Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Mar 20.
Published in final edited form as: Neuron. 2019 Mar 20;101(6):1003–1015. doi: 10.1016/j.neuron.2019.02.027

Harnessing Immunoproteostasis to Treat Neurodegenerative Disorders

Todd E Golde 1
PMCID: PMC6594693  NIHMSID: NIHMS1523577  PMID: 30897353

Abstract

Immunoproteostasis is a term used to reflect interactions between the immune system and the proteinopathies that are presumptive “triggers” of many neurodegenerative disorders. The study of immunoproteostasis is bolstered by several observations. Mutations or rare variants in genes expressed in microglial cells, known to regulate immune functions, or both, can cause, or alter risk for, various neurodegenerative disorders. Additionally, genetic association studies identify numerous loci harboring genes which encode proteins of known immune function that alter risk of developing Alzheimer’s disease (AD) and other neurodegenerative proteinopathies. Further, preclinical studies reveal beneficial effects and liabilities of manipulating immune pathways in various neurodegenerative disease models. Although there are concerns that manipulation of the immune system may cause more harm than good, there is considerable interest in developing immune modulatory therapies for neurodegenerative disorders. Herein, I highlight the promise and challenges of harnessing immunoproteostasis to treat neurodegenerative proteinopathies.

Keywords: Neurodegenertive Disease, Immune System, Therapeutics, Immunoproteostasis, Genetic Risk


Despite major advances in understanding key initiating events underlying various neurodegenerative disorders, there are limited efficacious treatment options for patients (Cummings et al., 2014; Golde, 2016). Approved therapies are primarily symptomatic in nature and do not dramatically modify disease course. With rare exceptions such as deep brain stimulation or dopamine replacement strategies for Parkinson’s disease (PD), the clinical impact of such therapies is underwhelming.

Over the last two decades, there have been huge investments towards the development of therapies for neurodegenerative diseases that target the proteins which aggregate and accumulate (i.e., the proteinopathy). Clinical data generated to date, primarily with respect to altering amyloid β protein (Aβ) deposition in Alzheimer’s disease (AD), indicates such therapies that are intended to target the triggering proteinopathy may have limited efficacy in symptomatic patients (Doody et al., 2014; Egan et al., 2018; Sevigny et al., 2016). Indeed, there is growing recognition and evidence from clinical trials that targeting the proteinopathies will have the best chance for showing efficacy if treatment is initiated well before disease-defining symptoms appear (Bateman et al., 2017; Golde et al., 2013; Reiman et al., 2011; Sperling et al., 2014). There is cautious optimism that such therapies intended to target early steps in the disease process, when tested in primary or secondary prevention trials, will have major impact on disease course; however, the timelines for possible regulatory approval and widespread deployment of effective prophylactic therapies remain uncertain (Golde, 2016).

Indeed, there is a bleak outlook for individuals who are currently in the symptomatic phase of a neurodegenerative disorder, as well as those who might develop these diseases in the near future because of advanced age, genetic risk or environmental influences. Given this unmet medical need, it is imperative to identify novel strategies to treat neurodegenerative disorders that can significantly alter the disease course, even if initiated in symptomatic patients. Development of such therapies is almost certain to be challenging given our imperfect understanding of the pathological events downstream of the triggering proteinopathy. Herein, I examine the rationale for one of these potential strategies, namely, targeting immunoproteostasis (Chakrabarty et al., 2015). My colleagues and I have used this term previously to capture the complex, multidirectional, interplay between the immune system and the underlying triggering neurodegenerative proteinopathy. As much of the attention in the field is centered on immunoproteostasis in AD, this Perspective will focus on the efforts to understand and harness the immune system for therapeutic benefit in AD, although illustrative examples from other neurodegenerative diseases are discussed. The Perspective is also primarily focused on the role of innate immunity; nevertheless, the more limited evidence regarding adaptive immune responses in neurodegenerative disorders will be briefly discussed.

There has been long-standing interest in determining the role of the immune system in neurodegenerative proteinopathies. Initial studies in this area largely focused on a descriptive association of the reactive gliosis and a cataloguing of alterations in expression or accumulation of innate immune signaling molecules in the brains of individuals who died from neurodegenerative disease (Akiyama et al., 2000; Eikelenboom et al., 1998; Heneka et al., 2015; Rogers et al., 1996). Although the general gestalt has been that pro-inflammatory activation of the innate immune system within the brain is harmful, a much more nuanced role of the immune system in neurodegenerative disease is warranted. Perhaps the most cautionary note in this vein, is that clinical trials of anti-inflammatory agents in AD have either shown no benefit or, in some cases, actually showed anti-inflammatory agents may have worsened decline (Aisen et al., 2003; Breitner et al., 2011; de Jong et al., 2008; Group, 2015; Group et al., 2007; Reines et al., 2004; Soininen et al., 2007). The side effects of anti-inflammatory therapy in these studies were also quite marked in these elderly trial participants. Thus, these trials may actually have done more harm than good.

The origins of the hypothesis that pro-inflammatory immune stimuli are harmful in AD.

The hypothesis that proinflammatory drivers played a harmful role in AD led to clinical trials of anti-inflammatory agents. Thus, it is useful to reevaluate the data and interpretation of the data that led to development of that hypothesis. The presence of gliosis (“activated” microglial cells and a robust astrocytosis) in the AD brain was a foundational observation on which the “proinflammatory stimuli are bad in AD” hypothesis was built (Akiyama et al., 2000). As a reactive gliosis is present in most, if not all, neurodegenerative disorders, these observations still contribute to the concept that proinflammatory factors are harmful in other neurodegenerative disorders.

For AD, another factor was the comparison of AD to Serum Amyloid A (SAA) amyloidosis. In SAA amyloidoisis, inflammation is essential for SAA amyloid deposition in the periphery, as pro-inflammatory stimuli rapidly and massively upregulate SAA mRNA and protein expression in the liver (Malle and De Beer, 1996; Sack, 2018). Following the cloning of the amyloid protein precursor (APP) gene that encodes Aβ and resides on human chromosome 21, a series of genetic and modeling studies emerged indicating that simply increased levels of APP or Aβ could lead to Aβ deposition (reviewed in (Hardy and Selkoe, 2002)). The most straightforward example of this is in human trisomy 21 (Downs’s syndrome). Individuals with trisomy 21 develop Aβ amyloid deposition relatively early in life and other features of AD pathology later (Delabar et al., 1987; Mann, 1988; Masters et al., 1985). This accelerated Aβ deposition is linked directly to increased expression of APP and increased Aβ levels. Thus, when subsequent data emerged showing that various proinflammatory stimuli could increase APP and Aβ levels, at least in cell culture systems (Griffin et al., 1998), the proinflammatory stimuli drives AD pathology by increasing APP and Aβ hypothesis emerged.

In parallel studies, several groups were beginning to evaluate immune molecules present within the brain (Akiyama et al., 2000; Eikelenboom et al., 1998; McGeer et al., 1994). These studies, likely biased towards evaluating immune factors normally associated with activating the immune response (e.g., complement, inflammatory cytokines), revealed that numerous acute phase reactants and other immune molecules accumulate within the AD brain. Further, these factors were either associated directly with amyloid plaques, present in the reactive microglial surrounding these plaques, or in reactive astrocytes. Additional studies demonstrated higher levels of peripheral proinflammatory cytokines in AD, or at least all-cause dementia, results that are now further supported by several meta-analyses (Koyama et al., 2013; Swardfager et al., 2010) . Perhaps the final and pivotal pieces of support for the proinflammatory hypothesis emerged when i) non-steroidal anti-inflammatory drugs (NSAIDs) use was reproducibly associated with decreased risk for AD (Breitner et al., 1994; Breitner et al., 1995; in ‘t Veld et al., 1998; McGeer et al., 1996; Stewart et al., 1997; Zandi and Breitner, 2001) and II) preclinical studies in mouse models suggested that select NSAIDs might lower Aβ amyloid pathology in mice (Jantzen et al., 2002; Lim et al., 2000). Despite the fact that NSAIDs use contains some risk, especially for elderly individuals, the aforementioned clinical trials were initiated in symptomatic AD and, subsequently, a prevention study.

Therapeutic clinical trials of NSAIDs in individuals with symptomatic AD did not mimic the setting that was likely to account for the possible epidemiologic association of decreased risk for development of AD (>2 year history of NSAID use at an indeterminate time prior to diagnosis of dementia) (in ‘t Veld et al., 1998; Stewart et al., 1997). Further, the specific NSAIDs used in those trials were not necessarily the drugs most likely to account for the epidemiologic association. Thus, many questions, but few firm conclusions, emerge from a retrospective examination of these studies. Notably, the initial epidemiologic association that drove the studies on NSAID use and AD risk was preceded by studies showing reduced risk of individuals with rheumatoid arthritis (RA) and AD (McGeer et al., 1996). This raises the issue of reverse causation. Did the well-catalogued proinflammatory status of individuals with RA, or other peripheral conditions associated with chronic long-term activation of the immune system, and not the NSAID per se, drive the association with reduced risk for AD? Indeed, individuals with a chronic inflammatory condition likely represent a large portion of those with the >2 year history of NSAID use most reproducibly associated with reduced risk AD. Further, NSAIDs typically only minimally alter the immune phenotype and those NSAIDs linked to the epidemiologic data are poorly brain penetrant (Eriksen et al., 2003; Kukar et al., 2005). Finally, additional data emerged that at least some NSAIDs could have other effects (e.g., selectively lowering production of the longer, more pathogenic forms of Aβ) that might account for their apparent benefit in preclinical models (Kukar et al., 2005; Weggen et al., 2001).

Whether the epidemiologic data indicates a possible protective role of NSAID use or the underlying conditions such as RA that led to long-term NSAID use, represents an unsolved dilemma for the field. A protective role for NSAIDs in AD, possibly through an anti-inflammatory mechanism of action, remains a marginally plausible hypothesis, but only in the setting of primary prevention. A protective role of the underlying condition that necessitate long-term NSAID use would conversely suggest that the proinflammatory state is protective. In other words, depending on how one views the epidemiologic data, one ends up with the exact opposite conclusion for how to move forward with a therapy modulating the immune system in AD.

Can we resolve this dilemma?

This retrospective analysis serves as an exemplar of the challenges faced with respect to developing and testing immune modulatory therapies for AD and other neurodegenerative disease based on our current knowledge. Indeed, long-term therapeutic modulation of the immune system in the elderly will almost invariably lead to a higher incidence of side effects than what might be observed with that same therapy in younger individuals, no matter whether the therapy is intended to be immune activating or suppressing. Many of these potential side effects will be due to effects on the peripheral immune status. Potential for untoward side effects might be reduced by therapeutics with a selective effect on immune activation states in the central nervous system (CNS), but such targeted approaches are largely conceptual at this time. Perhaps, anti-Aβ antibodies with activating Fc domains that preferentially bind Aβ aggregates in the brain, focally activate microglial cells, and reduce Aβ loads, serve as examples of such therapies (Ostrowitzki et al., 2017; Sevigny et al., 2016).

The current ambiguity with respect to directionality is highly problematic. If a therapy moves the activation state of the immune system in the wrong direction, then the therapy may actually accelerate the underlying neurodegenerative disease process. An emerging concept in the field is that a given immune activation state or immune manipulation may have different consequences on brain function, positive or negative dependent on the state of disease. This postulate is supported by limited, but nonetheless provocative, preclinical model data. For example, CX3CR1 or TREM2 knockout has been shown to have differential effects on amyloid and tau pathologies (Bhaskar et al., 2010; Lee et al., 2010; Leyns et al., 2017; Sayed et al., 2018). In another case, IFNγ overexpression has been shown to have a positive effect on amyloid pathology, but a negative impact on neuronal health (Chakrabarty et al., 2010; Chakrabarty et al., 2011). If bi-directional effects of an immune manipulation are conserved in humans, such data would suggest that it might be possible to intervene with an immune modulation at a defined disease stage (e.g., amyloid positive, tau negative) (Jack et al., 2016). Such stage-dependent treatment theoretically makes some sense, but is complicated by the fact that disease-state is heterogeneous in almost all neurodegenerative disorders across various CNS regions. Further, the knockout studies in mice that support this concept of “stage-specific” targeting of the immune system do not reflect a real-world intervention that would likely begin long after the pathology has emerged. Additionally, a thorough mechanistic understanding of the biology underlying these differential effects is lacking. For all of these reasons, clinical development of immune interventions that have beneficial effects on one pathology, but harmful effects on another in preclinical models should proceed very cautiously. It will be challenging to predict the likelihood that beneficial effects would outweigh negative effects.

With these caveats and concerns in mind, there are a number of paths forward that can help inform whether i) there are truly targets in the immune system that would provide disease modification in neurodegenerative proteinopathies and ii) determine directionality with respect to altering these targets. In the following sections. I will discuss these complimentary paths and review the progress that has been made in these areas.

Functional genetics studies can inform the directionality of immune therapies.

A key step in both identifying potential targets for immune therapies and whether agonism or antagonism of the target is desirable is to better understand how variants and mutations in immune genes implicated in altering risk for neurodegenerative disease alter immune function (see Table 1 and 2). AD and PD genome wide association studies (GWAS) and more recent meta-analyses of these GWAS implicate a number of loci that contain immune genes in mediating risk (Chang et al., 2017; Hamza et al., 2010; Hill-Burns et al., 2011; International Genomics of Alzheimer’s Disease, 2015; Lambert et al., 2010; Lambert et al., 2009; Nalls et al., 2014; Reitz et al., 2013; Wissemann et al., 2013; Witoelar et al., 2017). Unfortunately, there remains limited biological insight into how variants at these loci alter immune function, though some clues are emerging (Bradshaw et al., 2013; Brouwers et al., 2012; Carrasquillo et al., 2017; Huang et al., 2017; Katsumata et al., 2018; Siddiqui et al., 2017). Indeed, in many cases, though an immune gene is embedded within the region implicated by the GWAS association, the locus implicated is typically larger than a single gene and may contain both genes with known or purported immune function and genes with other functions. Of course, there are also examples outside of neurodegenerative disease where a disease-associated polymorphism, can have long-range effects outside of the region of association (Smemo et al., 2014; Tung et al., 2014). Further, even though evidence of association with the HLA locus is cited as genetic support for immune link to AD and PD, numerous non-immune genes are imbedded within the human HLA locus – potentially confounding this assertion. In addition, though the HLA associations are used as supporting rationale for studying adaptive immunity in these diseases, both MHC class I and class II are constitutively expressed on cells in the brain and these molecules can play roles in innate immune signaling (Frei et al., 2010; Shatz, 2009). Despite these caveats, given the large number of loci in both AD and PD, which contain genes that regulate immune function, it is reasonable to pursue studies designed to link genetic variation with functional variation in immune function.

Table 1. Candidate genes with known or possible immune function in loci implicated by GWAS in AD and PD.A.

GeneB Disease Evidence and Strength of Evidence for immune function in the brainC
CR1 AD Strong, complement receptor, minimal expression in CNS, soluble form present in plasma and CSF, association region includes the CR1L gene, a homologue of CR1
INPP5D AD Strong, highly expressed in microglial cells, Is activated by immunoreceptor tyrosine-based inhibition motif receptors, acts as an intrinsic brake on activation signaling
HLA-DRB1 AD Strong, one chain of the heterodimeric MHC class II molecule, highly expressed in microglial cells, region of association includes other MHC genes and non MHC genes, involved in antigen presentation, but may play a role in innate immune responses
TREM2 AD Strong, an immunoreceptor tyrosine-based activation motif receptor, highly expressed on microglial cells (see main text for more details)
MS4A6A AD Strong, strongly expressed in microglial in the brain and in peripheral immune cells, but sparse functional data, other MS4A family members are within the region of association
SP1 AD Strong, a transcription factor highly expressed in myeloid cells and microglial cells in the brain. C1qTNF4 a molecule with cytokine like functions is also within the region ofassociation.
BIN1 AD Strong (peripherally) Weak (CNS), evidence for relatively high levels of RNA expression in microglial cells, indirectly influences function of immune cells in the periphery by regulating of expression of the immunomodulatory enzyme indoleamine 2,3-dioxygenase (IDO). IDO mRNA is barely detectable in the brain.
CLU AD Strong, a secreted chaperone, regulates Aβ deposition, regulates complement activity among many other activities, intracellular form can regulate NFKB
CD33 AD Strong, an immunoreceptor tyrosine-based inhibition motif receptor that binds to sialic acid residues, highly expressed on microglial cells, activation of CD33 opposes activation of ITAM receptors such as TREM2.
ABI3 AD Moderate, expressed selectively on microglial cells, may function as part of the WAVE complex, implicated in regulation of cell migration, rare variants associated with increased risk for AD (see Table 2)
ADAM10 AD Moderate, cell surface proteins with a unique structure possessing both potential adhesion and protease domains, functionally studied for its effects as an α-secretase that cleaves APP and TNF-α, appears to be expressed highly in human microglial/macrophages, this and other α-secretases are known to regulate immune responses through their sheddase activity
APOE AD Moderate, highly expressed in microglial, APOE genotype, clear role in regulation of peripheral immune responses, interaction with Aβ aggregates can alter clearance of Aβ by microglial cells
CASS4 AD Moderate, Docking protein that plays a role in tyrosine kinase-based signaling related to cell adhesion and cell spreading, selectively expressed in the brain in microglial cells.
ABCA7 AD Moderate, widely expressed on many CNS cell types, can regulate microglial phagocytosis, deletions in ABCA7 have been associated with AD risk in African Americans
SORL1 AD Weak, a multifunctional endocytic receptor, functionally studied in neurons for its effects on Aβ production, but appears to be expressed highly in human microglial/macrophages.
ALPK2 AD Weak, a kinase, low expression in CNS cells but expressed in human microglial/macrophages
PICALM AD Weak, a clathrin assembly protein expressed in many CNS cell-types but expressed strongly in microglial cells.
CD2AP AD Weak, widely expressed in CNS cell types, has been shown to play a role in T-cell function
Il1R2 PD Strong, binds interleukin alpha (IL1A), interleukin beta (IL1B), and interleukin 1 receptor, type I(IL1R1/IL1RA), and acts as a decoy receptor that inhibits the activity of its ligands,
highly expressed on microglial cells
TLR9 PD Strong, a member of the Toll-like receptor (TLR) family, which plays a fundamental role in pathogen recognition and activation of innate immunity, highly expressed on mouse microglial cells
STAB1 PD Strong, a large, transmembrane receptor protein which may function in angiogenesis, lymphocyte homing, cell adhesion, or receptor scavenging, highly expressed on microglial cells
HLA-DRB6 PD Strong, one chain of the heterodimeric MHC class II molecule, highly expressed in microglial cells, region of association includes other MHC genes and non-MHC genes, involved in antigen presentation, but may play a role in innate immune responses. This gene has been called a pseudogene, though there is evidence that it can generate a protein.
HLA-DQA1 PD Strong, one chain of the heterodimeric MHC class II molecule, highly expressed in microglial cells, region of association includes other MHC genes and non MHC genes, involved in antigen presentation, but may play a role in innate immune responses
CTSB PD Strong, a lysosomal protease, highly expressed in microglial cells, plays a role in antigen processing and protein turnover.
GCH1 PD Moderate, a member of the GTP cyclohydrolase family, highly expressed on microglial cells,
CD38 PD Moderate, act as a receptor for cells in the immune system, primarily expressed by astrocyte in the brain.
CRHR1 PD Moderate, Corticotropin Releasing Hormone Receptor 1 G-protein coupled receptor that binds neuropeptides of the corticotropin releasing hormone family that are major regulators of the hypothalamic-pituitary-adrenal pathway. This receptor regulates corticosteroid levels via the HPA axis, but is also present on immune cells and CRH has been shown to directly modulate immune cell function.
GBA PD Weak, a lysosomal membrane protein that cleaves the beta-glucosidic linkage of glycosylceramide expressed on many CNS cell types but highly expressed in mouse microglial cells
SIPA1L2 PD Weak, a member of the signal-induced proliferation-associated 1 like family, highly expressed on microglial cells.
ARHGAP27 PD Weak, member of a large family of proteins that activate Rho-type guanosine triphosphate (GTP) metabolizing enzymes, highly expressed on mouse microglia cells.
B

These refer to the candidate immune genes within the region of association, in many cases there are other non-immune genes within the region of association.

C

Evidence for immune function based on data in genecards (https://www.genecards.org/) and pubmed https://www.ncbi.nlm.nih.gov/pubmed/ is highlighted, expression data is based on (Zhang et al., 2014; Zhang et al., 2016). The classification of evidence for function in the immune system in these disease (Strong, Moderate, Weak), is somewhat subjective but based on current evidence for functional role in the immune system and RNA expression data.

Table 2. Mutations and variants in immune genes that cause or alter riskA for neurodegenertive disease.

Gene Disease Mutation/Variant
CSF1R hereditary diffuse leukoencephalopathy with neuroaxonal spheroids (HDLS) Numerous, more than 50 pathogenic variants, including missense, frameshift and non-sense mutations, but also deletions and splice-site mutations, all located in the intracellular tyrosine kinase domain, strongly support haploinsufficiency and partial loss of function in disease mechanism. Highly expressed on microglial cells and myeloid cells.
GRN frontotemporal dementia (FTD) Numerous, heterozygous result in haploinsufficiency and thus partial loss of function through nonsense mediated RNA decay. Highly expressed by microglial cells.
GRN neuronal ceroid lipofuscinoses (NCL) Numerous, homozygous or compound heterozygous mutations cause near complete loss of function
TREM2 Nasu-Hakola disease (NHD), also referred to as polycystic lipomembranous osteodysplasia with sclerosing leukoencephalopathy (PLOSL), also FTD Numerous, homozygous or compound heterozygous mutations cause near complete loss of function. At least 11 mutations cause PLOSL/FTD. Not all mutation have been shown to have the osteodysplasia with the phenotype in those individuals largely FTD. Functionally mutations that cause PLOSL/FTD appear to be loss of function. Highly expressed on microglial cells.
TYROBP NHD/PLOSL Multiple, homozygous or compound heterozygous mutations cause near complete loss of function. Mutations include deletions, insertions and frameshift, resulting in either no or truncated non-functional TYROB. Strongly expressed on Microglial Cells
TREM2 Increased Risk for AD R47H and R62H TREM2 variants are associated with partial loss of function. Other variants may also be associated with altering AD risk, but studies showing these associations have not been as widely replicated
ABI3 Increased Risk for AD S209F variants is associted with increased risk for AD. Expressed selectively on microglial cells, may function as part of the WAVE complex, implicated in regulation of cell migration. Little data on functional effects of variant.
PLCG2 Decreased risk for AD P522R variant is associted with increased risk for AD. Initial data suggest that this variant slightly increases PLCG2 activity. Selectively expressed ion microglial cells in the brain and in many cells in the periphery.
A

Numerous other variants in immune genes have been associated with AD, this table focuses only on the ones for which there is wide consensus in the field that these alter disease risk.

Potentially more immediately informative with respect to causality and directionality are studies that i) link genes with function in the immune system to rare familial forms of neurodegenerative disease and ii) rare functional variants in genes whose mRNAs are selectively expressed in microglia or other immune cells that are associated with risk for AD. Examples of mutations in immune genes that cause neurodegeneration are i) colony stimulating factor-1 receptor (CSF1R) mutations that cause dominantly inherited hereditary diffuse leukoencephalopathy with neuroaxonal spheroids (HDLS) (Mitsui et al., 2012; Rademakers et al., 2011; Stabile et al., 2016) ii) progranulin (GRN) mutations that cause of frontotemporal dementia (FTD) (Baker et al., 2006; Cruts et al., 2006) or neuronal ceroid lipofuscinoses (NCL) (Smith et al., 2012) and iii) triggering receptor expressed on myeloid cells 2 (TREM2) or TYRO Protein Tyrosine Kinase Binding Protein (TYROBP) mutations that cause Nasu-Hakola disease (NHD), also referred to as polycystic lipomembranous osteodysplasia with sclerosing leukoencephalopathy (PLOSL) (Paloneva et al., 1993; Paloneva et al., 2002; Soragna et al., 2003). Examples of coding variants in immune genes that alter risk for a neurodegenerative disease are the AD-associated variants in TREM2, phospholipase C gamma 2 (PLCG2), and ABI family member 3 (ABI3) genes (Guerreiro et al., 2013; Jonsson et al., 2013; Sims et al., 2017). Like TREM2, PLCG2 and ABI3 are thought to be selectively expressed on microglia in the CNS and monocytes/macrophages and other immune cells in the periphery. Notably, whereas TREM2 and variants are associated with increased AD-risk, the PLCG2 variant is protective (Sims et al., 2017).

Though our understanding of how these various mutations alter protein function is evolving, the weight of evidence suggests that those associated with risk for neurodegeneration result in loss of, or reduced protein function or signaling and, consequently, promote reduced microglial activity, dysfunction, and neurodegeneration. Functional effects of mutations in GRN and TREM2 have been most extensively studied. Heterozygous, mutations in GRN which encodes the secreted protein progranulin (PRGN), clearly result in haploinsufficiency, due to non-sense mediated RNA decay and this leads to FTD (Baker et al., 2006; Cruts et al., 2006). In contrast, homozygous and compound heterozygous mutations that result in even further reductions in PRGN levels, cause NCL (Smith et al., 2012). Thus, it is clear that GRN mutations are loss-of-function and that the relative loss of function, from partial to near complete, results in distinct forms of neurodegeneration. Unfortunately, it remains unclear whether loss of PRGN leads to degeneration due to effects on microglial function, paracrine effect on neurons or other CNS cells, or some combination of the two (Arrant et al., 2018; Filiano et al., 2013; Kao et al., 2017; Lui et al., 2016; Martens et al., 2012).

Because i) it is associated with a rare form of neurodegeneration (PLOSL) and a more common form (AD) and ii) the AD risk variants have a large impact on disease risk, functional impacts of the disease-associated variants in TREM2 have been intensively studied (Ulrich et al., 2017). TREM2 is member of a family of type 1 membrane protein, triggering receptors expressed on myeloid cells (TREMs). TREM2 is distinct in that it is highly expressed in brain microglia. It contains an intracellular immunoreceptor tyrosine-based activation motif that conducts signals following binding to a variety of ligands such as various lipids and lipoproteins through interaction with the intracellular adaptor TYROBP (Ulrich et al., 2017). These studies show that the mutations that cause PLOSL have more distinct effects on protein function than do the variants that cause AD (Filipello et al., 2018; Kleinberger et al., 2014; Korvatska et al., 2015; Lee et al., 2018; Lessard et al., 2018; Neumann and Takahashi, 2007; Schlepckow et al., 2017; Song et al., 2018; Xiang et al., 2018; Zhao et al., 2018). Indeed, there is emerging consensus that PLOSL-associated TREM2 mutations result in defective protein maturation and in almost complete loss of function, whereas distinct AD-associated TREM2 variants result in subtle, partial loss-of-function. Consistent with these findings are data showing that a variant in the TREM locus is associated with decreased risk for AD and increased expression of TREM2. Additionally, CSF1R mutations, that cause disease in a heterozygous state, appear to be loss-of function, though the mechanisms that result in decreased function are still debated (Mitsui et al., 2012; Rademakers et al., 2011; Stabile et al., 2016). The AD associated PLCG2 variant is somewhat unique as it is associated with protection from disease. Further, it is one of the few classically druggable proteins identified in AD to date. Emerging data suggests that the variant results in slightly increased phospholipase activity compared to the wild-type enzyme that would be predicted to enhance immune signaling, though this data will need to be confirmed and extended (Magno et al., 2018).

Collectively, these genetic studies begin to paint a picture whereby decreased function of activating immune receptors or signaling molecules results in disease. In contrast, and with far fewer examples, promoting immune activation appears to be protective. Whether these functional associations will hold up as more data emerges is uncertain; however, there clearly is enough data to suggest that the long-standing association of neuroinflammation and immune activation as a driving, solely pathogenic force in neurodegeneration is misplaced. There are simply too many examples where loss of immune protein function and signaling that is functionally linked to less immune mobilization appears to trigger neurodegeneration.

Account for possible differences in immune function between human and mouse models.

Mouse models of neurodegenerative proteinopathies have limitations, but they have provided pivotal support for the hypothesis that many neurodegenerative disorders are fundamentally proteinopathies (Dawson et al., 2018). Although many of the alterations in immune cells and proteins documented in human neurodegenerative disease are noted in the counterpart rodent models, there are important differences in immune repertoire, responses and priming between rodent and human (Heneka et al., 2015; Labzin et al., 2018). Such differences need to be accounted for in order for observations made in the rodent model to be used to inform how a given manipulation of the immune system might alter the human disease or even how a given genetic alteration causes disease.

There are several examples of neurodegenerative diseases where genetic knockout of the mouse counterpart of the human gene does not produce a close phenocopy of the human disease. One example is the heterozygous Grn (Grn+/−) mice, which develop very mild phenotypes that do not mimic human FTD (Lui et al., 2016; Minami et al., 2014). In contrast, the full knockout (Grn−/−), produces a model of NCL that closely phenocopies the human disease (Martens et al., 2012). Another example is mice lacking Trem2, which exhibit select immune phenotypes and osteoclastogenesis, but do not closely phenocopy the neurodegenerative aspects of human PLOSL (Otero et al., 2012). Similarly, mice lacking Tyrobp that do show brain and bone abnormalities do not model the overt phenotype of human PLOSL (Haure-Mirande et al., 2017; Nataf et al., 2005). Differences in immune repertoire between mice and humans can impact the understanding how genetic variants alter risk for select human neurodegenerative diseases. For example, there are considerable differences between humans and mice in the major histocompatibility (MHC/HLA) locus, which has been genetically associated with AD and PD. Further, both the TREM and the MS4A locus, which are both implicated by GWAS in AD, are quite divergent between mice and humans (Colonna, 2003; Liang et al., 2001).

Efforts to standardize housing conditions and limit pathogen exposure means that our mouse models are typically reared for generations in relatively sterile environments that restrict immunologic priming, including the priming of microglial cells in the brain (Erny et al., 2015; Perry and Holmes, 2014). Variation in normal microbiome and pathogen exposure could, in theory, alter immunoproteostasis in unpredictable ways, not only through direct effects on the brain, but also through effects on peripheral immune cells (Prinz and Priller, 2017). Though recent studies do show that altering the microbiome can influence immunoproteostasis in model systems, for the most part, the effects observed to date are rather modest (Harach et al., 2017; Minter et al., 2017; Sampson et al., 2016).

Concerted efforts and careful study, including development of mice with humanized immune genes or gene loci and exposure of mouse models to more diverse microbiomes, will likely provide much needed insight into how some immune genes may modulate risk for neurodegenerative disease and how environmental factors may contribute to the neurodegenerative phenotype; however, these types of studies are challenging and may still not overcome all the inherent limitations of modeling complex human neurodegenerative disorders that typically occur in late life in mouse models with much shorter lifespans and only partially conserved physiology.

A need for more human data.

Given the aforementioned limitations of mouse model studies, when possible, we should try to study how variants in immune genes alter the immune system in living humans. Most of the immune genes implicated in neurodegenerative disorders are expressed in peripheral immune cells. Further, in some instances, there are peripheral manifestations associated with the neurodegenerative disease, for example, bone cysts in PLOSL (Paloneva et al., 1993). In parallel to modeling studies, there should be a concerted effort to catalogue peripheral immune function in humans that carry these variants and also to look for other signs of altered immune function (e.g, look for bone cysts in those who carry TREM2 AD risk variants). This phenotyping might rapidly provide data on how variants in immune genes implicated in neurodegeneration alter immune function in living humans and serve as translational biomarkers. There is, of course, continued value in looking at human postmortem brain using multi-omic approaches to provide a systems level “compendium” of changes in immune gene expression and protein levels; however, such studies have limited ability to stage changes in immune system within the disease continuum and are confounded by many factors including the inability to distinguish effects that may be the result of the agonal state or long-standing disease state.

One example of how data from such studies could be transformational would be study of how the AD-associated PLCG2 variants alter peripheral immune function (Sims et al., 2017). PLCG2 is a potentially tractable drug-target, but more information on how the AD-associated variant alters both enzymatic and immune function is needed. Notably, other coding region variants in PLCG2 have been linked to peripheral immune disorders. A PLCG2 mutation (p.Ser707Tyr) causes an autosomal dominant inflammatory disorder (Zhou et al., 2012). This mutation is hypermorphic with respect to enzyme activity, strongly increasing phospholipase activity relative to wild type enzyme. In contrast, exon skipping due to genetic deletions in PLCG2 result in a distinct peripheral immune disorder characterized by cold-induced urticaria, antibody deficiency, and susceptibility to infection and autoimmunity (Ombrello et al., 2012). These PLCG2 deletions, located within an autoinhibitory domain, result in constitutive phospholipase activity, but diminished cellular signaling at 37°C and enhanced signaling at lower temperatures. Given the initial data that the AD-associated variant is also a slightly hypermorphic with respect to enzyme activity (Magno et al., 2018), one might postulate that carriers of this variant also have a mild autoimmune disorder and altered responsiveness of peripheral immune cells.

There are challenges to such human studies. In the case of PLCG2, the AD-associated variant is rare (allele frequency <1%). To identify individuals with and without AD, who harbor this variant would require a significant investment. There would also need to be careful consideration for what assays, clinical assessments and other studies might be most informative. In contrast to rare variants, more common variants implicated by GWAS might be studied as well; however, as most of those variants have rather weak effects on disease risk, functional effects of such genetic variation may be muted and difficult to detect.

Much of our data on immune responses in human neurodegenerative disease comes from cross-sectional autopsy studies, which have inherent limitations. In contrast to mouse models, we have limited insight into the evolution of the immune response during disease progression. Studies using a positron emission tomography (PET) ligands that binds translocator protein 18 kDa (TSPO) which is strongly upregulated in “activated” microglial have some utility (Cerami et al., 2017; Edison et al., 2018); however, due to increases in TSPO binding as a simple consequence of age, ability to discriminate between diseased brain and normal brain in the aged is limited. Cerebrospinal fluid (CSF)-based biomarker studies have also suggested that there may be various distinct phases of the immune responses in AD, with different markers peaking at different stages of disease (Liu et al., 2018; Piccio et al., 2016; Rauchmann et al., 2018; Suarez-Calvet et al., 2016a; Suarez-Calvet et al., 2018; Suarez-Calvet et al., 2016b); however, there remains a critical need to develop more sensitive and specific PET- and CSF-based markers that can provide a more global assessment of immune activation state in the living brain and immune function as disease progresses.

Revisit epidemiological studies to look at how incidence of neurodegenerative disease is influenced by inflammatory conditions and immune modulatory medicines.

Given the rather uncertain impact and concerns over interpretation of epidemiologic studies relating to NSAID use and risk of AD, there may be some hesitancy to simply revisit this type of study; however, there are good reasons to reexamine a broader association between inflammatory disorders, immune modulatory therapy and neurodegenerative diseases. There are now many widely used immunomodulatory drugs that have much more profound functional effects on the immune system than NSAIDs. Immunosuppressive drugs, such as etaneracept, adilimumab, natalizumab and others, have potentially been used for long-enough periods that some insight into whether they influence risk for various neurodegenerative disorders may be gleaned from large-scale epidemiologic studies (Miller et al., 2003; Moreland et al., 1999; Weinblatt et al., 2003). As these are all immunosuppressive agents and are used to treat various pro-inflammatory immune disorders, one could argue that the same reverse causation confound regarding whether it is the disease itself or the treatment that might be associated with any alteration in risk; however, these biologic agents are not universally applied therapeutics. Thus, theoretically, any confound could be lessened by stratifying risk of neurodegeneration based on both underlying disease and type of medication used. A second reason is the emergence of many large databases, where millions or even tens of millions of individual’s medical data is housed and could be mined to evaluate this issue, potentially increasing statistical power to the point where there was much more confidence in the data that is generated. A final reason is simply that some of the therapies such as natalizumab (Miller et al., 2003), which binds α4 integrin and blocks T-cell infiltration from the periphery through the blood brain barrier, might have an unexpected impact, positive or negative, on neurodegenerative disease. Although there is some preclinical data implicating T-cells in modulation of some neurodegenerative disorders (Marsh et al., 2016), there is only sparse genetic and pathological data linking T-cells to most forms of neurodegeneration. Highly powered, appropriately controlled epidemiologic data could result in highly translatable insights that may never be resolved by studies in model systems.

Use genetics to broadly inform immune therapies.

There has been increasing emphasis in the pharmaceutical industry to only pursue genetically validated targets for disease modifying therapies. This is especially true for CNS disorders where therapeutic successes have been extremely limited. With respect to AD, the convergence of genetic data implicating the immune system has dramatically increased interest in the development of immune modulating therapies, specifically those that might target TREM2. Though TREM2 is a potentially tractable therapeutic target, development of therapeutics that increase CNS TREM2 function and signaling, which has emerged as the desired therapeutic mechanism of action, will not be trivial. Indeed, antagonism of a type 1 membrane receptor with a plethora of endogenous ligands that is also rapidly shed from the membrane poses both pharmacologic and pharmacokinetic challenges (Ulland and Colonna, 2018).

An alternative to a strict focus on proteins or other factors directly implicated by genetic studies is to broadly understand the signaling pathways implicated through genetics. Guided by this experimentally deduced functional understanding, it may be possible to identify more tractable targets that are likely to provide a similar therapeutic effect. A simplistic example relating to TREM2, is that TREM2 and other immunoreceptor tyrosine-based activation motif (ITAM) based signaling receptor function is often opposed by immunoreceptor tyrosine-based inhibition motif (ITIM) based signaling receptors. Inhibition of an ITIM based receptor, which opposes TREM2 signaling theoretically could result in a similar functional effect as activation of TREM2 (Barrow and Trowsdale, 2006; Linnartz and Neumann, 2013). As receptor antagonism or inhibition of downstream signaling is often easier to achieve pharmacologically than agonism; rigorous mechanistic studies of immune pathways in the brain that build off of genetic associations are almost certain to reveal additional, potentially tractable, therapeutic targets.

We also need to think about the therapeutic implications of preclinical studies in the context of human genetics. For example, as noted above, it appears that partial loss of function of CSF1R can cause a neurodegenerative disorder. In contrast, several preclinical studies indicate that small molecule CSF1R inhibition may have beneficial effects in APP and ALS mouse models (Elmore et al., 2018; Martinez-Muriana et al., 2016; Olmos-Alonso et al., 2016; Sosna et al., 2018; Spangenberg et al., 2016). The juxtaposing of these findings would certainly suggest that therapeutic studies of CSF1R inhibition in AD, or any neurodegenerative condition, should only proceed with appropriate caution and rigorous evaluation of potential liabilities, but also reinforce the need to truly understand how the variants within CSF1R cause disease.

Develop a more rigorous, translational preclinical road map for development of immune therapies targeting neurodegenerative disease.

Given the imperfect nature of many of our mouse models of neurodegenerative disease, their predictive value with respect to development of novel disease modifying therapies has been drawn into question. Despite these translational shortcomings, which in many cases are not necessarily an inherent defect with the model itself, these models have enabled us to understand critical aspects of disease pathogenesis and progression (Dawson et al., 2018). At least for the foreseeable future, these models are the best we can do with respect to providing a realistic system to explore immune modulating therapies designed to alter immunoproteostasis. In the future, it may be possible to develop ex vivo models, perhaps using differentiated human induced pluripotent stem cells (iPSCs) (Abud et al., 2017; Douvaras et al., 2017; Garcia-Reitboeck et al., 2018; Haenseler et al., 2017; Park et al., 2018; Takata et al., 2017). However, it is difficult to envision how one can recreate all of the complex interactions between various cells in the brain, the blood brain barrier, the circulatory system, the peripheral immune system, and environmental factors from iPSCs in a dish. Nevertheless, there is exciting work in this area and valuable insights may emerge from such studies in the future (Berry et al., 2018).

Because a given immune manipulation may have effects that are contextually dependent, the net effect may be dependent on both the timing with respect to disease stage and the intensity and length of the manipulation. This means that intensive preclinical studies are needed. Indeed, an interventional preclinical study with a single therapeutic dose that only looks at effects on a limited set of phenotypes and in one treatment paradigm is probably not sufficient to reliably predict efficacy in a different disease stage, or reveal possible side effects. Further, emerging data that an immune modulation may have different effects on different human neurodegenerative disease models indicates that preclinical studies of immune modulation should include multiple diseases. During these studies, there should be concerted efforts to develop translational theragnostic biomarkers that inform on both target engagement and potential efficacy. Some monitoring of the peripheral immune status is also warranted, as well as a survey of possible immune related effects. With increased rigor and more systematic preclinical studies, it may be possible to identify immune targeting therapies that can safely provide disease modification in select neurodegenerative disorders.

Notably, mouse-modeling studies conducted to date often reveal conflicting results of effects of modulating innate immune activation states in neurodegenerative disease models (reviewed in (Czirr and Wyss-Coray, 2012). Numerous studies show beneficial effects of an immune suppressing manipulation and harmful effects of an immune activating manipulation, but multiple studies show the opposite effects, or show that that the outcome is dependent on the disease model; for select recent examples of such conflicting data see (Ayers et al., 2014; Chakrabarty et al., 2015; Guillot-Sestier et al., 2015; Heneka et al., 2013; Venegas et al., 2017; Wendeln et al., 2018)). Future studies, which more systematically evaluate how a given immune manipulation alters the phenotype, as described above, will hopefully help build consensus regarding how these preclinical studies inform immune modulatory therapeutic approaches to neurodegenerative disease.

Change our lexicon regarding the role of the immune system in neurodegenerative disease.

Because of the i) potentially misplaced assertion that neuroinflammation is a pro-inflammatory state that is invariably harmful, and ii) the growing recognition that immune activation states are incredibly complex and only rarely, dichotomously, pro- or anti-inflammatory in nature, perhaps, in the future, there should be an effort to limit the use of term neuroinflammation. Similarly, as suggested by others previously, blanket descriptors of immune activation states in the brain as either pro- or anti-inflammatory, or M1 or M2, are probably not particularly useful (Graeber et al., 2011; Masgrau et al., 2017; Ransohoff, 2016). Perhaps as an alternative, we should simply be more precise in describing the immune response and resultant phenotype with respect to a particular disease stage or immune targeting intervention. With ever-increasing technological advances that make systems biology levels studies more feasible, this should become a best practice in the field.

Summary

In planetary science, the concept of a “Goldilocks zone” is used to describe planetary orbits that are potentially compatible with life - neither too close to the sun, “too hot”, or too far away, “too cold”. A similar concept likely applies to immunoproteostasis (see Figure 1). Although functional genetics clues are emerging that suggest that we may want to activate the immune system for therapeutic benefit in some neurodegenerative disorders, it is not at all clear if such an inference is generalizable. As manipulation of the immune system has potential for harm as well as benefit, caution and due diligence is warranted before testing novel immune therapies designed to treat a neurodegenerative disorder in humans. No matter whether “activation” or “inactivation” appears to be desirable effect, it is almost certain that pushing the system too far in one direction has potential to do no more harm than good.

Acknowledgments

Supported by grants from the NIH (U01AG046139 R01AG018454, P50AG047266).

Disclosures

TEG is a cofounder of Lacerta Inc. and on the SAB for Promis Neuroscience Inc.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  1. Abud EM, Ramirez RN, Martinez ES, Healy LM, Nguyen CHH, Newman SA, Yeromin AV, Scarfone VM, Marsh SE, Fimbres C, et al. (2017). iPSC-Derived Human Microglia-like Cells to Study Neurological Diseases. Neuron 94, 278–293 e279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Aisen PS, Schafer KA, Grundman M, Pfeiffer E, Sano M, Davis KL, Farlow MR, Jin S, Thomas RG, Thal LJ, et al. (2003). Effects of rofecoxib or naproxen vs placebo on Alzheimer disease progression: a randomized controlled trial. JAMA 289, 2819–2826. [DOI] [PubMed] [Google Scholar]
  3. Akiyama H, Barger S, Barnum S, Bradt B, Bauer J, Cole GM, Cooper NR, Eikelenboom P, Emmerling M, Fiebich BL, et al. (2000). Inflammation and Alzheimer’s disease. Neurobiol Aging 21, 383–421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Arrant AE, Filiano AJ, Patel AR, Hoffmann MQ, Boyle NR, Kashyap SN, Onyilo VC, Young AH, and Roberson ED (2018). Reduction of microglial progranulin does not exacerbate pathology or behavioral deficits in neuronal progranulin-insufficient mice. Neurobiol Dis 124, 152–162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Ayers JI, Fromholt S, Sinyavskaya O, Siemienski Z, Rosario AM, Li A, Crosby KW, Cruz PE, DiNunno NM, Janus C, et al. (2014). Widespread and efficient transduction of spinal cord and brain following neonatal AAV injection and potential disease modifying effect in ALS mice. Mol Ther [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Baker M, Mackenzie IR, Pickering-Brown SM, Gass J, Rademakers R, Lindholm C, Snowden J, Adamson J, Sadovnick AD, Rollinson S, et al. (2006). Mutations in progranulin cause tau-negative frontotemporal dementia linked to chromosome 17. Nature 442, 916–919. [DOI] [PubMed] [Google Scholar]
  7. Barrow AD, and Trowsdale J (2006). You say ITAM and I say ITIM, let’s call the whole thing off: the ambiguity of immunoreceptor signalling. Eur J Immunol 36, 1646–1653. [DOI] [PubMed] [Google Scholar]
  8. Bateman RJ, Benzinger TL, Berry S, Clifford DB, Duggan C, Fagan AM, Fanning K, Farlow MR, Hassenstab J, McDade EM, et al. (2017). The DIAN-TU Next Generation Alzheimer’s prevention trial: Adaptive design and disease progression model. Alzheimers Dement 13, 8–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Berry BJ, Smith AST, Young JE, and Mack DL (2018). Advances and Current Challenges Associated with the Use of Human Induced Pluripotent Stem Cells in Modeling Neurodegenerative Disease. Cells Tissues Organs, 1–19. [DOI] [PubMed] [Google Scholar]
  10. Bhaskar K, Konerth M, Kokiko-Cochran ON, Cardona A, Ransohoff RM, and Lamb BT (2010). Regulation of tau pathology by the microglial fractalkine receptor. Neuron 68, 19–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Bradshaw EM, Chibnik LB, Keenan BT, Ottoboni L, Raj T, Tang A, Rosenkrantz LL, Imboywa S, Lee M, Von Korff A, et al. (2013). CD33 Alzheimer’s disease locus: altered monocyte function and amyloid biology. Nat Neurosci 16, 848–850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Breitner JC, Baker LD, Montine TJ, Meinert CL, Lyketsos CG, Ashe KH, Brandt J, Craft S, Evans DE, Green RC, et al. (2011). Extended results of the Alzheimer’s disease anti-inflammatory prevention trial. Alzheimers Dement 7, 402–411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Breitner JC, Gau BA, Welsh KA, Plassman BL, McDonald WM, Helms MJ, and Anthony JC (1994). Inverse association of anti-inflammatory treatments and Alzheimer’s disease: initial results of a co-twin control study. Neurology 44, 227–232. [DOI] [PubMed] [Google Scholar]
  14. Breitner JC, Welsh KA, Helms MJ, Gaskell PC, Gau BA, Roses AD, Pericak-Vance MA, and Saunders AM (1995). Delayed onset of Alzheimer’s disease with nonsteroidal anti-inflammatory and histamine H2 blocking drugs. Neurobiol Aging 16, 523–530. [DOI] [PubMed] [Google Scholar]
  15. Brouwers N, Van Cauwenberghe C, Engelborghs S, Lambert JC, Bettens K, Le Bastard N, Pasquier F, Montoya AG, Peeters K, Mattheijssens M, et al. (2012). Alzheimer risk associated with a copy number variation in the complement receptor 1 increasing C3b/C4b binding sites. Mol Psychiatry 17, 223–233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Carrasquillo MM, Allen M, Burgess JD, Wang X, Strickland SL, Aryal S, Siuda J, Kachadoorian ML, Medway C, Younkin CS, et al. (2017). A candidate regulatory variant at the TREM gene cluster associates with decreased Alzheimer’s disease risk and increased TREML1 and TREM2 brain gene expression. Alzheimers Dement 13, 663–673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Cerami C, Iaccarino L, and Perani D (2017). Molecular Imaging of Neuroinflammation in Neurodegenerative Dementias: The Role of In Vivo PET Imaging. Int J Mol Sci 18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Chakrabarty P, Ceballos-Diaz C, Beccard A, Janus C, Dickson D, Golde TE, and Das P (2010). IFN-gamma promotes complement expression and attenuates amyloid plaque deposition in amyloid beta precursor protein transgenic mice. J Immunol 184, 5333–5343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Chakrabarty P, Ceballos-Diaz C, Lin WL, Beccard A, Jansen-West K, McFarland NR, Janus C, Dickson D, Das P, and Golde TE (2011). Interferon-gamma induces progressive nigrostriatal degeneration and basal ganglia calcification. Nat Neurosci 14, 694–696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Chakrabarty P, Li A, Ceballos-Diaz C, Eddy JA, Funk CC, Moore B, DiNunno N, Rosario AM, Cruz PE, Verbeeck C, et al. (2015). IL-10 alters immunoproteostasis in APP mice, increasing plaque burden and worsening cognitive behavior. Neuron 85, 519–533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Chang D, Nalls MA, Hallgrimsdottir IB, Hunkapiller J, van der Brug M, Cai F, International Parkinson’s Disease Genomics, C., andMe Research, T., Kerchner GA, Ayalon G, et al. (2017). A meta-analysis of genome-wide association studies identifies 17 new Parkinson’s disease risk loci. Nat Genet 49, 1511–1516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Colonna M (2003). TREMs in the immune system and beyond. Nat Rev Immunol 3, 445–453. [DOI] [PubMed] [Google Scholar]
  23. Cruts M, Gijselinck I, van der Zee J, Engelborghs S, Wils H, Pirici D, Rademakers R, Vandenberghe R, Dermaut B, Martin JJ, et al. (2006). Null mutations in progranulin cause ubiquitin-positive frontotemporal dementia linked to chromosome 17q21. Nature 442, 920–924. [DOI] [PubMed] [Google Scholar]
  24. Cummings JL, Morstorf T, and Zhong K (2014). Alzheimer’s disease drug-development pipeline: few candidates, frequent failures. Alzheimers Res Ther 6, 37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Czirr E, and Wyss-Coray T (2012). The immunology of neurodegeneration. J Clin Invest 122, 1156–1163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Dawson TM, Golde TE, and Lagier-Tourenne C (2018). Animal models of neurodegenerative diseases. Nat Neurosci 21, 1370–1379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. de Jong D, Jansen R, Hoefnagels W, Jellesma-Eggenkamp M, Verbeek M, Borm G, and Kremer B (2008). No effect of one-year treatment with indomethacin on Alzheimer’s disease progression: a randomized controlled trial. PLoS One 3, e1475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Delabar J-M, Goldgaber D, Lamour Y, Nicole A, Huret J-L, de Grouchy J, Brown P, Gajdusek DC, and Sinet P-M (1987). ß amyloid gene duplication in Alzheimer’s disease and karyotypically normal Down syndrome. Science 235, 1390–1392. [DOI] [PubMed] [Google Scholar]
  29. Doody RS, Thomas RG, Farlow M, Iwatsubo T, Vellas B, Joffe S, Kieburtz K, Raman R, Sun X, Aisen PS, et al. (2014). Phase 3 trials of solanezumab for mild-to-moderate Alzheimer’s disease. N Engl J Med 370, 311–321. [DOI] [PubMed] [Google Scholar]
  30. Douvaras P, Sun B, Wang M, Kruglikov I, Lallos G, Zimmer M, Terrenoire C, Zhang B, Gandy S, Schadt E, et al. (2017). Directed Differentiation of Human Pluripotent Stem Cells to Microglia. Stem Cell Reports 8, 1516–1524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Edison P, Donat CK, and Sastre M (2018). In vivo Imaging of Glial Activation in Alzheimer’s Disease. Front Neurol 9, 625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Egan MF, Kost J, Tariot PN, Aisen PS, Cummings JL, Vellas B, Sur C, Mukai Y, Voss T, Furtek C, et al. (2018). Randomized Trial of Verubecestat for Mild-to-Moderate Alzheimer’s Disease. N Engl J Med 378, 1691–1703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Eikelenboom P, Rozemuller JM, and van Muiswinkel FL (1998). Inflammation and Alzheimer’s disease: relationships between pathogenic mechanisms and clinical expression. Exp Neurol 154, 89–98. [DOI] [PubMed] [Google Scholar]
  34. Elmore MRP, Hohsfield LA, Kramar EA, Soreq L, Lee RJ, Pham ST, Najafi AR, Spangenberg EE, Wood MA, West BL, et al. (2018). Replacement of microglia in the aged brain reverses cognitive, synaptic, and neuronal deficits in mice. Aging Cell 17, e12832. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Eriksen JL, Sagi SA, Smith TE, Weggen S, Das P, McLendon DC, Ozols VV, Jessing KW, Zavitz KH, Koo EH, et al. (2003). NSAIDs and enantiomers of flurbiprofen target gamma-secretase and lower Abeta 42 in vivo. J Clin Invest 112, 440–449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Erny D, Hrabe de Angelis AL, Jaitin D, Wieghofer P, Staszewski O, David E, Keren-Shaul H, Mahlakoiv T, Jakobshagen K, Buch T, et al. (2015). Host microbiota constantly control maturation and function of microglia in the CNS. Nat Neurosci 18, 965–977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Filiano AJ, Martens LH, Young AH, Warmus BA, Zhou P, Diaz-Ramirez G, Jiao J, Zhang Z, Huang EJ, Gao FB, et al. (2013). Dissociation of frontotemporal dementia-related deficits and neuroinflammation in progranulin haploinsufficient mice. J Neurosci 33, 5352–5361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Filipello F, Morini R, Corradini I, Zerbi V, Canzi A, Michalski B, Erreni M, Markicevic M, Starvaggi-Cucuzza C, Otero K, et al. (2018). The Microglial Innate Immune Receptor TREM2 Is Required for Synapse Elimination and Normal Brain Connectivity. Immunity 48, 979–991 e978. [DOI] [PubMed] [Google Scholar]
  39. Frei R, Steinle J, Birchler T, Loeliger S, Roduit C, Steinhoff D, Seibl R, Buchner K, Seger R, Reith W, et al. (2010). MHC class II molecules enhance Toll-like receptor mediated innate immune responses. PLoS One 5, e8808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Garcia-Reitboeck P, Phillips A, Piers TM, Villegas-Llerena C, Butler M, Mallach A, Rodrigues C, Arber CE, Heslegrave A, Zetterberg H, et al. (2018). Human Induced Pluripotent Stem Cell-Derived Microglia-Like Cells Harboring TREM2 Missense Mutations Show Specific Deficits in Phagocytosis. Cell Rep 24, 2300–2311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Golde TE (2016). Overcoming translational barriers impeding development of Alzheimer’s disease modifying therapies. J Neurochem 139 Suppl 2, 224–236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Golde TE, Borchelt DR, Giasson BI, and Lewis J (2013). Thinking laterally about neurodegenerative proteinopathies. J Clin Invest 123, 1847–1855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Graeber MB, Li W, and Rodriguez ML (2011). Role of microglia in CNS inflammation. FEBS Lett 585, 3798–3805. [DOI] [PubMed] [Google Scholar]
  44. Griffin WS, Sheng JG, Royston MC, Gentleman SM, McKenzie JE, Graham DI, Roberts GW, and Mrak RE (1998). Glial-neuronal interactions in Alzheimer’s disease: the potential role of a ‘cytokine cycle’ in disease progression. Brain Pathol 8, 65–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Group A-FR (2015). Follow-up evaluation of cognitive function in the randomized Alzheimer’s Disease Anti-inflammatory Prevention Trial and its Follow-up Study. Alzheimers Dement 11, 216–225 e211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Group AR, Lyketsos CG, Breitner JC, Green RC, Martin BK, Meinert C, Piantadosi S, and Sabbagh M (2007). Naproxen and celecoxib do not prevent AD in early results from a randomized controlled trial. Neurology 68, 1800–1808. [DOI] [PubMed] [Google Scholar]
  47. Guerreiro R, Wojtas A, Bras J, Carrasquillo M, Rogaeva E, Majounie E, Cruchaga C, Sassi C, Kauwe JS, Younkin S, et al. (2013). TREM2 variants in Alzheimer’s disease. N Engl J Med 368, 117–127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Guillot-Sestier MV, Doty KR, Gate D, Rodriguez J Jr., Leung BP, Rezai-Zadeh K, and Town T (2015). Il10 deficiency rebalances innate immunity to mitigate Alzheimer-like pathology. Neuron 85, 534–548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Haenseler W, Sansom SN, Buchrieser J, Newey SE, Moore CS, Nicholls FJ, Chintawar S, Schnell C, Antel JP, Allen ND, et al. (2017). A Highly Efficient Human Pluripotent Stem Cell Microglia Model Displays a Neuronal-Co-culture-Specific Expression Profile and Inflammatory Response. Stem Cell Reports 8, 1727–1742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Hamza TH, Zabetian CP, Tenesa A, Laederach A, Montimurro J, Yearout D, Kay DM, Doheny KF, Paschall J, Pugh E, et al. (2010). Common genetic variation in the HLA region is associated with late-onset sporadic Parkinson’s disease. Nat Genet 42, 781–785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Harach T, Marungruang N, Duthilleul N, Cheatham V, Mc Coy KD, Frisoni G, Neher JJ, Fak F, Jucker M, Lasser T, et al. (2017). Reduction of Abeta amyloid pathology in APPPS1 transgenic mice in the absence of gut microbiota. Sci Rep 7, 41802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Hardy J, and Selkoe DJ (2002). The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. science 297, 353–356. [DOI] [PubMed] [Google Scholar]
  53. Haure-Mirande JV, Audrain M, Fanutza T, Kim SH, Klein WL, Glabe C, Readhead B, Dudley JT, Blitzer RD, Wang M, et al. (2017). Deficiency of TYROBP, an adapter protein for TREM2 and CR3 receptors, is neuroprotective in a mouse model of early Alzheimer’s pathology. Acta Neuropathol 134, 769–788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Heneka MT, Carson MJ, El Khoury J, Landreth GE, Brosseron F, Feinstein DL, Jacobs AH, Wyss-Coray T, Vitorica J, Ransohoff RM, et al. (2015). Neuroinflammation in Alzheimer’s disease. Lancet Neurol 14, 388–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Heneka MT, Kummer MP, Stutz A, Delekate A, Schwartz S, Vieira-Saecker A, Griep A, Axt D, Remus A, Tzeng TC, et al. (2013). NLRP3 is activated in Alzheimer’s disease and contributes to pathology in APP/PS1 mice. Nature 493, 674–678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Hill-Burns EM, Factor SA, Zabetian CP, Thomson G, and Payami H (2011). Evidence for more than one Parkinson’s disease-associated variant within the HLA region. PLoS One 6, e27109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Huang KL, Marcora E, Pimenova AA, Di Narzo AF, Kapoor M, Jin SC, Harari O, Bertelsen S, Fairfax BP, Czajkowski J, et al. (2017). A common haplotype lowers PU.1 expression in myeloid cells and delays onset of Alzheimer’s disease. Nat Neurosci 20, 1052–1061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. in ‘t Veld BA, Launer LJ, Hoes AW, Ott A, Hofman A, Breteler MM, and Stricker BH (1998). NSAIDs and incident Alzheimer’s disease. The Rotterdam Study [see comments]. Neurobiol Aging 19, 607–611. [DOI] [PubMed] [Google Scholar]
  59. International Genomics of Alzheimer’s Disease, C. (2015). Convergent genetic and expression data implicate immunity in Alzheimer’s disease. Alzheimers Dement 11, 658–671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Jack CR Jr., Bennett DA, Blennow K, Carrillo MC, Feldman HH, Frisoni GB, Hampel H, Jagust WJ, Johnson KA, Knopman DS, et al. (2016). A/T/N: An unbiased descriptive classification scheme for Alzheimer disease biomarkers. Neurology 87, 539–547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Jantzen PT, Connor KE, DiCarlo G, Wenk GL, Wallace JL, Rojiani AM, Coppola D, Morgan D, and Gordon MN (2002). Microglial activation and beta -amyloid deposit reduction caused by a nitric oxide-releasing nonsteroidal anti-inflammatory drug in amyloid precursor protein plus presenilin-1 transgenic mice. J Neurosci 22, 2246–2254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Jonsson T, Stefansson H, Steinberg S, Jonsdottir I, Jonsson PV, Snaedal J, Bjornsson S, Huttenlocher J, Levey AI, Lah JJ, et al. (2013). Variant of TREM2 associated with the risk of Alzheimer’s disease. N Engl J Med 368, 107–116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Kao AW, McKay A, Singh PP, Brunet A, and Huang EJ (2017). Progranulin, lysosomal regulation and neurodegenerative disease. Nat Rev Neurosci 18, 325–333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Katsumata Y, Nelson PT, Estus S, Alzheimer’s Disease Neuroimaging I. , and Fardo DW (2018). Translating Alzheimer’s disease-associated polymorphisms into functional candidates: a survey of IGAP genes and SNPs. Neurobiol Aging 74, 135–146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Kleinberger G, Yamanishi Y, Suarez-Calvet M, Czirr E, Lohmann E, Cuyvers E, Struyfs H, Pettkus N, Wenninger-Weinzierl A, Mazaheri F, et al. (2014). TREM2 mutations implicated in neurodegeneration impair cell surface transport and phagocytosis. Sci Transl Med 6, 243ra286. [DOI] [PubMed] [Google Scholar]
  66. Korvatska O, Leverenz JB, Jayadev S, McMillan P, Kurtz I, Guo X, Rumbaugh M, Matsushita M, Girirajan S, Dorschner MO, et al. (2015). R47H Variant of TREM2 Associated With Alzheimer Disease in a Large Late-Onset Family: Clinical, Genetic, and Neuropathological Study. JAMA Neurol 72, 920–927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Koyama A, O’Brien J, Weuve J, Blacker D, Metti AL, and Yaffe K (2013). The role of peripheral inflammatory markers in dementia and Alzheimer’s disease: a meta-analysis. J Gerontol A Biol Sci Med Sci 68, 433–440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Kukar T, Murphy MP, Eriksen JL, Sagi SA, Weggen S, Smith TE, Ladd T, Khan MA, Kache R, Beard J, et al. (2005). Diverse compounds mimic Alzheimer disease-causing mutations by augmenting Abeta42 production. Nat Med 11, 545–550. [DOI] [PubMed] [Google Scholar]
  69. Labzin LI, Heneka MT, and Latz E (2018). Innate Immunity and Neurodegeneration. Annu Rev Med 69, 437–449. [DOI] [PubMed] [Google Scholar]
  70. Lambert JC, Grenier-Boley B, Chouraki V, Heath S, Zelenika D, Fievet N, Hannequin D, Pasquier F, Hanon O, Brice A, et al. (2010). Implication of the immune system in Alzheimer’s disease: evidence from genome-wide pathway analysis. J Alzheimers Dis 20, 1107–1118. [DOI] [PubMed] [Google Scholar]
  71. Lambert JC, Heath S, Even G, Campion D, Sleegers K, Hiltunen M, Combarros O, Zelenika D, Bullido MJ, Tavernier B, et al. (2009). Genome-wide association study identifies variants at CLU and CR1 associated with Alzheimer’s disease. Nat Genet 41, 1094–1099. [DOI] [PubMed] [Google Scholar]
  72. Lee CYD, Daggett A, Gu X, Jiang LL, Langfelder P, Li X, Wang N, Zhao Y, Park CS, Cooper Y, et al. (2018). Elevated TREM2 Gene Dosage Reprograms Microglia Responsivity and Ameliorates Pathological Phenotypes in Alzheimer’s Disease Models. Neuron 97, 1032–1048 e1035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Lee S, Varvel NH, Konerth ME, Xu G, Cardona AE, Ransohoff RM, and Lamb BT (2010). CX3CR1 deficiency alters microglial activation and reduces beta-amyloid deposition in two Alzheimer’s disease mouse models. Am J Pathol 177, 2549–2562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Lessard CB, Malnik SL, Zhou Y, Ladd TB, Cruz PE, Ran Y, Mahan TE, Chakrabaty P, Holtzman DM, Ulrich JD, et al. (2018). High-affinity interactions and signal transduction between Abeta oligomers and TREM2. EMBO Mol Med 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Leyns CEG, Ulrich JD, Finn MB, Stewart FR, Koscal LJ, Remolina Serrano J, Robinson GO, Anderson E, Colonna M, and Holtzman DM (2017). TREM2 deficiency attenuates neuroinflammation and protects against neurodegeneration in a mouse model of tauopathy. Proc Natl Acad Sci U S A 114, 11524–11529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Liang Y, Buckley TR, Tu L, Langdon SD, and Tedder TF (2001). Structural organization of the human MS4A gene cluster on Chromosome 11q12. Immunogenetics 53, 357–368. [DOI] [PubMed] [Google Scholar]
  77. Lim GP, Yang F, Chu T, Chen P, Beech W, Teter B, Tran T, Ubeda O, Ashe KH, Frautschy SA, et al. (2000). Ibuprofen suppresses plaque pathology and inflammation in a mouse model for Alzheimer’s disease. J Neurosci 20, 5709–5714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Linnartz B, and Neumann H (2013). Microglial activatory (immunoreceptor tyrosine-based activation motif)- and inhibitory (immunoreceptor tyrosine-based inhibition motif)-signaling receptors for recognition of the neuronal glycocalyx. Glia 61, 37–46. [DOI] [PubMed] [Google Scholar]
  79. Liu D, Cao B, Zhao Y, Huang H, McIntyre RS, Rosenblat JD, and Zhou H (2018). Soluble TREM2 changes during the clinical course of Alzheimer’s disease: A meta-analysis. Neurosci Lett 686, 10–16. [DOI] [PubMed] [Google Scholar]
  80. Lui H, Zhang J, Makinson SR, Cahill MK, Kelley KW, Huang HY, Shang Y, Oldham MC, Martens LH, Gao F, et al. (2016). Progranulin Deficiency Promotes Circuit-Specific Synaptic Pruning by Microglia via Complement Activation. Cell 165, 921–935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Magno L, B., L.C., Matrtins M, Cruz PE, Katan M, Bilsland J, CHakrabarty P, Golde TE, and WHiting P (2018). Alzheimer’s disease Phospholipase C-gamma-2 (PLCG2) protective variant is a functional hypermorph. bioRxiv [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Malle E, and De Beer FC (1996). Human serum amyloid A (SAA) protein: a prominent acute-phase reactant for clinical practice. Eur J Clin Invest 26, 427–435. [DOI] [PubMed] [Google Scholar]
  83. Mann DM (1988). Alzheimer’s disease and Down’s syndrome. Histopathology 13, 125–137. [DOI] [PubMed] [Google Scholar]
  84. Marsh SE, Abud EM, Lakatos A, Karimzadeh A, Yeung ST, Davtyan H, Fote GM, Lau L, Weinger JG, Lane TE, et al. (2016). The adaptive immune system restrains Alzheimer’s disease pathogenesis by modulating microglial function. Proc Natl Acad Sci U S A 113, E1316–1325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Martens LH, Zhang J, Barmada SJ, Zhou P, Kamiya S, Sun B, Min SW, Gan L, Finkbeiner S, Huang EJ, et al. (2012). Progranulin deficiency promotes neuroinflammation and neuron loss following toxin-induced injury. J Clin Invest 122, 3955–3959. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Martinez-Muriana A, Mancuso R, Francos-Quijorna I, Olmos-Alonso A, Osta R, Perry VH, Navarro X, Gomez-Nicola D, and Lopez-Vales R (2016). CSF1R blockade slows the progression of amyotrophic lateral sclerosis by reducing microgliosis and invasion of macrophages into peripheral nerves. Sci Rep 6, 25663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Masgrau R, Guaza C, Ransohoff RM, and Galea E (2017). Should We Stop Saying ‘Glia’ and ‘Neuroinflammation’? Trends Mol Med 23, 486–500. [DOI] [PubMed] [Google Scholar]
  88. Masters CL, Simms G, Weinman NA, Multhaup G, McDonald BL, and Beyreuther K (1985). Amyloid plaque core protein in Alzheimer disease and Down syndrome. Proc Natl Acad Sci USA 82, 4245–4249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. McGeer PL, Rogers J, and McGeer EG (1994). Neuroimmune mechanisms in Alzheimer disease pathogenesis [see comments]. [Review]. Alzheimer Disease & Associated Disorders 8, 149–158. [DOI] [PubMed] [Google Scholar]
  90. McGeer PL, Schulzer M, and McGeer EG (1996). Arthritis and anti-inflammatory agents as possible protective factors for Alzheimer’s disease: a review of 17 epidemiologic studies [see comments]. Neurology 47, 425–432. [DOI] [PubMed] [Google Scholar]
  91. Miller DH, Khan OA, Sheremata WA, Blumhardt LD, Rice GP, Libonati MA, Willmer-Hulme AJ, Dalton CM, Miszkiel KA, O’Connor PW, et al. (2003). A controlled trial of natalizumab for relapsing multiple sclerosis. N Engl J Med 348, 15–23. [DOI] [PubMed] [Google Scholar]
  92. Minami SS, Min SW, Krabbe G, Wang C, Zhou Y, Asgarov R, Li Y, Martens LH, Elia LP, Ward ME, et al. (2014). Progranulin protects against amyloid beta deposition and toxicity in Alzheimer’s disease mouse models. Nat Med 20, 1157–1164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Minter MR, Hinterleitner R, Meisel M, Zhang C, Leone V, Zhang X, Oyler-Castrillo P, Zhang X, Musch MW, Shen X, et al. (2017). Antibiotic-induced perturbations in microbial diversity during post-natal development alters amyloid pathology in an aged APPSWE/PS1DeltaE9 murine model of Alzheimer’s disease. Sci Rep 7, 10411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Mitsui J, Matsukawa T, Ishiura H, Higasa K, Yoshimura J, Saito TL, Ahsan B, Takahashi Y, Goto J, Iwata A, et al. (2012). CSF1R mutations identified in three families with autosomal dominantly inherited leukoencephalopathy. Am J Med Genet B Neuropsychiatr Genet 159B, 951–957. [DOI] [PubMed] [Google Scholar]
  95. Moreland LW, Schiff MH, Baumgartner SW, Tindall EA, Fleischmann RM, Bulpitt KJ, Weaver AL, Keystone EC, Furst DE, Mease PJ, et al. (1999). Etanercept therapy in rheumatoid arthritis. A randomized, controlled trial. Ann Intern Med 130, 478–486. [DOI] [PubMed] [Google Scholar]
  96. Nalls MA, Pankratz N, Lill CM, Do CB, Hernandez DG, Saad M, DeStefano AL, Kara E, Bras J, Sharma M, et al. (2014). Large-scale meta-analysis of genome-wide association data identifies six new risk loci for Parkinson’s disease. Nat Genet 46, 989–993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Nataf S, Anginot A, Vuaillat C, Malaval L, Fodil N, Chereul E, Langlois JB, Dumontel C, Cavillon G, Confavreux C, et al. (2005). Brain and bone damage in KARAP/DAP12 loss-of-function mice correlate with alterations in microglia and osteoclast lineages. Am J Pathol 166, 275–286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Neumann H, and Takahashi K (2007). Essential role of the microglial triggering receptor expressed on myeloid cells-2 (TREM2) for central nervous tissue immune homeostasis. J Neuroimmunol 184, 92–99. [DOI] [PubMed] [Google Scholar]
  99. Olmos-Alonso A, Schetters ST, Sri S, Askew K, Mancuso R, Vargas-Caballero M, Holscher C, Perry VH, and Gomez-Nicola D (2016). Pharmacological targeting of CSF1R inhibits microglial proliferation and prevents the progression of Alzheimer’s-like pathology. Brain 139, 891–907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Ombrello MJ, Remmers EF, Sun G, Freeman AF, Datta S, Torabi-Parizi P, Subramanian N, Bunney TD, Baxendale RW, Martins MS, et al. (2012). Cold urticaria, immunodeficiency, and autoimmunity related to PLCG2 deletions. N Engl J Med 366, 330–338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Ostrowitzki S, Lasser RA, Dorflinger E, Scheltens P, Barkhof F, Nikolcheva T, Ashford E, Retout S, Hofmann C, Delmar P, et al. (2017). A phase III randomized trial of gantenerumab in prodromal Alzheimer’s disease. Alzheimers Res Ther 9, 95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Otero K, Shinohara M, Zhao H, Cella M, Gilfillan S, Colucci A, Faccio R, Ross FP, Teitelbaum SL, Takayanagi H, et al. (2012). TREM2 and beta-catenin regulate bone homeostasis by controlling the rate of osteoclastogenesis. J Immunol 188, 2612–2621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Paloneva J, Autti T, Hakola P, and Haltia MJ (1993). Polycystic Lipomembranous Osteodysplasia with Sclerosing Leukoencephalopathy (PLOSL) [PubMed] [Google Scholar]
  104. Paloneva J, Manninen T, Christman G, Hovanes K, Mandelin J, Adolfsson R, Bianchin M, Bird T, Miranda R, Salmaggi A, et al. (2002). Mutations in two genes encoding different subunits of a receptor signaling complex result in an identical disease phenotype. Am J Hum Genet 71, 656–662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Park J, Wetzel I, Marriott I, Dreau D, D’Avanzo C, Kim DY, Tanzi RE, and Cho H (2018). A 3D human triculture system modeling neurodegeneration and neuroinflammation in Alzheimer’s disease. Nat Neurosci 21, 941–951. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Perry VH, and Holmes C (2014). Microglial priming in neurodegenerative disease. Nat Rev Neurol 10, 217–224. [DOI] [PubMed] [Google Scholar]
  107. Piccio L, Deming Y, Del-Aguila JL, Ghezzi L, Holtzman DM, Fagan AM, Fenoglio C, Galimberti D, Borroni B, and Cruchaga C (2016). Cerebrospinal fluid soluble TREM2 is higher in Alzheimer disease and associated with mutation status. Acta Neuropathol 131, 925–933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Prinz M, and Priller J (2017). The role of peripheral immune cells in the CNS in steady state and disease. Nat Neurosci 20, 136–144. [DOI] [PubMed] [Google Scholar]
  109. Rademakers R, Baker M, Nicholson AM, Rutherford NJ, Finch N, Soto-Ortolaza A, Lash J, Wider C, Wojtas A, DeJesus-Hernandez M, et al. (2011). Mutations in the colony stimulating factor 1 receptor (CSF1R) gene cause hereditary diffuse leukoencephalopathy with spheroids. Nat Genet 44, 200–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Ransohoff RM (2016). A polarizing question: do M1 and M2 microglia exist? Nat Neurosci 19, 987–991. [DOI] [PubMed] [Google Scholar]
  111. Rauchmann BS, Schneider-Axmann T, Alexopoulos P, Perneczky R, and Alzheimer’s Disease Neuroimaging, I. (2018). CSF soluble TREM2 as a measure of immune response along the Alzheimer’s disease continuum. Neurobiol Aging 74, 182–190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Reiman EM, Langbaum JB, Fleisher AS, Caselli RJ, Chen K, Ayutyanont N, Quiroz YT, Kosik KS, Lopera F, and Tariot PN (2011). Alzheimer’s Prevention Initiative: a plan to accelerate the evaluation of presymptomatic treatments. J Alzheimers Dis 26 Suppl 3, 321–329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Reines SA, Block GA, Morris JC, Liu G, Nessly ML, Lines CR, Norman BA, Baranak CC, and Rofecoxib Protocol 091 Study, G. (2004). Rofecoxib: no effect on Alzheimer’s disease in a 1-year, randomized, blinded, controlled study. Neurology 62, 66–71. [DOI] [PubMed] [Google Scholar]
  114. Reitz C, Jun G, Naj A, Rajbhandary R, Vardarajan BN, Wang LS, Valladares O, Lin CF, Larson EB, Graff-Radford NR, et al. (2013). Variants in the ATP-binding cassette transporter (ABCA7), apolipoprotein E 4,and the risk of late-onset Alzheimer disease in African Americans. JAMA 309, 1483–1492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Rogers J, Webster S, Lue LF, Brachova L, Civin WH, Emmerling M, Shivers B, Walker D, and McGeer P (1996). Inflammation and Alzheimer’s disease pathogenesis. Neurobiol Aging 17, 681–686. [DOI] [PubMed] [Google Scholar]
  116. Sack GH Jr. (2018). Serum amyloid A - a review. Mol Med 24, 46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Sampson TR, Debelius JW, Thron T, Janssen S, Shastri GG, Ilhan ZE, Challis C, Schretter CE, Rocha S, Gradinaru V, et al. (2016). Gut Microbiota Regulate Motor Deficits and Neuroinflammation in a Model of Parkinson’s Disease. Cell 167, 1469–1480 e1412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Sayed FA, Telpoukhovskaia M, Kodama L, Li Y, Zhou Y, Le D, Hauduc A, Ludwig C, Gao F, Clelland C, et al. (2018). Differential effects of partial and complete loss of TREM2 on microglial injury response and tauopathy. Proc Natl Acad Sci U S A 115, 10172–10177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Schlepckow K, Kleinberger G, Fukumori A, Feederle R, Lichtenthaler SF, Steiner H, and Haass C (2017). An Alzheimer-associated TREM2 variant occurs at the ADAM cleavage site and affects shedding and phagocytic function. EMBO Mol Med 9, 1356–1365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Sevigny J, Chiao P, Bussiere T, Weinreb PH, Williams L, Maier M, Dunstan R, Salloway S, Chen T, Ling Y, et al. (2016). The antibody aducanumab reduces Abeta plaques in Alzheimer’s disease. Nature 537, 50–56. [DOI] [PubMed] [Google Scholar]
  121. Shatz CJ (2009). MHC class I: an unexpected role in neuronal plasticity. Neuron 64, 40–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Siddiqui SS, Springer SA, Verhagen A, Sundaramurthy V, Alisson-Silva F, Jiang W, Ghosh P, and Varki A (2017). The Alzheimer’s disease-protective CD33 splice variant mediates adaptive loss of function via diversion to an intracellular pool. J Biol Chem 292, 15312–15320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Sims R, van der Lee SJ, Naj AC, Bellenguez C, Badarinarayan N, Jakobsdottir J, Kunkle BW, Boland A, Raybould R, Bis JC, et al. (2017). Rare coding variants in PLCG2, ABI3, and TREM2 implicate microglial-mediated innate immunity in Alzheimer’s disease. Nat Genet 49, 1373–1384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Smemo S, Tena JJ, Kim KH, Gamazon ER, Sakabe NJ, Gomez-Marin C, Aneas I, Credidio FL, Sobreira DR, Wasserman NF, et al. (2014). Obesity-associated variants within FTO form long-range functional connections with IRX3. Nature 507, 371–375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Smith KR, Damiano J, Franceschetti S, Carpenter S, Canafoglia L, Morbin M, Rossi G, Pareyson D, Mole SE, Staropoli JF, et al. (2012). Strikingly different clinicopathological phenotypes determined by progranulin-mutation dosage. Am J Hum Genet 90, 1102–1107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Soininen H, West C, Robbins J, and Niculescu L (2007). Long-term efficacy and safety of celecoxib in Alzheimer’s disease. Dement Geriatr Cogn Disord 23, 8–21. [DOI] [PubMed] [Google Scholar]
  127. Song WM, Joshita S, Zhou Y, Ulland TK, Gilfillan S, and Colonna M (2018). Humanized TREM2 mice reveal microglia-intrinsic and -extrinsic effects of R47H polymorphism. J Exp Med 215, 745–760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Soragna D, Papi L, Ratti MT, Sestini R, Tupler R, and Montalbetti L (2003). An Italian family affected by Nasu-Hakola disease with a novel genetic mutation in the TREM2 gene. J Neurol Neurosurg Psychiatry 74, 825–826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Sosna J, Philipp S, Albay R 3rd, Reyes-Ruiz JM, Baglietto-Vargas D, LaFerla FM, and Glabe CG (2018). Early long-term administration of the CSF1R inhibitor PLX3397 ablates microglia and reduces accumulation of intraneuronal amyloid, neuritic plaque deposition and pre-fibrillar oligomers in 5XFAD mouse model of Alzheimer’s disease. Mol Neurodegener 13, 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Spangenberg EE, Lee RJ, Najafi AR, Rice RA, Elmore MR, Blurton-Jones M, West BL, and Green KN (2016). Eliminating microglia in Alzheimer’s mice prevents neuronal loss without modulating amyloid-beta pathology. Brain 139, 1265–1281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Sperling RA, Rentz DM, Johnson KA, Karlawish J, Donohue M, Salmon DP, and Aisen P (2014). The A4 study: stopping AD before symptoms begin? Sci Transl Med 6, 228fs213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Stabile C, Taglia I, Battisti C, Bianchi S, and Federico A (2016). Hereditary diffuse leukoencephalopathy with axonal spheroids (HDLS): update on molecular genetics. Neurol Sci 37, 1565–1569. [DOI] [PubMed] [Google Scholar]
  133. Stewart WF, Kawas C, Corrada M, and Metter EJ (1997). Risk of Alzheimer’s disease and duration of NSAID use. Neurology 48, 626–632. [DOI] [PubMed] [Google Scholar]
  134. Suarez-Calvet M, Araque Caballero MA, Kleinberger G, Bateman RJ, Fagan AM, Morris JC, Levin J, Danek A, Ewers M, Haass C, et al. (2016a). Early changes in CSF sTREM2 in dominantly inherited Alzheimer’s disease occur after amyloid deposition and neuronal injury. Sci Transl Med 8, 369ra178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Suarez-Calvet M, Capell A, Araque Caballero MA, Morenas-Rodriguez E, Fellerer K, Franzmeier N, Kleinberger G, Eren E, Deming Y, Piccio L, et al. (2018). CSF progranulin increases in the course of Alzheimer’s disease and is associated with sTREM2, neurodegeneration and cognitive decline. EMBO Mol Med [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Suarez-Calvet M, Kleinberger G, Araque Caballero MA, Brendel M, Rominger A, Alcolea D, Fortea J, Lleo A, Blesa R, Gispert JD, et al. (2016b). sTREM2 cerebrospinal fluid levels are a potential biomarker for microglia activity in early-stage Alzheimer’s disease and associate with neuronal injury markers. EMBO Mol Med 8, 466–476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Swardfager W, Lanctot K, Rothenburg L, Wong A, Cappell J, and Herrmann N (2010). A meta-analysis of cytokines in Alzheimer’s disease. Biol Psychiatry 68, 930–941. [DOI] [PubMed] [Google Scholar]
  138. Takata K, Kozaki T, Lee CZW, Thion MS, Otsuka M, Lim S, Utami KH, Fidan K, Park DS, Malleret B, et al. (2017). Induced-Pluripotent-Stem-Cell-Derived Primitive Macrophages Provide a Platform for Modeling Tissue-Resident Macrophage Differentiation and Function. Immunity 47, 183–198 e186. [DOI] [PubMed] [Google Scholar]
  139. Tung YCL, Yeo GSH, O’Rahilly S, and Coll AP (2014). Obesity and FTO: Changing Focus at a Complex Locus. Cell Metab 20, 710–718. [DOI] [PubMed] [Google Scholar]
  140. Ulland TK, and Colonna M (2018). TREM2 - a key player in microglial biology and Alzheimer disease. Nat Rev Neurol 14, 667–675. [DOI] [PubMed] [Google Scholar]
  141. Ulrich JD, Ulland TK, Colonna M, and Holtzman DM (2017). Elucidating the Role of TREM2 in Alzheimer’s Disease. Neuron 94, 237–248. [DOI] [PubMed] [Google Scholar]
  142. Venegas C, Kumar S, Franklin BS, Dierkes T, Brinkschulte R, Tejera D, Vieira-Saecker A, Schwartz S, Santarelli F, Kummer MP, et al. (2017). Microglia-derived ASC specks cross-seed amyloid-beta in Alzheimer’s disease. Nature 552, 355–361. [DOI] [PubMed] [Google Scholar]
  143. Weggen S, Eriksen JL, Das P, Sagi SA, Wang R, Pietrzik CU, Findlay KA, Smith TE, Murphy MP, Bulter T, et al. (2001). A subset of NSAIDs lower amyloidogenic Abeta42 independently of cyclooxygenase activity. Nature 414, 212–216. [DOI] [PubMed] [Google Scholar]
  144. Weinblatt ME, Keystone EC, Furst DE, Moreland LW, Weisman MH, Birbara CA, Teoh LA, Fischkoff SA, and Chartash EK (2003). Adalimumab, a fully human anti-tumor necrosis factor alpha monoclonal antibody, for the treatment of rheumatoid arthritis in patients taking concomitant methotrexate: the ARMADA trial. Arthritis Rheum 48, 35–45. [DOI] [PubMed] [Google Scholar]
  145. Wendeln AC, Degenhardt K, Kaurani L, Gertig M, Ulas T, Jain G, Wagner J, Hasler LM, Wild K, Skodras A, et al. (2018). Innate immune memory in the brain shapes neurological disease hallmarks. Nature 556, 332–338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Wissemann WT, Hill-Burns EM, Zabetian CP, Factor SA, Patsopoulos N, Hoglund B, Holcomb C, Donahue RJ, Thomson G, Erlich H, et al. (2013). Association of Parkinson disease with structural and regulatory variants in the HLA region. Am J Hum Genet 93, 984–993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Witoelar A, Jansen IE, Wang Y, Desikan RS, Gibbs JR, Blauwendraat C, Thompson WK, Hernandez DG, Djurovic S, Schork AJ, et al. (2017). Genome-wide Pleiotropy Between Parkinson Disease and Autoimmune Diseases. JAMA Neurol 74, 780–792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Xiang X, Piers TM, Wefers B, Zhu K, Mallach A, Brunner B, Kleinberger G, Song W, Colonna M, Herms J, et al. (2018). The Trem2 R47H Alzheimer’s risk variant impairs splicing and reduces Trem2 mRNA and protein in mice but not in humans. Mol Neurodegener 13, 49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Zandi PP, and Breitner JC (2001). Do NSAIDs prevent Alzheimer’s disease? And, if so, why? The epidemiological evidence. Neurobiol Aging 22, 811–817. [DOI] [PubMed] [Google Scholar]
  150. Zhang Y, Chen K, Sloan SA, Bennett ML, Scholze AR, O’Keeffe S, Phatnani HP, Guarnieri P, Caneda C, Ruderisch N, et al. (2014). An RNA-sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex. J Neurosci 34, 11929–11947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Zhang Y, Sloan SA, Clarke LE, Caneda C, Plaza CA, Blumenthal PD, Vogel H, Steinberg GK, Edwards MS, Li G, et al. (2016). Purification and Characterization of Progenitor and Mature Human Astrocytes Reveals Transcriptional and Functional Differences with Mouse. Neuron 89, 37–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Zhao Y, Wu X, Li X, Jiang LL, Gui X, Liu Y, Sun Y, Zhu B, Pina-Crespo JC, Zhang M, et al. (2018). TREM2 Is a Receptor for beta-Amyloid that Mediates Microglial Function. Neuron 97, 1023–1031 e1027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Zhou Q, Lee GS, Brady J, Datta S, Katan M, Sheikh A, Martins MS, Bunney TD, Santich BH, Moir S, et al. (2012). A hypermorphic missense mutation in PLCG2, encoding phospholipase Cgamma2, causes a dominantly inherited autoinflammatory disease with immunodeficiency. Am J Hum Genet 91, 713–720. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES