Citation: Tara P. Hurst, Caroline Coleman-Vaughan, Indu Patwal, Justin V. McCarthy. Regulated intramembrane proteolysis, innate immunity and therapeutic targets in Alzheimer’s disease[J]. AIMS Molecular Science, 2016, 3(2): 138-157. doi: 10.3934/molsci.2016.2.138
[1] | Mariacristina De Luca, Kevin Pels, Susana Moleirinho, Graziella Curtale . The epigenetic landscape of innate immunity. AIMS Molecular Science, 2017, 4(1): 110-139. doi: 10.3934/molsci.2017.1.110 |
[2] | Peter X. Shaw, Travis Stiles, Christopher Douglas, Daisy Ho, Wei Fan, Hongjun Du, Xu Xiao . Oxidative stress, innate immunity, and age-related macular degeneration. AIMS Molecular Science, 2016, 3(2): 196-221. doi: 10.3934/molsci.2016.2.196 |
[3] | María Pilar Carrera-González, María Jesús Ramírez-Expósito, Carmen Guerrero-González, José Manuel Martínez-Martos . Alzheimer's disease: Is there a relationship between brain renin-angiotensin system, estradiol and glucose transporter-4 (GLUT-4)?. AIMS Molecular Science, 2023, 10(1): 37-51. doi: 10.3934/molsci.2023004 |
[4] | Rajkumar Bavaharini, Chaitanya Sree Somala, Konda Mani Saravanan, Thirunavukarasou Anand . Neurogranin in Alzheimer's Disease: Roles in synaptic function, pathology, and potential as a diagnostic biomarker. AIMS Molecular Science, 2024, 11(4): 330-350. doi: 10.3934/molsci.2024020 |
[5] | Rosanna Parlato, Holger Bierhoff . Role of nucleolar dysfunction in neurodegenerative disorders: a game of genes?. AIMS Molecular Science, 2015, 2(3): 211-224. doi: 10.3934/molsci.2015.3.211 |
[6] | Morgan Robinson, Brenda Yasie Lee, Zoya Leonenko . Drugs and drug delivery systems targeting amyloid-β in Alzheimer's disease. AIMS Molecular Science, 2015, 2(3): 332-358. doi: 10.3934/molsci.2015.3.332 |
[7] | Mairembam Stelin Singh, PV Parvati Sai Arun, Mairaj Ahmed Ansari . Unveiling common markers in COVID-19: ADAMTS2, PCSK9, and OLAH emerged as key differential gene expression profiles in PBMCs across diverse disease conditions. AIMS Molecular Science, 2024, 11(2): 189-205. doi: 10.3934/molsci.2024011 |
[8] | Marco Feligioni, Serena Marcelli, Erin Knock, Urooba Nadeem, Ottavio Arancio, Paul E. Fraser . SUMO modulation of protein aggregation and degradation. AIMS Molecular Science, 2015, 2(4): 382-410. doi: 10.3934/molsci.2015.4.382 |
[9] | Chisato Kinoshita, Koji Aoyama, Toshio Nakaki . microRNA as a new agent for regulating neuronal glutathione synthesis and metabolism. AIMS Molecular Science, 2015, 2(2): 124-143. doi: 10.3934/molsci.2015.2.124 |
[10] | Seidu A. Richard, Wu Min, Zhaoliang Su, Hua-Xi Xu . Epochal neuroinflammatory role of high mobility group box 1 in central nervous system diseases. AIMS Molecular Science, 2017, 4(2): 185-218. doi: 10.3934/molsci.2017.2.185 |
Abbreviations: Aβ, amyloid-β peptide; AD, Alzheimer’s disease; APP, amyloid precursor protein; APH-1, anterior pharynx-defective 1; CNS, central nervous system; CTD, carboxyl terminal domain; CCR2, chemokine (C-C motif) receptor 2; CSF-1, colony stimulating factor-1; COX, cyclooxygenase; ErbB4, epidermal growth factor receptor; FDA, federal drug administration; FAD, familial Alzheimer’s disease; GSK-3β, glycogen synthase kinase 3β; GWAS, genome-wide association studies; I-CLiPs, intramembrane-cleaving proteases; IL-1R1, interleukin-1 receptor, type I; IL-1RII, interleukin-1 receptor, type II; interleukin; IFN, interferon; ICD, intracellular domain; LPS, lipopolysaccharide; MCP-1/CCL2, monocytes chemotactic protein-1; MCI, mild cognitively impaired; MAC, membrane attack complex; NFTs, neurofibrillary tangles; NMDA, N-methyl-D-aspartate; NSAIDs, non-steroidal anti-inflammatory drugs; PEN-2, presenilin enhancer 2; p75NTR, p75 neurotrophin receptor; PS1, presenilin-1; PS2, presenilin-2; RIP, regulated intramembrane proteolysis; SLE, systemic lupus erythrematosis; SPP, signal peptide peptidase; TNFα, tumour necrosis factor-α; TNFR-I, tumour necrosis factor receptor type I; TLR, Toll-like receptor; TMD, transmembrane domain.
Alzheimer’s disease (AD) is an age-related dementia that is characterized by the presence of amyloid protein (Aβ) plaques formed from the cleavage of the amyloid precursor protein (APP), formation of neurofibrillary tangles (NFTs), neuroinflammation and neuronal loss. The cleavage of APP is a sequential two-step process, with the initial proteolysis by α- or β-secretase (BACE) releasing the APP ectodomain into the extracellular milieu (Figure 1). This is followed by a second cleavage event of the membrane-anchored carboxyl-terminal domains of APP by the γ-secretase proteases [1], generating an APP intracellular domain (AICD) and Aβ peptides of various sizes. Mutations in two related genes, PSEN1 and PSEN2, are associated with familial AD [2], and these genes were subsequently found to encode the highly homologous presenilin (PS1 and PS2) proteins, respectively [3]. Mutations in PS1 are associated with amyloidogenesis in AD from the outset, in particular with production of the longer cleavage product of APP, Aβ42 [3].
According to the amyloid hypothesis, aberrant accumulation or defective clearance of the Aβ peptides leads to plaque formation and the onset of AD pathology, reviewed in [4]. While the Aβ hypothesis is not the only mechanism proposed to explain the pathology of AD, it is clear that there is a role for the Aβ peptides in the disease and this is likely due to detrimental effects on neuronal survival as a result of immune responsiveness or disturbances in calcium signalling, reviewed in [5,6]. In addition, there is evidence that the presence of Aβ peptides could be causally linked to the formation of NFTs via activation of apoptotic signalling pathways [7]. Thus, the presence of excess Aβ peptides could lead to the formation of the NFTs, providing a causal link between these two hallmarks of AD pathogenesis and supporting the Aβ hypothesis. Since the accumulation of aggregated Aβ, generated as a result of γ-secretase activities, is associated with the pathogenesis and progression of disease, both the γ-secretase proteases and Aβ peptides serve as points of entry in the development of AD therapeutics. In this review, we discuss the γ-secretase complexes and highlight the emerging importance of the immune system in the pathogenesis of AD. The challenges of designing therapeutics to γ-secretase due to the plethora of substrates other than APP are discussed, along with promising new immunotherapies to the Aβ peptide that are in clinical trials.
The majority of AD cases are considered to be sporadic, with a non-Mendelian pattern of inheritance and a complex etiology [8,9]. However, genetic risk factors causing early- and late-onset FAD are well documented, where mutations in APP and both PS1 and PS2 are strongly associated with early-onset FAD [2]. In addition to several risk factors, including aging and prior traumatic brain injury, inheritable polymorphisms are causally linked to AD pathogenesis, as reviewed in [8,9]. The search for susceptibility loci makes use of population studies, such as genome-wide association studies (GWAS) and more recent, exome and genome sequencing studies to identify common genetic variants associated with sporadic late-onset AD, reviewed elsewhere [8,9,10]. These studies have helped identify other loci that have a strong, weak or rare AD risk-association, and have also helped to delineate some biological processes that may be involved in the pathogenesis of AD, including the innate immune system and inflammation, cholesterol metabolism and endosomal vesicle recycling [10,11]. So far, such studies have identified the ε4-allele of the apolipoprotein E (APOE) gene, CLU (which encodes clusterin), the sorting protein-related receptor gene (SORL1), https://www.aimspress.com/aimspress-data/aimsmoles/2016/2/PICALM (also known as CALM) encodes phosphatidylinositol-binding clathrin assembly protein, which is involved in clathrin-mediated endocytosis) and CR1 (which encodes the major receptor of C3b, a protein involved in complement activation) as a critical risk factor in sporadic late-onset AD [8,11]. It is therefore possible that the production of and response to Aβ peptides is associated with both sporadic and familial occurrences of AD.
Much more progress has been made in understanding the autosomal dominant inheritance of mutations in the presenilin genes that are associated with early-onset familial AD [2]. While this represents only a small proportion of AD patients, the research into FAD mutations has elucidated the molecular mechanisms of Aβ peptide production and Aβ peptides appear to be a common feature of both sporadic and familial AD. Although much is now known about the molecular mechanisms of Aβ production, the initial factor that leads to the release and subsequent accumulation of these peptides is not fully understood, although a genetic predisposition for aberrant APP cleavage is indicated. One hypothesis is that infections with viruses that replicate within cells of the central nervous system (CNS) could be a danger signal that triggers the onset of AD. Herpes viruses are able to remain latent in neurons for years until viral replication is reactivated by changes in the brain, such as those due to aging, reviewed in [12]. Reactivation of latent herpes simplex virus 1 (HSV1) has been found to cause local inflammation and neurotoxicity, as well as accumulation of Aβ [13], and HSV1 DNA is associated with Aβ plaques in AD patients [14]. HSV-1 infection has also been found to alter autophagosomal processing of Aβ and to inhibit the non-amyloidogenic pathway of APP cleavage [15]. Interestingly, incidences of sporadic AD were found to be associated with a cluster of genes in a GWAS [12] and this association was confirmed by meta-analyses of other GWASs [16]. The genes identified by these analyses were all known to be associated with viral infection and replication, particularly with that of herpes viruses, reviewed in [12,17]. There is thus some evidence for an association between reactivation of latent viruses and AD, underscoring the importance of the immune response in the pathogenesis of this disease.
The γ-secretase protease is a multi-protein complex consisting of PS1 or PS2, nicastrin, anterior pharynx defective-1 (APH-1) and presenilin enhancer-2 (PEN-2), reviewed in [18]. The unconditional requirement for each of these four integral membrane proteins for γ-secretase activity was verified following genetic ablation or RNAi knockdown of one or the other of the components, as well as by genetic reconstitution of γ-secretase activity in Saccharomyces cerevisiae, which lacks endogenous γ-secretase, [19]. All four proteins associate with each other and their co-expression resulted in increased γ-secretase activity in mammalian cells [20], Drosophila and S. cerevisiae, reviewed in [18,20,21]. The multi-protein identity of the γ-secretase proteases was subsequently corroborated by several studies reporting the purification of the active γ-secretase protease complexes [22,23,24]. Of these γ- secretase components, several variants exist of the presenilins (PS1 and PS2) and APH1 due to multiple genes and to alternative splicing thereof. In all species examined, there are two PSEN genes (PSEN1 and PSEN2); while in humans there are also two Aph-1 genes, Aph-1a and Aph-1b, which are alternatively spliced [25]. Further, in rodents, gene duplication of Aph-1b produces a third gene, Aph-1c [25]. Furthermore, it has now been demonstrated that multiple combinations of the four proteins can exist, depending upon which PSEN and APH1 gene product is included therein, and that γ-secretase complexes have different functions based on their precise co-factor composition since the gene products of PSEN and APH1 are not redundant [24,25,26].
While the presenilins were mainly examined in the context of APP cleavage and AD pathogenesis, they have been found to have many substrates other than APP, as well as biological functions independent of γ- secretase, reviewed elsewhere [27,28,29]. It is becoming apparent that the mechanism of cleavage of transmembrane proteins by membrane-embedded enzymes like γ-secretase is widely-employed in cell biology, with such enzymes classed as intramembrane-cleaving proteases or I-CLiPs, as reviewed in [30]. Further, this sequential proteolysis is now called regulated intramembrane proteolysis, and has emerged as a highly conserved signalling system that involves the cleavage of certain type I transmembrane proteins, reviewed in [31]. This mechanism allows the direct transfer of an extracellular signal into the cytoplasm or nucleus via the production of functional intracellular domains (ICDs), several of which have been found to regulate gene expression [32]. This novel mechanism is distinct from the classic membrane-to-nucleus signalling model via a cascade of post-translational modifications and intracellular messengers. To date, over 100 different γ-secretase substrates have been identified, including several cytokines and cytokine receptors TNFR1 [33], IL-1R1 [34], IL-1R2 [35], IL-6R [36], CX3CL1 and CXCL16 [37], indicating a generic role in the regulation of receptor-mediated signalling pathways and highlighting the importance of γ- secretase in the regulation of growth factor and cytokine signalling, reviewed in [29].
The link between γ-secretase and the immune system has been demonstrated by the phenotypic characterization of in vivo models of presenilin deletion or mutation. Since deletion of PSEN1 is embryonic lethal [38], transgenic mouse models have been developed to circumvent this effect, including PS1 null mice that have been rescued with a PS1 transgene [39] and brain region-specific gene knockout techniques based on the Cre/LoxP system [40,41,42]. In addition, the selective knockout of one or more alleles of the presenilins, with the retention of at least one functional copy, has been used since the early days of presenilin research to define its function with the Psen1+/−Psen2−/− mouse having the most severe reduction in presenilin alleles that permits post-natal survival [43]. The phenotype of Psen1+/−Psen2−/− “partial deficient” mice is normal up to approximately six months, when the majority of the mice develop skin and autoimmune defects similar to those observed in systemic lupus erythematosus (SLE) [44]. Thus, PS1 deficiency is associated with an abnormal immune response and the development of an autoimmune phenotype. The requirement for the presenilins in adaptive immune system function has since been demonstrated in lymphocytes [45,46,47]. Deficiency in presenilins antagonized T-cell homeostasis and signalling [45,46], while presenilin-deficient B-cells were defective in responsiveness to LPS and B-cell receptor-induced proliferation and signal transduction [47]. From these studies, it is clear that the presenilins have essential functions in the immune system in general, outside of the CNS. Within the brain, studies using conditional knockouts of the presenilins identified a role for it in negatively regulating the inflammatory response mediated by microglia [48,49]. Further, it was found that PS2, rather than PS1, was critical in inhibiting microglia-mediated inflammation, with IFNγ selectively up-regulating PS2 expression and with PS2 deficiency resulting in enhanced cytokine expression [49]. Interestingly, it has recently been found that TLR activation in macrophages regulates the γ-secretase-mediated cleavage of the colony-stimulating factor 1 (CSF-1) receptor, which is required for monocyte/macrophage differentiation [50]. Although this work was done in macrophages, a similar system could be in operation in microglia and the activation of TLRs in response to Aβ peptides could lead to microglial activation via promotion of CSF-1 receptor proteolysis by TACE and γ- secretase. From the data, a model is emerging whereby the functions of TLRs and γ-secretase in inflammation and microglial activation combine to regulate the inflammatory response in the brain in general and to Aβ peptides in particular. Dysfunction of this system could be fundamental to the pathophysiology of AD.
Further to this, γ-secretase-mediated cleavage of innate immune receptors has recently been shown to be important in cytokine signalling. The pro-inflammatory cytokine IL-1 and its receptors, IL-1RI and IL-1RII, have long been associated with AD [51]. While IL-1RI is essential for signal transduction to NF-κB activation following IL-1 binding, IL-1RII is a decoy receptor that binds to excess ligand and does not transmit a signal, as reviewed in [52]. Both of these receptors can be found in a membrane-bound and soluble form, both forms of which are known to have biological functions [52]. In an early study, elevated levels of the soluble form of IL-1RII were found in the cerebrospinal fluid of AD patients [51]. Thus, the finding that IL-1RII was subject to cleavage by α- and β- secretase followed by intramembrane proteolysis by γ-secretase [35] fits with the observed elevation of soluble Il-1RII in AD [51]. Work in our laboratory has shown that the type I IL-1R was also subject to cleavage by metalloproteases and γ-secretase [34]. We demonstrated that the IL-1RI ectodomain could be cleaved constitutively and that this cleavage was enhanced by activation of metalloproteases as well as exogenous addition of IL-1 [34]. Ectodomain shedding released a soluble IL-1RI into the extracellular milieu, while the remaining membrane-bound CTD was a substrate for further cleavage by γ-secretase to liberate an IL-1RI ICD into the cytoplasm. Importantly, we demonstrated that inhibition of γ-secretase activity antagonized IL-1-induced cytokine secretion, suggesting that γ-secretase regulated cytokine responsiveness [34].
The effect of the presenilins on IL-1 signalling could be mediated by its interaction with downstream signalling molecules. The over-expression of TNF receptor-associated factor 6 (TRAF6) or interleukin-1 receptor-associated kinase 2 (IRAK2) enhanced the cleavage of IL-1RI, suggesting that these adaptors up-regulated γ-secretase activity [34,53]. This observation is consistent with the finding that TRAF6 and IRAK2 are also novel interaction partners for the presenilins [34,54]. In addition to the effects on signalling adaptors and cascades, the γ-secretase cleavage of IL-1RI could directly affect gene expression. As with other γ-secretase substrates, it is feasible that the ICD of IL-1RI could act as a transcription factor. There are nuclear localization signals within the ICD and IL-1RI has previously been reported to localize to the nucleus [55]. It thus remains to be determined if the induction of cytokine gene expression following IL-1 treatment is due, in part, to ICD transactivation as well as to the usual signal transduction pathway. IL-1 signalling is complex, with numerous regulatory mechanisms, and it is possible that proteolysis of the receptors following ligand binding simply adds another level of regulation to this system. In addition to IL-1R, other cytokine receptors could be regulated by γ- secretase. There is evidence that TNFR1 and TNFR2 are subject to ectodomain shedding [56] and we recently demonstrated that TNFR1 is also cleaved by γ-secretase [33]. The physiological role of the presenilins in signalling pathways downstream of IL-1R and TNFR1 is not yet fully known but it is feasible that it acts as a scaffold or chaperone for adaptor proteins facilitating the spatial segregation of divergent signalling events arising from each receptor. Several lines of evidence suggest a role for the presenilins in trafficking within the cell and the presenilins are known to localize to the endosome [57,58]. Thus, it is possible that the presenilins help to recruit adaptors to such organelles and this could be critical for signal transduction given the endosomal localization of certain innate immune receptors, such as the TLRs in the phagosomes of microglia.
In addition to TNFR1, IL-1RI and IL1-RII, several additional γ-secretase substrates are receptors with important biological functions in the immune system. The type I interferons (IFNs) bind surface receptors (IFNARs) and induce a signalling cascade via the Janus kinase (JAK)-signal-transducer and activator of transcription [59] pathway to induce IFN-stimulated gene (ISG) expression, reviewed in [60]. At least one IFN receptor, IFNAR2, was found to be proteolytically cleaved by γ-secretase and the ICD was shown to repress gene transcription [61]. In addition to receptors, regulated intramembrane proteolysis of other cell surface transmembrane proteins has been described. There are a number of chemokines that are expressed in a transmembrane form, including CX3CL1 (fractalkine) and CXCL16, which are cleaved by α-secretase and γ-secretase [37]. Ectodomain shedding has been found to be required for chemokine recruitment of immune cells and in microglial activation, as reviewed in [62]. The mucin-type sialoglycoprotein, leukosialin or CD43, was cleaved during neutrophil activation in a γ-secretase-dependent manner and generation of the ICDs was found to be required for neutrophil adhesion [63]. Unlike in previous instances of regulated intramembrane proteolysis, CD43 is first cleaved by cathepsin G, not a metalloprotease [63]. The γ-secretase-generated CD44 ICD has been shown to localize to the nucleus of macrophages in which it promotes the activation of NF-κB and macrophage fusion [64]. Major histocompatibility class I (MHC I) proteins are found on most nucleated cells and are essential in many immune processes, including antigen presentation to T cells [65]. An MHC class I protein, human leukocyte antigen-A2 (HLA-A2) was found to be cleaved by α-secretase and then by γ-secretase [65]. Unlike other ICDs generated by γ-secretase cleavage, the HLA-A2 ICD was very unstable and rapidly degraded [65], supporting the hypothesis that regulated intramembrane proteolysis of some transmembrane proteins could serve to clear them from the membrane rather than in signal transduction. There is evidence that both ectodomain shedding and γ-secretase cleavage are critical in normal immune system function.
Although amyloid plaques in the brains of AD patients are primarily composed of Aβ, other molecules including activated complement fragments have been identified in close association with the plaques. In the CNS, components of the complement system are synthesized by microglia, astrocytes and neurons and contribute to the local inflammatory response, reviewed in [66,67]. There are conflicting reports of beneficial and detrimental effects of activation of complement in AD. A lot of this confusion arises from different AD mouse models used to study complement by either knocking out complement genes or using inhibitors of complement components, reviewed in [68]. For example, the complement component C1q has long been associated with AD since it was found to co- localize with amyloid deposits in the brains of AD patients [69]. In mouse models of AD (Tg2576 and APP/PS1), deletion of the C1q gene reduced the severity of AD neuropathology and this suggested that C1q expression was detrimental in AD [70]. However, the binding of Aβ peptides by C1q also enhanced phagocytosis of these peptides by microglia, which express the receptor for C1q and can thus bind to C1q-bound Aβ peptides [69]. Subsequently, C1q was found to help selectively remove apoptotic neurons and to suppress pro-inflammatory cytokine production in an in vitro model using primary rat neurons and microglia [71]. This suggests that C1q could be involved in a mechanism to limit the inflammatory response within the brain.
In order to resolve the conflicting roles of complement in AD, the model system must be chosen carefully. Importantly, the differences between mice and humans could limit the usefulness of mouse models in this area. For example, it has been found that Aβ42 peptides are also bound by the complement component C3b, which then binds to the complement receptor type I (CR1) on erythrocytes and is subsequently cleared from the circulation by macrophages in the liver [72]. Importantly, mice do not have complement receptors on their erythrocytes and thus would not utilize this mode of Aβ clearance, reviewed in [73]. Moreover, while mice have complement receptors on other cells, such as macrophages and neutrophils, murine complement is poorly activated by human Aβ peptides [74]. Thus, differences in murine and human complement biology must temper results of AD mouse models geared towards looking at complement activation. Interestingly, polymorphisms in the above-mentioned complement receptor type I (CR1) gene locus have also been identified as a risk factor for sporadic AD, reviewed in [8]. Further, herpes viruses are known to modulate complement function and to target complement- associated proteins (CR1 and APOJ) that are also linked to sporadic AD, reviewed in [12]. Complement is thus likely to be important in the clearance of Aβ peptides by promoting uptake by liver macrophages or by enhancing phagocytosis by microglia.
Microglia and astrocytes are the immune effector cells within the CNS and they are considered to be “activated” when they exhibit altered morphology and express specific marker proteins, such as IBA-1in the case of microglia [75]. The recruitment of microglia and astrocytes to the Aβ plaques results in localized inflammation around the plaques indicating that the glial cells are producing pro-inflammatory mediators following activation, reviewed in [76]. This is consistent with the finding that activation of microglia and, to a lesser extent, astrocytes is mediated by Toll-like receptors (TLRs) [77]. TLRs are normally activated in response to microbial ligands and TLR signalling results in the induction of cytokine and chemokine production, however TLRs have also been found to respond to endogenous ligands produced following tissue damage, which are referred to as danger-associated molecular patterns or DAMPs, reviewed in [78,79]. It is thus feasible that either the Aβ peptides, or other molecules produced as a result of the cytotoxic effect of Aβ on surrounding cells, are sensed as DAMPs by microglia and astrocytes. Microglia and astrocytes express TLRs, particularly TLRs 1-4 and TLR9, and TLR signalling is activated in response to uptake of Aβ peptides [77,79]. In particular, TLR2, TLR4 and TLR9 have been implicated in Aβ responsiveness since ligands for these TLRs enhanced phagocytic uptake of Aβ peptides [80,81,82]. TLRs are thus critical for activating glial cells and enhancing microglial phagocytosis, as further supported by research in AD mouse models. In one AD mouse model that was also homozygous for a destructive mutation of TLR4, increased diffuse and fibrillar Aβ deposits were observed that were not seen in TLR4 wild-type AD mice [82]. Also, the administration of an acute injection of the TLR4 ligand, lipopolysaccharide (LPS), in transgenic mice reduced Aβ plaque burden, suggesting that TLR4 activation facilitated Aβ uptake [83,84]. TLR2 is the most highly expressed TLR in microglia and a number of studies have found it to be critical for the response to Aβ peptides, in particular since microglia from TLR2−/− mice were unable to respond to Aβ42 [85,86,87]. Deficiency of TLR2 in knockout mice also accelerated cognitive decline [85]. Further, it has been found that Aβ peptides activate typical TLR signalling pathways via MyD88 and NF-κB [86]. In an AD mouse model, knock-down of MyD88 expression reduced inflammation while also accelerating cognitive deficits, indicating that TLR signalling via MyD88 had a protective function in AD [88]. It has also been found that an NF-κB-regulated microRNA (miRNA-146a) is upregulated in response to Aβ [89]. It is thus feasible that TLRs are involved in the sensing of Aβ by microglia and that TLR-induced signalling could be implicated in AD.
One outcome of TLR signalling is the induction of pro-inflammatory cytokine production and elevated levels of these cytokines are observed in response to Aβ peptides. In general, pro-inflammatory cytokines are produced in response to a stimulus but their production is curtailed by anti-inflammatory signals. In AD, it is hypothesized that inflammation, while initially beneficial, if sustained can result in a self-propagating cycle that contributes to neurodegeneration, reviewed in [90]. In transgenic mice expressing wild-type or mutant APP, Aβ plaques were associated with activated microglia and astrocytes, as well as increased levels of the cytokines TNFα, IL-1β and IL-6 [75]. Studies using human brain tissue and in vitro, cell-based systems have shown that Aβ-activated glial cells over- produce pro-inflammatory cytokines and that this can lead to the degeneration of neurons [91,92]. Further, the deletion of either of the cytokine receptors TNFR-I or interferon (IFN)-γ receptor type I in transgenic mouse models resulted in decreased Aβ plaque formation, inflammation and prevented learning and memory deficits [93]. Several studies have identified potentially pathological consequences of IL-1β signalling in AD, including increased expression and proteolysis of APP [94], tau phosphorylation [95] and secretion of phospholipase A2-IIA (sPLA2-IIA) from astrocytes [96]. While these studies indicate that pro-inflammatory cytokine production in response to Aβ peptides could exacerbate the symptoms of AD, other work has shown a protective effect of inflammation. Cytokines can stimulate uptake of Aβ by microglia that are otherwise unable to phagocytose the peptides in the large quantities seen in AD [97]. Additional to this, sustained overexpression of IL-1β in the hippocampus of APPswe/PS1ΔE9 mouse model of AD resulted in activated microglia and astrocytes, increased levels of TNFα and IL-6 and decreased amyloid plaque load by enhancing Aβ phagocytosis [98]. It is thus possible that the induction of cytokines does enhance Aβ uptake and clearance. However, the continued presence of Aβ peptides in the CNS, which could be sensed as DAMPs by microglial TLRs, could lead to a perpetual activation of the TLRs and continuous pro-inflammatory cytokine release. The failure to down-regulate cytokine production appears to be able to induce some of the effects associated with AD but there is also evidence for improved Aβ clearance following IL-1β stimulation.
The persistence of Aβ peptides is likely due to a defect in the uptake of these peptides by microglia, such as that observed in the brains of AD patients and transgenic AD mouse models [76,97]. It is feasible that the normal function of microglia is altered in AD by an unknown mechanism, particularly since bone-marrow-derived phagocytes recruited into the CNS following irradiation in mouse models are able to clear Aβ plaques [97]. It has been suggested that the plaques in human AD patients are more insoluble than those in AD mouse models, making it more difficult for the microglia to clear the plaques, reviewed in [99]. In a model referred to as “frustrated phagocytosis”, microglia respond to the presence of difficult-to-phagocytose material by enhanced secretion of inflammatory molecules, including potentially cytotoxic cytokines [99]. Further, a study found that inflammation and enhanced phagocytosis actually increased Aβ production [74]. Interestingly, IFNγ can stimulate γ-secretase activity within the phagosomes of macrophages and induce the cleavage of other phagosome proteins, such as CD44 [100]. This suggests that one effect of pro-inflammatory cytokine secretion is to enhance γ-secretase activity, ostensibly to activate phagocytosis but this could also have a detrimental effect in further enhancing APP cleavage. Thus, in AD, defective γ-secretase function could lead to abnormal phagocytosis and ineffectual clearance of Aβ peptides, as well as aberrant proteolysis of APP.
In addition to cytokines, activated microglia and astrocytes also secrete chemokines, proteins that recruit more immune cells to the site of inflammation; and up-regulation of chemokines and their receptors is associated with AD [97]. The deletion or inhibition of monocyte chemotactic protein-1 (MCP-1/CCL2) in an AD mouse model was associated with lower Aβ levels, as well as reduced microglia and astrocyte recruitment and cognitive impairment [101]. Consistent with this, when CCL2 was over-expressed in transgenic mice, it was found to have a deleterious effect, including increased cognitive impairment and Aβ production [101]. In contrast, deletion of the receptor (CC-chemokine receptor 2 or CCR2) for MCP-1/CCL2 in an AD mouse model had deleterious effects and promoted the rapid onset of disease symptoms and death [102]. The absence of the CCR2 impaired microglial activation and responsiveness to Aβ, which is hypothesized to account for the early onset of disease [102]. These findings appear to contradict one another but it has been suggested that they reflect distinct mechanisms of action. The CCR2 deficiency is associated with earlier onset of AD symptoms and this is hypothesized to be due to the role of CCR2 in recruiting macrophages from the circulation into the brain to clear Aβ plaques [101]. Thus, CCR2 plays a protective role in the early stages of AD by promoting the clearance of Aβ peptides, while the over-expression of MCP-1/CCL2 during the course of the disease could exacerbate AD symptoms. These results support the hypothesis of inflammation induced by microglia and astrocytes in response to Aβ accumulation.
While there is currently no cure for AD, several pharmacological and lifestyle-based therapies are available to manage the most common symptoms. Research into the underlying biochemical and genetic causes of AD have however allowed the development of therapeutics aimed at the molecular basis of the disease. During the last decade, the pharmaceutical industry has focused on reducing Aβ formation and preventing the deposition and accumulation of Aβ in amyloid plaques. Given the importance of γ-secretase in the production of Aβ40 and Aβ42, this enzyme has been the focus of intensive research in order to identify compounds that could inhibit or modulate its activity, reviewed in [103]. These potential therapeutics target active and allosteric sites of γ-secretase [104] or modify the substrate so as to reduce Aβ42 formation or aggregation, reviewed in [105]. Several small molecules can inhibit γ- secretase activity in vitro [106,107,108,109], however some studies have raised concerns that off-target effects associated with inhibition of γ-secretase cleavage of other substrates, including Notch, could interfere with critical cell signalling events in cellular homeostasis, haematopoiesis and cellular adhesion [110]. Nonetheless, some of these compounds have been put forward for clinical trials, the current state of which has been reviewed elsewhere [111,112,113]. One of the most promising AD therapeutics was the γ-secretase inhibitor semagacestat (LY450139), since results from initial clinical trials showed that it could cross the blood-brain barrier and reduce Aβ levels in the brain [114]. However, phase III clinical trials were halted in 2010 as a result of the drug worsening cognitive symptoms in patients, as well as being associated with a higher risk of developing skin cancer [114]. MK-0752 was also abandoned since Aβ levels rebounded and even exceeded baseline levels. Another prominent γ-secretase inhibitor, avagacestat (BMS-708163) [115], was discontinued during a second phase II trial when Bristol-Myers Squibb announced that it would halt all clinical development of avagacestat for AD. The recent failure of these and other compounds highlight the desperation of scientists to produce an AD therapeutic, but also highlights our limited understanding of the γ-secretase proteases as a therapeutic target in AD treatment [103,116]. The presence of several γ-secretase protease complexes, and multiple substrates means that off-target side effects or even the induction of new pathologies, such as skin cancers observed in the semagacestat trial, is likely [114,117]. This development is not surprising in light of the pleiotropic functions of γ-secretase, in particular the reported involvement of the presenilins and γ-secretase proteases in several cancers and with reduced PS1 levels associated with hyper-proliferation of the epidermis in mice [118]. One of the chief concerns with γ-secretase inhibitors is with respect to the concomitant inhibition of Notch cleavage, since this is essential in the differentiation of cell lineages in adults, such as the maturation of B- and T- lymphocytes, reviewed in [119]. Thus, there are fears that inhibition of γ-secretase proteases could impair vital biological processes even while inhibiting the cleavage of APP and Aβ formation [114].
Given the difficulties associated with inhibition of γ-secretase, other avenues for the treatment of AD are being pursued. One of the most promising is antibody-based immunotherapies, reviewed in [114,120,121]. Both active and passive immunotherapies have been investigated for their efficacy in clearing Aβ peptides from the blood and thereby reducing plaque sizes [122]. Active immunization involves the administration of Aβ peptides to the circulation in order to trigger an immune response, including the production of anti-Aβ antibodies. In contrast, passive immunization involves the delivery of anti-Aβ antibodies. The merit of Aβ immunotherapies is thought to lie in the removal of the Aβ peptides by either opsonization, which enhances phagocytosis, or the creation of a peripheral Aβ sink to withdraw peptides from aggregates within the brain [112]. An alternative mechanism is that the anti-Aβ monoclonal m266 sequestered monomeric Aβ peptides in the brain, preventing plaque formation, though it is not known what happens to the sequestered monomers [123].
Immunization with Aβ peptides was found to attenuate the pathology of AD in mouse models [124,125]. Aβ immunization has been tested in humans and some benefits have been observed in terms of reduced cerebral Aβ levels and stabilized or improved cognition [126]. While there is evidence of clinical benefit of these therapies, there have been complications with at least one trial reporting some cases of meningoencephalitis and without having any protective effect against the neurotoxic peptides [122]. Administration of the Aβ peptides generates an immune response that includes activation of T cells and can lead to inflammation [112]. The use of passive immunotherapies is thought to be safer and less likely to induce a T cell-mediated immune response [122]. Modifications of anti-Aβ antibodies for passive immunization have been reported to improve the safety of immunotherapy, including the use of antibody fragments [127,128] and humanized monoclonal antibodies [129]. Another alternative method of active immunization, that of Aβ-encoding DNA that is conjugated to gold particles and delivered by gene gun injection, has been found to elicit an antibody response without also inducing T cell proliferation or IFNγ and IL-17 secretion, thereby reducing the risk of inflammation in the brain [130]. A number of passive and active Aβ immunotherapeutic strategies are currently being tested in phase II and III clinical trials [122], including the humanized monoclonal antibody Bapineuzumab that produced disappointing results in two-phase III trials [85,131,132]. Developments in the area of Aβ therapeutics have been addressed in a number of recent reviews [112,120,121,133]. Overall antibody-based immunotherapies have failed to show a significant clinical benefit in patients with mild to moderate AD, leading to the proposal that therapeutic intervention is occurring too late in these patient groups and treatment should commence earlier when patients are at the asymptomatic stages of the disease. Likewise, these studies highlight the poorly understood relationship between the immune system and AD and encourage research aimed at increasing our understanding of the role of immune responses in AD and their impact on immunotherapy approaches in AD. Finally, evaluation of the efficacy of AD therapeutics is hindered by the lack of stage-specific biomarkers that are easy to measure. While Aβ peptides, plaque formation and hyperphosphorylated tau are reliable biomarkers, they are present in the CNS and so are monitored by either cerebrospinal fluid withdrawal or the use of brain imaging techniques [134]. These methods are invasive, costly and might not be useful predictive markers for the development of AD since they are only elevated following disease onset and not in all cases of AD [134]. Current research in AD biomarkers has identified differences in the immune system protein profiles of normal and AD patients and these proteins could be used to develop a test for the early stages of AD [134,135,136].
There is clearly a role for the immune response in AD, with markers of inflammation and complement activation being closely associated with the Aβ plaques. It is interesting to find that the critical enzyme implicated in AD is also important in immune system function and regulation. This suggests a close association between aberrant APP cleavage and immune system dysfunction in the AD brain since presenilin mutations could be the cause of both. While only a few cytokine receptors are yet known to be γ-secretase substrates, it is possible that more remain to be discovered. Further, a function is assumed but has not been demonstrated for the ICDs produced following γ-secretase cleavage. Given the gene transactivation observed with the ICDs of Notch and APP, it is feasible that the ICD of IL-1RI is also functional. This would be consistent with nuclear localization of this ICD. If this were a general mechanism employed by other innate immune receptors, it would greatly alter the current understanding of signalling via adaptor recruitment and cascades of post-translational modifications. The importance of γ-secretase in the immune response underscores the pleiotropic functions of this enzyme complex that render it difficult to treat AD by γ-secretase inhibitors. The best therapeutic prospects at the moment lie with modulation of γ-secretase activity by certain NSAIDs, as well as with the use of monoclonal antibodies to bind to excess Aβ peptides to remove them from the system.
The authors declare no conflict of interest.
This work was supported and funded by grants from Science Foundation Ireland (02/IN1/B218 and 09/IN.1/B2624) and the Irish Research Council for Science, Engineering and Technology (RS/2012/407). Due to space limitations, we apologize to authors whose relevant work we did not refer to directly.
[1] |
De Strooper B, Saftig P, Craessaerts K, et al. (1998) Deficiency of presenilin-1 inhibits the normal cleavage of amyloid precursor protein. Nature 391: 387-390. doi: 10.1038/34910
![]() |
[2] |
Jankowsky JL, Fadale DJ, Anderson J, et al. (2004) Mutant presenilins specifically elevate the levels of the 42 residue beta-amyloid peptide in vivo: evidence for augmentation of a 42-specific gamma secretase. Hum Mol Genet 13: 159-170. doi: 10.1093/hmg/ddh019
![]() |
[3] |
Clark RF, Hutton M, Fuldner M, et al. (1995) The structure of the presenilin 1 (S182) gene and identification of six novel mutations in early onset AD families. Nat Genet 11: 219-222. doi: 10.1038/ng1095-219
![]() |
[4] |
Hardy J, Selkoe DJ (2002) The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics. Science 297: 353-356. doi: 10.1126/science.1072994
![]() |
[5] |
Weksler ME, Gouras G, Relkin NR, et al. (2005) The immune system, amyloid-beta peptide, and Alzheimer's disease. Immunol Rev 205: 244-256. doi: 10.1111/j.0105-2896.2005.00264.x
![]() |
[6] |
Hass MR, Sato C, Kopan R, et al. (2009) Presenilin: RIP and beyond. Semin Cell Dev Biol 20: 201-210. doi: 10.1016/j.semcdb.2008.11.014
![]() |
[7] |
de Calignon A, Fox LM, Pitstick R, et al. (2010) Caspase activation precedes and leads to tangles. Nature 464: 1201-1204. doi: 10.1038/nature08890
![]() |
[8] |
Sleegers K, Lambert JC, Bertram L, et al. (2010) The pursuit of susceptibility genes for Alzheimer's disease: progress and prospects. Trends Genet 26: 84-93. doi: 10.1016/j.tig.2009.12.004
![]() |
[9] |
Bertram L, Tanzi RE (2008) Thirty years of Alzheimer's disease genetics: the implications of systematic meta-analyses. Nat Rev Neurosci 9: 768-778. doi: 10.1038/nrn2494
![]() |
[10] |
Selkoe DJ, Hardy J (2016) The amyloid hypothesis of Alzheimer's disease at 25 years. EMBO Mol Med2016: e201606210. doi: 10.15252/emmm.201606210
![]() |
[11] |
Jones L, Holmans PA, Hamshere ML, et al. (2010) Genetic evidence implicates the immune system and cholesterol metabolism in the aetiology of Alzheimer's disease. PLoS One 5: e13950. doi: 10.1371/journal.pone.0013950
![]() |
[12] |
Porcellini E, Carbone I, Ianni M, et al. (2010) Alzheimer's disease gene signature says: beware of brain viral infections. Immun Ageing 7: 16. doi: 10.1186/1742-4933-7-16
![]() |
[13] |
Wozniak MA, Itzhaki RF, Shipley SJ, et al. (2007) Herpes simplex virus infection causes cellular beta-amyloid accumulation and secretase upregulation. Neurosci Lett 429: 95-100. doi: 10.1016/j.neulet.2007.09.077
![]() |
[14] |
Wozniak MA, Mee AP, Itzhaki RF (2009) Herpes simplex virus type 1 DNA is located within Alzheimer's disease amyloid plaques. J Pathol 217: 131-138. doi: 10.1002/path.2449
![]() |
[15] |
Santana S, Recuero M, Bullido MJ, et al. (2012) Herpes simplex virus type I induces the accumulation of intracellular beta-amyloid in autophagic compartments and the inhibition of the non-amyloidogenic pathway in human neuroblastoma cells. Neurobiol Aging 33: 430.e419-433. doi: 10.1016/j.neurobiolaging.2010.12.010
![]() |
[16] |
Seshadri S, Fitzpatrick AL, Ikram MA, et al. (2010) Genome-wide analysis of genetic loci associated with Alzheimer disease. JAMA 303: 1832-1840. doi: 10.1001/jama.2010.574
![]() |
[17] |
Carter CJ (2010) Alzheimer's Disease: A Pathogenetic Autoimmune Disorder Caused by Herpes Simplex in a Gene-Dependent Manner. Int J Alzheimers Dis 2010: 140539. doi: 10.4061/2010/140539
![]() |
[18] |
Tolia A, De Strooper B (2009) Structure and function of γ-secretase. Semin Cell Dev Biol 20: 211-218. doi: 10.1016/j.semcdb.2008.10.007
![]() |
[19] |
Edbauer D, Winkler E, Regula JT, et al. (2003) Reconstitution of gamma-secretase activity. Nat Cell Biol 5: 486-488. doi: 10.1038/ncb960
![]() |
[20] |
Kimberly WT, LaVoie MJ, Ostaszewski BL, et al. (2003) Gamma-secretase is a membrane protein complex comprised of presenilin, nicastrin, Aph-1, and Pen-2. Proc Natl Acad Sci U S A 100: 6382-6387. doi: 10.1073/pnas.1037392100
![]() |
[21] |
Takasugi N, Tomita T, Hayashi I, et al. (2003) The role of presenilin cofactors in the gamma-secretase complex. Nature 422: 438-441. doi: 10.1038/nature01506
![]() |
[22] |
Fraering PC, Ye W, Strub JM, et al. (2004) Purification and characterization of the human gamma-secretase complex. Biochemistry 43: 9774-9789. doi: 10.1021/bi0494976
![]() |
[23] |
Winkler E, Hobson S, Fukumori A, et al. (2009) Purification, pharmacological modulation, and biochemical characterization of interactors of endogenous human gamma-secretase. Biochemistry 48: 1183-1197. doi: 10.1021/bi801204g
![]() |
[24] |
Sato T, Diehl TS, Narayanan S, et al. (2007) Active gamma-secretase complexes contain only one of each component. J Biol Chem 282: 33985-33993. doi: 10.1074/jbc.M705248200
![]() |
[25] |
Hebert SS, Serneels L, Dejaegere T, et al. (2004) Coordinated and widespread expression of gamma-secretase in vivo: evidence for size and molecular heterogeneity. Neurobiol Dis 17: 260-272. doi: 10.1016/j.nbd.2004.08.002
![]() |
[26] |
Shirotani K, Edbauer D, Prokop S, et al. (2004) Identification of distinct gamma-secretase complexes with different APH-1 variants. J Biol Chem 279: 41340-41345. doi: 10.1074/jbc.M405768200
![]() |
[27] |
Beel AJ, Sanders CR (2008) Substrate specificity of gamma-secretase and other intramembrane proteases. Cell Mol Life Sci 65: 1311-1334. doi: 10.1007/s00018-008-7462-2
![]() |
[28] |
Coen K, Annaert W (2010) Presenilins: how much more than gamma-secretase?! Biochem Soc Trans 38: 1474-1478. doi: 10.1042/BST0381474
![]() |
[29] |
McCarthy JV, Twomey C, Wujek P (2009) Presenilin-dependent regulated intramembrane proteolysis and γ-secretase activity. Cell Mol Life Sci 66: 1534-1555. doi: 10.1007/s00018-009-8435-9
![]() |
[30] |
Wolfe MS (2009) Intramembrane-cleaving Proteases. J Biol Chem 284: 13969-13973. doi: 10.1074/jbc.R800039200
![]() |
[31] |
Brown MS, Ye J, Rawson RB, et al. (2000) Regulated intramembrane proteolysis: a control mechanism conserved from bacteria to humans. Cell 100: 391-398. doi: 10.1016/S0092-8674(00)80675-3
![]() |
[32] |
McCarthy JV, Twomey C, Wujek P (2009) Presenilin-dependent regulated intramembrane proteolysis and gamma-secretase activity. Cell Mol Life Sci 66: 1534-1555. doi: 10.1007/s00018-009-8435-9
![]() |
[33] |
Chhibber-Goel J, Coleman-Vaughan C, Agrawal V, et al. (2016) gamma-Secretase Activity Is Required for Regulated Intramembrane Proteolysis of Tumor Necrosis Factor (TNF) Receptor 1 and TNF-mediated Pro-apoptotic Signalling. J Biol Chem 291: 5971-5985. doi: 10.1074/jbc.M115.679076
![]() |
[34] | Elzinga BM, Twomey C, Powell JC, et al. (2009) Interleukin-1 receptor type 1 is a substrate for gamma-secretase-dependent regulated intramembrane proteolysis. J Biol Chem 284: 1394-1409. |
[35] |
Kuhn PH, Marjaux E, Imhof A, et al. (2007) Regulated intramembrane proteolysis of the interleukin-1 receptor II by alpha-, beta-, and gamma-secretase. J Biol Chem 282: 11982-11995. doi: 10.1074/jbc.M700356200
![]() |
[36] |
Chalaris A, Gewiese J, Paliga K, et al. (2010) ADAM17-mediated shedding of the IL6R induces cleavage of the membrane stub by gamma-secretase. Biochim Biophys Acta 1803: 234-245. doi: 10.1016/j.bbamcr.2009.12.001
![]() |
[37] |
Schulte A, Schulz B, Andrzejewski MG, et al. (2007) Sequential processing of the transmembrane chemokines CX3CL1 and CXCL16 by alpha- and gamma-secretases. Biochem Biophys Res Commun 358: 233-240. doi: 10.1016/j.bbrc.2007.04.100
![]() |
[38] |
Donoviel DB, Hadjantonakis AK, Ikeda M, et al. (1999) Mice lacking both presenilin genes exhibit early embryonic patterning defects. Genes Dev 13: 2801-2810. doi: 10.1101/gad.13.21.2801
![]() |
[39] |
Xia X, Qian S, Soriano S, et al. (2001) Loss of presenilin 1 is associated with enhanced beta-catenin signalling and skin tumorigenesis. Proc Natl Acad Sci U S A 98: 10863-10868. doi: 10.1073/pnas.191284198
![]() |
[40] |
Guo Q, Fu W, Sopher BL, et al. (1999) Increased vulnerability of hippocampal neurons to excitotoxic necrosis in presenilin-1 mutant knock-in mice. Nat Med 5: 101-106. doi: 10.1038/4789
![]() |
[41] |
Feng R, Rampon C, Tang YP, et al. (2001) Deficient neurogenesis in forebrain-specific presenilin-1 knockout mice is associated with reduced clearance of hippocampal memory traces. Neuron 32: 911-926. doi: 10.1016/S0896-6273(01)00523-2
![]() |
[42] |
Wang R, Dineley KT, Sweatt JD, et al. (2004) Presenilin 1 familial Alzheimer's disease mutation leads to defective associative learning and impaired adult neurogenesis. Neuroscience 126: 305-312. doi: 10.1016/j.neuroscience.2004.03.048
![]() |
[43] |
Herreman A, Hartmann D, Annaert W, et al. (1999) Presenilin 2 deficiency causes a mild pulmonary phenotype and no changes in amyloid precursor protein processing but enhances the embryonic lethal phenotype of presenilin 1 deficiency. Proc Natl Acad Sci U S A 96: 11872-11877. doi: 10.1073/pnas.96.21.11872
![]() |
[44] |
Tournoy J, Bossuyt X, Snellinx A, et al. (2004) Partial loss of presenilins causes seborrheic keratosis and autoimmune disease in mice. Hum Mol Genet 13: 1321-1331. doi: 10.1093/hmg/ddh151
![]() |
[45] |
Maraver A, Tadokoro CE, Badura ML, et al. (2007) Effect of presenilins in the apoptosis of thymocytes and homeostasis of CD8+ T cells. Blood 110: 3218-3225. doi: 10.1182/blood-2007-01-070359
![]() |
[46] |
Laky K, Fowlkes BJ (2007) Presenilins regulate αβ T cell development by modulating TCR signalling. J Exp Med 204: 2115-2129. doi: 10.1084/jem.20070550
![]() |
[47] |
Yagi T, Giallourakis C, Mohanty S, et al. (2008) Defective signal transduction in B lymphocytes lacking presenilin proteins. Proc Natl Acad Sci U S A 105: 979-984. doi: 10.1073/pnas.0707755105
![]() |
[48] |
Beglopoulos V, Sun X, Saura CA, et al. (2004) Reduced beta-amyloid production and increased inflammatory responses in presenilin conditional knock-out mice. J Biol Chem 279: 46907-46914. doi: 10.1074/jbc.M409544200
![]() |
[49] |
Jayadev S, Case A, Eastman AJ, et al. (2010) Presenilin 2 Is the Predominant γ-Secretase in Microglia and Modulates Cytokine Release. PLoS ONE 5: e15743. doi: 10.1371/journal.pone.0015743
![]() |
[50] |
Glenn G, van der Geer P (2008) Toll-like receptors stimulate regulated intramembrane proteolysis of the CSF-1 receptor through Erk activation. FEBS Lett 582: 911-915. doi: 10.1016/j.febslet.2008.02.029
![]() |
[51] |
Garlind A, Brauner A, Hojeberg B, et al. (1999) Soluble interleukin-1 receptor type II levels are elevated in cerebrospinal fluid in Alzheimer's disease patients. Brain Res 826: 112-116. doi: 10.1016/S0006-8993(99)01092-6
![]() |
[52] |
Dinarello CA (2011) Interleukin-1 in the pathogenesis and treatment of inflammatory diseases. Blood 117: 3720-3732. doi: 10.1182/blood-2010-07-273417
![]() |
[53] |
Twomey C, Qian S, McCarthy JV (2009) TRAF6 promotes ubiquitination and regulated intramembrane proteolysis of IL-1R1. Biochem Biophys Res Commun 381: 418-423. doi: 10.1016/j.bbrc.2009.02.051
![]() |
[54] |
Powell JC, Twomey C, Jain R, et al. (2009) Association between Presenilin-1 and TRAF6 modulates regulated intramembrane proteolysis of the p75NTR neurotrophin receptor. J Neurochem 108: 216-230. doi: 10.1111/j.1471-4159.2008.05763.x
![]() |
[55] | Curtis BM, Widmer MB, deRoos P, et al. (1990) IL-1 and its receptor are translocated to the nucleus. J Immunol 144: 1295-1303. |
[56] |
Levine SJ (2008) Molecular Mechanisms of Soluble Cytokine Receptor Generation. J Biol Chem 283: 14177-14181. doi: 10.1074/jbc.R700052200
![]() |
[57] |
Frykman S, Hur JY, Franberg J, et al. (2010) Synaptic and endosomal localization of active gamma-secretase in rat brain. PLoS One 5: e8948. doi: 10.1371/journal.pone.0008948
![]() |
[58] | Meckler X, Checler F (2016) Presenilin 1 and Presenilin 2 Target gamma-Secretase Complexes to Distinct Cellular Compartments. J Biol ChemM115: 708297. |
[59] |
Doody RS, Raman R, Farlow M, et al. (2013) A phase 3 trial of semagacestat for treatment of Alzheimer's disease. N Engl J Med 369: 341-350. doi: 10.1056/NEJMoa1210951
![]() |
[60] |
Goodbourn S, Didcock L, Randall RE (2000) Interferons: cell signalling, immune modulation, antiviral response and virus countermeasures. J Gen Virol 81: 2341-2364. doi: 10.1099/0022-1317-81-10-2341
![]() |
[61] |
Saleh AZ, Fang AT, Arch AE, et al. (2004) Regulated proteolysis of the IFNaR2 subunit of the interferon-alpha receptor. Oncogene 23: 7076-7086. doi: 10.1038/sj.onc.1207955
![]() |
[62] |
Garton KJ, Gough PJ, Raines EW (2006) Emerging roles for ectodomain shedding in the regulation of inflammatory responses. J Leukoc Biol 79: 1105-1116. doi: 10.1189/jlb.0106038
![]() |
[63] |
Mambole A, Baruch D, Nusbaum P, et al. (2008) The cleavage of neutrophil leukosialin (CD43) by cathepsin G releases its extracellular domain and triggers its intramembrane proteolysis by presenilin/gamma-secretase. J Biol Chem 283: 23627-23635. doi: 10.1074/jbc.M710286200
![]() |
[64] |
Cui W, Ke JZ, Zhang Q, et al. (2006) The intracellular domain of CD44 promotes the fusion of macrophages. Blood 107: 796-805. doi: 10.1182/blood-2005-05-1902
![]() |
[65] |
Carey BW, Kim DY, Kovacs DM (2007) Presenilin/gamma-secretase and alpha-secretase-like peptidases cleave human MHC Class I proteins. Biochem J 401: 121-127. doi: 10.1042/BJ20060847
![]() |
[66] |
Bonifati DM, Kishore U (2007) Role of complement in neurodegeneration and neuroinflammation. Mol Immunol 44: 999-1010. doi: 10.1016/j.molimm.2006.03.007
![]() |
[67] |
Veerhuis R, Nielsen HM, Tenner AJ (2011) Complement in the brain. Mol Immunol 48: 1592-1603. doi: 10.1016/j.molimm.2011.04.003
![]() |
[68] |
Fonseca MI, Chu SH, Berci AM, et al. (2011) Contribution of complement activation pathways to neuropathology differs among mouse models of Alzheimer's disease. J Neuroinflammation 8: 4. doi: 10.1186/1742-2094-8-4
![]() |
[69] |
Webster SD, Galvan MD, Ferran E, et al. (2001) Antibody-Mediated Phagocytosis of the Amyloid β-Peptide in Microglia Is Differentially Modulated by C1q. J Immunol 166: 7496-7503. doi: 10.4049/jimmunol.166.12.7496
![]() |
[70] |
Fonseca MI, Zhou J, Botto M, et al. (2004) Absence of C1q leads to less neuropathology in transgenic mouse models of Alzheimer's disease. J Neurosci 24: 6457-6465. doi: 10.1523/JNEUROSCI.0901-04.2004
![]() |
[71] |
Fraser DA, Pisalyaput K, Tenner AJ (2010) C1q enhances microglial clearance of apoptotic neurons and neuronal blebs, and modulates subsequent inflammatory cytokine production. J Neurochem 112: 733-743. doi: 10.1111/j.1471-4159.2009.06494.x
![]() |
[72] |
Rogers J, Li R, Mastroeni D, et al. (2006) Peripheral clearance of amyloid beta peptide by complement C3-dependent adherence to erythrocytes. Neurobiol Aging 27: 1733-1739. doi: 10.1016/j.neurobiolaging.2005.09.043
![]() |
[73] |
Akiyama H, Barger S, Barnum S, et al. (2000) Inflammation and Alzheimer's disease. Neurobiol Aging 21: 383-421. doi: 10.1016/S0197-4580(00)00124-X
![]() |
[74] |
Spitzer P, Herrmann M, Klafki H-W, et al. (2010) Phagocytosis and LPS alter the maturation state of β-amyloid precursor protein and induce different Aβ peptide release signatures in human mononuclear phagocytes. J Neuroinflammation 7: 59-59. doi: 10.1186/1742-2094-7-59
![]() |
[75] |
Morgan D, Gordon MN, Tan J, et al. (2005) Dynamic complexity of the microglial activation response in transgenic models of amyloid deposition: implications for Alzheimer therapeutics. J Neuropathol Exp Neurol 64: 743-753. doi: 10.1097/01.jnen.0000178444.33972.e0
![]() |
[76] |
Streit WJ (2004) Microglia and Alzheimer's disease pathogenesis. J Neurosci Res 77: 1-8. doi: 10.1002/jnr.20093
![]() |
[77] | Trudler D, Farfara D, Frenkel D (2010) Toll-Like Receptors Expression and Signalling in Glia Cells in Neuro-Amyloidogenic Diseases: Towards Future Therapeutic Application. Mediators of Inflammation 2010: 12. |
[78] |
Kumar H, Kawai T, Akira S (2011) Pathogen recognition by the innate immune system. Int Rev Immunol 30: 16-34. doi: 10.3109/08830185.2010.529976
![]() |
[79] |
Carty M, Bowie AG (2011) Evaluating the role of Toll-like receptors in diseases of the central nervous system. Biochem Pharmacol 81: 825-837. doi: 10.1016/j.bcp.2011.01.003
![]() |
[80] |
Chen F, Hasegawa H, Schmitt-Ulms G, et al. (2006) TMP21 is a presenilin complex component that modulates [gamma]-secretase but not [epsiv]-secretase activity. Nature 440: 1208-1212. doi: 10.1038/nature04667
![]() |
[81] | Iribarren P, Chen K, Hu J, et al. (2005) CpG-containing oligodeoxynucleotide promotes microglial cell uptake of amyloid beta 1-42 peptide by up-regulating the expression of the G-protein- coupled receptor mFPR2. FASEBJ 19: 2032-2034. |
[82] |
Jin JJ, Kim HD, Maxwell JA, et al. (2008) Toll-like receptor 4-dependent upregulation of cytokines in a transgenic mouse model of Alzheimer's disease. J Neuroinflammation 5: 23. doi: 10.1186/1742-2094-5-23
![]() |
[83] |
Herber DL, Mercer M, Roth LM, et al. (2007) Microglial activation is required for Abeta clearance after intracranial injection of lipopolysaccharide in APP transgenic mice. J Neuroimmune Pharmacol 2: 222-231. doi: 10.1007/s11481-007-9069-z
![]() |
[84] |
DiCarlo G, Wilcock D, Henderson D, et al. (2001) Intrahippocampal LPS injections reduce Abeta load in APP+PS1 transgenic mice. Neurobiol Aging 22: 1007-1012. doi: 10.1016/S0197-4580(01)00292-5
![]() |
[85] |
Richard KL, Filali M, Prefontaine P, et al. (2008) Toll-like receptor 2 acts as a natural innate immune receptor to clear amyloid beta 1-42 and delay the cognitive decline in a mouse model of Alzheimer's disease. J Neurosci 28: 5784-5793. doi: 10.1523/JNEUROSCI.1146-08.2008
![]() |
[86] |
Jana M, Palencia CA, Pahan K (2008) Fibrillar amyloid-beta peptides activate microglia via TLR2: implications for Alzheimer's disease. J Immunol 181: 7254-7262. doi: 10.4049/jimmunol.181.10.7254
![]() |
[87] |
Frank S, Copanaki E, Burbach GJ, et al. (2009) Differential regulation of toll-like receptor mRNAs in amyloid plaque-associated brain tissue of aged APP23 transgenic mice. Neurosci Lett 453: 41-44. doi: 10.1016/j.neulet.2009.01.075
![]() |
[88] |
Michaud J-P, Richard KL, Rivest S (2011) MyD88-adaptor protein acts as a preventive mechanism for memory deficits in a mouse model of Alzheimer's disease. Mol Neurodegener 6: 5-5. doi: 10.1186/1750-1326-6-5
![]() |
[89] |
Cui JG, Li YY, Zhao Y, et al. (2010) Differential regulation of interleukin-1 receptor-associated kinase-1 (IRAK-1) and IRAK-2 by microRNA-146a and NF-kappaB in stressed human astroglial cells and in Alzheimer disease. J Biol Chem 285: 38951-38960. doi: 10.1074/jbc.M110.178848
![]() |
[90] |
Standridge JB (2006) Vicious cycles within the neuropathophysiologic mechanisms of Alzheimer's disease. Curr Alzheimer Res 3: 95-108. doi: 10.2174/156720506776383068
![]() |
[91] |
Lue LF, Walker DG, Rogers J (2001) Modeling microglial activation in Alzheimer's disease with human postmortem microglial cultures. Neurobiol Aging 22: 945-956. doi: 10.1016/S0197-4580(01)00311-6
![]() |
[92] | Yan Q, Zhang J, Liu H, et al. (2003) Anti-inflammatory drug therapy alters beta-amyloid processing and deposition in an animal model of Alzheimer's disease. J Neurosci 23: 7504-7509. |
[93] |
Yamamoto M, Kiyota T, Horiba M, et al. (2007) Interferon-γ and Tumor Necrosis Factor-α Regulate Amyloid-β Plaque Deposition and β-Secretase Expression in Swedish Mutant APP Transgenic Mice. Am J Pathol 170: 680-692. doi: 10.2353/ajpath.2007.060378
![]() |
[94] |
Kong Q, Peterson TS, Baker O, et al. (2009) Interleukin-1beta enhances nucleotide-induced and alpha-secretase-dependent amyloid precursor protein processing in rat primary cortical neurons via up-regulation of the P2Y(2) receptor. J Neurochem 109: 1300-1310. doi: 10.1111/j.1471-4159.2009.06048.x
![]() |
[95] |
Sheng JG, Zhu SG, Jones RA, et al. (2000) Interleukin-1 Promotes Expression and Phosphorylation of Neurofilament and tau Proteins in Vivo. Exp Neurol 163: 388-391. doi: 10.1006/exnr.2000.7393
![]() |
[96] |
Moses GS, Jensen MD, Lue LF, et al. (2006) Secretory PLA2-IIA: a new inflammatory factor for Alzheimer's disease. J Neuroinflammation 3: 28. doi: 10.1186/1742-2094-3-28
![]() |
[97] |
Rezai-Zadeh K, Gate D, Gowing G, et al. (2011) How to Get from Here to There: Macrophage Recruitment in Alzheimer's Disease. Curr Alzheimer Res 8: 156-163. doi: 10.2174/156720511795256017
![]() |
[98] |
Shaftel SS, Kyrkanides S, Olschowka JA, et al. (2007) Sustained hippocampal IL-1 beta overexpression mediates chronic neuroinflammation and ameliorates Alzheimer plaque pathology. J Clin Invest 117: 1595-1604. doi: 10.1172/JCI31450
![]() |
[99] |
Rogers J, Strohmeyer R, Kovelowski CJ, et al. (2002) Microglia and inflammatory mechanisms in the clearance of amyloid beta peptide. Glia 40: 260-269. doi: 10.1002/glia.10153
![]() |
[100] |
Jutras I, Laplante A, Boulais J, et al. (2005) Gamma-secretase is a functional component of phagosomes. J Biol Chem 280: 36310-36317. doi: 10.1074/jbc.M504069200
![]() |
[101] |
Kiyota T, Yamamoto M, Xiong H, et al. (2009) CCL2 Accelerates Microglia-Mediated Aβ Oligomer Formation and Progression of Neurocognitive Dysfunction. PLoS ONE 4: e6197. doi: 10.1371/journal.pone.0006197
![]() |
[102] |
El Khoury J, Toft M, Hickman SE, et al. (2007) Ccr2 deficiency impairs microglial accumulation and accelerates progression of Alzheimer-like disease. Nat Med 13: 432-438. doi: 10.1038/nm1555
![]() |
[103] |
De Strooper B, Chavez Gutierrez L (2015) Learning by failing: ideas and concepts to tackle gamma-secretases in Alzheimer's disease and beyond. Annu Rev Pharmacol Toxicol 55: 419-437. doi: 10.1146/annurev-pharmtox-010814-124309
![]() |
[104] | Beher D, Clarke EE, Wrigley JD, et al. (2004) Selected non-steroidal anti-inflammatory drugs and their derivatives target gamma-secretase at a novel site. Evidence for an allosteric mechanism. J Biol Chem 279: 43419-43426. |
[105] |
Kukar T, Golde TE (2008) Possible mechanisms of action of NSAIDs and related compounds that modulate gamma-secretase cleavage. Curr Top Med Chem 8: 47-53. doi: 10.2174/156802608783334042
![]() |
[106] |
Wong GT, Manfra D, Poulet FM, et al. (2004) Chronic treatment with the gamma-secretase inhibitor LY-411,575 inhibits beta-amyloid peptide production and alters lymphopoiesis and intestinal cell differentiation. J Biol Chem 279: 12876-12882. doi: 10.1074/jbc.M311652200
![]() |
[107] |
Jack C, Berezovska O, Wolfe MS, et al. (2001) Effect of PS1 deficiency and an APP gamma-secretase inhibitor on Notch1 signalling in primary mammalian neurons. Brain Res Mol Brain Res 87: 166-174. doi: 10.1016/S0169-328X(01)00010-9
![]() |
[108] |
Geling A, Steiner H, Willem M, et al. (2002) A gamma-secretase inhibitor blocks Notch signalling in vivo and causes a severe neurogenic phenotype in zebrafish. EMBO Rep 3: 688-694. doi: 10.1093/embo-reports/kvf124
![]() |
[109] | Micchelli CA, Esler WP, Kimberly WT, et al. (2003) Gamma-secretase/presenilin inhibitors for Alzheimer's disease phenocopy Notch mutations in Drosophila. FASEB J 17: 79-81. |
[110] |
Henley DB, Sundell KL, Sethuraman G, et al. (2014) Safety profile of semagacestat, a gamma-secretase inhibitor: IDENTITY trial findings. Curr Med Res Opin 30: 2021-2032. doi: 10.1185/03007995.2014.939167
![]() |
[111] |
Panza F, Frisardi V, Imbimbo BP, et al. (2010) REVIEW: gamma-Secretase inhibitors for the treatment of Alzheimer's disease: The current state. CNS Neurosci Ther 16: 272-284. doi: 10.1111/j.1755-5949.2010.00164.x
![]() |
[112] |
Frisardi V, Solfrizzi V, Imbimbo PB, et al. (2010) Towards disease-modifying treatment of Alzheimer's disease: drugs targeting beta-amyloid. Curr Alzheimer Res 7: 40-55. doi: 10.2174/156720510790274400
![]() |
[113] |
Bergmans BA, De Strooper B (2010) gamma-secretases: from cell biology to therapeutic strategies. Lancet Neurol 9: 215-226. doi: 10.1016/S1474-4422(09)70332-1
![]() |
[114] | Samson K (2010) NerveCenter: Phase III Alzheimer trial halted: Search for therapeutic biomarkers continues. Ann Neurol 68: A9-a12. |
[115] |
Gillman KW, Starrett JE, Jr., Parker MF, et al. (2010) Discovery and Evaluation of BMS-708163, a Potent, Selective and Orally Bioavailable gamma-Secretase Inhibitor. ACS Med Chem Lett 1: 120-124. doi: 10.1021/ml1000239
![]() |
[116] |
De Strooper B (2014) Lessons from a failed gamma-secretase Alzheimer trial. Cell 159: 721-726. doi: 10.1016/j.cell.2014.10.016
![]() |
[117] |
Cummings J (2010) What Can Be Inferred from the Interruption of the Semagacestat Trial for Treatment of Alzheimer's Disease? Biol Psychiatry 68: 876-878. doi: 10.1016/j.biopsych.2010.09.020
![]() |
[118] |
Qyang Y, Chambers SM, Wang P, et al. (2004) Myeloproliferative disease in mice with reduced presenilin gene dosage: effect of gamma-secretase blockage. Biochemistry 43: 5352-5359. doi: 10.1021/bi049826u
![]() |
[119] |
Imbimbo BP (2008) Therapeutic potential of gamma-secretase inhibitors and modulators. Curr Top Med Chem 8: 54-61. doi: 10.2174/156802608783334015
![]() |
[120] |
Pul R, Dodel R, Stangel M (2011) Antibody-based therapy in Alzheimer's disease. Expert Opin Biol Ther 11: 343-357. doi: 10.1517/14712598.2011.552884
![]() |
[121] |
Sarazin M, Dorothee G, de Souza LC, et al. (2013) Immunotherapy in Alzheimer's disease: do we have all the pieces of the puzzle? Biol Psychiatry 74: 329-332. doi: 10.1016/j.biopsych.2013.04.011
![]() |
[122] |
Brodaty H, Breteler MM, Dekosky ST, et al. (2011) The world of dementia beyond 2020. J Am Geriatr Soc 59: 923-927. doi: 10.1111/j.1532-5415.2011.03365.x
![]() |
[123] |
Yamada K, Yabuki C, Seubert P, et al. (2009) Abeta immunotherapy: intracerebral sequestration of Abeta by an anti-Abeta monoclonal antibody 266 with high affinity to soluble Abeta. J Neurosci 29: 11393-11398. doi: 10.1523/JNEUROSCI.2021-09.2009
![]() |
[124] |
Schenk D, Barbour R, Dunn W, et al. (1999) Immunization with amyloid-[beta] attenuates Alzheimer-disease-like pathology in the PDAPP mouse. Nature 400: 173-177. doi: 10.1038/22124
![]() |
[125] |
Lemere CA, Spooner ET, Leverone JF, et al. (2003) Amyloid-beta immunization in Alzheimer's disease transgenic mouse models and wildtype mice. Neurochem Res 28: 1017-1027. doi: 10.1023/A:1023203122036
![]() |
[126] |
Fu HJ, Liu B, Frost JL, et al. (2010) Amyloid-beta immunotherapy for Alzheimer's disease. CNS Neurol Disord Drug Targets 9: 197-206. doi: 10.2174/187152710791012017
![]() |
[127] | Robert R, Dolezal O, Waddington L, et al. (2009) Engineered antibody intervention strategies for Alzheimer's disease and related dementias by targeting amyloid and toxic oligomers. Protein Eng Des Sel 22: 199-208. |
[128] |
Marin-Argany M, Rivera-Hernandez G, Marti J, et al. (2011) An anti-Abeta (amyloid beta) single-chain variable fragment prevents amyloid fibril formation and cytotoxicity by withdrawing Abeta oligomers from the amyloid pathway. Biochem J 437: 25-34. doi: 10.1042/BJ20101712
![]() |
[129] |
Panza F, Frisardi V, Imbimbo BP, et al. (2011) Anti-beta-amyloid immunotherapy for Alzheimer's disease: focus on bapineuzumab. Curr Alzheimer Res 8: 808-817. doi: 10.2174/156720511798192718
![]() |
[130] |
Lambracht-Washington D, Qu BX, Fu M, et al. (2011) DNA immunization against amyloid beta 42 has high potential as safe therapy for Alzheimer's disease as it diminishes antigen-specific Th1 and Th17 cell proliferation. Cell Mol Neurobiol 31: 867-874. doi: 10.1007/s10571-011-9680-7
![]() |
[131] |
Salloway S, Sperling R, Brashear HR (2014) Phase 3 trials of solanezumab and bapineuzumab for Alzheimer's disease. N Engl J Med 370: 1460. doi: 10.1056/NEJMc1402193
![]() |
[132] |
Salloway S, Sperling R, Fox NC, et al. (2014) Two phase 3 trials of bapineuzumab in mild-to-moderate Alzheimer's disease. N Engl J Med 370: 322-333. doi: 10.1056/NEJMoa1304839
![]() |
[133] |
Krishnamurthy PK, Sigurdsson EM (2011) Therapeutic applications of antibodies in non-infectious neurodegenerative diseases. New Biotechnol 28: 511-517. doi: 10.1016/j.nbt.2011.03.020
![]() |
[134] |
Britschgi M, Wyss-Coray T (2009) Blood protein signature for the early diagnosis of Alzheimer disease. Arch Neurol 66: 161-165. doi: 10.1001/archneurol.2008.530
![]() |
[135] |
Lindberg C, Chromek M, Ahrengart L, et al. (2005) Soluble interleukin-1 receptor type II, IL-18 and caspase-1 in mild cognitive impairment and severe Alzheimer's disease. Neurochem Int 46: 551-557. doi: 10.1016/j.neuint.2005.01.004
![]() |
[136] |
Reddy MM, Wilson R, Wilson J, et al. (2011) Identification of Candidate IgG Biomarkers for Alzheimer's Disease via Combinatorial Library Screening. Cell 144: 132-142. doi: 10.1016/j.cell.2010.11.054
![]() |
1. | Aonghus J. McCarthy, Caroline Coleman-Vaughan, Justin V. McCarthy, Regulated intramembrane proteolysis: emergent role in cell signalling pathways, 2017, 45, 0300-5127, 1185, 10.1042/BST20170002 | |
2. | Caroline Coleman-Vaughan, Arijit Mal, Abhijit De, Justin V. McCarthy, 2017, Chapter 3, 978-981-10-6140-0, 47, 10.1007/978-981-10-6141-7_3 |