Next Article in Journal
An Anti-Invasive Role for Mdmx through the RhoA GTPase under the Control of the NEDD8 Pathway
Previous Article in Journal
Regulation of Enterocyte Brush Border Membrane Primary Na-Absorptive Transporters in Human Intestinal Organoid-Derived Monolayers
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

β-Amyloids and Immune Responses Associated with Alzheimer’s Disease

by
Elizaveta Kolobova
1,2,
Irina Petrushanko
1,
Vladimir Mitkevich
1,
Alexander A Makarov
1 and
Irina L Grigorova
1,2,*
1
Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
2
Institute of Translational Medicine, Pirogov Russian National Research Medical University, 117513 Moscow, Russia
*
Author to whom correspondence should be addressed.
Cells 2024, 13(19), 1624; https://doi.org/10.3390/cells13191624
Submission received: 2 September 2024 / Revised: 24 September 2024 / Accepted: 25 September 2024 / Published: 28 September 2024
(This article belongs to the Section Cellular Aging)

Abstract

:
Alzheimer’s disease (AD) is associated with the accumulation of β-amyloids (Aβs) and the formation of Aβ plaques in the brain. Various structural forms and isoforms of Aβs that have variable propensities for oligomerization and toxicity and may differentially affect the development of AD have been identified. In addition, there is evidence that β-amyloids are engaged in complex interactions with the innate and adaptive immune systems, both of which may also play a role in the regulation of AD onset and progression. In this review, we discuss what is currently known about the intricate interplay between β-amyloids and the immune response to Aβs with a more in-depth focus on the possible roles of B cells in the pathogenesis of AD.

1. Introduction

Alzheimer’s disease (AD) is a neurodegenerative disease characterized by progressive neuronal death and cerebral atrophy, and, as of today, is the most common neurodegenerative pathology [1]. The two major factors associated with AD are senile plaques formed from β-amyloids (Aβs) and neurofibrillary tangles formed by hyperphosphorylated tau proteins accumulating in AD patients’ brains. Several different models (such as the amyloid hypothesis, tau hyperphosphorylation, chronic inflammation, genetic theories, the microbiotic theory, etc.) aimed at explaining the onset of AD have been proposed (discussed in detail in reviews [2,3,4,5,6,7,8,9]). Currently, the amyloid cascade hypothesis, which suggests that the abnormal accumulation of Aβ may be the main trigger of the disease, is the most popular [10]. AD is currently viewed not as an isolated pathology of the central nervous system but as a systemic disease with important roles being played by both the innate and adaptive immune systems, as has been suggested by multiple lines of evidence [11,12,13,14,15,16,17,18,19]. However, the interactions between one of the main actors of the progression of AD, Aβ, and the innate and adaptive immune responses, as well as the effect their interplay has on the development and progression of the disease, are not fully understood.
While the accumulation of extracellular Aβ plaques in the brain is characteristic of AD, Aβ can exist in various structural forms (e.g., monomers, oligomers, and fibrillar structures), which differ in solubility and can have different effects on the pathogenesis of AD [20,21,22]. In addition, there are some mutations [23,24,25,26,27,28,29] and post-translational modifications of the Aβ N-terminus that could affect the rate of pathology development (Table 1) [30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49]. Various forms of Aβ are liable to differ in their propensity for triggering neurotoxicity and neuroinflammation, both of which may affect the accumulation of Aβ in the brain [31,35,39,40,41,43,45,46]. Accumulating Aβ can also trigger an adaptive immune response by T cells and B cells, and an autoantibody response that may in turn affect the progression of the disease (Figure 1) [17,50,51,52,53,54]. However, the propensity of various Aβ structures to trigger immune responses that may in turn promote or impede AD development and associated pathology is poorly understood. In this review, we highlight what is currently known about various forms of Aβ and their ability to trigger pathology, as well as immune responses. We also provide more in-depth analysis of the B cells’ role in the regulation of AD progression. Based on an analysis of the published data, we attempt to outline the gaps in the current understanding of the Aβ–immune system interactions and their role in AD.
The accumulation of various forms of β-amyloid in the brain parenchyma leads to the activation of resident innate immune cells (astrocytes and microglia) that then promote chronic neuroinflammation and AD-associated pathology. This neuroinflammation can be either further augmented by B cells that secrete pro-inflammatory interleukins (e.g., IL-6, IL-12, IL-15), or suppressed by regulatory B cells that produce anti-inflammatory factors (e.g., IL-10 and IL-35). IL-35 may also inhibit the expression of β-secretase, thereby limiting the production of Aβ from APP and accumulation of β-amyloids. Neuroinflammation can be also positively or negatively regulated by T cells that gain access into the brain parenchyma.
In AD patients, Aβ can induce T cell responses that may be influenced by B cells in antigen-dependent or independent ways. Different forms of β-amyloid may drain from the brain into the SLO, where they may be acquired by dendritic cells (DCs) that could stimulate Aβ-specific T cells that recognize MHC/Aβ peptide complexes presented on DCs via their T cell receptors (TCRs). β-amyloids also activate Aβ-specific B cells by interactions with their B cell receptors (BCRs). Activated B cells engulf BCR/antigen complexes, proteolyze them, and, similarly to DCs, can present MHCII/peptide complexes for specific recognition by helper T cells. These interactions may promote the stimulation of Aβ-specific helper T cells and acquisition of T cell help by the antigen-presenting B cells leading to the generation of Aβ-specific memory B cells and long-lived plasma cells.
Aβ-specific autoantibodies secreted by plasma cells may access the brain parenchyma directly and/or interfere with β-amyloid peptide transport from the blood and lymphatic systems into the brain. In these ways, Aβ-specific antibodies may qualitatively and quantitatively alter the composition of β-amyloid deposits in the brain and possibly modulate the progression of AD.

2. The Role of β-Amyloid and Its Forms in the Pathogenesis of AD

The molecular basis for the occurrence of AD is currently ambiguous. However, the disease is associated with the accumulation of Aβ fibrillary plaques consisting of Aβ peptides [55]. Aβ peptides are formed by the β- and γ-secretase-dependent endoproteolytic cleavage of amyloid-β precursor protein (APP), which is expressed in neurons located in the brain, glial cells, endothelial cells, and platelets [56]. The length of the resulting peptides varies from 37 to 43 amino acid residues [55,57,58], but the predominating forms are Aβ1–40 and Aβ1–42 [59,60]. Although Aβ is a normal component of biological fluids (in picomolar concentrations) [61], abnormalities in Aβ accumulation and aggregation are associated with AD. For example, mutations in the PSEN1 (presenilin-1) and PSEN2 (presenilin-2) genes alter the proteolytic cleavage of APP, thereby provoking the accumulation of Aβ1–42, a peptide more prone to aggregation than Aβ1–40 [62]. In addition, specific mutations in the genes encoding APP are a risk factor for the development of AD at young to middle ages, likely due to the increased propensity of Aβ mutants to aggregate [24,26,27] (Table 1).
Table 1. The most frequent/well-studied Aβ mutations and post-translational modifications in the metal-binding region (N-terminus) of Aβ that affect its oligomerization and aggregation.
Table 1. The most frequent/well-studied Aβ mutations and post-translational modifications in the metal-binding region (N-terminus) of Aβ that affect its oligomerization and aggregation.
Mutation in AβEffects
A2V Increased Aβ production [63,64]
Acceleration of Aβ aggregation [64,65,66]
English (H6R)Increased Zn2+-dependent oligomerization of Aβ [23]
Increased neurotoxicity of Aβ oligomers [24,25]
Acceleration of Aβ fibril formation [26]
Tottori (D7N)Acceleration of Aβ aggregation [27]
Increased oligomerization of Aβ [24]
Increased neurotoxicity of Aβ oligomers [24]
Acceleration of Aβ fibril formation [26]
Taiwan (D7H)Increased Aβ production [28,29]
Increased Aβ42/40 ratio [28]
Increased Zn2+/Cu2+-dependent oligomerization of Aβ [28,29]
Increased neurotoxicity of Aβ oligomers [28,29]
Post-translational modification of AβEffects
Iso-D7-Aβ (isomerized Asp7 residue)Increased Zn2+-dependent oligomerization of Aβ [34,36,38,43,48,49]
Increased neurotoxicity of Aβ oligomers [43]
Age-related accumulation [44]
3pE-Aβ(pyroglutamate modified glutamic acid residue)Acceleration of Aβ aggregation [31,46]
Increased oligomerization of Aβ [31,45,47]
Increased neurotoxicity of Aβ oligomers [31,35,45,46]
Age-related accumulation [32]
pS8-Aβ(phosphorylated Ser8 residue)Acceleration of Aβ aggregation [39,40,41]
Increased oligomerization of Aβ [39,40,41]
Increased neurotoxicity of Aβ oligomers [39,40,41]
Age-related accumulation in APP-PS1 tg mice [39,40]
Inhibition of Zn2+-dependent oligomerization of Aβ [30]
Prevention of Na+/K+ ATPase inhibition [30]
Reduction of amyloid plaques in APP-PS1 tg mice [30,37]
Excessive accumulation of β-amyloid in the brains of AD patients may be caused by increased production of Aβ. Alternatively, Aβ buildup might occur due to an insufficiently rapid rate of its clearance from the extracellular space by phagocytosis (mediated by astrocytes and glia cells) and degradation (by proteases secreted by astrocytes) [14,67,68]. Another process affecting Aβ brain concentration is its transportation between the brain, bloodstream, and lymphatic system. Any change in the dynamic equilibrium maintained by the production, removal, and transportation of Aβ can lead to its accumulation and aggregation [69,70] (Figure 1). However, the extent to which each of the above processes influences the development of AD remains unclear.
Aβ is present in mammals in various forms, including the monomeric, dimeric, oligomeric, and fibrillar structures, which contribute in various ways to the pathology leading to AD. For an extended period, AD research has focused on senile plaques in the brain that mainly consist of insoluble Aβ aggregates in the extracellular space. The formation and accumulation of plaques was originally thought to cause vascular damage, neuronal cell death, and progression of dementia [71]. However, neuronal death has been observed in patients’ brain regions devoid of plaques, suggesting no direct correlation between the manifestation of the disease and the number of plaques. Moreover, senile plaques have been found in people without clear manifestations of cognitive impairment. These findings suggest that insoluble Aβ aggregates are not likely to be the main cause for neurodegeneration [20]. More recent studies have linked cognitive impairment to the presence of large amounts of soluble Aβ oligomers [21,22], the most toxic of Aβ species, which leads to cell necrosis or apoptosis through various mechanisms.

2.1. Possible Mechanisms of Aβ Oligomer Toxicity

Based on studies comparing the effects of various types of Aβ aggregates on cellular membranes (reviewed in [72]), Aβ oligomers are generally considered to be more cytotoxic than fibrils. Soluble Aβ oligomers may contribute to neuronal death in a number of different ways including the disruption of cellular membranes, dysregulation of ion flows, pathological engagement of membrane receptors, induction of oxidative stress, and AD-related target protein phosphorylation [73]. Importantly, post-translational modifications of Aβ (Table 1) can significantly alter its cytotoxic effects [33].

2.1.1. Disruption of Cellular Membranes

Under certain conditions, Aβ oligomers can form pores in cellular membranes, thereby disrupting their ion permeability [74]. Some studies suggest that Aβ monomers and dimers can directly insert themselves into membranes, where they can form toxic pores by interacting with Aβ oligomers in the membrane [75]. In this respect, it is important to take note of the lipid-chaperone hypothesis (LCH), which suggests that the transport of intrinsically disordered proteins (IDPs) from the aqueous phase into the lipid bilayer may be facilitated by their binding to free phospholipids in the aqueous phase [76]. The LCH model suggests that lipid dysregulation may be contributing to the formation of toxic pores by Aβ peptides, as well as other IDPs (including islet amyloid polypeptides (IAPPs), a-synuclein, amylin, and β-synuclein) that are associated with various human diseases [77,78,79]. While the LCH hypothesis requires further experimental validation, taken that the ɛ4 allele of apolipoprotein E (the main extracellular lipid and cholesterol transporter in the brain that could also bind Aβ [80]) is a prominent genetic risk factor for AD [81], the possible role of lipid association with Aβ and oligomers should not be disregarded.

2.1.2. Dysregulation of Ion Flows

Disruption in ion homeostasis is one of the key factors in AD [82,83,84]. Impaired Na+/K+ [85,86,87,88] and calcium homeostasis [89] in AD has been observed not only in neurons, but also in other cell types [90]. The disturbance of ion homeostasis in Aβ-exposed cells may be caused by impaired membrane barrier properties (as discussed above) and/or due to the direct interaction of Aβ with ion-transporting proteins and receptors regulating ion entry (reviewed in [91]). Aβ has been shown to directly bind and inhibit Na+/K+ ATPases, leading to an impaired sodium–potassium gradient in neuronal cells [85,87,92,93]. This results in the disruption of normal neuronal activity, calcium overload, and cell death [85,87,88,94,95]. In addition, Aβs promote the activation of N-methyl-D-aspartate (NMDA) receptors leading to calcium entry and an increase in its intracellular concentration [96]. Aβ oligomers have also been shown to increase mitochondrial Ca2+ [97], thereby promoting mitochondrial dysfunction, cell death [98], and synaptic failure (reviewed in [99]).

2.1.3. Pathological Engagement of Membrane Receptors

Some of the cytotoxic effects occur as a consequence of Aβ interaction with multiple membrane receptors [100], including the NR2B-containing NMDA receptor [101]; α7-nicotinic acetylcholine receptor [102,103]; PrPc receptor, that may regulate the synaptic function of hippocampal neurons [104,105,106]; the receptor for advanced glycation end products (RAGE), which carries Aβ from the blood to the brain across the blood–brain barrier [107]; and other receptors [100].
These receptors can bind monomeric, oligomeric, and fibrillar forms of Aβ [100]. Moreover, both Aβ monomers and oligomers can interact with some receptors [100], likely contributing to both their physiological and pathological functions. Another possibility is that Aβ oligomerization occurs following its binding to surface receptors, as has been proposed for Aβ interactions with Na+/K+ ATPases [30]. The oligomerization of Aβ seeded on Na+/K+ ATPases leads to the inhibition of the enzyme’s activity, thereby disrupting the normal functioning of neurons [85,93].
Importantly, the interactions of Aβ isoforms containing some post-translational modifications with membrane receptors differ from those of its unmodified form, which leads to increased neurotoxicity [108,109] (Table 1).

2.1.4. Induction of Oxidative Stress

In addition to the scenarios described above, Aβ oligomers have been shown to promote cytotoxicity by inducing oxidative stress in neuronal cells, leading to, among other things, lipid peroxidation and the disruption of cell membrane barrier properties [110,111]. The precise molecular mechanism of oxidative stress induction in neurons by Aβ is unclear. However, it is at least partially mediated by decreased intracellular glutathione, which is the main thiol and cellular antioxidant [112,113,114]. Alterations in intracellular redox status lead to redox-dependent modifications of proteins (nitrosylation and glutathionylation) and changes in their functions [115,116,117]. For example, increased nitrosylation of cyclin-dependent kinase 5 (Cdk5) that is associated with AD promotes Cdk5 activation and contributes to NMDAR-mediated neuronal dendritic spine loss that is induced by Aβ [118]. Importantly, post-translational modifications of Aβ can significantly alter the effect that Aβ has on the redox status of cells [43,108,119,120] (Table 1).

2.1.5. AD-Related Target Protein Phosphorylation

In addition to the accumulation of Aβ, the hyperphosphorylation of tau proteins that promotes the formation of intracellular neurofibrillary tangles and neuritic dystrophy is characteristic for AD. However, a number of studies suggest that accumulation of the Aβ oligomers precedes the hyperphosphorylation of tau proteins in AD (reviewed in ([10])). Importantly, Aβ1–42 oligomers have been shown to induce the increased phosphorylation of several target proteins in cultures of neuronal cell lines (also characteristic for AD), including the hyperphosphorylation of tau proteins and other proteins involved into microtubule (MT) assembly [121,122,123]. While the mechanism of this regulation is not fully understood, Aβ has been shown to alter the activities of phosphatidylinositol-3-kinase, Akt, and protein kinase C (PKC) that, through their downstream target glycogen synthase kinase-3β (GSK3β), may regulate the phosphorylation of tau proteins [124].
Of specific interest is that some common post-translational modifications of Aβ promote the increased phosphorylation of proteins in neuronal cell lines, including the MT-regulation-associated tau protein, tubulins, and matrin 3 [123].
Based on the studies described above, various forms of Aβ can differentially affect mechanisms that regulate cytotoxicity, with Aβ oligomers and some post-translational modifications of Aβ likely pathologically contributing to the development of neurotoxicity associated with AD.

2.2. Scenarios of Aβ Oligomerization and Post-Translational Modifications

Aβ oligomers can form in various ways. One possible scenario is for Aβ peptides, which are folded into two parallel β-sheets (residues 12–24 and 30–40), to form oligomers via intermolecular interactions with each other within the 25–29 bended regions, which subsequently assemble into Aβ fibrils and β-fold structures [125,126]. Inside these fibrillar structures, the middle and C-terminus regions of Aβ peptides (that could be tentatively divided into Aβ12–29 and Aβ30–40) are located within the protein complexes, while their N-termini are exposed [53,126,127]. Other scenarios of Aβ structural oligomerization occur in the presence of transition metal ions (e.g., Zn2+, Cu2+, Fe2+, Al3+) and depend on Aβ mutations, post-translational modifications, as well as other factors [29,128,129]. Aβ1–42 is among the most aggregation-prone peptides that forms oligomers, and is a major component of amyloid plaques [130,131,132]. In addition, some post-translational modifications of Aβ peptides are known to accelerate their oligomerization (Table 1).
One of the most common post-translational modifications of β-amyloids is Aβ with an isomerized Asp7 residue (Iso-D7-Aβ) [133] (Table 1). The Iso-D7-Aβ form possesses a significantly higher propensity for oligomerization than Aβ1–42, and depends on the binding of zinc ions to the Aβ1–16 metal-binding domain [34,36,38,43,48,49]. The coordination of Zn2+ promotes the dimerization and oligomerization of Aβ peptides via segment Aβ11–14 [134]. The Iso-D7-Aβ isoform has been shown to induce greater neurotoxicity in cells compared to intact Aβ1–42 [43]. In transgenic nematodes (that express human Aβ), Iso-D7-Aβ induces the zinc-dependent oligomerization of endogenous Aβ, which leads to a significant reduction in animal lifespan [135]. It also accelerates cerebral amyloidosis upon intravenous administration into mouse models of AD [136].
The presence of Iso-D7-Aβ in the brains of AD patients was first described in 1993 [137]. The isoform was detected both in extracellular Aβ deposits and in cerebral blood vessels [133,138]. The amount of Iso-D7-Aβ in the brain was shown to be positively correlated with patients’ ages [44], suggesting either age-dependent increases in the formation of the isomerized form of Aβ [139] or a decrease in its deisomerization, degradation, phagocytosis, or transport. Based on the increased ability of Iso-D7-Aβ to promote Aβ oligomerization and induce neurotoxicity, age-dependent accumulation of Iso-D7-Aβ in the brain and blood may contribute to AD development.
In addition to Iso-D7-Aβ, there are other post-translational modifications within the Aβ1–16 metal-binding domain that may influence the development of AD (Table 1). One of the most well-studied is the pyroglutamate-modified form of Aβ (3pE-Aβ), which is truncated of the first two amino-acids. 3pE-Aβ also accumulates in the brain tissue with age [32]. It is generally accepted that 3pE-Aβ accelerates the formation of Aβ oligomers [31,45,47] and Aβ aggregation [31,46], and increases Aβ toxicity to neurons [31,35,45,46] (Table 1). Another post-translational modification that has been reported to exert an effect on Aβ oligomerization is Aβ phosphorylated on the serine 8 residue (pS8-Aβ) (Table 1). There is some evidence that pS8-Aβ increases the oligomerization and aggregation of Aβ, as well as the neurotoxicity of Aβ oligomers [39,40,41]. However, other studies indicate the inhibition of Zn2+-dependent oligomerization [30] and reduced amyloid deposition in APP-PS1 transgenic mice upon intravenous administration of pS8-Aβ [37] (Table 1 and Table 2).
In addition to post-translational modifications, the differential activity of α/β/ɣ-secretases and other enzymes can generate shortened forms of Aβ that may also influence AD pathogenesis [147].
While various forms of Aβ accumulate in humans in the process of normal aging and in AD patients, the understanding of their interaction with the innate immune system and ability to promote neuroinflammation and regulate progression of the disease is currently limited.

3. The Interaction of Aβ with the Innate Immune System and the Pathogenesis of Alzheimer’s Disease

Neuroinflammation is believed to play a major role in the pathology associated with AD [15,148,149,150]. Protein aggregates, dead neurons, and various pathogens can trigger the innate immune cells of the brain (microglia and astrocytes) to migrate to the site of damage and initiate neuroinflammation [13,14,15,19,151]. Importantly, neuroinflammation may have a dual function by playing a neuroprotective role during the acute phase of the immune response, but contributing to pathological processes during chronic activation of the involved cells [152]. It is possible that chronic activation of microglia and astrocytes is supported by cell death induced by soluble Aβ oligomers and Iso-D7-Aβ or other post-translationally modified forms of Aβ (Figure 1, Table 1). Neutrophils and other phagocytes may also potentially contribute to AD-associated neuroinflammation while regulating Aβ aggregation and accumulation. On one hand, neutrophils could inhibit Aβ aggregation and neurotoxicity by secreting neutrophil granule proteins [153]. On the other hand, they may contribute to oxidative stress that negatively affects Aβ phagocytosis [154,155].
An analysis of brain tissue RNA from AD patients and controls suggested an elevated signature of monocytes, MO macrophages, and dendritic cells and a decreased presence of resting NK cells in AD patients [156]. In addition, reduced prevalence and cytotoxicity of NK cells in AD patients have been reported [157]. In that respect, of interest is the ongoing phase II trial of SNK01 as an AD therapy (where patients’ NK cells are expanded and activated ex vivo), which has shown promising results in phase I trials [158]. Further studies are desirable to better dissect the role of NK cells in the regulation of inflammation and development/progression of AD. In addition, while Aβ has been shown to impact both monocytes and neutrophils, whether Aβ could also exert a direct action on NK cells remains unclear (reviewed in [159]).
Overall, the relationship between neuroinflammation and the accumulation of Aβ in AD is very complex, as in some cases the phagocytosis of Aβ by innate immune cells [160] may also promote their inflammatory response, while in other cases neuroinflammation appears to trigger the accumulation of Aβ [148].
In addition to the well-established role of innate immunity in the pathogenesis of AD, there is evidence suggesting the involvement of the adaptive immune system in regulating the disease. This regulation may depend, at least in part, on the antigen-specific response of T and B lymphocytes to Aβ that has been detected in humans and will be discussed below.

4. The Interaction of Aβ with the Adaptive Immune System and the Pathogenesis of Alzheimer’s Disease

Antigens from the brain parenchyma can enter the lymphatic system and, by means of perivascular drainage, the glymphatic system, or lymphatic drainage of cerebrospinal fluid (CSF), reach the deep cervical lymph nodes [161,162], where they can activate antigen-specific T and B cells (Figure 1). Thus, there is a possibility that Aβ peptides and their soluble multimeric forms in the brain may enter cervical lymph nodes and induce the activation of Aβ-specific lymphocytes. An adaptive immune response to Aβ may also take place in other secondary lymphoid organs (SLOs) in the case of more systemic spread of the autoantigens.

4.1. Aβ-Specific T Cells in AD

Possible mechanisms of T cell participation in AD onset and progression have been addressed in multiple studies [11,16,17]. T cells with an activated phenotype accumulate in the brain parenchyma of AD patients (as well as in some mouse models of AD) [163,164,165], suggesting their likely contribution to the regulation of AD. The accumulating T cells are enriched in the hippocampal region, where the main histologic changes associated with AD are manifested, including the excessive deposition of β-amyloid and hyperphosphorylation of tau proteins, as well as neuronal death [166]. A recent study also showed an accumulation of T cell receptor (TCR) clonal CD8 effector memory T cells (TEMRA) in the CSF of AD patients, some of which had specificity to the Epstein–Barr virus [167]. Locally present effector T cells are likely to promote neuroinflammation in the brain. However, in some cases they may limit neuroinflammation [17,50,51]. In addition, neuroinflammation should be negatively controlled by the regulatory T cells that represent about 10–30% of all CD4+ T cells in a normal human brain [168]. Which subpopulations of T cells play the dominant role in immunoregulation in the brain of patients with AD, and what their antigen specificity is, is still unclear (Figure 1).
Consistent with the Aβ-mediated activation of antigen-specific lymphocytes, increased reactivity to Aβ in CD4 and CD8 T cells derived from AD patients has been reported [169,170,171,172]. One of these studies suggested that Aβ1–42 is more immunogenic for human T cells than Aβ1–40 and, at the same time, that Aβ16–30 epitopes elicit the highest number of T cell clones responding to it [171]. However, the role of Aβ-specific T cells in the pathogenesis of AD in humans is unclear.
Based on studies in various murine models of AD (Table 2), Aβ-specific CD4 T cells can crosstalk with microglia and either promote or impede the development of the disease [50,173]. Aβ-specific T cells generated in mice have been shown to induce both pro-inflammatory activation (for Th1 and Th17 cells) and more regulatory action (for Th2 cells) in cocultured mixed glial cells [51]. When adoptively transferred into older (6–7 months old) APP/PS1 transgenic mice (Table 2), Aβ-specific T cells infiltrated mouse brains, where IFNγ-producing Th1-like T cells promoted the activation of glia and deposition of Aβ, thereby significantly accelerating disease development [174]. Interestingly, the infection of APP/PS1 mice with Bordetella pertussis induced significant infiltration of the endogenous IFNγ- and IL-17-producing T cells in the brains of mice, also eliciting glial inflammation and Aβ deposition [175].
However, in another mouse model of AD, Aβ-specific IFNγ-producing Th1 cells facilitated the clearance of Aβ deposits by microglia. In this model, APP-Tg J20 mice (Table 2) were directly immunized with the Aβ10–24 peptide in adjuvant. While immunization with the immunodominant Aβ peptide generated a robust Aβ-specific Th1 response and no Aβ-specific Abs, T cells gained access into the brains only in APP Tg mice crossed to SJL mice expressing IFN-γ in the central nervous system (CNS) under a myelin basic protein (MBP) promoter (Table 2). The immunization of 9-month-old APP/IFN-γ double Tg mice led to an accumulation of CD4+ T cells and macrophages in the hippocampus, and to some extent in the cortex and cerebellum, resulting in temporary meningoencephalitis, the activation of microglia, and elevated expression of the CD86 costimulatory molecule by microglia at the sites of Aβ plaques [146]. It should be noted that immunization with Aβ1–42 in an adjuvant also resulted in meningoencephalitis in about 6% of AD patients in a human clinical trial [176]. The Th1-induced pro-inflammatory response in the brains of APP/IFN-γ mice lasted for a month and led to the enhanced clearance of Aβ due to glial cell uptake [146]. Similar effects were observed in the 5XFAD mouse model (Table 2), where Aβ-specific CD4+ TH1 cells abrogated AD-like pathology via a subpopulation of microglia that expressed the major histocompatibility complex class II (MHC II) [50].
Overall, the studies described above suggest that in some mouse models Aβ-specific T cells can access the brain where they interact with local glia and microglia and play a role in the AD-like disease development. However, why IFN-γ-producing Aβ-specific Th1 cells are associated with enhanced Aβ deposition in some mouse models and increased clearance of Aβ plaques in other models is unclear. In addition, whether mouse-model-derived conclusions about Aβ-specific T cells may be relevant to human AD pathology remains an open question.
To summarize all of the above, while elevated levels of Aβ-specific T cells have been detected in the blood and an accumulation of T cells (including some clonally expanded CD8 T cells) was found in the brains and CSF of patients with AD, the presence of Aβ-specific CD4 and CD8 T cells in the brains of AD patients has not been yet reported. Moreover, the function of Aβ-specific T cells in the regulation of brain inflammation, Aβ deposition, as well as cytotoxic responses in human AD patients remains to be established. In addition to the more direct mechanisms of action described above, Aβ-specific CD4+ T cells may be involved in providing help to Aβ-specific B cells that may also play an important role in the regulation of AD.

4.2. Aβ-Specific and Other B Cells in AD

A number of studies have reported the presence of IgG autoantibodies to Aβ in the blood and CSF of both healthy volunteers and AD patients [127,177,178,179,180]. This indicates the involvement of Aβ-specific B cells in the immune response, during which they differentiate into plasma cells secreting autoantibodies to Aβ (Figure 1). While the activation of Aβ-specific B cells and formation of Aβ-specific autoantibodies may play a role in the onset and progression of AD, their interplay with various structural forms of Aβ in the regulation of AD is ambiguous.

4.2.1. Aβ-Specific Antibodies

One of the important questions for AD diagnostics and the analysis of its regulation is whether AD patients have an elevated or reduced proportion of autoantibodies to Aβ. Interestingly, some works reported reduced titers of IgG to Aβ1–42 in AD patients compared to healthy donors [177,179], while no difference was detected in other studies [127,180]. The observed discrepancy may be due to different methods of Aβ-specific antibody detection or differences between the AD groups selected for analysis [127,177,179,180].
As indicated above, β-amyloid can exist in the body in various multimeric forms. All forms may contribute to the pathogenesis of AD and induce a humoral response to different epitopes of β-amyloid formations [127,181]. Antibodies can target Aβ peptides at the N-terminal, the middle, and the C-terminal sites. Additional structural epitopes for antibody binding may arise or become hidden due to Aβ oligomerization and aggregation. For example, the increased formation of fibrillar Aβ may lead to decreased surface exposure of the middle and C-terminus regions of engaged Aβ peptides [127]. Consistent with this hypothesis, one study reported increased autoantibodies to Aβ1–12 and decreased Aβ19–30- and Aβ25–36-specific IgG in the blood and CSF of AD patients compared to healthy donors and patients with preclinical AD. Moreover, increased antibody titers to the N-terminus of Aβ correlated with increased amyloid burden in the brain and cognitive decline in AD patients [127]. The observed increase in autoantibodies to the N-terminus of Aβ likely reflects the accumulation of fibrillar amyloids in AD patients, in which the N-terminus is accessible while other Aβ regions are more buried for B cell activation and the formation of new autoantibodies.
How the availability of autoantibodies to different epitopes of Aβ influences the development of AD has been examined in the PDAPP mouse model of AD [53] (Table 2). PDAPP transgenic mice were immunized with a T cell epitope from ovalbumin bound to either different N-terminal Aβ peptides (Aβ1–5, Aβ3–9, or Aβ5–11), or to a fragment derived from the middle region of the peptide (Aβ15–24). The immunization of mice with each of the three N-terminal peptides significantly reduced the amount of amyloid deposits in the brain. In addition, Aβ3–9 and Aβ5–11 significantly reduced neuronal dystrophy and death. In contrast, immunization with an antigen including Aβ15–24 did not reduce amyloid load and did not protect against neuronal dystrophy or death [53]. The length of Aβ3–9 and Aβ5–11 peptides is insufficient for their presentation in complex with major histocompatibility complexes (MHCI and/or MHCII) and T cell activation. On this basis, it can be hypothesized that in this mouse model of AD, the antibodies to the N-terminus, but not to the mid-domain epitopes, influence the amount of Aβ deposited in the brain [53]. While immunization with complexes that include epitopes of N-terminal Aβ peptides had a therapeutic effect on AD mice [52,53], studies performed on the immunization of AD patients with antigens containing these Aβ epitopes did not have a positive effect on the course of the disease [182,183].
In addition to the studies described above, the functional activity of antibodies to different epitopes of Aβ was investigated for a number of monoclonal antibodies (mAbs) developed for AD therapy (including Bapinezumab, Lecanemab, Crenezumab, Ponezumab, Gantenerumab, and Solanezumab), which can bind to monomeric, oligomeric, or fibrillar forms of Aβ1–42 [184]. In studies conducted on mice, these drugs exerted therapeutic effects without causing significant side effects. Moreover, it has been demonstrated that, when passing through the blood–brain barrier (BBB), anti-Aβ mAbs may inhibit the toxic oligomerization of Aβ, promote the phagocytic clearance of microglia plaques in brain parenchyma [185], and/or exert anti-inflammatory effects by reducing the production of pro-inflammatory cytokines [186]. However, testing of these mAbs, including Bapineuzumab (a mAb specific to the Aβ1–5 epitope), Lecanemab (a mAb specific to the N-terminus of Aβ), and Solanezumab or Crenezumab (mAbs specific to the middle-domain Aβ16–26 or Aβ13–24 epitopes) in clinical trials has not led to any significant improvement or very minor positive effects (for Lecanemab) in patients’ conditions. Moreover, some of these drugs (e.g., Bapineuzumab) resulted in life-threatening side effects such as vasogenic edema, microbleeding, and cerebral ischemia [184]. It is believed that the occurrence of such side effects may be caused by the binding of antibodies to β-amyloid deposits in the blood vessels of the brain [187].
There are at least two possible explanations for the modest effects observed after the administration of mAbs to Aβ in humans with AD. First, it is possible that better control of pathogenic Aβ oligomers by mAbs cannot reverse pathological neurodegeneration during the later stages of disease progression (when AD is usually detected in human patients). Second, the difference in the effects observed in mice and humans could be due to more difficulty in antibodies accessing the brain parenchyma from the blood in humans compared to mice, even with impaired permeability of the BBB induced by the development of AD [188]. In that case, it is conceivable that if Aβ-specific plasma cells infiltrate the brain, the local production of antibodies to Aβ could have a greater impact on the course of AD. It should be noted that a postmortem analysis of brain sections showed increased amounts of IgA in the brain parenchyma of AD patients [189]. However, the local presence of plasma cells and specificity of the antibodies in the brain parenchyma of AD patients is still unclear.
It is also unclear whether AD-afflicted and/or normal elderly patients, who often accumulate elevated levels of Iso-D7-Aβ and 3pE-Aβ in the brain, have autoantibodies that specifically recognize these forms of Aβ. The role of the Iso-D7-Aβ-specific mAb with no cross-reactivity to the intact form of Aβ has been examined once on the 5xFAD mouse model of AD [190] (Table 2). Administration of this mAb resulted in a reduction in the total amount of unmodified Aβ in the brains of mice to the same extent as the administration of a control mAb specific to the N-terminus of Aβ. Moreover, in both cases, an improvement in cognitive function was observed in the mice as compared to the control group [139]. In addition, multiple studies have been devoted to the development of antibodies or vaccines selective against 3pE-Aβ. To date, there are monoclonal mouse antibodies (TAP01, 9D5, 3B8, 07/01, BAMB31, mE8) capable of binding both oligomers [191,192,193] and larger deposits of 3pE-Aβ in plaques [192,194]. Some of these antibodies exerted positive effects on cognitive function [191,192,195] and reduced the deposition of amyloids in various mouse models of AD, while causing no significant side effects in mice [191,192,193,194,195,196,197]. Subsequently, a humanized IgG1 mAb to 3pE-Aβ (Donanemab) has been developed based on the murine mE8-IgG2a. Donanemab has shown positive results in trials in AD patients, especially at the early stages of the disease. Although the safety of this drug has been questionable [198,199], Donanemab was recently approved for use by the United States Food and Drug Administration [200].
Based on the fact that Iso-D7-Aβ and 3pE-Aβ are peculiar triggers of Aβ1–40 and Aβ1–42 aggregation, it can be assumed that antibodies specific to these forms of Aβ may have a preventive effect against the excessive formation of toxic Aβ oligomers in the brain without targeting intact Aβ and its physiological functions [139].
Based on the results of the studies described above, the role of autoantibodies to Aβ in the regulation of AD in humans remains ambiguous. It is also not known whether autoantibodies to more toxic and oligomerization-promoting Aβ forms, such as Iso-D7-Aβ and 3pE-Aβ, are formed in healthy adults and AD patients and, if so, whether they may influence the regulation of the disease.

4.2.2. B Cells

While B cells are essential for the formation of autoantibodies to Aβ, their direct role in the pathogenesis of AD is rarely considered and remains a matter of debate. Initial studies have shown a decrease in the total number of circulating B cells in the peripheral blood of patients with AD, both in absolute numbers and as a percentage of peripheral blood mononuclear cells (PBMCs) [201,202]. On the other hand, an analysis of the B cell receptor (BCR) repertoire of B cells from the peripheral blood of AD patients demonstrated enrichment of some BCR clones as compared to healthy donors of the same age [54]. These results suggest that patients with AD have an accumulation of certain antigen-specific B cells involved in the immune response [54]. Based on the previously described accumulation of autoantibodies to the N-terminus of Aβ in a group of patients with more severe AD [127], a corresponding increase in the circulating memory B cells specific to the N-terminus of Aβ could be hypothesized. However, to date, the antigen-specificity of B cell clones accumulating in AD patients, as well as their specificity to various forms of Aβ, including the most common post-translational modifications of Aβ, is not known. In addition, the direct role of B cells in the pathogenesis of AD is unclear (Figure 1).
A recent study has shown that B cell deficiency in different mouse models of AD (3xTgAD, APP/PS1, 5xFAD) leads to a decrease in amyloid deposits at the base of the hippocampus and improved cognitive function [203] (Table 2). This study was conducted both in B cell-knocked-out mice (BKO) and in mice where B cells were transiently depleted by administration of antibodies to CD20 or B220 [203]. In the first case, B cell-deficient mice were obtained by crossing JHT mice that do not develop mature B cells [203] with 3xTgAD [204] or APP/PS1 [205] mice. Both B cell-deficient mouse models of AD had reduced amounts of amyloid deposits in the brain and improved cognitive abilities (at 50–60 weeks in 3xTgAD-BKO mice and 20–35 weeks in APP/PS1-BKO mice) as compared to animals of the same background in which B cells were not knocked out. In addition, α-CD20 or α-B220-mediated B cell elimination in 3xTgAD (60–70 weeks old) and 5xFAD (35–47 weeks old) mice showed reduced deposition of amyloid plaques in the brains of mice two months after the start of therapy. The results suggest that B cell elimination leads to the inhibition of AD and improved cognitive function. However, it is unclear what kind of B cells exacerbate disease progression in mouse models of AD, including their antigen specificity, mechanism of action, and the role they play in the brain parenchyma [203].In addition to autoantibody production, which may influence the development of AD in mice, several other possible mechanisms of B cell action may exacerbate AD development. One possibility is that autoantigen-specific B cells stimulate T cells, pathogenic for AD development, by presenting autoantigenic peptides (e.g., from Aβ) in complex with MHCs on their surface (Figure 1). The B cell-dependent activation of T cells can occur either distantly (in SLO) or locally (in the brain parenchyma) in the case of the colocalization of antigen-specific B and T cells. While the B cell-dependent activation of T cells in SLO is more likely due to scarce B cell presence in the brain, one study reported an increase in the numbers of B cells in the brain parenchyma of 5xFAD female mice [203]. In addition to the antigen-specific activation of cognate T cells, B cells may influence T cell responses in other ways [206,207]. B cells may also contribute to AD development through the hypersecretion of pro-inflammatory mediators, which, in turn, may contribute to the activation of microglia in the brain [19] and neuroinflammation (Figure 1).
In addition to stimulating the innate and T cell responses, B cells may also play a role in suppressing the inflammation and inhibiting the development of pathological processes. Some subpopulations of B cells (e.g., regulatory B cells) are known to express anti-inflammatory cytokines such as IL-10, IL-35, and TGFβ, which tend to limit the immune response and reduce the risk of autoimmune diseases [208]. Interestingly, it was recently shown that in 3-month-old 5xFAD mice, which had already begun to show AD pathology, IL-35 produced by B cells inhibited β-secretase (BACE1), which is responsible for the formation of β-amyloid from APP [209] (Figure 1). Thus, even in murine models of AD, B cells can play different, sometimes opposing, roles at different stages of disease progression. Further studies are required to address the role of B cells, including Aβ-specific B cells, in human AD pathology.
To summarize the above, B cells represent a heterogeneous population of cells with different antigen specificities, localizations, and functions [203]. An assessment of the contribution of B cells to the development of Alzheimer’s disease requires a detailed analysis of B cell subsets circulating in the blood and localizing into the brain at different stages of pathology development, and of their specificity to Aβ and other autoantigens.

5. Concluding Notes

In addition to the well-examined role of β-amyloids in AD pathogenesis, multiple studies have suggested the important role of the immune system in its regulation. However, at this time, there are outstanding gaps in the understanding of the immune response to Aβ and how it affects the predisposition of healthy adults to AD or progression of the ongoing disease. This includes insufficient information on (i) how various pathogenic forms of Aβ affect the innate immune system and inflammation, (ii) the involvement of various subsets of Aβ-specific CD8 and CD4 T cells in the regulation of the disease, (iii) the autoantigen-reactivity and function of B cell clones that accumulate in AD patients and their specificity to various forms of Aβ, (iv) the presence of autoantibodies to more toxic and oligomerization-promoting Aβ forms in healthy adults and AD patients and their role in the control of the disease, (v) the presence of local plasma cells in the brain parenchyma of AD patients, and (vi) the specificity of antibodies in the brain parenchyma to Aβ and other antigens. Resolving these questions is important for dissecting the interplay between various forms of Aβ and the immune system in the regulation of AD and could aid in the development of novel diagnostic and therapeutic tools for this debilitating disease.

Author Contributions

E.K.: conceptualization, writing—original draft; I.P.: conceptualization, writing—review/editing; V.M.: conceptualization, writing—review/editing; A.A.M.: funding, supervision, writing—review/editing; I.L.G.: conceptualization, supervision, writing—review/editing. All authors have read and agreed to the published version of the manuscript.

Funding

The author(s) declare that financial support was received for the research, authorship, and/or publication of this article. This research was funded by the Russian Science Foundation (Grant No. #19-74-30007).

Acknowledgments

We thank Alexander Novoselov for technical assistance with this manuscript.

Conflicts of Interest

The authors declare that this research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Abbreviations

AD, Alzheimer’s disease; Aβ, β-amyloid; APP, amyloid-β precursor protein; BCR, B cell receptor; PSEN1, presenilin-1; PSEN2, presenilin-2; 3pE-Aβ, pyroglutamate-modified form of Aβ; Iso-D7-Aβ, Aβ with an isomerized Asp7 residue; pS8-Aβ, Aβ with a phosphorylated serine 8 residue; CSF, cerebrospinal fluid; IL, interleukin; SLOs, secondary lymphoid organs; TCR, T cell receptor; MHCI and/or MHCII, major histocompatibility complexes; BBB, blood–brain barrier; mAbs, monoclonal antibodies; PBMCs, peripheral blood mononuclear cells.

References

  1. Dementia. Available online: https://www.who.int/news-room/fact-sheets/detail/dementia (accessed on 20 June 2024).
  2. Angelucci, F.; Cechova, K.; Amlerova, J.; Hort, J. Antibiotics, gut microbiota, and Alzheimer’s disease. J. Neuroinflamm. 2019, 16, 108. [Google Scholar] [CrossRef] [PubMed]
  3. Gao, Y.; Tan, L.; Yu, J.-T.; Tan, L. Tau in Alzheimer’s Disease: Mechanisms and Therapeutic Strategies. Curr. Alzheimer Res. 2018, 15, 283–300. [Google Scholar] [CrossRef] [PubMed]
  4. Medeiros, R.; Baglietto-Vargas, D.; LaFerla, F.M. The Role of Tau in Alzheimer’s Disease and Related Disorders. CNS Neurosci. Ther. 2010, 17, 514–524. [Google Scholar] [CrossRef]
  5. Muralidar, S.; Ambi, S.V.; Sekaran, S.; Thirumalai, D.; Palaniappan, B. Role of tau protein in Alzheimer’s disease: The prime pathological player. Int. J. Biol. Macromol. 2020, 163, 1599–1617. [Google Scholar] [CrossRef] [PubMed]
  6. Naseri, N.N.; Wang, H.; Guo, J.; Sharma, M.; Luo, W. The complexity of tau in Alzheimer’s disease. Neurosci. Lett. 2019, 705, 183–194. [Google Scholar] [CrossRef]
  7. Newcombe, E.A.; Camats-Perna, J.; Silva, M.L.; Valmas, N.; Huat, T.J.; Medeiros, R. Inflammation: The link between comorbidities, genetics, and Alzheimer’s disease. J. Neuroinflamm. 2018, 15, 276. [Google Scholar] [CrossRef] [PubMed]
  8. Sun, Q.; Xie, N.; Tang, B.; Li, R.; Shen, Y. Alzheimer’s Disease: From Genetic Variants to the Distinct Pathological Mechanisms. Front. Mol. Neurosci. 2017, 10, 319. [Google Scholar] [CrossRef]
  9. Weber, C.; Dilthey, A.; Finzer, P. The role of microbiome-host interactions in the development of Alzheimer’s disease. Front. Cell. Infect. Microbiol. 2023, 13, 1151021. [Google Scholar] [CrossRef]
  10. Selkoe, D.J.; Hardy, J. The amyloid hypothesis of Alzheimer’s disease at 25 years. EMBO Mol. Med. 2016, 8, 595–608. [Google Scholar] [CrossRef]
  11. Asamu, M.O.; Oladipo, O.O.; Abayomi, O.A.; Adebayo, A.A. Alzheimer’s disease: The role of T lymphocytes in neuroinflammation and neurodegeneration. Brain Res. 2023, 1821, 148589. [Google Scholar] [CrossRef]
  12. Bettcher, B.M.; Tansey, M.G.; Dorothée, G.; Heneka, M.T. Peripheral and central immune system crosstalk in Alzheimer disease—A research prospectus. Nat. Rev. Neurol. 2021, 17, 689–701. [Google Scholar] [CrossRef] [PubMed]
  13. Chen, X.; Holtzman, D.M. Emerging roles of innate and adaptive immunity in Alzheimer’s disease. Immunity 2022, 55, 2236–2254. [Google Scholar] [CrossRef] [PubMed]
  14. Hansen, D.V.; Hanson, J.E.; Sheng, M. Microglia in Alzheimer’s disease. J. Cell Biol. 2018, 217, 459–472. [Google Scholar] [CrossRef] [PubMed]
  15. Heneka, M.T.; Carson, M.J.; El Khoury, J.; Landreth, G.E.; Brosseron, F.; Feinstein, D.L.; Jacobs, A.H.; Wyss-Coray, T.; Vitorica, J.; Ransohoff, R.M.; et al. Neuroinflammation in Alzheimer’s Disease. Lancet Neurol. 2015, 14, 388–405. [Google Scholar] [CrossRef]
  16. McManus, R.M. The Role of Immunity in Alzheimer’s Disease. Adv. Biol. 2022, 6, e2101166. [Google Scholar] [CrossRef]
  17. McManus, R.M.; Mills, K.H.G.; Lynch, M.A. T Cells-Protective or Pathogenic in Alzheimer’s Disease? J. Neuroimmune Pharmacol. 2015, 10, 547–560. [Google Scholar] [CrossRef]
  18. Wu, K.-M.; Zhang, Y.-R.; Huang, Y.-Y.; Dong, Q.; Tan, L.; Yu, J.-T. The role of the immune system in Alzheimer’s disease. Ageing Res. Rev. 2021, 70, 101409. [Google Scholar] [CrossRef] [PubMed]
  19. Wyatt-Johnson, S.K.; Brutkiewicz, R.R. The Complexity of Microglial Interactions With Innate and Adaptive Immune Cells in Alzheimer’s Disease. Front. Aging Neurosci. 2020, 12, 592359. [Google Scholar] [CrossRef]
  20. Erten-Lyons, D.; Woltjer, R.L.; Dodge, H.; Nixon, R.; Vorobik, R.; Calvert, J.F.; Leahy, M.; Montine, T.; Kaye, J. Factors associated with resistance to dementia despite high Alzheimer disease pathology. Neurology 2009, 72, 354–360. [Google Scholar] [CrossRef] [PubMed]
  21. Hsiao, K.; Chapman, P.; Nilsen, S.; Eckman, C.; Harigaya, Y.; Younkin, S.; Yang, F.; Cole, G. Correlative memory deficits, Abeta elevation, and amyloid plaques in transgenic mice. Science 1996, 274, 99–102. [Google Scholar] [CrossRef] [PubMed]
  22. Nguyen, P.H.; Ramamoorthy, A.; Sahoo, B.R.; Zheng, J.; Faller, P.; Straub, J.E.; Dominguez, L.; Shea, J.-E.; Dokholyan, N.V.; De Simone, A.; et al. Amyloid Oligomers: A Joint Experimental/Computational Perspective on Alzheimer’s Disease, Parkinson’s Disease, Type II Diabetes, and Amyotrophic Lateral Sclerosis. Chem. Rev. 2021, 121, 2545–2647. [Google Scholar] [CrossRef] [PubMed]
  23. Kozin, S.A.; Kulikova, A.A.; Istrate, A.N.; Tsvetkov, P.O.; Zhokhov, S.S.; Mezentsev, Y.V.; Kechko, O.I.; Ivanov, A.S.; Polshakov, V.I.; Makarov, A.A. The English (H6R) familial Alzheimer’s disease mutation facilitates zinc-induced dimerization of the amyloid-β metal-binding domain. Metallomics 2015, 7, 422–425. [Google Scholar] [CrossRef]
  24. Ono, K.; Condron, M.M.; Teplow, D.B. Effects of the English (H6R) and Tottori (D7N) Familial Alzheimer Disease Mutations on Amyloid β-Protein Assembly and Toxicity. J. Biol. Chem. 2010, 285, 23186–23197. [Google Scholar] [CrossRef] [PubMed]
  25. Ono, K.; Yamada, M. Low-n oligomers as therapeutic targets of Alzheimer’s disease. J. Neurochem. 2011, 117, 19–28. [Google Scholar] [CrossRef] [PubMed]
  26. Hori, Y.; Hashimoto, T.; Wakutani, Y.; Urakami, K.; Nakashima, K.; Condron, M.M.; Tsubuki, S.; Saido, T.C.; Teplow, D.B.; Iwatsubo, T. The Tottori (D7N) and English (H6R) familial Alzheimer disease mutations accelerate Abeta fibril formation without increasing protofibril formation. J. Biol. Chem. 2007, 282, 4916–4923. [Google Scholar] [CrossRef]
  27. Xu, L.; Chen, Y.; Wang, X. Dual effects of familial Alzheimer’s disease mutations (D7H, D7N, and H6R) on amyloid β peptide: Correlation dynamics and zinc binding. Proteins 2014, 82, 3286–3297. [Google Scholar] [CrossRef]
  28. Chen, W.-T.; Hong, C.-J.; Lin, Y.-T.; Chang, W.-H.; Huang, H.-T.; Liao, J.-Y.; Chang, Y.-J.; Hsieh, Y.-F.; Cheng, C.-Y.; Liu, H.-C.; et al. Amyloid-Beta (Aβ) D7H Mutation Increases Oligomeric Aβ42 and Alters Properties of Aβ-Zinc/Copper Assemblies. PLoS ONE 2012, 7, e35807. [Google Scholar] [CrossRef]
  29. Kechko, O.I.; Adzhubei, A.A.; Tolstova, A.P.; Indeykina, M.I.; Popov, I.A.; Zhokhov, S.S.; Gnuchev, N.V.; Mitkevich, V.A.; Makarov, A.A.; Kozin, S.A. Molecular Mechanism of Zinc-Dependent Oligomerization of Alzheimer’s Amyloid-β with Taiwan (D7H) Mutation. Int. J. Mol. Sci. 2023, 24, 11241. [Google Scholar] [CrossRef]
  30. Barykin, E.P.; Petrushanko, I.Y.; Kozin, S.A.; Telegin, G.B.; Chernov, A.S.; Lopina, O.D.; Radko, S.P.; Mitkevich, V.A.; Makarov, A.A. Phosphorylation of the Amyloid-Beta Peptide Inhibits Zinc-Dependent Aggregation, Prevents Na,K-ATPase Inhibition, and Reduces Cerebral Plaque Deposition. Front. Mol. Neurosci. 2018, 11, 302. [Google Scholar] [CrossRef]
  31. Cruceta, L.; Sun, Y.; Kenyaga, J.M.; Ostrovsky, D.; Rodgers, A.; Vugmeyster, L.; Yao, L.; Qiang, W. Modulation of aggregation and structural polymorphisms of β-amyloid fibrils in cellular environments by pyroglutamate-3 variant cross-seeding. J. Biol. Chem. 2023, 299, 105196. [Google Scholar] [CrossRef]
  32. De Kimpe, L.; van Haastert, E.S.; Kaminari, A.; Zwart, R.; Rutjes, H.; Hoozemans, J.J.M.; Scheper, W. Intracellular accumulation of aggregated pyroglutamate amyloid beta: Convergence of aging and Aβ pathology at the lysosome. Age 2013, 35, 673–687. [Google Scholar] [CrossRef]
  33. Barykin, E.P.; Mitkevich, V.A.; Kozin, S.A.; Makarov, A.A. Amyloid β Modification: A Key to the Sporadic Alzheimer’s Disease? Front. Genet. 2017, 8, 58. [Google Scholar] [CrossRef] [PubMed]
  34. Faller, P.; Hureau, C. Bioinorganic chemistry of copper and zinc ions coordinated to amyloid-beta peptide. Dalton Trans. Camb. Engl. 2009, 7, 1080–1094. [Google Scholar] [CrossRef] [PubMed]
  35. Hassan, R.; Abedin, F.; Tatulian, S.A. Structure of unmodified and pyroglutamylated amyloid beta peptides in lipid membranes. Biophys. J. 2022, 121, 328a. [Google Scholar] [CrossRef]
  36. Istrate, A.N.; Tsvetkov, P.O.; Mantsyzov, A.B.; Kulikova, A.A.; Kozin, S.A.; Makarov, A.A.; Polshakov, V.I. NMR solution structure of rat aβ(1-16): Toward understanding the mechanism of rats’ resistance to Alzheimer’s disease. Biophys. J. 2012, 102, 136–143. [Google Scholar] [CrossRef]
  37. Kozin, S.A.; Barykin, E.P.; Telegin, G.B.; Chernov, A.S.; Adzhubei, A.A.; Radko, S.P.; Mitkevich, V.A.; Makarov, A.A. Intravenously Injected Amyloid-β Peptide With Isomerized Asp7 and Phosphorylated Ser8 Residues Inhibits Cerebral β-Amyloidosis in AβPP/PS1 Transgenic Mice Model of Alzheimer’s Disease. Front. Neurosci. 2018, 12, 518. [Google Scholar] [CrossRef]
  38. Kozin, S.A.; Zirah, S.; Rebuffat, S.; Hoa, G.H.; Debey, P. Zinc binding to Alzheimer’s Abeta(1-16) peptide results in stable soluble complex. Biochem. Biophys. Res. Commun. 2001, 285, 959–964. [Google Scholar] [CrossRef]
  39. Kumar, S.; Wirths, O.; Theil, S.; Gerth, J.; Bayer, T.A.; Walter, J. Early intraneuronal accumulation and increased aggregation of phosphorylated Abeta in a mouse model of Alzheimer’s disease. Acta Neuropathol. 2013, 125, 699–709. [Google Scholar] [CrossRef]
  40. Kumar, S.; Rezaei-Ghaleh, N.; Terwel, D.; Thal, D.R.; Richard, M.; Hoch, M.; Mc Donald, J.M.; Wüllner, U.; Glebov, K.; Heneka, M.T.; et al. Extracellular phosphorylation of the amyloid β-peptide promotes formation of toxic aggregates during the pathogenesis of Alzheimer’s disease. EMBO J. 2011, 30, 2255–2265. [Google Scholar] [CrossRef]
  41. Kumar, S.; Walter, J. Phosphorylation of amyloid beta (Aβ) peptides—A trigger for formation of toxic aggregates in Alzheimer’s disease. Aging 2011, 3, 803–812. [Google Scholar] [CrossRef]
  42. Kummer, M.P.; Hermes, M.; Delekarte, A.; Hammerschmidt, T.; Kumar, S.; Terwel, D.; Walter, J.; Pape, H.-C.; König, S.; Roeber, S.; et al. Nitration of tyrosine 10 critically enhances amyloid β aggregation and plaque formation. Neuron 2011, 71, 833–844. [Google Scholar] [CrossRef] [PubMed]
  43. Mitkevich, V.A.; Petrushanko, I.Y.; Yegorov, Y.E.; Simonenko, O.V.; Vishnyakova, K.S.; Kulikova, A.A.; Tsvetkov, P.O.; Makarov, A.A.; Kozin, S.A. Isomerization of Asp7 leads to increased toxic effect of amyloid-β42 on human neuronal cells. Cell Death Dis. 2013, 4, e939. [Google Scholar] [CrossRef] [PubMed]
  44. Moro, M.L.; Phillips, A.S.; Gaimster, K.; Paul, C.; Mudher, A.; Nicoll, J.A.R.; Boche, D. Pyroglutamate and Isoaspartate modified Amyloid-Beta in ageing and Alzheimer’s disease. Acta Neuropathol. Commun. 2018, 6, 3. [Google Scholar] [CrossRef] [PubMed]
  45. Nussbaum, J.M.; Schilling, S.; Cynis, H.; Silva, A.; Swanson, E.; Wangsanut, T.; Tayler, K.; Wiltgen, B.; Hatami, A.; Rönicke, R.; et al. Prion-like Behavior and Tau-dependent Cytotoxicity of Pyroglutamylated β-Amyloid. Nature 2012, 485, 651–655. [Google Scholar] [CrossRef] [PubMed]
  46. Russo, C.; Violani, E.; Salis, S.; Venezia, V.; Dolcini, V.; Damonte, G.; Benatti, U.; D’Arrigo, C.; Patrone, E.; Carlo, P.; et al. Pyroglutamate-modified amyloid beta-peptides—AbetaN3(pE)—Strongly affect cultured neuron and astrocyte survival. J. Neurochem. 2002, 82, 1480–1489. [Google Scholar] [CrossRef] [PubMed]
  47. Schlenzig, D.; Rönicke, R.; Cynis, H.; Ludwig, H.-H.; Scheel, E.; Reymann, K.; Saido, T.; Hause, G.; Schilling, S.; Demuth, H.-U. N-Terminal pyroglutamate formation of Aβ38 and Aβ40 enforces oligomer formation and potency to disrupt hippocampal long-term potentiation. J. Neurochem. 2012, 121, 774–784. [Google Scholar] [CrossRef] [PubMed]
  48. Tsvetkov, P.O.; Kulikova, A.A.; Golovin, A.V.; Tkachev, Y.V.; Archakov, A.I.; Kozin, S.A.; Makarov, A.A. Minimal Zn2+ binding site of amyloid-β. Biophys. J. 2010, 99, L84–L86. [Google Scholar] [CrossRef] [PubMed]
  49. Zirah, S.; Kozin, S.A.; Mazur, A.K.; Blond, A.; Cheminant, M.; Ségalas-Milazzo, I.; Debey, P.; Rebuffat, S. Structural changes of region 1-16 of the Alzheimer disease amyloid beta-peptide upon zinc binding and in vitro aging. J. Biol. Chem. 2006, 281, 2151–2161. [Google Scholar] [CrossRef]
  50. Mittal, K.; Eremenko, E.; Berner, O.; Elyahu, Y.; Strominger, I.; Apelblat, D.; Nemirovsky, A.; Spiegel, I.; Monsonego, A. CD4 T Cells Induce A Subset of MHCII-Expressing Microglia that Attenuates Alzheimer Pathology. iScience 2019, 16, 298–311. [Google Scholar] [CrossRef]
  51. McQuillan, K.; Lynch, M.A.; Mills, K.H.G. Activation of mixed glia by Aβ-specific Th1 and Th17 cells and its regulation by Th2 cells. Brain. Behav. Immun. 2010, 24, 598–607. [Google Scholar] [CrossRef]
  52. Sigurdsson, E.M.; Scholtzova, H.; Mehta, P.D.; Frangione, B.; Wisniewski, T. Immunization with a Nontoxic/Nonfibrillar Amyloid-β Homologous Peptide Reduces Alzheimer’s Disease-Associated Pathology in Transgenic Mice. Am. J. Pathol. 2001, 159, 439–447. [Google Scholar] [CrossRef]
  53. Bard, F.; Barbour, R.; Cannon, C.; Carretto, R.; Fox, M.; Games, D.; Guido, T.; Hoenow, K.; Hu, K.; Johnson-Wood, K.; et al. Epitope and isotype specificities of antibodies to β-amyloid peptide for protection against Alzheimer’s disease-like neuropathology. Proc. Natl. Acad. Sci. USA 2003, 100, 2023–2028. [Google Scholar] [CrossRef]
  54. Park, J.-C.; Noh, J.; Jang, S.; Kim, K.H.; Choi, H.; Lee, D.; Kim, J.; Chung, J.; Lee, D.Y.; Lee, Y.; et al. Association of B cell profile and receptor repertoire with the progression of Alzheimer’s disease. Cell Rep. 2022, 40, 111391. [Google Scholar] [CrossRef]
  55. Cras, P.; Kawai, M.; Lowery, D.; Gonzalez-DeWhitt, P.; Greenberg, B.; Perry, G. Senile plaque neurites in Alzheimer disease accumulate amyloid precursor protein. Proc. Natl. Acad. Sci. USA 1991, 88, 7552. [Google Scholar] [CrossRef] [PubMed]
  56. Lahiri, D.K.; Ge, Y.-W. Role of the APP promoter in Alzheimer’s disease: Cell type-specific expression of the beta-amyloid precursor protein. Ann. N. Y. Acad. Sci. 2004, 1030, 310–316. [Google Scholar] [CrossRef] [PubMed]
  57. Benilova, I.; Karran, E.; De Strooper, B. The toxic Aβ oligomer and Alzheimer’s disease: An emperor in need of clothes. Nat. Neurosci. 2012, 15, 349–357. [Google Scholar] [CrossRef] [PubMed]
  58. Chow, V.W.; Mattson, M.P.; Wong, P.C.; Gleichmann, M. An Overview of APP Processing Enzymes and Products. Neuromolecular Med. 2010, 12, 1–12. [Google Scholar] [CrossRef]
  59. Andreasen, N.; Hesse, C.; Davidsson, P.; Minthon, L.; Wallin, A.; Winblad, B.; Vanderstichele, H.; Vanmechelen, E.; Blennow, K. Cerebrospinal fluid beta-amyloid(1-42) in Alzheimer disease: Differences between early- and late-onset Alzheimer disease and stability during the course of disease. Arch. Neurol. 1999, 56, 673–680. [Google Scholar] [CrossRef]
  60. Roher, A.E.; Lowenson, J.D.; Clarke, S.; Woods, A.S.; Cotter, R.J.; Gowing, E.; Ball, M.J. beta-Amyloid-(1-42) is a major component of cerebrovascular amyloid deposits: Implications for the pathology of Alzheimer disease. Proc. Natl. Acad. Sci. USA 1993, 90, 10836–10840. [Google Scholar] [CrossRef]
  61. Masters, C.L.; Selkoe, D.J. Biochemistry of amyloid β-protein and amyloid deposits in Alzheimer disease. Cold Spring Harb. Perspect. Med. 2012, 2, a006262. [Google Scholar] [CrossRef]
  62. De Strooper, B.; Iwatsubo, T.; Wolfe, M.S. Presenilins and γ-secretase: Structure, function, and role in Alzheimer Disease. Cold Spring Harb. Perspect. Med. 2012, 2, a006304. [Google Scholar] [CrossRef] [PubMed]
  63. Kimura, A.; Hata, S.; Suzuki, T. Alternative Selection of β-Site APP-Cleaving Enzyme 1 (BACE1) Cleavage Sites in Amyloid β-Protein Precursor (APP) Harboring Protective and Pathogenic Mutations within the Aβ Sequence. J. Biol. Chem. 2016, 291, 24041–24053. [Google Scholar] [CrossRef] [PubMed]
  64. Di Fede, G.; Catania, M.; Morbin, M.; Rossi, G.; Suardi, S.; Mazzoleni, G.; Merlin, M.; Giovagnoli, A.R.; Prioni, S.; Erbetta, A.; et al. A recessive mutation in the APP gene with dominant-negative effect on amyloidogenesis. Science 2009, 323, 1473–1477. [Google Scholar] [CrossRef] [PubMed]
  65. Benilova, I.; Gallardo, R.; Ungureanu, A.-A.; Castillo Cano, V.; Snellinx, A.; Ramakers, M.; Bartic, C.; Rousseau, F.; Schymkowitz, J.; De Strooper, B. The Alzheimer disease protective mutation A2T modulates kinetic and thermodynamic properties of amyloid-β (Aβ) aggregation. J. Biol. Chem. 2014, 289, 30977–30989. [Google Scholar] [CrossRef]
  66. Hatami, A.; Monjazeb, S.; Milton, S.; Glabe, C.G. Familial Alzheimer’s Disease Mutations within the Amyloid Precursor Protein Alter the Aggregation and Conformation of the Amyloid-β Peptide. J. Biol. Chem. 2017, 292, 3172–3185. [Google Scholar] [CrossRef]
  67. Miners, J.S.; Barua, N.; Kehoe, P.G.; Gill, S.; Love, S. Aβ-degrading enzymes: Potential for treatment of Alzheimer disease. J. Neuropathol. Exp. Neurol. 2011, 70, 944–959. [Google Scholar] [CrossRef]
  68. Vilchez, D.; Saez, I.; Dillin, A. The role of protein clearance mechanisms in organismal ageing and age-related diseases. Nat. Commun. 2014, 5, 5659. [Google Scholar] [CrossRef] [PubMed]
  69. Yoon, S.-S.; Jo, S.A. Mechanisms of Amyloid-β Peptide Clearance: Potential Therapeutic Targets for Alzheimer’s Disease. Biomol. Ther. 2012, 20, 245–255. [Google Scholar] [CrossRef]
  70. Mucke, L. Neuroscience: Alzheimer’s disease. Nature 2009, 461, 895–897. [Google Scholar] [CrossRef]
  71. Hardy, J.A.; Higgins, G.A. Alzheimer’s disease: The amyloid cascade hypothesis. Science 1992, 256, 184–185. [Google Scholar] [CrossRef]
  72. Viles, J.H. Imaging Amyloid-β Membrane Interactions: Ion-Channel Pores and Lipid-Bilayer Permeability in Alzheimer’s Disease. Angew. Chem. Int. Ed. 2023, 62, e202215785. [Google Scholar] [CrossRef]
  73. Paranjape, G.S.; Gouwens, L.K.; Osborn, D.C.; Nichols, M.R. Isolated Amyloid-β(1–42) Protofibrils, But Not Isolated Fibrils, Are Robust Stimulators of Microglia. ACS Chem. Neurosci. 2012, 3, 302–311. [Google Scholar] [CrossRef]
  74. Kayed, R.; Lasagna-Reeves, C.A. Molecular mechanisms of amyloid oligomers toxicity. J. Alzheimer’s Dis. 2013, 33 (Suppl. S1), S67–S78. [Google Scholar] [CrossRef]
  75. Jang, H.; Connelly, L.; Arce, F.T.; Ramachandran, S.; Kagan, B.L.; Lal, R.; Nussinov, R. Mechanisms for the Insertion of Toxic, Fibril-like β-Amyloid Oligomers into the Membrane. J. Chem. Theory Comput. 2013, 9, 822–833. [Google Scholar] [CrossRef]
  76. La Rosa, C.; Scalisi, S.; Lolicato, F.; Pannuzzo, M.; Raudino, A. Lipid-assisted protein transport: A diffusion-reaction model supported by kinetic experiments and molecular dynamics simulations. J. Chem. Phys. 2016, 144, 184901. [Google Scholar] [CrossRef]
  77. Eskandari, H.; Ghanadian, M.; Noleto-Dias, C.; Lomax, C.; Tawfike, A.; Christiansen, G.; Sutherland, D.S.; Ward, J.L.; Mohammad-Beigi, H.; Otzen, D.E. Inhibitors of α-Synuclein Fibrillation and Oligomer Toxicity in Rosa damascena: The All-Pervading Powers of Flavonoids and Phenolic Glycosides. ACS Chem. Neurosci. 2020, 11, 3161–3173. [Google Scholar] [CrossRef]
  78. Scollo, F.; Tempra, C.; Lolicato, F.; Sciacca, M.F.M.; Raudino, A.; Milardi, D.; La Rosa, C. Phospholipids Critical Micellar Concentrations Trigger Different Mechanisms of Intrinsically Disordered Proteins Interaction with Model Membranes. J. Phys. Chem. Lett. 2018, 9, 5125–5129. [Google Scholar] [CrossRef]
  79. Tempra, C.; Scollo, F.; Pannuzzo, M.; Lolicato, F.; La Rosa, C. A unifying framework for amyloid-mediated membrane damage: The lipid-chaperone hypothesis. Biochim. Biophys. Acta Proteins Proteom. 2022, 1870, 140767. [Google Scholar] [CrossRef]
  80. Liu, C.-C.; Liu, C.-C.; Kanekiyo, T.; Xu, H.; Bu, G. Apolipoprotein E and Alzheimer disease: Risk, mechanisms and therapy. Nat. Rev. Neurol. 2013, 9, 106–118. [Google Scholar] [CrossRef]
  81. Bertram, L.; Lill, C.M.; Tanzi, R.E. The genetics of Alzheimer disease: Back to the future. Neuron 2010, 68, 270–281. [Google Scholar] [CrossRef]
  82. Griffioen, G. Calcium Dyshomeostasis Drives Pathophysiology and Neuronal Demise in Age-Related Neurodegenerative Diseases. Int. J. Mol. Sci. 2023, 24, 13243. [Google Scholar] [CrossRef]
  83. Hattori, N.; Kitagawa, K.; Higashida, T.; Yagyu, K.; Shimohama, S.; Wataya, T.; Perry, G.; Smith, M.A.; Inagaki, C. Cl-ATPase and Na+/K+-ATPase activities in Alzheimer’s disease brains. Neurosci. Lett. 1998, 254, 141–144. [Google Scholar] [CrossRef] [PubMed]
  84. Vitvitsky, V.M.; Garg, S.K.; Keep, R.F.; Albin, R.L.; Banerjee, R. Na+ and K+ ion imbalances in Alzheimer’s disease. Biochim. Biophys. Acta 2012, 1822, 1671–1681. [Google Scholar] [CrossRef] [PubMed]
  85. Dickey, C.A.; Gordon, M.N.; Wilcock, D.M.; Herber, D.L.; Freeman, M.J.; Morgan, D. Dysregulation of Na+/K+ ATPase by amyloid in APP+PS1 transgenic mice. BMC Neurosci. 2005, 6, 7. [Google Scholar] [CrossRef]
  86. Kairane, C.; Mahlapuu, R.; Ehrlich, K.; Zilmer, M.; Soomets, U. The effects of different antioxidants on the activity of cerebrocortical MnSOD and Na,K-ATPase from post mortem Alzheimer’s disease and age-matched normal brains. Curr. Alzheimer Res. 2014, 11, 79–85. [Google Scholar] [CrossRef]
  87. Kreutz, F.; Scherer, E.B.; Ferreira, A.G.K.; Petry, F.D.S.; Pereira, C.L.; Santana, F.; de Souza Wyse, A.T.; Salbego, C.G.; Trindade, V.M.T. Alterations on Na+,K+-ATPase and acetylcholinesterase activities induced by amyloid-β peptide in rat brain and GM1 ganglioside neuroprotective action. Neurochem. Res. 2013, 38, 2342–2350. [Google Scholar] [CrossRef] [PubMed]
  88. Zhang, L.-N.; Sun, Y.-J.; Pan, S.; Li, J.-X.; Qu, Y.-E.; Li, Y.; Wang, Y.-L.; Gao, Z.-B. Na+-K+-ATPase, a potent neuroprotective modulator against Alzheimer disease. Fundam. Clin. Pharmacol. 2013, 27, 96–103. [Google Scholar] [CrossRef]
  89. LaFerla, F.M. Calcium dyshomeostasis and intracellular signalling in Alzheimer’s disease. Nat. Rev. Neurosci. 2002, 3, 862–872. [Google Scholar] [CrossRef]
  90. Liu, R.; Collier, J.M.; Abdul-Rahman, N.-H.; Capuk, O.; Zhang, Z.; Begum, G. Dysregulation of Ion Channels and Transporters and Blood-Brain Barrier Dysfunction in Alzheimer’s Disease and Vascular Dementia. Aging Dis. 2024, 15, 1748–1770. [Google Scholar] [CrossRef]
  91. Mroczko, B.; Groblewska, M.; Litman-Zawadzka, A.; Kornhuber, J.; Lewczuk, P. Cellular Receptors of Amyloid β Oligomers (AβOs) in Alzheimer’s Disease. Int. J. Mol. Sci. 2018, 19, 1884. [Google Scholar] [CrossRef]
  92. Ohnishi, T.; Yanazawa, M.; Sasahara, T.; Kitamura, Y.; Hiroaki, H.; Fukazawa, Y.; Kii, I.; Nishiyama, T.; Kakita, A.; Takeda, H.; et al. Na, K-ATPase α3 is a death target of Alzheimer patient amyloid-β assembly. Proc. Natl. Acad. Sci. USA 2015, 112, E4465–E4474. [Google Scholar] [CrossRef]
  93. Petrushanko, I.Y.; Mitkevich, V.A.; Anashkina, A.A.; Adzhubei, A.A.; Burnysheva, K.M.; Lakunina, V.A.; Kamanina, Y.V.; Dergousova, E.A.; Lopina, O.D.; Ogunshola, O.O.; et al. Direct interaction of beta-amyloid with Na,K-ATPase as a putative regulator of the enzyme function. Sci. Rep. 2016, 6, 27738. [Google Scholar] [CrossRef] [PubMed]
  94. Gu, Q.B.; Zhao, J.X.; Fei, J.; Schwarz, W. Modulation of Na+,K+ pumping and neurotransmitter uptake by beta-amyloid. Neuroscience 2004, 126, 61–67. [Google Scholar] [CrossRef]
  95. Mark, R.J.; Hensley, K.; Butterfield, D.A.; Mattson, M.P. Amyloid beta-peptide impairs ion-motive ATPase activities: Evidence for a role in loss of neuronal Ca2+ homeostasis and cell death. J. Neurosci. 1995, 15, 6239–6249. [Google Scholar] [CrossRef]
  96. Alberdi, E.; Sánchez-Gómez, M.V.; Cavaliere, F.; Pérez-Samartín, A.; Zugaza, J.L.; Trullas, R.; Domercq, M.; Matute, C. Amyloid beta oligomers induce Ca2+ dysregulation and neuronal death through activation of ionotropic glutamate receptors. Cell Calcium 2010, 47, 264–272. [Google Scholar] [CrossRef]
  97. Kaminsky, Y.G.; Tikhonova, L.A.; Kosenko, E.A. Critical analysis of Alzheimer’s amyloid-beta toxicity to mitochondria. Front. Biosci. Landmark Ed. 2015, 20, 173–197. [Google Scholar] [CrossRef]
  98. Morkuniene, R.; Cizas, P.; Jankeviciute, S.; Petrolis, R.; Arandarcikaite, O.; Krisciukaitis, A.; Borutaite, V. Small Aβ1-42 oligomer-induced membrane depolarization of neuronal and microglial cells: Role of N-methyl-D-aspartate receptors. J. Neurosci. Res. 2015, 93, 475–486. [Google Scholar] [CrossRef] [PubMed]
  99. Sayehmiri, F.; Motamedi, F.; Batool, Z.; Naderi, N.; Shaerzadeh, F.; Zoghi, A.; Rezaei, O.; Khodagholi, F.; Pourbadie, H.G. Mitochondrial plasticity and synaptic plasticity crosstalk; in health and Alzheimer’s disease. CNS Neurosci. Ther. 2024, 30, e14897. [Google Scholar] [CrossRef]
  100. Jarosz-Griffiths, H.H.; Noble, E.; Rushworth, J.V.; Hooper, N.M. Amyloid-β Receptors: The Good, the Bad, and the Prion Protein. J. Biol. Chem. 2016, 291, 3174–3183. [Google Scholar] [CrossRef]
  101. Li, S.; Jin, M.; Koeglsperger, T.; Shepardson, N.E.; Shankar, G.M.; Selkoe, D.J. Soluble Aβ oligomers inhibit long-term potentiation through a mechanism involving excessive activation of extrasynaptic NR2B-containing NMDA receptors. J. Neurosci. 2011, 31, 6627–6638. [Google Scholar] [CrossRef]
  102. Parri, H.R.; Hernandez, C.M.; Dineley, K.T. Research update: Alpha7 nicotinic acetylcholine receptor mechanisms in Alzheimer’s disease. Biochem. Pharmacol. 2011, 82, 931–942. [Google Scholar] [CrossRef]
  103. Puzzo, D.; Privitera, L.; Leznik, E.; Fà, M.; Staniszewski, A.; Palmeri, A.; Arancio, O. Picomolar amyloid-beta positively modulates synaptic plasticity and memory in hippocampus. J. Neurosci. 2008, 28, 14537–14545. [Google Scholar] [CrossRef] [PubMed]
  104. Freir, D.B.; Nicoll, A.J.; Klyubin, I.; Panico, S.; Mc Donald, J.M.; Risse, E.; Asante, E.A.; Farrow, M.A.; Sessions, R.B.; Saibil, H.R.; et al. Interaction between prion protein and toxic amyloid β assemblies can be therapeutically targeted at multiple sites. Nat. Commun. 2011, 2, 336. [Google Scholar] [CrossRef] [PubMed]
  105. Laurén, J.; Gimbel, D.A.; Nygaard, H.B.; Gilbert, J.W.; Strittmatter, S.M. Cellular prion protein mediates impairment of synaptic plasticity by amyloid-beta oligomers. Nature 2009, 457, 1128–1132. [Google Scholar] [CrossRef] [PubMed]
  106. Um, J.W.; Nygaard, H.B.; Heiss, J.K.; Kostylev, M.A.; Stagi, M.; Vortmeyer, A.; Wisniewski, T.; Gunther, E.C.; Strittmatter, S.M. Alzheimer amyloid-β oligomer bound to postsynaptic prion protein activates Fyn to impair neurons. Nat. Neurosci. 2012, 15, 1227–1235. [Google Scholar] [CrossRef] [PubMed]
  107. Origlia, N.; Bonadonna, C.; Rosellini, A.; Leznik, E.; Arancio, O.; Yan, S.S.; Domenici, L. Microglial receptor for advanced glycation end product-dependent signal pathway drives beta-amyloid-induced synaptic depression and long-term depression impairment in entorhinal cortex. J. Neurosci. 2010, 30, 11414–11425. [Google Scholar] [CrossRef]
  108. Barykin, E.P.; Garifulina, A.I.; Kruykova, E.V.; Spirova, E.N.; Anashkina, A.A.; Adzhubei, A.A.; Shelukhina, I.V.; Kasheverov, I.E.; Mitkevich, V.A.; Kozin, S.A.; et al. Isomerization of Asp7 in Beta-Amyloid Enhances Inhibition of the α7 Nicotinic Receptor and Promotes Neurotoxicity. Cells 2019, 8, 771. [Google Scholar] [CrossRef] [PubMed]
  109. Varshavskaya, K.B.; Petrushanko, I.Y.; Mitkevich, V.A.; Barykin, E.P.; Makarov, A.A. Post-translational modifications of beta-amyloid alter its transport in the blood-brain barrier in vitro model. Front. Mol. Neurosci. 2024, 17, 1362581. [Google Scholar] [CrossRef]
  110. Cheignon, C.; Tomas, M.; Bonnefont-Rousselot, D.; Faller, P.; Hureau, C.; Collin, F. Oxidative stress and the amyloid beta peptide in Alzheimer’s disease. Redox Biol. 2018, 14, 450–464. [Google Scholar] [CrossRef]
  111. Ebenezer, P.J.; Weidner, A.M.; LeVine, H.; Markesbery, W.R.; Murphy, M.P.; Zhang, L.; Dasuri, K.; Fernandez-Kim, S.O.; Bruce-Keller, A.J.; Gavilán, E.; et al. Neuron specific toxicity of oligomeric amyloid-β: Role for JUN-kinase and oxidative stress. J. Alzheimer’s Dis. 2010, 22, 839–848. [Google Scholar] [CrossRef]
  112. Aoyama, K. Glutathione in the Brain. Int. J. Mol. Sci. 2021, 22, 5010. [Google Scholar] [CrossRef]
  113. Chiang, G.C.; Mao, X.; Kang, G.; Chang, E.; Pandya, S.; Vallabhajosula, S.; Isaacson, R.; Ravdin, L.D.; Alzheimer’s Disease Neuroimaging Initiative; Shungu, D.C. Relationships among Cortical Glutathione Levels, Brain Amyloidosis, and Memory in Healthy Older Adults Investigated In Vivo with 1H-MRS and Pittsburgh Compound-B PET. AJNR Am. J. Neuroradiol. 2017, 38, 1130–1137. [Google Scholar] [CrossRef] [PubMed]
  114. Mandal, P.K.; Saharan, S.; Tripathi, M.; Murari, G. Brain glutathione levels—A novel biomarker for mild cognitive impairment and Alzheimer’s disease. Biol. Psychiatry 2015, 78, 702–710. [Google Scholar] [CrossRef] [PubMed]
  115. Anjo, S.I.; He, Z.; Hussain, Z.; Farooq, A.; McIntyre, A.; Laughton, C.A.; Carvalho, A.N.; Finelli, M.J. Protein Oxidative Modifications in Neurodegenerative Diseases: From Advances in Detection and Modelling to Their Use as Disease Biomarkers. Antioxidants 2024, 13, 681. [Google Scholar] [CrossRef] [PubMed]
  116. Dyer, R.R.; Ford, K.I.; Robinson, R.A.S. The roles of S-nitrosylation and S-glutathionylation in Alzheimer’s disease. Methods Enzymol. 2019, 626, 499–538. [Google Scholar] [CrossRef]
  117. Tramutola, A.; Lanzillotta, C.; Perluigi, M.; Butterfield, D.A. Oxidative stress, protein modification and Alzheimer disease. Brain Res. Bull. 2017, 133, 88–96. [Google Scholar] [CrossRef]
  118. Qu, J.; Nakamura, T.; Cao, G.; Holland, E.A.; McKercher, S.R.; Lipton, S.A. S-Nitrosylation activates Cdk5 and contributes to synaptic spine loss induced by beta-amyloid peptide. Proc. Natl. Acad. Sci. USA 2011, 108, 14330–14335. [Google Scholar] [CrossRef] [PubMed]
  119. Barykin, E.P.; Petrushanko, I.Y.; Burnysheva, K.M.; Makarov, A.A.; Mitkevich, V.A. Isomerization of Asp7 increases the toxic effects of amyloid β and its phosphorylated form in SH-SY5Y neuroblastoma cells. Mol. Biol. 2016, 50, 863–869. [Google Scholar] [CrossRef]
  120. Petrovskaya, A.V.; Tverskoi, A.M.; Barykin, E.P.; Varshavskaya, K.B.; Dalina, A.A.; Mitkevich, V.A.; Makarov, A.A.; Petrushanko, I.Y. Distinct Effects of Beta-Amyloid, Its Isomerized and Phosphorylated Forms on the Redox Status and Mitochondrial Functioning of the Blood-Brain Barrier Endothelium. Int. J. Mol. Sci. 2022, 24, 183. [Google Scholar] [CrossRef]
  121. Henriques, A.G.; Müller, T.; Oliveira, J.M.; Cova, M.; da Cruz E Silva, C.B.; da Cruz E Silva, O.A.B. Altered protein phosphorylation as a resource for potential AD biomarkers. Sci. Rep. 2016, 6, 30319. [Google Scholar] [CrossRef]
  122. Shankar, G.M.; Li, S.; Mehta, T.H.; Garcia-Munoz, A.; Shepardson, N.E.; Smith, I.; Brett, F.M.; Farrell, M.A.; Rowan, M.J.; Lemere, C.A.; et al. Amyloid-β protein dimers isolated directly from Alzheimer’s brains impair synaptic plasticity and memory. Nat. Med. 2008, 14, 837–842. [Google Scholar] [CrossRef]
  123. Zatsepina, O.G.; Kechko, O.I.; Mitkevich, V.A.; Kozin, S.A.; Yurinskaya, M.M.; Vinokurov, M.G.; Serebryakova, M.V.; Rezvykh, A.P.; Evgen’ev, M.B.; Makarov, A.A. Amyloid-β with isomerized Asp7 cytotoxicity is coupled to protein phosphorylation. Sci. Rep. 2018, 8, 3518. [Google Scholar] [CrossRef] [PubMed]
  124. Henriques, A.G.; Oliveira, J.M.; Carvalho, L.P.; da Cruz E Silva, O.A.B. Aβ Influences Cytoskeletal Signaling Cascades with Consequences to Alzheimer’s Disease. Mol. Neurobiol. 2015, 52, 1391–1407. [Google Scholar] [CrossRef] [PubMed]
  125. Busch, L.; Eggert, S.; Endres, K.; Bufe, B. The Hidden Role of Non-Canonical Amyloid β Isoforms in Alzheimer’s Disease. Cells 2022, 11, 3421. [Google Scholar] [CrossRef] [PubMed]
  126. Yang, H.; Li, J.; Li, X.; Ma, L.; Hou, M.; Zhou, H.; Zhou, R. Based on molecular structures: Amyloid-β generation, clearance, toxicity and therapeutic strategies. Front. Mol. Neurosci. 2022, 15, 927530. [Google Scholar] [CrossRef] [PubMed]
  127. Liu, Y.-H.; Wang, J.; Li, Q.-X.; Fowler, C.J.; Zeng, F.; Deng, J.; Xu, Z.-Q.; Zhou, H.-D.; Doecke, J.D.; Villemagne, V.L.; et al. Association of naturally occurring antibodies to β-amyloid with cognitive decline and cerebral amyloidosis in Alzheimer’s disease. Sci. Adv. 2021, 7, eabb0457. [Google Scholar] [CrossRef] [PubMed]
  128. Abelein, A. Metal Binding of Alzheimer’s Amyloid-β and Its Effect on Peptide Self-Assembly. Acc. Chem. Res. 2023, 56, 2653–2663. [Google Scholar] [CrossRef]
  129. Chia, S.; Cataldi, R.L.; Ruggeri, F.S.; Limbocker, R.; Condado-Morales, I.; Pisani, K.; Possenti, A.; Linse, S.; Knowles, T.P.J.; Habchi, J.; et al. A Relationship between the Structures and Neurotoxic Effects of Aβ Oligomers Stabilized by Different Metal Ions. ACS Chem. Neurosci. 2024, 15, 1125–1134. [Google Scholar] [CrossRef]
  130. Iwatsubo, T.; Odaka, A.; Suzuki, N.; Mizusawa, H.; Nukina, N.; Ihara, Y. Visualization of A beta 42(43) and A beta 40 in senile plaques with end-specific A beta monoclonals: Evidence that an initially deposited species is A beta 42(43). Neuron 1994, 13, 45–53. [Google Scholar] [CrossRef]
  131. Finder, V.H.; Glockshuber, R. Amyloid-beta aggregation. Neurodegener. Dis. 2007, 4, 13–27. [Google Scholar] [CrossRef]
  132. Suzuki, N.; Cheung, T.T.; Cai, X.D.; Odaka, A.; Otvos, L.; Eckman, C.; Golde, T.E.; Younkin, S.G. An increased percentage of long amyloid beta protein secreted by familial amyloid beta protein precursor (beta APP717) mutants. Science 1994, 264, 1336–1340. [Google Scholar] [CrossRef]
  133. Shimizu, T.; Watanabe, A.; Ogawara, M.; Mori, H.; Shirasawa, T. Isoaspartate formation and neurodegeneration in Alzheimer’s disease. Arch. Biochem. Biophys. 2000, 381, 225–234. [Google Scholar] [CrossRef]
  134. Kozin, S.A.; Mezentsev, Y.V.; Kulikova, A.A.; Indeykina, M.I.; Golovin, A.V.; Ivanov, A.S.; Tsvetkov, P.O.; Makarov, A.A. Zinc-induced dimerization of the amyloid-β metal-binding domain 1-16 is mediated by residues 11-14. Mol. Biosyst. 2011, 7, 1053–1055. [Google Scholar] [CrossRef]
  135. Mitkevich, V.A.; Barykin, E.P.; Eremina, S.; Pani, B.; Katkova-Zhukotskaya, O.; Polshakov, V.I.; Adzhubei, A.A.; Kozin, S.A.; Mironov, A.S.; Makarov, A.A.; et al. Zn-dependent β-amyloid aggregation and its reversal by the tetrapeptide HAEE. Aging Dis. 2022, 13, 1–10. [Google Scholar] [CrossRef]
  136. Kozin, S.A.; Cheglakov, I.B.; Ovsepyan, A.A.; Telegin, G.B.; Tsvetkov, P.O.; Lisitsa, A.V.; Makarov, A.A. Peripherally applied synthetic peptide isoAsp7-Aβ(1-42) triggers cerebral β-amyloidosis. Neurotox. Res. 2013, 24, 370–376. [Google Scholar] [CrossRef] [PubMed]
  137. Roher, A.E.; Lowenson, J.D.; Clarke, S.; Wolkow, C.; Wang, R.; Cotter, R.J.; Reardon, I.M.; Zürcher-Neely, H.A.; Heinrikson, R.L.; Ball, M.J. Structural alterations in the peptide backbone of beta-amyloid core protein may account for its deposition and stability in Alzheimer’s disease. J. Biol. Chem. 1993, 268, 3072–3083. [Google Scholar] [CrossRef]
  138. Fonseca, M.I.; Head, E.; Velazquez, P.; Cotman, C.W.; Tenner, A.J. The presence of isoaspartic acid in beta-amyloid plaques indicates plaque age. Exp. Neurol. 1999, 157, 277–288. [Google Scholar] [CrossRef] [PubMed]
  139. Gnoth, K.; Piechotta, A.; Kleinschmidt, M.; Konrath, S.; Schenk, M.; Taudte, N.; Ramsbeck, D.; Rieckmann, V.; Geissler, S.; Eichentopf, R.; et al. Targeting isoaspartate-modified Aβ rescues behavioral deficits in transgenic mice with Alzheimer’s disease-like pathology. Alzheimer’s Res. Ther. 2020, 12, 149. [Google Scholar] [CrossRef]
  140. Games, D.; Adams, D.; Alessandrini, R.; Barbour, R.; Berthelette, P.; Blackwell, C.; Carr, T.; Clemens, J.; Donaldson, T.; Gillespie, F. Alzheimer-type neuropathology in transgenic mice overexpressing V717F beta-amyloid precursor protein. Nature 1995, 373, 523–527. [Google Scholar] [CrossRef]
  141. Mucke, L.; Masliah, E.; Yu, G.Q.; Mallory, M.; Rockenstein, E.M.; Tatsuno, G.; Hu, K.; Kholodenko, D.; Johnson-Wood, K.; McConlogue, L. High-level neuronal expression of abeta 1-42 in wild-type human amyloid protein precursor transgenic mice: Synaptotoxicity without plaque formation. J. Neurosci. 2000, 20, 4050–4058. [Google Scholar] [CrossRef]
  142. Radde, R.; Bolmont, T.; Kaeser, S.A.; Coomaraswamy, J.; Lindau, D.; Stoltze, L.; Calhoun, M.E.; Jäggi, F.; Wolburg, H.; Gengler, S.; et al. Aβ42-driven cerebral amyloidosis in transgenic mice reveals early and robust pathology. EMBO Rep. 2006, 7, 940–946. [Google Scholar] [CrossRef]
  143. Oakley, H.; Cole, S.L.; Logan, S.; Maus, E.; Shao, P.; Craft, J.; Guillozet-Bongaarts, A.; Ohno, M.; Disterhoft, J.; Van Eldik, L.; et al. Intraneuronal β-Amyloid Aggregates, Neurodegeneration, and Neuron Loss in Transgenic Mice with Five Familial Alzheimer’s Disease Mutations: Potential Factors in Amyloid Plaque Formation. J. Neurosci. 2006, 26, 10129–10140. [Google Scholar] [CrossRef] [PubMed]
  144. Oddo, S.; Caccamo, A.; Shepherd, J.D.; Murphy, M.P.; Golde, T.E.; Kayed, R.; Metherate, R.; Mattson, M.P.; Akbari, Y.; LaFerla, F.M. Triple-transgenic model of Alzheimer’s disease with plaques and tangles: Intracellular Abeta and synaptic dysfunction. Neuron 2003, 39, 409–421. [Google Scholar] [CrossRef] [PubMed]
  145. Renno, T.; Taupin, V.; Bourbonnière, L.; Verge, G.; Tran, E.; De Simone, R.; Krakowski, M.; Rodriguez, M.; Peterson, A.; Owens, T. Interferon-gamma in progression to chronic demyelination and neurological deficit following acute EAE. Mol. Cell. Neurosci. 1998, 12, 376–389. [Google Scholar] [CrossRef]
  146. Monsonego, A.; Imitola, J.; Petrovic, S.; Zota, V.; Nemirovsky, A.; Baron, R.; Fisher, Y.; Owens, T.; Weiner, H.L. Aβ-induced meningoencephalitis is IFN-γ-dependent and is associated with T cell-dependent clearance of Aβ in a mouse model of Alzheimer’s disease. Proc. Natl. Acad. Sci. USA 2006, 103, 5048–5053. [Google Scholar] [CrossRef]
  147. Kummer, M.P.; Heneka, M.T. Truncated and modified amyloid-beta species. Alzheimer’s Res. Ther. 2014, 6, 28. [Google Scholar] [CrossRef]
  148. Leng, F.; Edison, P. Neuroinflammation and microglial activation in Alzheimer disease: Where do we go from here? Nat. Rev. Neurol. 2021, 17, 157–172. [Google Scholar] [CrossRef] [PubMed]
  149. Thakur, S.; Dhapola, R.; Sarma, P.; Medhi, B.; Reddy, D.H. Neuroinflammation in Alzheimer’s Disease: Current Progress in Molecular Signaling and Therapeutics. Inflammation 2023, 46, 1–17. [Google Scholar] [CrossRef] [PubMed]
  150. Sobue, A.; Komine, O.; Yamanaka, K. Neuroinflammation in Alzheimer’s disease: Microglial signature and their relevance to disease. Inflamm. Regen. 2023, 43, 26. [Google Scholar] [CrossRef]
  151. Kettenmann, H.; Hanisch, U.K.; Noda, M.; Verkhratsky, A. Physiology of microglia. Physiol. Rev. 2011, 91, 461–553. [Google Scholar] [CrossRef]
  152. Kim, Y.S.; Joh, T.H. Microglia, major player in the brain inflammation: Their roles in the pathogenesis of Parkinson’s disease. Exp. Mol. Med. 2006, 38, 333–347. [Google Scholar] [CrossRef]
  153. Kasus-Jacobi, A.; Washburn, J.L.; Land, C.A.; Pereira, H.A. Neutrophil Granule Proteins Inhibit Amyloid Beta Aggregation and Neurotoxicity. Curr. Alzheimer Res. 2021, 18, 414–427. [Google Scholar] [CrossRef] [PubMed]
  154. Jairani, P.S.; Aswathy, P.M.; Krishnan, D.; Menon, R.N.; Verghese, J.; Mathuranath, P.S.; Gopala, S. Apolipoprotein E Polymorphism and Oxidative Stress in Peripheral Blood-Derived Macrophage-Mediated Amyloid-Beta Phagocytosis in Alzheimer’s Disease Patients. Cell. Mol. Neurobiol. 2019, 39, 355–369. [Google Scholar] [CrossRef] [PubMed]
  155. Bianca, V.D.; Dusi, S.; Bianchini, E.; Dal Prà, I.; Rossi, F. beta-amyloid activates the O-2 forming NADPH oxidase in microglia, monocytes, and neutrophils. A possible inflammatory mechanism of neuronal damage in Alzheimer’s disease. J. Biol. Chem. 1999, 274, 15493–15499. [Google Scholar] [CrossRef]
  156. Liu, Z.; Li, H.; Pan, S. Discovery and Validation of Key Biomarkers Based on Immune Infiltrates in Alzheimer’s Disease. Front. Genet. 2021, 12, 658323. [Google Scholar] [CrossRef]
  157. Qi, C.; Liu, F.; Zhang, W.; Han, Y.; Zhang, N.; Liu, Q.; Li, H. Alzheimer’s disease alters the transcriptomic profile of natural killer cells at single-cell resolution. Front. Immunol. 2022, 13, 1004885. [Google Scholar] [CrossRef]
  158. NKGen Biotech’s SNK01 NK Cell Therapy Cleared to Start Phase 2 Clinical Trial in Alzheimer’s Disease | NKGen Biotech. 2024. Available online: https://nkgenbiotech.com/nkgen-biotechs-snk01-nk-cell-therapy-cleared-to-start-phase-2-clinical-trial-in-alzheimers-disease/ (accessed on 24 September 2024).
  159. Shi, M.; Chu, F.; Zhu, F.; Zhu, J. Peripheral blood amyloid-β involved in the pathogenesis of Alzheimer’s disease via impacting on peripheral innate immune cells. J. Neuroinflamm. 2024, 21, 5. [Google Scholar] [CrossRef] [PubMed]
  160. Wang, X. A Bridge Between the Innate Immunity System and Amyloid-β Production in Alzheimer’s Disease. Neurosci. Bull. 2021, 37, 898–901. [Google Scholar] [CrossRef]
  161. Xin, S.-H.; Tan, L.; Cao, X.; Yu, J.-T.; Tan, L. Clearance of Amyloid Beta and Tau in Alzheimer’s Disease: From Mechanisms to Therapy. Neurotox. Res. 2018, 34, 733–748. [Google Scholar] [CrossRef]
  162. Tarasoff-Conway, J.M.; Carare, R.O.; Osorio, R.S.; Glodzik, L.; Butler, T.; Fieremans, E.; Axel, L.; Rusinek, H.; Nicholson, C.; Zlokovic, B.V.; et al. Clearance systems in the brain-implications for Alzheimer disease. Nat. Rev. Neurol. 2015, 11, 457–470. [Google Scholar] [CrossRef] [PubMed]
  163. Unger, M.S.; Li, E.; Scharnagl, L.; Poupardin, R.; Altendorfer, B.; Mrowetz, H.; Hutter-Paier, B.; Weiger, T.M.; Heneka, M.T.; Attems, J.; et al. CD8+ T-cells infiltrate Alzheimer’s disease brains and regulate neuronal- and synapse-related gene expression in APP-PS1 transgenic mice. Brain. Behav. Immun. 2020, 89, 67–86. [Google Scholar] [CrossRef]
  164. McManus, R.M.; Heneka, M.T. T cells in Alzheimer’s disease: Space invaders. Lancet Neurol. 2020, 19, 285–287. [Google Scholar] [CrossRef] [PubMed]
  165. Togo, T.; Akiyama, H.; Iseki, E.; Kondo, H.; Ikeda, K.; Kato, M.; Oda, T.; Tsuchiya, K.; Kosaka, K. Occurrence of T cells in the brain of Alzheimer’s disease and other neurological diseases. J. Neuroimmunol. 2002, 124, 83–92. [Google Scholar] [CrossRef]
  166. Rao, Y.L.; Ganaraja, B.; Murlimanju, B.V.; Joy, T.; Krishnamurthy, A.; Agrawal, A. Hippocampus and its involvement in Alzheimer’s disease: A review. 3 Biotech 2022, 12, 55. [Google Scholar] [CrossRef] [PubMed]
  167. Gate, D.; Saligrama, N.; Leventhal, O.; Yang, A.C.; Unger, M.S.; Middeldorp, J.; Chen, K.; Lehallier, B.; Channappa, D.; De Los Santos, M.B.; et al. Clonally expanded CD8 T cells patrol the cerebrospinal fluid in Alzheimer’s disease. Nature 2020, 577, 399–404. [Google Scholar] [CrossRef] [PubMed]
  168. Pasciuto, E.; Burton, O.T.; Roca, C.P.; Lagou, V.; Rajan, W.D.; Theys, T.; Mancuso, R.; Tito, R.Y.; Kouser, L.; Callaerts-Vegh, Z.; et al. Microglia Require CD4 T Cells to Complete the Fetal-to-Adult Transition. Cell 2020, 182, 625–640.e24. [Google Scholar] [CrossRef]
  169. Lanuti, P.; Ciccocioppo, F.; Bonanni, L.; Marchisio, M.; Lachmann, R.; Tabet, N.; Pierdomenico, L.; Santavenere, E.; Catinella, V.; Iacone, A.; et al. Amyloid-specific T-cells differentiate Alzheimer’s disease from Lewy body dementia. Neurobiol. Aging 2012, 33, 2599–2611. [Google Scholar] [CrossRef] [PubMed]
  170. Monsonego, A.; Nemirovsky, A.; Harpaz, I. CD4 T cells in immunity and immunotherapy of Alzheimer’s disease. Immunology 2013, 139, 438–446. [Google Scholar] [CrossRef]
  171. Monsonego, A.; Zota, V.; Karni, A.; Krieger, J.I.; Bar-Or, A.; Bitan, G.; Budson, A.E.; Sperling, R.; Selkoe, D.J.; Weiner, H.L. Increased T cell reactivity to amyloid beta protein in older humans and patients with Alzheimer disease. J. Clin. Investig. 2003, 112, 415–422. [Google Scholar] [CrossRef]
  172. Saresella, M.; Calabrese, E.; Marventano, I.; Piancone, F.; Gatti, A.; Calvo, M.G.; Nemni, R.; Clerici, M. PD1 negative and PD1 positive CD4+ T regulatory cells in mild cognitive impairment and Alzheimer’s disease. J. Alzheimer’s Dis. 2010, 21, 927–938. [Google Scholar] [CrossRef]
  173. Machhi, J.; Yeapuri, P.; Lu, Y.; Foster, E.; Chikhale, R.; Herskovitz, J.; Namminga, K.L.; Olson, K.E.; Abdelmoaty, M.M.; Gao, J.; et al. CD4+ effector T cells accelerate Alzheimer’s disease in mice. J. Neuroinflamm. 2021, 18, 272. [Google Scholar] [CrossRef]
  174. Browne, T.C.; McQuillan, K.; McManus, R.M.; O’Reilly, J.-A.; Mills, K.H.G.; Lynch, M.A. IFN-γ Production by amyloid β-specific Th1 cells promotes microglial activation and increases plaque burden in a mouse model of Alzheimer’s disease. J. Immunol. 2013, 190, 2241–2251. [Google Scholar] [CrossRef] [PubMed]
  175. McManus, R.M.; Higgins, S.C.; Mills, K.H.G.; Lynch, M.A. Respiratory infection promotes T cell infiltration and amyloid-β deposition in APP/PS1 mice. Neurobiol. Aging 2014, 35, 109–121. [Google Scholar] [CrossRef]
  176. Orgogozo, J.-M.; Gilman, S.; Dartigues, J.-F.; Laurent, B.; Puel, M.; Kirby, L.C.; Jouanny, P.; Dubois, B.; Eisner, L.; Flitman, S.; et al. Subacute meningoencephalitis in a subset of patients with AD after Abeta42 immunization. Neurology 2003, 61, 46–54. [Google Scholar] [CrossRef]
  177. Song, M.S.; Mook-Jung, I.; Lee, H.J.; Min, J.Y.; Park, M.H. Serum anti-amyloid-beta antibodies and Alzheimer’s disease in elderly Korean patients. J. Int. Med. Res. 2007, 35, 301–306. [Google Scholar] [CrossRef] [PubMed]
  178. Cao, W.; Zheng, H. Peripheral immune system in aging and Alzheimer’s disease. Mol. Neurodegener. 2018, 13, 51. [Google Scholar] [CrossRef]
  179. Nagele, E.P.; Han, M.; Acharya, N.K.; DeMarshall, C.; Kosciuk, M.C.; Nagele, R.G. Natural IgG Autoantibodies Are Abundant and Ubiquitous in Human Sera, and Their Number Is Influenced By Age, Gender, and Disease. PLoS ONE 2013, 8, e60726. [Google Scholar] [CrossRef]
  180. Britschgi, M.; Olin, C.E.; Johns, H.T.; Takeda-Uchimura, Y.; LeMieux, M.C.; Rufibach, K.; Rajadas, J.; Zhang, H.; Tomooka, B.; Robinson, W.H.; et al. Neuroprotective natural antibodies to assemblies of amyloidogenic peptides decrease with normal aging and advancing Alzheimer’s disease. Proc. Natl. Acad. Sci. USA 2009, 106, 12145–12150. [Google Scholar] [CrossRef] [PubMed]
  181. Szabo, P.; Relkin, N.; Weksler, M.E. Natural human antibodies to amyloid beta peptide. Autoimmun. Rev. 2008, 7, 415–420. [Google Scholar] [CrossRef]
  182. Esposito, M.; Luccarini, I.; Cicatiello, V.; De Falco, D.; Fiorentini, A.; Barba, P.; Casamenti, F.; Prisco, A. Immunogenicity and therapeutic efficacy of phage-displayed beta-amyloid epitopes. Mol. Immunol. 2008, 45, 1056–1062. [Google Scholar] [CrossRef]
  183. Winblad, B.; Andreasen, N.; Minthon, L.; Floesser, A.; Imbert, G.; Dumortier, T.; Maguire, R.P.; Blennow, K.; Lundmark, J.; Staufenbiel, M.; et al. Safety, tolerability, and antibody response of active Aβ immunotherapy with CAD106 in patients with Alzheimer’s disease: Randomised, double-blind, placebo-controlled, first-in-human study. Lancet Neurol. 2012, 11, 597–604. [Google Scholar] [CrossRef]
  184. Jeremic, D.; Jiménez-Díaz, L.; Navarro-López, J.D. Past, present and future of therapeutic strategies against amyloid-β peptides in Alzheimer’s disease: A systematic review. Ageing Res. Rev. 2021, 72, 101496. [Google Scholar] [CrossRef]
  185. Suzuki, K.; Iwata, A.; Iwatsubo, T. The past, present, and future of disease-modifying therapies for Alzheimer’s disease. Proc. Jpn. Acad. Ser. B Phys. Biol. Sci. 2017, 93, 757–771. [Google Scholar] [CrossRef] [PubMed]
  186. Valera, E.; Spencer, B.; Masliah, E. Immunotherapeutic Approaches Targeting Amyloid-β, α-Synuclein, and Tau for the Treatment of Neurodegenerative Disorders. Neurother. J. Am. Soc. Exp. Neurother. 2016, 13, 179–189. [Google Scholar] [CrossRef] [PubMed]
  187. Boche, D.; Zotova, E.; Weller, R.O.; Love, S.; Neal, J.W.; Pickering, R.M.; Wilkinson, D.; Holmes, C.; Nicoll, J.A.R. Consequence of Abeta immunization on the vasculature of human Alzheimer’s disease brain. Brain J. Neurol. 2008, 131, 3299–3310. [Google Scholar] [CrossRef] [PubMed]
  188. Pardridge, W.M. Alzheimer’s disease drug development and the problem of the blood-brain barrier. Alzheimer’s Dement. J. Alzheimer’s Assoc. 2009, 5, 427–432. [Google Scholar] [CrossRef] [PubMed]
  189. Pocevičiūtė, D.; Nuñez-Diaz, C.; Roth, B.; Janelidze, S.; Giannisis, A.; Hansson, O.; Wennström, M. Increased plasma and brain immunoglobulin A in Alzheimer’s disease is lost in apolipoprotein E ε4 carriers. Alzheimer’s Res. Ther. 2022, 14, 117. [Google Scholar] [CrossRef] [PubMed]
  190. Forner, S.; Kawauchi, S.; Balderrama-Gutierrez, G.; Kramár, E.A.; Matheos, D.P.; Phan, J.; Javonillo, D.I.; Tran, K.M.; Hingco, E.; da Cunha, C.; et al. Systematic phenotyping and characterization of the 5xFAD mouse model of Alzheimer’s disease. Sci. Data 2021, 8, 270. [Google Scholar] [CrossRef]
  191. Wirths, O.; Erck, C.; Martens, H.; Harmeier, A.; Geumann, C.; Jawhar, S.; Kumar, S.; Multhaup, G.; Walter, J.; Ingelsson, M.; et al. Identification of Low Molecular Weight Pyroglutamate Aβ Oligomers in Alzheimer Disease. J. Biol. Chem. 2010, 285, 41517–41524. [Google Scholar] [CrossRef] [PubMed]
  192. Antonios, G.; Borgers, H.; Richard, B.C.; Brauß, A.; Meißner, J.; Weggen, S.; Pena, V.; Pillot, T.; Davies, S.L.; Bakrania, P.; et al. Alzheimer therapy with an antibody against N-terminal Abeta 4-X and pyroglutamate Abeta 3-X. Sci. Rep. 2015, 5, 17338. [Google Scholar] [CrossRef]
  193. Bakrania, P.; Hall, G.; Bouter, Y.; Bouter, C.; Beindorff, N.; Cowan, R.; Davies, S.; Price, J.; Mpamhanga, C.; Love, E.; et al. Discovery of a novel pseudo β-hairpin structure of N-truncated amyloid-β for use as a vaccine against Alzheimer’s disease. Mol. Psychiatry 2022, 27, 840–848. [Google Scholar] [CrossRef]
  194. Janssens, J.; Hermans, B.; Vandermeeren, M.; Barale-Thomas, E.; Borgers, M.; Willems, R.; Meulders, G.; Wintmolders, C.; Van den Bulck, D.; Bottelbergs, A.; et al. Passive immunotherapy with a novel antibody against 3pE-modified Aβ demonstrates potential for enhanced efficacy and favorable safety in combination with BACE inhibitor treatment in plaque-depositing mice. Neurobiol. Dis. 2021, 154, 105365. [Google Scholar] [CrossRef] [PubMed]
  195. Frost, J.; Liu, B.; Rahfeld, J.-U.; Kleinschmidt, M.; O’nuallain, B.; Le, K.; Lues, I.; Caldarone, B.; Schilling, S.; Demuth, H.-U.; et al. An anti-pyroglutamate-3 Aβ vaccine reduces plaques and improves cognition in APPswe/PS1ΔE9 mice. Neurobiol. Aging 2015, 36, 3187–3199. [Google Scholar] [CrossRef] [PubMed]
  196. DeMattos, R.B.; Lu, J.; Tang, Y.; Racke, M.M.; DeLong, C.A.; Tzaferis, J.A.; Hole, J.T.; Forster, B.M.; McDonnell, P.C.; Liu, F.; et al. A Plaque-Specific Antibody Clears Existing β-amyloid Plaques in Alzheimer’s Disease Mice. Neuron 2012, 76, 908–920. [Google Scholar] [CrossRef] [PubMed]
  197. Frost, J.L.; Liu, B.; Kleinschmidt, M.; Schilling, S.; Demuth, H.-U.; Lemere, C.A. Passive Immunization against Pyroglutamate-3 Amyloid-β Reduces Plaque Burden in Alzheimer-Like Transgenic Mice: A Pilot Study. Neurodegener. Dis. 2012, 10, 265–270. [Google Scholar] [CrossRef] [PubMed]
  198. Mintun, M.A.; Lo, A.C.; Duggan Evans, C.; Wessels, A.M.; Ardayfio, P.A.; Andersen, S.W.; Shcherbinin, S.; Sparks, J.; Sims, J.R.; Brys, M.; et al. Donanemab in Early Alzheimer’s Disease. N. Engl. J. Med. 2021, 384, 1691–1704. [Google Scholar] [CrossRef]
  199. Sims, J.R.; Zimmer, J.A.; Evans, C.D.; Lu, M.; Ardayfio, P.; Sparks, J.; Wessels, A.M.; Shcherbinin, S.; Wang, H.; Monkul Nery, E.S.; et al. Donanemab in Early Symptomatic Alzheimer Disease: The TRAILBLAZER-ALZ 2 Randomized Clinical Trial. JAMA 2023, 330, 512–527. [Google Scholar] [CrossRef]
  200. Grill, J.; Sajjadi, S.A.; Sultzer, D. FDA Approves Donanemab—UCI MIND. Available online: https://mind.uci.edu/donanemab/ (accessed on 26 August 2024).
  201. Busse, M.; Michler, E.; von Hoff, F.; Dobrowolny, H.; Hartig, R.; Frodl, T.; Busse, S. Alterations in the Peripheral Immune System in Dementia. J. Alzheimer’s Dis. 2017, 58, 1303–1313. [Google Scholar] [CrossRef] [PubMed]
  202. Pellicanò, M.; Bulati, M.; Buffa, S.; Barbagallo, M.; Di Prima, A.; Misiano, G.; Picone, P.; Di Carlo, M.; Nuzzo, D.; Candore, G.; et al. Systemic immune responses in Alzheimer’s disease: In vitro mononuclear cell activation and cytokine production. J. Alzheimer’s Dis. 2010, 21, 181–192. [Google Scholar] [CrossRef]
  203. Kim, K.; Wang, X.; Ragonnaud, E.; Bodogai, M.; Illouz, T.; DeLuca, M.; McDevitt, R.A.; Gusev, F.; Okun, E.; Rogaev, E.; et al. Therapeutic B-cell depletion reverses progression of Alzheimer’s disease. Nat. Commun. 2021, 12, 2185. [Google Scholar] [CrossRef] [PubMed]
  204. Shekari, A.; Fahnestock, M. Cholinergic neurodegeneration in Alzheimer disease mouse models. Handb. Clin. Neurol. 2021, 182, 191–209. [Google Scholar] [CrossRef]
  205. Li, H.; Wei, Y.; Wang, Z.; Wang, Q. Application of APP/PS1 transgenic mouse model for Alzheimers disease. J Alzheimer’s Park. 2015, 5, 10–4172. [Google Scholar]
  206. Van Meerhaeghe, T.; Néel, A.; Brouard, S.; Degauque, N. Regulation of CD8 T cell by B-cells: A narrative review. Front. Immunol. 2023, 14, 1125605. [Google Scholar] [CrossRef] [PubMed]
  207. Ahn, J.J.; Abu-Rub, M.; Miller, R.H. B Cells in Neuroinflammation: New Perspectives and Mechanistic Insights. Cells 2021, 10, 1605. [Google Scholar] [CrossRef] [PubMed]
  208. Iyer, S.S.; Cheng, G. Role of Interleukin 10 Transcriptional Regulation in Inflammation and Autoimmune Disease. Crit. Rev. Immunol. 2012, 32, 23–63. [Google Scholar] [CrossRef] [PubMed]
  209. Feng, W.; Zhang, Y.; Ding, S.; Chen, S.; Wang, T.; Wang, Z.; Zou, Y.; Sheng, C.; Chen, Y.; Pang, Y.; et al. B lymphocytes ameliorate Alzheimer’s disease-like neuropathology via interleukin-35. Brain. Behav. Immun. 2023, 108, 16–31. [Google Scholar] [CrossRef]
Figure 1. A model of Aβ and immune system participation in AD pathogenesis. Notes: blue arrows—stimulation, red arrows—inhibition; solid lines—established (acknowledged) regulatory pathways, dashed lines—potential (possible) regulatory pathways.
Figure 1. A model of Aβ and immune system participation in AD pathogenesis. Notes: blue arrows—stimulation, red arrows—inhibition; solid lines—established (acknowledged) regulatory pathways, dashed lines—potential (possible) regulatory pathways.
Cells 13 01624 g001
Table 2. Mouse models of Alzheimer’s disease discussed in this review.
Table 2. Mouse models of Alzheimer’s disease discussed in this review.
Mouse Model NR2B-Containing NMDA ReceptorGene
(Mutation)
Aβ PathologyTau PathologyNeuronal LossCognitive ImpairmentFirst Described
PDAPPAPP
(V717F)
6 mo. +--3 mo. +1995 [140]
APP-Tg J20APP
(K670/671NL, V717F)
8 mo. +-3 mo. +4 mo. +[141]
APP/PS1APP
(KM670/671NL)
6 mo. +-8 mo. +12 mo. +2006 [142]
PSEN1
(delta9)
5xFADAPP
(KM670/671NL,
I716V, V717I)
8 mo. +-6 mo. +4 mo. +2006 [143]
PSEN1
(M146L, L286V)
3xTgADAPP
(KM670/671NL)
6 mo. +12 mo. +unknown4 mo. +2003 [144]
MAPT 0N4R (P301L)
Psen
(M146V knock-in)
SJL miceexpressing IFN-γ under the MBP promoter[145]
APP/IFN-γ Tghomozygous IFN-γ-Tg mice were bred with APP-Tg J20 mice[146]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kolobova, E.; Petrushanko, I.; Mitkevich, V.; Makarov, A.A.; Grigorova, I.L. β-Amyloids and Immune Responses Associated with Alzheimer’s Disease. Cells 2024, 13, 1624. https://doi.org/10.3390/cells13191624

AMA Style

Kolobova E, Petrushanko I, Mitkevich V, Makarov AA, Grigorova IL. β-Amyloids and Immune Responses Associated with Alzheimer’s Disease. Cells. 2024; 13(19):1624. https://doi.org/10.3390/cells13191624

Chicago/Turabian Style

Kolobova, Elizaveta, Irina Petrushanko, Vladimir Mitkevich, Alexander A Makarov, and Irina L Grigorova. 2024. "β-Amyloids and Immune Responses Associated with Alzheimer’s Disease" Cells 13, no. 19: 1624. https://doi.org/10.3390/cells13191624

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop