• Open Access
    Review

    Innate and adaptive glial cell responses in Alzheimer’s disease

    Ankita Singh
    Tiratha Raj Singh *

    Explor Neuroprot Ther. 2023;3:90–104 DOI: https://doi.org/10.37349/ent.2023.00039

    Received: July 05, 2022 Accepted: January 20, 2023 Published: April 03, 2023

    Academic Editor: Jose Miguel Soria, CEU Cardenal Herrera University, Spain

    This article belongs to the special issue Glia and Neuroprotection

    Abstract

    Alzheimer’s disease (AD), which affects around twenty-seven million people globally, is an aging-related neurodegenerative condition characterized by the extracellular deposition of misfolded amyloid-β (Aβ) peptides and the intracellular production of neurofibrillary tangles (NFTs) AD results from the death of certain groups of neurons in the brain while appearing to have no impact on neighboring neurons. It is progressive and incurable. Therefore, the pathophysiology of afflicted populations and the development of intervention measures to stop neuronal cell death have been the main areas of attention for delineating therapeutic options. Proinflammatory cytokines are responsible for the stimulation of inflammatory responses and are mostly generated by activated macrophages in the brain. This review discusses how glial cells and innate and adaptive immune responses have a critical role in AD. It also provides information about microglial activation through the cluster of differentiation 40 (CD40) ligation and CD40L. CD40L ligation of microglial CD40 results in a large increase in tumor necrosis factor-α (TNF-α) production. Cultured cortical neuronal injury is caused when microglia are activated by CD40 ligation in the presence of interferon-γ (IFN-γ). This injury is significantly reduced by blocking the CD40 pathway or neutralising TNF-α. The management of AD would require integrating all available information about the innate and adaptive immune response affecting AD-related neuronal death.

    Keywords

    Alzheimer’s disease, neurodegenerative disease, aging, innate, adaptive, glia

    Introduction

    Alzheimer’s disease (AD) is a neurodegenerative ailment that is irreversible. It is characterized by the progressive degradation of brain areas necessary for memory and learning. The condition gradually worsens over months or years, affecting one’s memory, reasoning, judgment, communication ability, and even the ability to perform simple chores [1]. AD is thought to have been discovered for the first time in 1906, when a German psychiatrist, Alois Alzheimer, performed an autopsy on a woman with language and memory difficulties [2]. Abnormal structures of senile plaques and neurofibrillary tangles (NFTs) across the cerebral cortex of the patient were the first signs of AD [2]. As people grow older, their cognitive abilities change more frequently, and being older is the leading cause of cognitive decline, mild cognitive impairment, and age-related dementias like AD. It is believed that specific AD pathologies, such as the development of intracellular NFTs made up of tau and naturally occurring proteins that clump abnormally to form plaques that accumulate between neurons and impair cell function, start to form decades before symptoms appear [3], and lead to cortical atrophy and ventricular enlargement, which results in around a 35% reduction in the total mass of the brain. Early in the disease, the para-hippocampal areas are essential for the formation of new memories, but as it worsens, patients suffer from neuronal and synaptic loss [4]. AD is the most common cause of dementia in the world, it follows by frontotemporal lobar degeneration (FTLD), and cerebrovascular illness. Lewy bodies are protein accumulations that form in nerve cells in the parts of the brain responsible for thought, movement, and memory [5]. At least 60–70% of cases of dementia are caused by AD. Research suggests that AD appears to affect women more frequently than men [6, 7]. However, the reason for this fact is not known. AD is the sixth-largest cause of mortality in the USA and one of the most prevalent neurological conditions worldwide [8, 9].

    Diversity of the nervous system

    One of the most complex organs in the body is the brain. Aging is a significant cause of neurological conditions like AD [10, 11]. Most research into diseases caused by protein misfolding has been concentrated on the nervous system for many years. Glial cells are increasingly recognized as significant participants in neurodegeneration. Several diseases caused by protein misfolding [including amyotrophic lateral sclerosis (ALS), Parkinson’s disease (PD), and AD] show glial cell activation along with the deposition of misfolded proteins, yet their exact function in the neurological disease phase is unidentified. A large body of evidence indicates that misfolded proteins alone will not cause neuronal damage [12]. Glial cells were discovered to play a role in neurodegenerative diseases as early as 1910 [13]. As a result, it’s only recently been found that a significant study focus has switched back to improving the understanding of glial cells. Recent developments in identifying the stages involved in dementia have revealed that glial cells play a significant part in the process, even though much of the early research concentrated on the clinical stages of the condition [11].

    Neurodegenerative disorders and innate immunity

    In the central nervous system (CNS), the innate immune system comprises microglia and astrocytes. Microglia act as scouts for potentially hazardous substances or events, whereas astrocytes are primarily responsible for neurotrophic support and tissue homeostasis [14, 15]. Microglia become activated when immune-suppressive signals are blocked and pathogen-associated molecular patterns or endogenous damage-associated molecular patterns (DAMPs) are present. Activated microglia can develop various phenotypes based on environmental inputs [15]. According to preclinical studies in animal models, microglia are linked to the progression of neurodegenerative disorders such as PD and AD [16]. Based on environmental factors, microglia can become traditionally activated, causing a higher or smaller inflammatory process [17]. After activation, microglia can help remove the primary cause of inflammation by controlling the inflammatory response and promoting healing by secreting neurotrophins. Astrocytes, on the other hand, might increase inflammation, causing neurotoxicity [1820]. In neurological illnesses, microglial heterogeneity appears, and it may affect disease activities [21, 22]. Activating toll-like receptors (TLRs) results in phagocytosis, cytokine synthesis, and cell activation. These receptors are hypothesized to contribute to innate immunity and increase activity within the nervous system [23]. TLRs-2, 4, and 9, have been linked to the approval of abnormal protein collection and may play defensive microglia roles in the early phases of AD. Also, the form of inflammatory response that such receptors trigger depends on the stage of the illness, which is frequently benign in the initial phases but harmful later on [24, 25]. In AD, the activation of three complement pathways produces complement 5 (C5) convertase, which cleaves C5 into C5a and C5b. While C5a acts as an anaphylatoxin, C5b binds to C6, C7, C8, and multiple copies of C9 to form C5b-9, also known as the membrane attack complex [26], which co-localizes with amyloid plaques and tangles [27]. Microglial activation is also aided by neurons. The neuron-microglia connection includes the fractalkine [CX3C chemokine ligand 1 (CX3CL1)]-fractalkine receptor (CX3C chemokine receptor 1 (CX3CR1)] route, which restricts microglial activation. The microglia become active and hyperresponsive after the neuron is injured [28]. Microglial and astrocyte activation orchestration of non-cell-autonomous death is a common pathogenic feature of neurodegenerative diseases [29]. Hence, it is evident that microglia and astrocyte cells maintain the innate immune system in the CNS. Microglia cells are activated by a number of molecules and may contribute to neurological disorders.

    Neurodegenerative disorders and adaptive immunity

    Cytokines released by the activated glia, such as interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α), increase the endothelial production of adhesion molecules and enable lymphocyte infiltration in an inflammatory environment. When cells reach the injury site, they contribute to the inflammatory response and increase neuronal death [30]. Since B lymphocytes are found in the CNS, cells of patients with neurological disorders, and experimental animals with these ailments, it appears that humoral immunity plays a supportive role in neurodegeneration [31]. In a study, the humoral immune response in ALS patients was examined. Serum immunoglobulin G (IgG) concentrations increased significantly, while IgM levels remained below normal limits. According to the scientists, ALS serum antibody levels “show distinct immunological profiles associated with predominant humoral or cellular immune responses” and could indicate an autoimmune phase in disease development [32]. T regulatory cells (Tregs) are a group of T cluster of differentiation 4+ (CD4+) cells that help to moderate or reduce inflammation [33]. Tregs have the power to restrict T effector cell function and potentially start the apoptotic process. CD4+ CD25+ Tregs support the survival of neurons while suppressing microglial activation. This impact may be caused by blocking T helper type 17 (Th17) pathways. Upregulation of neurotrophic factors, including glial cell-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF), is one of the other hallmarks of Tregs [34]. Reduced inflammation protects neurons but also makes it more difficult to remove debris, misfolded proteins, and viral reservoirs [35]. Activated glial cells release IL-1β and TNF-α which promote lymphocyte infiltration and neuronal death. In ALS, IgG levels rise while IgM levels remain stable, indicating an autoimmune phase. Tregs restrict the function of T effector cells and start apoptosis, which is regulated by cell-derived factors and BDNF.

    Innate immunological response to AD

    AD patients’ blood and cerebrospinal fluid (CSF) contain antibodies that can be used to identify human senile plaques in the CNS. These plaques, also known as amyloid-β (Aβ) deposition and NFTs, cause an innate immune response that may be pathogenic, as opposed to the inflammatory response observed in AD [36, 37]. The innate immune system recognizes non-self-initiated ligands that express highly conserved pattern recognition receptors (PRRs). Complement receptors, TLRs, and scavenger receptors (SRs) are glial cells’ most frequently expressed molecules. To aid in the removal of debris, the glial cells of the immune system contain a large number of receptors, of which it is important to be aware [3841]. The complement system partly mediates the local inflammatory response brought on by Aβ deposition and NFTs in AD. Astrocytes, microglia, and neurons produce complement system components in the CNS. The majority of amyloid deposits in AD patients’ brains improve with complement component 1q (C1q) [4245]. The C1q gene has a distinct mechanism for attracting reactive glial cells by producing pro-inflammatory cytokines and oxidative byproducts like nitric oxide. It is found near the Aβ plaque, causing inflammatory processes, neuronal malfunction, and, eventually, degeneration. Reduced neuropathology is caused by the deletion of this gene in transgenic AD mice [46, 47]. The complement system protects against neurological disorders by removing aggregated and toxic proteins linked to them. As a result, increased phagocytosis of Aβ by microglial cells can be induced by activating the complement system [48]. For the efficient removal of Aβ without causing brain injury and, hence, for successful treatment outcomes, balanced complement activation is probably necessary [49].

    Chemokines are essential for controlling the innate immune response in the CNS. Chemokines draw in immune cells from the blood or brain. Chemokines may be produced by glial cells, particularly microglia and astrocytes in the CNS [50]. In the CNS, glial cells, mainly microglia, and astrocytes, are thought to be a key source of chemokines. Some chemokine receptors have already been found to be up-regulated in the AD brain, and studies have connected their activity to the activation of glial cells [50]. Antibodies found in the blood and CSF of AD patients may trigger a pathogenic immune response similar to the inflammatory response seen in the disease. TLRs and SRs are found on glial cells and many others to aid in the removal of debris. The CNS complement system is in charge of AD C1q—pulling reactive glial cells found near Aβ plaques, which cause neuronal malfunction and degeneration. Chemokines are also important molecules produced by glial cells that are found to activate them.

    Signaling via the innate immune system

    When bacterial infections associated with molecular patterns activate innate immune sensors in microglia, this sets off a series of signaling pathways that culminate in a planned response to the pathogens discovered. Microglia might not be able to distinguish between oligomeric or fibrillar Aβ and an invading pathogen. Fibrillar and aggregated Aβ can activate TLR2, TLR4, and TLR6 as well as their co-receptors to support signaling through the innate immune system. These receptors are present on microglia and participate in the activation of cells by various Aβ-types. Additionally, the development of DAMPs as well as the activation of microglia by Aβ have been linked to the SR-A and the ion channel Ca2+-activated K 3.1 (KCa3.1) [51]. The host uses the majority of the signaling routes that are triggered by neurodegenerative stimulation to combat infections. The immune system reacts to microorganisms by starting a proteolytic cascade that controls the synthesis of cytokines from the highly pro-inflammatory IL-1β family. The inflammasome apoptosis speck-like protein (ASC) binds to sensor molecules from the pyrin and hematopoietic expression, interferon (IFN)-inducible nature, and nuclear localization domain-containing (PYHIN) or nod-like receptor (NLR) families, which subsequently multimerize and activate caspase-1 to produce the inflammasome [52, 53]. Amyloid precursor protein (APP)/presenilin 1 (PS1) mice lacking NLR family pyrin domain containing 3 (NLRP3), caspase-1, or ASC are protected from developing AD by a protective mechanism that involves the production of IL-1β in the brain triggered by Aβ and increased Aβ clearance by microglia. The reduction of synaptic plasticity is entirely prevented by Aβ, which can affect the development of dendritic spines regulated by neurotrophic factor and tropomyosin-related kinase B (TRKB) [54, 55]. The NLRP3 inflammasome is a key player in both brain inflammation and typical age-related systemic inflammatory responses [56]. The brain’s inflammatory reaction can create a deadly feed-forward cycle. The immune system’s activation in response to Aβ and proinflammatory cytokines has been shown to hinder the clearance of Aβ and neuronal debris [57]. Phenotypic alterations in microglia work together to cause cognitive impairment in patients. The finding that IL-1 receptor-associated kinase 4 (IRAK4), a crucial kinase downstream of TLRs, can be disrupted to improve Aβ clearance and change microglial cells’ inflammatory phenotype from pro- to anti-inflammatory, is in line with the idea that pro-inflammatory molecules like the IL-1β family of cytokines or substances that mimic TLRs can impair the release feature of microglia [58]. When microglia cells come into contact with bacterial molecular patterns, they activate a number of signaling pathways. Certain receptors are present on microglia cells for different Aβ-types. A decrease in synaptic plasticity may prevent the development of dendritic spines. IRAK4 can be disrupted to improve Aβ clearance and change the inflammatory phenotypes of microglial cells. A family of cytokines that mimic TLR can impair the release features of microglia.

    Immune system regulation by glial cells in AD

    The health and regulation of the brain depend heavily on glial cells. The blood-brain barrier (BBB) can become more permeable in response to stress, allowing T cells and other specialized immune cells to be drawn in from the periphery. The effectiveness of any possible immune intervention therapy for neurodegeneration depends on understanding the communication with glial and peripheral immune cells [36]. Both innate and adaptive immune responses linked to the damage and repair processes in AD are tabulated in Table 1 [59].

    Innate and adaptive immune response in AD

    DiseaseInnate immunityAdaptive immunity
    ADChemokines, cytokines, and complements are all expressed in amyloid plaques. In vitro, Aβ induces innate immune expression. Aβ clearance is aided by microgliaAs a therapeutic approach, activation of the adaptive immune system against Aβ peptide will aid clearance
    Display full size

    Endothelial cell and glial cell interaction in AD

    BBB is supported and maintained through the connections with glial cells, astrocytes, and endothelial cells. For mammalian brain homeostasis, the BBB is essential. The low-density lipoprotein (LDL) receptor-related protein 1 (LRP-1) and receptor for advanced glycation endproducts (RAGE) are the two major receptors used by BBB to control the transit of Aβ to and from the brain. RAGE transmits Aβ into the brain, and LRP carries Aβ out of the brain [6062]. The balance between the pathways for Aβ synthesis and clearance depends heavily on LRP. Reactive astrocytes and the endothelium of the capillaries in the brain express LRP; apolipoprotein E (ApoE)-containing lipoproteins are secreted by glial cells and play a crucial role in maintaining the CNS’s lipid homeostasis. The presence of ApoE3, which is a risk factor for AD, was found to be more protective against cognitive decline than Apo4, suggesting it may be responsible for LRP’s upregulation [6367].

    Abnormal angiogenesis, dysregulated LRP1/RAGE-mediated movement, and arterial dysfunction may lead to Aβ formation, neurovascular inflammation, brain hypoperfusion, and cerebrovascular regression [68]. After crossing the BBB, the circulating Aβ is quickly taken up by neurons, causing cellular stress. To suppress blood flow, endothelin-1, a potent vasoconstrictor, is expanded as a result of the RAGE-Aβ interaction [60]. In 80% of cases of AD, there is amyloid buildup around cerebral blood vessels, which can result in cerebral amyloid angiopathy (CAA). Aβ deposits in the cerebrovasculature are connected to changes in smooth muscle cells, endothelial cell thinning, and mitochondrial loss. Aβ stroke with hemosiderin deposits, numerous infarcts, and cognitive impairment are caused by the deposits, which also cause intraparenchymal and subarachnoid hemorrhage [69].

    Interaction of glial cells and T cells in AD

    Middle-aged people with AD have a weaker T cell response to Aβ than healthy older people and patients with AD. It has become essential for AD immunotherapy to understand T cell responses and adaptive immunity, even though there is no detectable T cell reaction in patients with AD [70]. Glial cells are crucial in promoting the T cell response and controlling how the immune response develops. Contact between glial cells and T cells occurs through a variety of receptors, and the first type of signal regulates how T cells mature [71]. Recent studies have demonstrated that the interaction between the T cell receptor CD40L and CD40, which is expressed on the surface of microglia, enhances pro-inflammatory microglial cell activation in reaction to Aβ [72]. Decreased levels of Aβ and Aβ plaque deposition are seen in transgenic mice overproducing Aβ but lacking CD40L. This discovery confirms the CD40-CD40L relationship for future therapeutic aims in AD and points to several potential mechanisms behind the reduction of AD pathology in response to CD40L. Signaling pathways that are started by CD95L, CD95L [Fas ligand (FasL)]/CD95 (Fas), perforin, and the TNF receptor 1 pathway help prevent brain inflammation as part of the defense mechanism. Glial cells may promote the death of the infiltrating T cells [73, 74]. T cell responses could be advantageous by assisting in the removal of Aβ, most likely through activating microglial cells. This glial and T cell connection could point to a new target for reducing brain inflammation [36]. This association may aid in the production of innovative immunotherapeutic treatments for AD.

    AD microglia activation following CD40 ligation: a change in innate to an adaptive response

    Non-steroidal anti-inflammatory drug (NSAID) use is also connected to a lower incidence of AD in humans, and administering NSAIDs to AD mice shows less amyloid plaque load and improved microglial activation. Because activated microglia secrete pro-inflammatory innate cytokines like TNF-α and IL-1β, which have been shown to increase the risk of neuronal damage, they may play an important role in the etiology of AD [7578]. When microglia are challenged with tagged Aβ-peptides, phagocytosis is promoted but soluble or fibrillar Aβ-peptide breakdown via SRs is low. Maxfield’s laboratory discovered that the primary SRs (type I and II) are responsible for Aβ absorption by microglial cells [7983].

    The CD40-CD40L connection may contribute to AD pathogenesis by boosting brain inflammation after excessive amounts of astrocyte-derived CD40L and microglia-associated CD40 were recently discovered in and around Aβ plaques in the brains of AD patients. In animal models of AD, genetically eliminating CD40 or administering an antibody significantly lowers amyloid plaques [8486]. The phagocytic response to Aβ alone was not associated with the production of the pro-inflammatory innate cytokines TNF-α, IL-6, or IL-1β, a finding that is consistent with when microglia are challenged with apoptotic cells and produce an anti-inflammatory, pro-phagocytic innate response [87, 88]. CD40L reduced the speed of microglia phagocytosis and chased them away from cells after an hour of culture in the absence or presence of CD40L stimulation, preventing them from breaking down Aβ peptides. This reaction was enhanced by pro-inflammatory cytokines known to enhance effector T cell function. IFN-γ and TNF-α both pro-inflammatory Th1 cytokines, inhibited Aβ phagocytosis, whereas pro-inflammatory IL-4 and IL-10 increased its clearance. By using this experimental method, it can be concluded that CD40L also slowed down the microglial clearance of the peptide [80, 82].

    CD40 ligation raises increases Th1-promoting cytokines such as IL-6, TNF-α, IL-2, and IFN-γ in co-cultured cells while decreasing innate (phagocytic) activation of microglia stimulated with Aβ. An antagonistic CD40 antibody could be added to stop these effects, proving that the CD40-CD40L interaction itself is what causes these effects. The activation profile of microglia is biased away from innate, phagocytic activation and towards adaptive, APC activation when they come into contact with Aβ in the setting of co-simulation (e.g., CD40L) [80, 89].

    Glial cells in neurodegenerative disease

    To better understand the complicated glial responses in disease, animal models of developing neurodegeneration will be used to study the relationships between numerous brain area alterations. Glial cell interactions in the context of neurodegeneration have been extremely useful in learning prion disorders. Prions cause a predictable, advancing neurological disease in mice, with symptoms similar to those seen in human prion diseases [11]. One critical study used laser microdissection to separate astrocytes from distinct areas of the brain at various phases of AD, as characterised by the Braak neurofibrillary stages [90]. Many changes in gene expression were associated with astrocyte cytoskeletal proteins, which are compatible with the progressive reactive astrogliosis seen in AD. Apoptosis and ubiquitin-mediated proteolysis-related genes were revealed to be differently activated earlier in AD [9193]. Glial cell activity changes as the disease progress in brain areas susceptible or resistant to neurodegeneration. Understanding the glia’s therapeutic potential should improve on gaining a better understanding of the glial response in various brain areas as the illness progresses [12]. The microglia in areas of the brain where the activity of a range of microglial markers is affected by neurological diseases such as AD and prion diseases differ from those in regions with no signs of dementia [9496]. The stages of glial activation may influence the initiation of neurodegenerative diseases as well as the severity of dementia. An IFN-related phenotype of microglia was found in the afflicted brain regions of a recent meta-analysis of AD [97].

    Interactions of glial cells in AD

    In the developmental stage of illness, microglia express a variety of innate immune activation genes. The impact of different microglial activation states on the neurodegenerative process has been discussed. Even though astrocytes exhibit a wide range of functional abnormalities as the disease progresses, the impact of astrogliosis and astrocytic dysfunction on neuronal health has been restricted for many years. It is well recognized that astrocytes express a variety of immune-related genes, suggesting that they may be a part of an organized immune-activated response to a neurodegenerative ailment. The process through which this glial communication occurs has been discovered in prion diseases. Several cytokines lead to astrogliosis, secreted by activated microglia in the early stages of illness.

    Consequently, chemokine expression is enhanced by astrocytes, and this, along with local microglial proliferation, causes microglia to multiply and become more active. Progressive astrogliosis results from reactive microglia secreting more cytokines into the surrounding environment. Glial activation doesn’t seem to decrease over time during disease; rather, it seems to gradually increase with time, showing that this feedback loop is a glial-specific perpetual cycle. According to the latest research, AD neurodegeneration is caused by the astrogliosis condition induced by cytokines like IL-1β. The therapeutic potential of focusing on the connection between glial cells in AD is highlighted by investigations that have found proof of microglial-mediated astrogliosis resolution (Figure 1) [11, 95]. A previous study found that when injected into mice, a prion-infected neuron or astrocyte culture had chemotactic traits that weren’t seen in control cultures [98]. This finding suggests that neurons, astrocytes, or both mediated microglial recruitment to the injection site. One other study found that the proinflammatory cytokine IL-1β is crucial for activating astrogliosis and that, during illness, IL-1β production is powerful in microglia populations (including NLRP3 inflammasome pathway activation) [99, 100]. Microglia express various genes for the cultivation of the innate immune system. Astrocytes express a variety of immune-related genes, so they are part of the immune-activated response to a neurodegenerative ailment. The astrogliosis condition induced by IL-1β causes AD neurodegeneration. Either neurons, astrocytes, or both mediate microglial cell recruitment to the infection site. IL-1β is essential for astrogliosis activation, and its production greatly increases during illness.

    Mechanism of disease-related glial cells. Microglial activation causes the release of several cytokines that cause astrogliosis in the early stages of the disease. In response, astrocytes upregulate chemokine expression, which, together with local microglial growth, increases microglia numbers and reactive activity. This feedback loop is a glial-specific perpetual cycle because activation states of the glia do not appear to resolve over time. P2Y1: purinergic receptor

    Note. Adapted from “Unravelling the glial response in the pathogenesis of Alzheimer’s disease,” by Alibhai JD, Diack AB, Manson JC. FASEB J. 2018;32:5766–77 (https://faseb.onlinelibrary.wiley.com/doi/full/10.1096/fj.201801360R). CC BY.

    Microglial activation is important for astrogliosis because it triggers the cytokine that releases IL-1β, which in turn triggers a chemokine response that attracts more microglia, during illness, working as a feedback connection loop with glial cells. TNF-α, C1q, and IL-1β can be secreted by activated microglia and cause astrogliosis implying that the glial link processes underpin AD and other neurological disorders [101, 102]. Additionally, functional impairment in activated astrocytes can result in neuronal cell death. Such a type of astrogliosis is known as annexin 1 (A1) because the activation is functionally similar to a “classic” microglial activation state. A1 astrocytes have been found in a variety of neurodegenerative diseases including AD. This research shows that resolving the activation of glial cells or disrupting glial cell contact during illness could be a valuable treatment strategy. Recent research suggests that microglia can create neuroprotection in the astrocytic phenotype by down-regulating the P2Y1 receptor, opening the possibility of glial-targeted therapeutic intervention [103].

    Heavy metal intake alters astroglial homeostasis and neuroprotective cascades such as the glutamate/gamma-aminobutyric acid (GABA)-glutamine shuttle, antioxidative machinery, and energy metabolism. Astrocytes are the main homeostatic cells in the CNS that defend neurons from all kinds of insults, particularly heavy metal accumulation. This makes them the primary target of heavy metal neurotoxicity [104]. There is no significant difference in cuprum (Cu), iron (Fe), or zinc (Zn) concentrations in large structures such as the corpus callosum, cortex, and striatum in Mouse Toronto KnockOut mice (MTKO) compared to age-matched controls [105]. MTKO mice retain approximately one-third as many Cu-rich aggregates, resulting in a decrease in periventricular Cu concentration. As a binding protein, metallothionein, a cysteine-rich protein important for metal homeostasis, was proposed [105]. Astrocytes have strong pro-inflammatory abilities. Astrocytes are thus emerging as key regulators of CNS inflammatory responses. Astrocytes play an important role in attracting and restricting CNS inflammation, which has significant implications for a variety of CNS disorders [106]. Astrocytes are the most abundant cell type in the brain and contribute to brain homeostasis in a variety of ways, such as buffering extracellular K+ and controlling neurotransmitter release. Astrocytes can also create the BBB and regulate the brain’s immune response [107].

    Genetic information on AD and the innate immune system

    AD is a neurodegenerative disease caused by gene mutations that influence amyloid formation, such as PS1, PS2, and APP. A small proportion of AD is autosomal dominantly inherited within families and appears as early dementia onset, often between the ages of 30 and 60 [108]. Over the age of 30, AD risk loci have been identified thus far through a combination of candidate genes, genetic linkage, whole genome/exome sequencing studies, and genome-wide association studies. Over ninety-nine, percent of cases of AD occur in people over the age of 65 and are known as late-onset AD. Genetics and age are the two most significant risk factors [109]. Over half of the different genes proven by functional genomics are involved in innate immune cell function and microglial function, such as the top 2 risk genes, triggering receptor expressed on myeloid cells 2 (TREM2) and ApoE. The epigenomic analysis revealed that AD genome-wide association study loci are selectively enriched in promoter sequences associated with innate immune mechanisms [110].

    Prospects of AD

    Immunotherapy has attracted the attention of pharmaceutical companies, clinicians, and researchers seeking to develop an effective treatment for AD. Several immunotherapies have been developed and tested in clinical trials to combat the pathology of amyloid or tau [111].

    The presence of extracellular senile plaques, which are made of Aβ peptide, and intracellular NFTs, which are made of tau protein, is a hallmark of AD. Targeting the aggregation process of Aβ and tau has been an active strategy for designing and developing therapeutic agents for pharmacological intervention in AD [112]. Researchers found that synaptic pathology and microgliosis may be the first signs of neuronal dysfunction caused by tauopathy in P301S mutant tau transgenic mice. The activation of glycogen synthase kinase-3β (GSK-3β) or cyclin-dependent kinase-5 (CDK-5) kinases resulted in the stimulation of microglial cells and the release of IL-1β [113]. The CNS is affected by various neurodegenerative and neuroinflammatory disorders that affect GSK-3β function [8].

    Conclusions

    Chronic inflammation, which is a hallmark of neurodegenerative disorders, is stimulated in both directions by innate and adaptive immunity. Inflammation always turns pathogenic and causes neuronal death, although it typically acts as a preventive strategy in the initial phases of the disease. Glial cells are the first line of defense against pathological anomalies that develop in neurodegenerative illnesses. Glial cells are crucial for keeping a healthy brain, additionally, they also safeguard and support the brain’s ability to recover from damage. Glial cell activation can be mediated and affected by a variety of signals and is one of the most important functions of the CNS. Glial cells’ aberrant behavior throughout aging may be caused by alterations in their proliferating process and may provide light on the development of fatal disorders like AD. Both microglia and astrocytes appear to provide a dual function in the disease process by repairing CNS damage. Knowledge of these long-lasting neurological illness mechanisms has advanced with the discovery of commonalities among the many protein-misfolding diseases. However, because each glial family exhibits such diversity, they continue to remain a riddle that holds the secret to their alteration. In summary, inflammation and its pathology could be targeted to manage neurological conditions, and more specifically, AD. The glial response could be a crucial factor in accomplishing this future goal.

    Abbreviations

    AD:

    Alzheimer’s disease

    ALS:

    amyotrophic lateral sclerosis

    ApoE:

    apolipoprotein E

    Aβ:

    amyloid-β

    BBB:

    blood-brain barrier

    C1q:

    component 1q

    C5:

    complement 5

    CD40:

    cluster of differentiation 40

    CNS:

    central nervous system

    Cu:

    cuprum

    IFN:

    interferon

    IL-1β:

    interleukin-1β

    LRP-1:

    low-density lipoprotein receptor-related protein 1

    NFTs:

    neurofibrillary tangles

    NLRP3:

    nod-like receptor family pyrin domain containing 3

    PS1:

    presenilin 1

    RAGE:

    receptor for advanced glycation endproducts

    SRs:

    scavenger receptors

    Th17:

    T helper type 17

    TLRs:

    toll-like receptors

    TNF-α:

    tumor necrosis factor-α

    Tregs:

    T regulatory cells

    Declarations

    Acknowledgments

    Thanks are due to CEHTI and the bioinformatics lab of the Department of Biotechnology and Bioinformatics, Jaypee University of Information technology, Solan, Himachal Pradesh, India for the technical help.

    Author contributions

    TRS: Conceptualization, Writing—review & editing. AS: Writing—original draft, Writing—review & editing. All authors read and approved the submitted version.

    Conflicts of interest

    The authors declare that they have no conflicts of interest.

    Ethical approval

    Not applicable.

    Consent to participate

    Not applicable.

    Consent to publication

    Not applicable.

    Availability of data and materials

    Not applicable.

    Funding

    Not applicable.

    Copyright

    © The Author(s) 2023.

    References

    Gleerup HS, Hasselbalch SG, Simonsen AH. Biomarkers for Alzheimer’s disease in saliva: a systematic review. Dis Markers. 2019;2019:4761054. [DOI] [PubMed] [PMC]
    Hardy J. A hundred years of Alzheimer’s disease research. Neuron. 2006;52:313. [DOI] [PubMed]
    Rajan KB, Wilson RS, Weuve J, Barnes LL, Evans DA. Cognitive impairment 18 years before clinical diagnosis of Alzheimer disease dementia. Neurology. 2015;85:898904. [DOI] [PubMed] [PMC]
    Alves L, Correia ASA, Miguel R, Alegria P, Bugalho P. Alzheimer’s disease: a clinical practice-oriented review. Front Neurol. 2012;3:63. [DOI] [PubMed] [PMC]
    Alzheimer’s Association. 2019 Alzheimer’s disease facts and figures. Alzheimers Dement. 2019;15:32187. [DOI]
    Silva MVF, Loures CMG, Alves LCV, de Souza LC, Borges KBG, Carvalho MDG. Alzheimer’s disease: risk factors and potentially protective measures. J Biomed Sci. 2019;26:33. [DOI] [PubMed] [PMC]
    Gunawardena IPC, Retinasamy T, Shaikh MF. Is Aducanumab for LMICs? Promises and challenges. Brain Sci. 2021;11:1547. [DOI] [PubMed] [PMC]
    Shukla R, Singh TR. High-throughput screening of natural compounds and inhibition of a major therapeutic target HsGSK-3β for Alzheimer’s disease using computational approaches. J Genet Eng Biotechnol. 2021;19:61. [DOI] [PubMed] [PMC]
    Kumar A, Singh TR. A new decision tree to solve the puzzle of Alzheimer’s disease pathogenesis through standard diagnosis scoring system. Interdiscip Sci. 2017;9:10715. [DOI] [PubMed]
    Kumar A, Bansal A, Singh TR. ABCD: Alzheimer’s disease biomarkers comprehensive database. 3 Biotech. 2019;9:351. [DOI] [PubMed] [PMC]
    Alibhai JD, Diack AB, Manson JC. Unravelling the glial response in the pathogenesis of Alzheimer’s disease. FASEB J. 2018;32:576677. [DOI] [PubMed]
    Alibhai J, Blanco RA, Barria MA, Piccardo P, Caughey B, Perry VH, et al. Distribution of misfolded prion protein seeding activity alone does not predict regions of neurodegeneration. PLoS Biol. 2016;14:e1002579. [DOI] [PubMed] [PMC]
    Heneka MT, Rodríguez JJ, Verkhratsky A. Neuroglia in neurodegeneration. Brain Res Rev. 2010;63:189211. [DOI] [PubMed]
    Zhao J, Su M, Lin Y, Liu H, He Z, Lai L. Administration of amyloid precursor protein gene deleted mouse ESC-derived thymic epithelial progenitors attenuates Alzheimer’s pathology. Front Immunol. 2020;11:1781. [DOI] [PubMed] [PMC]
    Hanisch UK, Kettenmann H. Microglia: active sensor and versatile effector cells in the normal and pathologic brain. Nat Neurosc. 2007;10:138794. [DOI] [PubMed]
    McCoy MK, Martinez TN, Ruhn KA, Szymkowski DE, Smith CG, Botterman BR, et al. Blocking soluble tumor necrosis factor signaling with dominant-negative tumor necrosis factor inhibitor attenuates loss of dopaminergic neurons in models of Parkinson’s disease. J Neurosci. 2006;26:936575. [DOI] [PubMed] [PMC]
    Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A, Locati M. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 2004;25:67786. [DOI] [PubMed]
    Hanisch UK. Microglia as a source and target of cytokines. Glia. 2002;40:14055. [DOI] [PubMed]
    Innes S, Pariante CM, Borsini A. Microglial-driven changes in synaptic plasticity: a possible role in major depressive disorder. Psychoneuroendocrinology. 2019;102:23647. [DOI] [PubMed]
    Heese K, Hock C, Otten U. Inflammatory signals induce neurotrophin expression in human microglial cells. J Neurochem. 1998;70:699707. [DOI] [PubMed]
    Colton CA, Mott RT, Sharpe H, Xu Q, van Nostrand WE, Vitek MP. Expression profiles for macrophage alternative activation genes in AD and in mouse models of AD. J Neuroinflammation. 2006;3:27. [DOI] [PubMed] [PMC]
    Carson MJ, Bilousova TV, Puntambekar SS, Melchior B, Doose JM, Ethell IM. A rose by any other name? The potential consequences of microglial heterogeneity during CNS health and disease. Neurotherapeutics. 2007;4:5719. [DOI] [PubMed] [PMC]
    Kawai T, Akira S. The role of pattern-recognition receptors in innate immunity: update on toll-like receptors. Nat Immunol. 2010;11:37384. [DOI] [PubMed]
    Buchanan MM, Hutchinson H, Watkins LR, Yin H. Toll-like receptor 4 in CNS pathologies. J Neurochem. 2010;114:1327. [DOI] [PubMed] [PMC]
    Rifkin IR, Leadbetter EA, Busconi L, Viglianti G, Marshak-Rothstein A. Toll-like receptors, endogenous ligands, and systemic autoimmune disease. Immunol Rev. 2005;204:2742. [DOI] [PubMed]
    Koopman JJE, van Essen MF, Rennke HG, de Vries APJ, van Kooten C. Deposition of the membrane attack complex in healthy and diseased human kidneys. Front Immunol. 2021;11:599974. [DOI] [PubMed] [PMC]
    Maier M, Peng Y, Jiang L, Seabrook TJ, Carroll MC, Lemere CA. Complement C3 deficiency leads to accelerated amyloid β plaque deposition and neurodegeneration and modulation of the microglia/macrophage phenotype in amyloid precursor protein transgenic mice. J Neurosci. 2008;28:633341. [DOI] [PubMed] [PMC]
    Cardona AE, Poprp EP, Sasse ME, Kostenko V, Cardona SM, Dijkstra IM, et al. Control of microglial neurotoxicity by the fractalkine receptor. Nat Neurosci. 2006;9:91724. [DOI] [PubMed]
    Ilieva H, Polymenidou M, Cleveland DW. Non-cell autonomous toxicity in neurodegenerative disorders: ALS and beyond. J Cell Biol. 2009;187:76172. [DOI] [PubMed] [PMC]
    Mosley RL, Hutter-Saunders JA, Stone DK, Gendelman HE. Inflammation and adaptive immunity in Parkinson’s disease. Cold Spring Harb Perspect Med. 2012;2:a009381. [DOI] [PubMed] [PMC]
    Bartos A, Fialová L, Sřvarcová J, Ripova D. Patients with Alzheimer disease have elevated intrathecal synthesis of antibodies against tau protein and heavy neurofilament. J Neuroimmunol. 2012;252:1005. [DOI] [PubMed]
    Saleh IA, Zesiewicz T, Xie Y, Sullivan KL, Miller AM, Kuzmin-Nichols N, et al. Evaluation of humoral immune response in adaptive immunity in ALS patients during disease progression. J Neuroimmunol. 2009;215:96101. [DOI] [PubMed]
    Romero-Ramos M, von Euler Chelpin M, Sanchez-Guajardo V. Vaccination strategies for Parkinson disease: induction of a swift attack or raising tolerance? Hum Vaccin Immunother. 2014;10:85267. [DOI] [PubMed] [PMC]
    Rodrigues MCO, Sanberg PR, Cruz LE, Garbuzova-Davis S. The innate and adaptive immunological aspects in neurodegenerative diseases. J Neuroimmunol. 2014;269:18. [DOI] [PubMed]
    Reynolds AD, Stone DK, Mosley RL, Gendelman HE. Proteomic studies of nitrated alpha-synuclein microglia regulation by CD4+CD25+ T cells. J Proteome Res. 2009;8:3497511. [DOI] [PubMed] [PMC]
    Farfara D, Lifshitz V, Frenkel D. Neuroprotective and neurotoxic properties of glial cells in the pathogenesis of Alzheimer’s disease. J Cell Mol Med. 2008;12:76280. [DOI] [PubMed] [PMC]
    Kellner J, Matschke J, Bernreuther C, Moch H, Ferrer I, Glatzel M. Autoantibodies against β-amyloid are common in Alzheimer’s disease and help control plaque burden. Ann Neurol. 2009;65:2431. [DOI] [PubMed]
    Husemann J, Loike JD, Anankov R, Febbraio M, Silverstein SC. Scavenger receptors in neurobiology and neuropathology: their role on microglia and other cells of the nervous system. Glia. 2002;40:195205. [DOI] [PubMed]
    Bsibsi M, Persoon-Deen C, Verwer RWH, Meeuwsen S, Ravid R, van Noort JM. Toll-like receptor 3 on adult human astrocytes triggers production of neuroprotective mediators. Glia. 2006;53:68895. [DOI] [PubMed]
    McGeer PL, McGeer EG. Inflammation, autotoxicity and Alzheimer disease. Neurobiol Aging. 2001;22:799809. [DOI] [PubMed]
    Gasque P, Dean YD, McGreal EP, Vanbeek J, Morgan BP. Complement components of the innate immune system in health and disease in the CNS. Immunopharmacology. 2000;49:17186. [DOI] [PubMed]
    Shukla R, Munjal NS, Singh TR. Identification of novel small molecules against GSK3β for Alzheimer’s disease using chemoinformatics approach. J Mol Graph Model. 2019;91:91104. [DOI] [PubMed]
    McGreal E, Gasque P. Structure-function studies of the receptors for complement C1q. Biochemic Soc Trans. 2002;30:10104. [DOI] [PubMed]
    Webster S, Rogers J. Relative efficacies of amyloid β peptide (Aβ) binding proteins in Aβ aggregation. J Neurosci Res. 1996;46:5866. [DOI] [PubMed]
    Panayiotou E, Fella E, Papacharalambous R, Malas S, Kyriakides T. The role of complement in ATTR amyloidosis: a new therapeutic avenue? Orphanet J Rare Dis. 2015;10:P3. [DOI]
    Bonifati DM, Kishore U. Role of complement in neurodegeneration and neuroinflammation. Mol Immunol. 2007;44:9991010. [DOI] [PubMed]
    McGeer PL, McGeer EG. Mechanisms of cell death in Alzheimer disease—immunopathology. J Neural Transm Suppl. 1998;54:15966. [DOI] [PubMed]
    Webster SD, Galvan MD, Ferran E, Garzon-Rodriguez W, Glabe CG, Tenner AJ. Antibody-mediated phagocytosis of the amyloid β-peptide in microglia is differentially modulated by C1q. J Immunol. 2001;166:7496503. [DOI] [PubMed]
    Wyss-Coray T, Yan F, Lin AHT, Lambris JD, Alexander JJ, Quigg RJ, et al. Prominent neurodegeneration and increased plaque formation in complement-inhibited Alzheimer’s mice. Proc Natl Acad Sci U S A. 2002;99:1083742. [DOI] [PubMed] [PMC]
    Xia MQ, Hyman BT. Chemokines/chemokine receptors in the central nervous system and Alzheimer’s disease. J Neurovirol. 1999;5:3241. [DOI] [PubMed]
    Heneka MT, Golenbock DT, Latz E. Innate immunity in Alzheimer’s disease. Nat Immunol. 2015;16:22936. [DOI] [PubMed]
    Latz E, Xiao TS, Stutz A. Activation and regulation of the inflammasomes. Nat Rev Immunol. 2013;13:397411. [DOI] [PubMed] [PMC]
    Halle A, Hornung V, Petzold GC, Stewart CR, Monks BG, Reinheckel T, et al. The NALP3 inflammasome is involved in the innate immune response to amyloid-β. Nat Immunol. 2008;9:85765. [DOI] [PubMed] [PMC]
    Yasuno F, Kosaka J, Ota M, Higuchi M, Ito H, Fujimura Y, et al. Increased binding of peripheral benzodiazepine receptor in mild cognitive impairment–dementia converters measured by positron emission tomography with [11C]DAA1106. Psychiatry Res. 2012;203:6774. [DOI] [PubMed]
    Tong L, Prieto GA, Kramár EA, Smith ED, Cribbs DH, Lynch G, et al. Brain-derived neurotrophic factor-dependent synaptic plasticity is suppressed by interleukin-1β via p38 mitogen-activated protein kinase. J Neurosci. 2012;32:1771424. [DOI] [PubMed] [PMC]
    Youm YH, Grant RW, McCabe LR, Albarado DC, Nguyen KY, Ravussin A, et al. Canonical Nlrp3 inflammasome links systemic low-grade inflammation to functional decline in aging. Cell Metab. 2013;18:51932. [DOI] [PubMed] [PMC]
    Heneka MT, Kummer MP, Stutz A, Delekate A, Schwartz S, Vieira-Saecker A, et al. NLRP3 is activated in Alzheimer’s disease and contributes to pathology in APP/PS1 mice. Nature. 2013;493:6748. [DOI] [PubMed] [PMC]
    Cameron B, Tse W, Lamb R, Li X, Lamb BT, Landreth GE. Loss of interleukin receptor-associated kinase 4 signaling suppresses amyloid pathology and alters microglial phenotype in a mouse model of Alzheimer’s disease. J Neurosci. 2012;32:1511223. [DOI] [PubMed] [PMC]
    Amor S, Woodroofe MN. Innate and adaptive immune responses in neurodegeneration and repair. Immunology. 2014;141:28791. [DOI] [PubMed] [PMC]
    Deane D, Wu Z, Zlokovic BV. RAGE (Yin) versus LRP (Yang) balance regulates Alzheimer amyloid β-peptide clearance through transport across the blood-brain barrier. Stroke. 2004;35:262831. [DOI] [PubMed]
    Deane R, Du Yan S, Submamaryan RK, LaRue B, Jovanovic S, Hogg E, et al. RAGE mediates amyloid-β peptide transport across the blood-brain barrier and accumulation in brain. Nat Med. 2003;9:90713. [DOI] [PubMed]
    Yang J, Aschner M. Developmental aspects of blood-brain barrier (BBB) and rat brain endothelial (RBE4) cells as in vitro model for studies on chlorpyrifos transport. Neurotoxicology. 2003;24:7415. [DOI] [PubMed]
    Levy-Lahad E, Bird TD. Genetic factors in Alzheimer’s disease: a review of recent advances. Ann Neurol. 1996;40:82940. [DOI] [PubMed]
    Hayashi H, Campenot RB, Vance DE, Vance JE. Apolipoprotein E-containing lipoproteins protect neurons from apoptosis via a signaling pathway involving low-density lipoprotein receptor-related protein-1. J Neurosci. 2007;27:193341. [DOI] [PubMed] [PMC]
    Han X. The role of apolipoprotein E in lipid metabolism in the central nervous system. Cell Mole Life Sci. 2004;61:1896906. [DOI] [PubMed]
    Deane R, Bell RD, Sagare A, Zlokovic BV. Clearance of amyloid-β peptide across the blood-brain barrier: implication for therapies in Alzheimer’s disease. CNS Neurol Disord Drug Targets. 2009;8:1630. [DOI] [PubMed] [PMC]
    Shibata M, Yamada S, Kumar SR, Calero M, Bading J, Frangione B, et al. Clearance of Alzheimer’s amyloid-β1-40 peptide from brain by LDL receptor-related protein-1 at the blood-brain barrier. J Clin Invest. 2000;106:148999. [DOI] [PubMed] [PMC]
    Deane R, Zlokovic BV. Role of the blood-brain barrier in the pathogenesis of Alzheimers disease. Curr Alzheimer Res. 2007;4:1917. [DOI] [PubMed]
    Weller RO, Nicoll JAR. Cerebral amyloid angiopathy: pathogenesis and effects on the ageing and Alzheimer brain. Neurol Res. 2003;25:6116. [DOI] [PubMed]
    Monsonego A, Zota V, Karni A, Krieger JI, Bar-Or A, Bitan G, et al. Increased T cell reactivity to amyloid β protein in older humans and patients with Alzheimer disease. J Clin Invest. 2003;112:41522. [DOI] [PubMed] [PMC]
    Chaouchi N, Wallon C, Taieb J, Auffredou MT, Tertian G, Lemoine FM, et al. Interferon-alpha-mediated prevention of in vitro apoptosis of chronic lymphocytic leukemia B cells: role of bcl-2 and c-myc. Clin Immunol Immunopathol. 1994;73:197204. [DOI] [PubMed]
    Tan J, Town T, Crawford F, Mori T, DelleDonne A, Crescentini R, et al. Role of CD40 ligand in amyloidosis in transgenic Alzheimer’s mice. Nat Neurosci. 2002;5:128893. [DOI] [PubMed]
    Magnus T, Chan A, Grauer O, Toyka KV, Gold R. Microglial phagocytosis of apoptotic inflammatory T cells leads to down-regulation of microglial immune activation. J Immunol. 2001;167:500410. [DOI] [PubMed]
    Gasque P, Jones J, Singhrao SK, Morgan BP. Identification of an astrocyte cell population from human brain that expresses perforin, a cytotoxic protein implicated in immune defense. J Exp Med. 1998;187:45160. [DOI] [PubMed] [PMC]
    Takada LT. Innate immunity and inflammation in Alzheimer’s disease pathogenesis. Arq Neuropsiquiatr. 2017;75:6078. [DOI] [PubMed]
    Meda L, Cassatella MA, Szendrei GI, Otvos L Jr, Baron P, Villalba M, et al. Activation of microglial cells by β-amyloid protein and interferon-γ. Nature. 1995;374:64750. [DOI] [PubMed]
    Lim GP, Chu T, Yang F, Beech W, Frautschy SA, Cole GM. The curry spice curcumin reduces oxidative damage and amyloid pathology in an Alzheimer transgenic mouse. J Neurosci. 2001;21:83707. [DOI] [PubMed] [PMC]
    McGeer EG, McGeer PL. The importance of inflammatory mechanisms in alzheimer disease. Exp Gerontol. 1998;33:3718. [DOI] [PubMed]
    Brazil MI, Chung H, Maxfield FR. Effects of incorporation of immunoglobulin G and complement component C1q on uptake and degradation of Alzheimer’s disease amyloid fibrils by microglia. J Bio Chem. 2000;275:169417. [DOI] [PubMed]
    Town T, Nikolic V, Tan J. The microglial “activation” continuum: from innate to adaptive responses. J Neuroinflammation. 2005;2:24. [DOI] [PubMed] [PMC]
    Paresce DM, Chung H, Maxfield FR. Slow degradation of aggregates of the Alzheimer’s disease amyloid β-protein by microglial cells. J Bio Chem. 1997;272:293907. [DOI] [PubMed]
    Paresce DM, Ghosh RN, Maxfield FR. Microglial cells internalize aggregates of the Alzheimer’s disease amyloid β-protein via a scavenger receptor. Neuron. 1996;17:55365. [DOI] [PubMed]
    Wang T, Town T, Alexopoulou L, Anderson JF, Fikrig E, Flavell RA. Toll-like receptor 3 mediates west nile virus entry into the brain causing lethal encephalitis. Nat Med. 2004;10:136673. [DOI] [PubMed]
    Tan J, Town T, Paris D, Mori T, Suo Z, Crawford F, et al. Microglial activation resulting from CD40-CD40L interaction after β-amyloid stimulation. Science. 1999;286:23525. [DOI] [PubMed]
    Calingasan NY, Erdely HA, Altar CA. Identification of CD40 ligand in Alzheimer’s disease and in animal models of Alzheimer’s disease and brain injury. Neurobiol Aging. 2002;23:319. [DOI] [PubMed]
    Town T, Tan J, Mullan M. CD40 signaling and Alzheimer’s disease pathogenesis. Neurochem Int. 2001;39:37180. [DOI] [PubMed]
    Townsend KP, Town T, Mori T, Lue LF, Shytle D, Sanberg PR, et al. CD40 signaling regulates innate and adaptive activation of microglia in response to amyloid β-peptide. Eur J Immunol. 2005;35:90110. [DOI] [PubMed]
    Minghetti L, Ajmone-Cat MA, de Berardinis MA, de Simone R. Microglial activation in chronic neurodegenerative diseases: roles of apoptotic neurons and chronic stimulation. Brain Res Brain Res Rev. 2005;48:2516. [DOI] [PubMed]
    Austin SA, Combs CK. Mechanisms of microglial activation by amyloid precursor protein and its proteolytic fragments. In: Lane TE, Carson M, Bergmann C, Wyss-Coray T, editors. Central nervous system diseases and inflammation. Boston: Springer; 2008. pp. 13–32.
    Simpson JE, Ince PG, Shaw PJ, Heath PR, Raman R, Garwood CJ, et al. Microarray analysis of the astrocyte transcriptome in the aging brain: relationship to Alzheimer’s pathology and APOE genotype. Neurobiol Aging. 2011;32:1795807. [DOI] [PubMed]
    Tan MG, Chua WT, Esiri MM, Smith AD, Vinters HV, Lai MK. Genome wide profiling of altered gene expression in the neocortex of Alzheimer’s disease. J Neurosci Res. 2010;88:115769. [DOI] [PubMed]
    Cooper-Knock J, Kirby J, Ferraiuolo L, Heath PR, Rattray M, Shaw PJ. Gene expression profiling in human neurodegenerative disease. Nat Rev Neurol. 2012;8:51830. [DOI] [PubMed]
    Miller JA, Woltjer RL, Goodenbour JM, Horvath S, Geschwind DH. Genes and pathways underlying regional and cell type changes in Alzheimer’s disease. Genome Med. 2013;5:48. [DOI] [PubMed] [PMC]
    Orre M, Kamphuis W, Osborn LM, Melief J, Kooijman L, Huitinga I, et al. Acute isolation and transcriptome characterization of cortical astrocytes and microglia from young and aged mice. Neurobiol Aging. 2014;35:114. [DOI] [PubMed]
    Vincenti JE, Murphy L, Grabert K, McColl BW, Cancellotti E, Freeman TC, et al. Defining the microglia response during the time course of chronic neurodegeneration. J Virol. 2016;90:300317. [DOI] [PubMed] [PMC]
    Srinivasan K, Friedman BA, Larson JL, Lauffer BE, Goldstein LD, Appling LL, et al. Untangling the brain’s neuroinflammatory and neurodegenerative transcriptional responses. Nat Commun. 2016;7:11295. [DOI] [PubMed] [PMC]
    Friedman BA, Srinivasan K, Ayalon G, Meilandt WJ, Lin H, Huntley MA, et al. Diverse brain myeloid expression profiles reveal distinct microglial activation states and aspects of Alzheimer’s disease not evident in mouse models. Cell Rep. 2018;22:83247. [DOI] [PubMed]
    Pennisi M, Crupi R, Di Paola R, Ontario ML, Bella R, Calabrese EJ, et al. Inflammasomes, hormesis, and antioxidants in neuroinflammation: role of NRLP3 in Alzheimer disease. J Neurosci Res. 2017;95:136072. [DOI] [PubMed]
    Schultz J, Schwarz A, Neidhold S, Burwinkel M, Riemer C, Simon D, et al. Role of interleukin-1 in prion disease-associated astrocyte activation. Am J Pathol. 2004;165:6718. [DOI] [PubMed] [PMC]
    Hennessy E, Griffin ÉW, Cunningham C. Astrocytes are primed by chronic neurodegeneration to produce exaggerated chemokine and cell infiltration responses to acute stimulation with the cytokines IL-1β and TNF-α. J Neurosci. 2015;35:841122. [DOI] [PubMed] [PMC]
    Shukla R, Singh TR. Identification of small molecules against cyclin dependent kinase-5 using chemoinformatics approach for Alzheimer’s disease and other tauopathies. J Biomol Struct Dyn. 2022;40:281527. [DOI] [PubMed]
    Liddelow SA, Guttenplan KA, Clarke LE, Bennett FC, Bohlen CJ, Schirmer L, et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature. 2017;541:4817. [DOI] [PubMed] [PMC]
    Shinozaki Y, Shibata K, Yoshida K, Shigetomi E, Gachet C, Ikenaka K, et al. Transformation of astrocytes to a neuroprotective phenotype by microglia via P2Y1 receptor downregulation. Cell Rep. 2017;19:115164. [DOI] [PubMed]
    Li B, Xia M, Zorec R, Parpura V, Verkhratsky A. Astrocytes in heavy metal neurotoxicity and neurodegeneration. Brain Res. 2021;1752:147234. [DOI] [PubMed] [PMC]
    Sullivan B, Robison G, Osborn J, Kay M, Thompson P, Davis K, et al. On the nature of the Cu-rich aggregates in brain astrocytes. Redox Biol. 2017;11:2319. [DOI] [PubMed] [PMC]
    Sofroniew MV. Astrocyte barriers to neurotoxic inflammation. Nat Rev Neurosci. 2015;16:24963. [DOI] [PubMed] [PMC]
    Gee JR, Keller JN. Astrocytes: regulation of brain homeostasis via apolipoprotein E. Int J Biochem Cell Biol. 2005;37:114550. [DOI] [PubMed]
    Rajasekhar K, Govindaraju T. Current progress, challenges and future prospects of diagnostic and therapeutic interventions in Alzheimer’s disease. RSC Adv. 2018;8:23780804. [DOI] [PubMed] [PMC]
    Fettelschoss A, Zabel F, Bachmann MF. Vaccination against Alzheimer disease: an update on future strategies. Hum Vaccin Immunother. 2014;10:84751. [DOI] [PubMed] [PMC]
    Ott PA. Immune-modified response criteria—an iterative learning process? Nat Rev Clinl Oncol. 2018;15:2678. [DOI] [PubMed]
    Pimenova AA, Raj T, Goate AM. Untangling genetic risk for Alzheimer’s disease. Biol Psychiatry. 2018;83:30010. [DOI] [PubMed] [PMC]
    Gjoneska E, Pfenning AR, Mathys H, Quon G, Kundaje A, Tsai LH, et al. Conserved epigenomic signals in mice and humans reveal immune basis of Alzheimer’s disease. Nature. 2015;518:3659. [DOI] [PubMed] [PMC]
    Rather MA, Khan A, Alshahrani S, Rashid H, Qadri M, Rashid S, et al. Inflammation and Alzheimer’s disease: mechanisms and therapeutic implications by natural products. Mediators Inflamm. 2021;2021:9982954. [DOI] [PubMed] [PMC]