism and learning difficulties
.
Eur. J. Hum. Genet.
17
,
37
43
[PubMed]
214.
Yilmaz
M.
,
Yalcin
E.
,
Presumey
J.
,
Aw
E.
,
Ma
M.
,
Whelan
C.W.
et al.
(
2021
)
Overexpression of schizophrenia susceptibility factor human complement C4A promotes excessive synaptic loss and behavioral changes in mice
.
Nat. Neurosci.
24
,
214
224
[PubMed]
215.
Comer
A.
,
Jinadasa
T.
,
Sriram
B.
,
Phadke
R.
,
Kretsge
L.
,
Nguyen
T.
et al.
(
2020
)
Increased expression of schizophrenia-associated gene C4 leads to hypoconnectivity of prefrontal cortex and reduced social interaction
.
PLoS Biol.
18
,
[PubMed]
216.
Woo
J.J.
,
Pouget
J.G.
,
Zai
C.C.
and
Kennedy
J.L.
(
2019
)
The complement system in schizophrenia: where are we now and what's next?
Mol. Psychiatry
25
,
114
130
[PubMed]
217.
Kopczynska
M.
,
Zelek
W.
,
Touchard
S.
,
Gaughran
F.
,
Di Forti
M.
,
Mondelli
V.
et al.
(
2019
)
Complement system biomarkers in first episode psychosis
.
Schizophr. Res.
204
,
16
22
[PubMed]
218.
Walss-Bass
C.
,
Lokesh
G.
,
Dyukova
E.
,
Gorenstein
D.
,
Roberts
D.
,
Velligan
D.
et al.
(
2019
)
X-Aptamer Technology Identifies C4A and ApoB in Blood as Potential Markers for Schizophrenia
.
Mol. Neuropsychiatry
5
,
52
59
219.
Asavapanumas
N.
,
Tradtrantip
L.
and
Verkman
A.
(
2021
)
Targeting the complement system in neuromyelitis optica spectrum disorder
.
Expert Opin. Biol. Ther.
21
,
1073
1086
[PubMed]
220.
Dhillon
S.
(
2018
)
Eculizumab: A review in generalized myasthenia gravis
.
Drugs
78
367
376
221.
Zelek
W.
and
Morgan
B.
(
2022
)
Targeting complement in neurodegeneration: challenges, risks, and strategies
.
Trends Pharmacol. Sci.
43
,
615
628
[PubMed]
222.
Drouin-Ouellet
J.
,
Sawiak
S.
,
Cisbani
G.
,
Lagacé
M.
,
Kuan
W.
,
Saint-Pierre
M.
et al.
(
2015
)
Cerebrovascular and blood-brain barrier impairments in Huntington's disease: Potential implications for its pathophysiology
.
Ann. Neurol.
78
,
160
77
[PubMed]
223.
Di Pardo
A.
,
Amico
E.
,
Scalabrì
F.
,
Pepe
G.
,
Castaldo
S.
,
Elifani
F.
et al.
(
2017
)
Impairment of blood-brain barrier is an early event in R6/2 mouse model of Huntington Disease
.
Sci. Rep.
7
,
224.
(
2023
)
Alexion: Pipeline | Alexion
.
https://alexion.com Accessed 29 July 2023
225.
Pardo-Moreno
T.
,
González-Acedo
A.
,
Rivas-Domínguez
A.
,
García-Morales
V.
,
García-Cozar
F.
,
Ramos-Rodríguez
J.
et al.
(
2022
)
Therapeutic Approach to Alzheimer's Disease: Current Treatments and New Perspectives
.
Pharmaceutics
14
,
[PubMed]
226.
Gupta
G.
and
Samant
N.
(
2021
)
Current druggable targets for therapeutic control of Alzheimer's disease
.
Contemporary Clin. Trials
109
,
227.
Imbimbo
B.
and
Watling
M.
(
2021
)
What have we learned from past failures of investigational drugs for Alzheimer's disease?
Expert Opin. Investig. Drugs
30
,
1175
1182
[PubMed]
228.
Karran
E.
and
De Strooper
B.
(
2022
)
The amyloid hypothesis in Alzheimer disease: new insights from new therapeutics
.
Nat. Rev. Drug Discovery
21
,
306
318
229.
Valiukas
Z.
,
Ephraim
R.
,
Tangalakis
K.
,
Davidson
M.
,
Apostolopoulos
V.
and
Feehan
J.
(
2022
)
Immunotherapies for Alzheimer's Disease-A Review
.
Vaccines
10
,
[PubMed]
230.
Haddad
H.
,
Malone
G.
,
Comardelle
N.
,
Degueure
A.
,
Kaye
A.
and
Kaye
A.
(
2022
)
Aducanumab, a Novel Anti-Amyloid Monoclonal Antibody, for the Treatment of Alzheimer's Disease: A Comprehensive Review
.
Health Psychol. Res.
10
,
231.
Kuller
L.
and
Lopez
O.
(
2021
)
ENGAGE and EMERGE: Truth and consequences?
Alzheimer's & Dementia: J. Alzheimer's Assoc.
17
,
692
695
232.
Alexander
G.
,
Emerson
S.
and
Kesselheim
A.
(
2021
)
Evaluation of Aducanumab for Alzheimer Disease: Scientific Evidence and Regulatory Review Involving Efficacy, Safety, and Futility
.
JAMA
325
1717
1718
233.
Andrews
J.
,
Desai
U.
,
Kirson
N.
,
Zichlin
M.
,
Ball
D.
and
Matthews
B.
(
2019
)
Disease severity and minimal clinically important differences in clinical outcome assessments for Alzheimer's disease clinical trials
.
Alzheimer's & Dementia (New York, N. Y.)
5
,
354
363
[PubMed]
234.
Knopman
D.
(
2023
)
Lecanemab reduces brain amyloid-β and delays cognitive worsening
.
Cell Reports Med.
4
,
[PubMed]
235.
Velazquez
P.
,
Cribbs
D.
,
Poulos
T.
and
Tenner
A.
(
1997
)
Aspartate residue 7 in amyloid beta-protein is critical for classical complement pathway activation: implications for Alzheimer's disease pathogenesis
.
Nat. Med.
3
,
77
9
[PubMed]
236.
Pavlovski
D.
,
Thundyil
J.
,
Monk
P.
,
Wetsel
R.
,
Taylor
S.
and
Woodruff
T.
(
2012
)
Generation of complement component C5a by ischemic neurons promotes neuronal apoptosis
.
FASEB J.: Off. Publ. Federation Am. Societies Exp. Biol.
26
,
3680
90
[PubMed]
237.
Hernandez
M.
,
Namiranian
P.
,
Nguyen
E.
,
Fonseca
M.
and
Tenner
A.
(
2017
)
C5a increases the injury to primary neurons elicited by fibrillar amyloid beta
.
ASN Neuro
9
,
[PubMed]
238.
Panayiotou
E.
,
Fella
E.
,
Andreou
S.
,
Papacharalambous
R.
,
Gerasimou
P.
,
Costeas
P.
et al.
(
2019
)
C5aR agonist enhances phagocytosis of fibrillar and non-fibrillar Aβ amyloid and preserves memory in a mouse model of familial Alzheimer's disease
.
PLoS ONE
14
,
239.
Hu
X.
,
Tomlinson
S.
and
Barnum
S.
(
2012
)
Targeted inhibition of complement using complement receptor 2-conjugated inhibitors attenuates EAE
.
Neurosci. Lett.
531
,
35
9
[PubMed]
240.
Alajangi
H.
,
Kaur
M.
,
Sharma
A.
,
Rana
S.
,
Thakur
S.
,
Chatterjee
M.
et al.
(
2022
)
Blood-brain barrier: emerging trends on transport models and new-age strategies for therapeutics intervention against neurological disorders
.
Mol. Brain
15
,
49
[PubMed]
241.
Henkel
J.
,
Beers
D.
,
Wen
S.
,
Bowser
R.
and
Appel
S.
(
2009
)
Decreased mRNA expression of tight junction proteins in lumbar spinal cords of patients with ALS
.
Neurology
72
,
1614
6
[PubMed]
242.
Zlokovic
B.
(
2008
)
The blood-brain barrier in health and chronic neurodegenerative disorders
.
Neuron
57
,
178
201
[PubMed]
243.
Alarcan
H.
,
Al Ojaimi
Y.
,
Lanznaster
D.
,
Escoffre
J.
,
Corcia
P.
,
Vourc'h
P.
et al.
(
2022
)
Taking Advantages of Blood-Brain or Spinal Cord Barrier Alterations or Restoring Them to Optimize Therapy in ALS?
J. Personalized Med.
12
,
1071
[PubMed]
244.
Kumar
R.
,
Claassen
D.
,
Mongan
A.
,
Hoehn
B.
,
Lin
P.
,
Cahir-Mcfarland
E.
et al.
(
2023
)
A Phase 2 Open-Label Study to Assess the Safety, Tolerability, Pharmacokinetics, and Pharmacodynamics of Intravenous ANX005 in Patients with, or at Risk of, Manifest Huntington's Disease (HD) (S32.009)
.
Neurology
245.
Zhang
W.
,
Chen
Y.
and
Pei
H.
(
2023
)
C1q and central nervous system disorders
.
Front. Immunol.
14
,
246.
(
2023
)
Biosciences A: Game-changing medicines for complement mediated diseases
.
247.
Dräger
N.
,
Sattler
S.
,
Huang
C.
,
Teter
O.
,
Leng
K.
,
Hashemi
S.
et al.
(
2022
)
A CRISPRi/a platform in human iPSC-derived microglia uncovers regulators of disease states
.
Nat. Neurosci.
25
,
1149
1162
[PubMed]
248.
Gasque
P.
,
Ischenko
A.
,
Legoedec
J.
,
Mauger
C.
,
Schouft
M.
and
Fontaine
M.
(
1993
)
Expression of the complement classical pathway by human glioma in culture. A model for complement expression by nerve cells
.
J. Biol. Chem.
268
,
25068
74
[PubMed]
249.
Kuraya
M.
,
Matsushita
M.
,
Endo
Y.
,
Thiel
S.
and
Fujita
T.
(
2003
)
Expression of H-ficolin/Hakata antigen, mannose-binding lectin-associated serine protease (MASP)-1 and MASP-3 by human glioma cell line T98G
.
Int. Immunol.
15
,
109
17
[PubMed]
250.
Walker
D.
and
McGeer
P.
(
1993
)
Complement gene expression in neuroblastoma and astrocytoma cell lines of human origin
.
Neurosci. Lett.
157
,
99
102
[PubMed]
251.
Veerhuis
R.
,
Janssen
I.
,
De Groot
C.
,
Van Muiswinkel
F.
,
Hack
C.
and
Eikelenboom
P.
(
1999
)
Cytokines associated with amyloid plaques in Alzheimer's disease brain stimulate human glial and neuronal cell cultures to secrete early complement proteins, but not C1-inhibitor
.
Exp. Neurol.
160
,
289
99
[PubMed]
252.
Gasque
P.
,
Julen
N.
,
Ischenko
A.
,
Picot
C.
,
Mauger
C.
,
Chauzy
C.
et al.
(
1992
)
Expression of complement components of the alternative pathway by glioma cell lines
.
J. Immunol. (Baltimore, Md: 1950)
149
,
1381
7
253.
Barnum
S.
,
Jones
J.
and
Benveniste
E.
(
1992
)
Interferon-gamma regulation of C3 gene expression in human astroglioma cells
.
J. Neuroimmunol.
38
,
275
82
[PubMed]
254.
Walker
D.G.
and
McGeer
P.L.
(
1992
)
Complement gene expression in human brain: comparison between normal and Alzheimer disease cases
.
Brain Res. Mol. Brain Res.
14
,
109
116
[PubMed]
255.
Walker
D.
,
Kim
S.
and
McGeer
P.
(
1998
)
Expression of complement C4 and C9 genes by human astrocytes
.
Brain Res.
809
,
31
8
[PubMed]
256.
Veerhuis
R.
,
Janssen
I.
,
Hoozemans
J.
,
De Groot
C.
,
Hack
C.
and
Eikelenboom
P.
(
1998
)
Complement C1-inhibitor expression in Alzheimer's disease
.
Acta Neuropathol. (Berl)
96
,
287
96
257.
Walker
D.
,
Yasuhara
O.
,
Patston
P.
,
McGeer
E.
and
McGeer
P.
(
1995
)
Complement C1 inhibitor is produced by brain tissue and is cleaved in Alzheimer disease
.
Brain Res.
675
,
75
82
[PubMed]
258.
Davoust
N.
,
Jones
J.
,
Stahel
P.
,
Ames
R.
and
Barnum
S.
(
1999
)
Receptor for the C3a anaphylatoxin is expressed by neurons and glial cells
.
Glia
26
,
201
11
[PubMed]
259.
Gasque
P.
,
Singhrao
S.
,
Neal
J.
,
Wang
P.
,
Sayah
S.
,
Fontaine
M.
et al.
(
1998
)
The receptor for complement anaphylatoxin C3a is expressed by myeloid cells and nonmyeloid cells in inflamed human central nervous system: analysis in multiple sclerosis and bacterial meningitis
.
J. Immunol. (Baltimore, Md: 1950)
160
,
3543
54
260.
Gasque
P.
,
Thomas
A.
,
Fontaine
M.
and
Morgan
B.
(
1996
)
Complement activation on human neuroblastoma cell lines in vitro: route of activation and expression of functional complement regulatory proteins
.
J. Neuroimmunol.
66
,
29
40
[PubMed]
261.
Yang
C.
,
Jones
J.
and
Barnum
S.
(
1993
)
Expression of decay-accelerating factor (CD55), membrane cofactor protein (CD46) and CD59 in the human astroglioma cell line, D54-MG, and primary rat astrocytes
.
J. Neuroimmunol.
47
,
123
32
[PubMed]
262.
Gordon
D.
,
Sadlon
T.
,
Wesselingh
S.
,
Russell
S.
,
Johnstone
R.
and
Purcell
D.
(
1992
)
Human astrocytes express membrane cofactor protein (CD46), a regulator of complement activation
.
J. Neuroimmunol.
36
,
199
208
[PubMed]
263.
Avery
V.
,
Adrian
D.
and
Gordon
D.
(
1993
)
Detection of mosaic protein mRNA in human astrocytes
.
Immunol. Cell Biol.
71
,
215
9
[PubMed]
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY). Open access for this article was enabled by the participation of Cardiff University in an all-inclusive Read & Publish agreement with Portland Press and the Biochemical Society under a transformative agreement with JISC.