Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 May 28.
Published in final edited form as: J Neural Transm (Vienna). 2017 Oct 23;125(5):809–826. doi: 10.1007/s00702-017-1795-7

Differential contribution of microglia and monocytes in neurodegenerative diseases

Caroline Baufeld 1, Elaine O’Loughlin 1, Narghes Calcagno 1, Charlotte Madore 1, Oleg Butovsky 1,2
PMCID: PMC7255107  NIHMSID: NIHMS1589225  PMID: 29063348

Abstract

Neuroinflammation is a hallmark of neurodegenerative diseases including Alzheimer’s disease (AD), Parkinson’s disease (PD), and amyotrophic lateral sclerosis (ALS). Microglia, the innate immune cells of the CNS, are the first to react to pathological insults. However, multiple studies have also demonstrated an involvement of peripheral monocytes in several neurodegenerative diseases. Due to the different origins of these two cell types, it is important to distinguish their role and function in the development and progression of these diseases. In this review, we will summarize and discuss the current knowledge of the differential contributions of microglia and monocytes in the common neurodegenerative diseases AD, PD, and ALS, as well as multiple sclerosis, which is now regarded as a combination of inflammatory processes and neurodegeneration. Until recently, it has been challenging to differentiate microglia from monocytes, as there were no specific markers. Therefore, the recent identification of specific molecular signatures of both cell types will help to advance our understanding of their differential contribution in neurodegenerative diseases.

Keywords: Microglia, Monocytes, Neurodegeneration, Neuroinflammation

Introduction

Neurodegeneration is a common pathological hallmark of many central nervous system (CNS) diseases and disorders. It results in disturbances of CNS homeostasis and neuronal function ultimately leading to neuronal death. Over the past few decades, research studies have focused on understanding and resolving the underlying mechanisms of neurodegeneration. The most known and studied neurodegenerative diseases include Alzheimer’s disease (AD), Parkinson’s disease (PD), and amyotrophic lateral sclerosis (ALS). In addition, this review will also cover multiple sclerosis (MS), which has long been considered primarily an autoimmune disease (Lassmann et al. 2012). However, studies have described axonal alterations and lesions in MS plaques that are associated with the clinical neuropathology (Ferguson et al. 1997; Trapp et al. 1998; Lassmann et al. 2012). Thus, it has been acknowledged that MS is a combination of inflammatory processes and neurodegeneration, typically at later stages of the disease (Chaudhuri 2013; Kawachi and Lassmann 2017).

Familial cases of neurodegenerative diseases have been very useful in identifying the genetic mutations that could increase an individual’s vulnerability to develop the disease. These include mutations in the superoxide dismutase 1 (SOD1) gene, which have been implicated in the origin of ALS, in the β-amyloid (Aβ) precursor protein in AD, or in α-synuclein for PD (Goate et al. 1991; Rosen et al. 1993; Polymeropoulos et al. 1997). Several genetic variations were also identified through genome-wide association studies (GWAS). However, many sporadic cases occur due to unknown factors. Although the causes of neurodegenerative diseases remain largely unknown, there is a growing amount of evidence showing that inflammatory and immune responses may contribute to disease progression.

Inflammation is a defense mechanism that counters diverse insults by removing or inhibiting pathogens (Wyss-Coray and Mucke 2002). In neurodegenerative diseases, inflammation results mainly from the formation of toxic endogenous protein aggregates. This inflammatory response can have beneficial effects by promoting tissue repair and the removal of cellular debris. However, it also results in the production of neurotoxic compounds that, in return, exacerbate neurodegenerative processes (Wyss-Coray and Mucke 2002; Glass et al. 2010). The response of the innate immune system includes immune cell activation and recruitment, phagocytosis of pathogens, clearance of cellular debris, and antigen presentation. It is well accepted that neuroinflammation is an underlying element of neurodegenerative disease such as ALS, AD, MS, and PD. To what extent central and peripheral immunity contribute to the development and progression of these diseases is not yet fully understood. This review will focus on CNS resident microglia and peripheral monocytes and give a broad overview over of what is known about their role in neurodegenerative diseases.

Microglia vs. monocytes: characterization and identity

Microglia are the brain’s innate immune cells and are considered to be the first responders to CNS injury. However, in certain disease conditions, the blood–brain barrier (BBB) is disrupted, allowing the entry of peripheral immune cells from the bloodstream (Carvey et al. 2009). The contribution of microglia vs. peripheral monocytes in neuroinflammatory and neurodegenerative diseases is still a matter of debate. The investigations have been hampered by the inability to distinguish the two cell types, since both can give rise to inflammatory macrophages in the CNS and are morphologically indistinguishable. Many of the findings in the past few decades are based on markers that are expressed by both peripheral monocytes and microglia(i.e., Iba1, CD45, and CD11b) (Prinz et al. 2011; Greter et al. 2015). The distinction between resident microglia and infiltrating monocytes is critical to determine the characteristics of each disease, the involvement of both cell types, and the potential modulation of their immune response. Tables 1, 2 summarize common and newly described markers for monocytes and microglia and how their expression profile change in neurodegenerative diseases.

Table 1.

Microglia markers in health and disease

Microglia Markers References
Homeostatic Cd11b+ Hickman et al. (2013), Butovsky et al. (2014), Asai et al. (2015), Greenhalgh et al. (2016), Zrzavy et al. (2017)
Cd45low
Cx3cr1+
F4/80+
Fcrls+
Iba1+
Olfml3+
P2ry12+
Sall1+
Siglec-H+
Tmem119+
Tgfbr+
Csf1r+
Neurodegenerative diseases Arg1↑ Chiu et al. (2013), Holtman et al. (2015), Keren-Shaul (2017), Krasemann (2017), Zrzavy et al. (2017)
Apoe ↑
Axl ↑
Ccl2 ↑
Cd45 ↑
Clec7a ↑
Gpnmb ↑
Msr1↑
Spp1↑
Cd11b →
F4/80 →
Fcrls →
Iba1 →
Tmem119 →
Csf1r↓
P2ry12 ↓
Siglec-H ↓
Sall1 ↓
Tgfbr↓

Arrows indicate changes in expression: ↑ increased, ↓ decreased, → unchanged

Table 2.

Monocyte markers in health and disease

Monocyte subsets Markers References
Classical inflammatory Ccr2high Mildner et al. (2007, 2009), Ingersoll et al. (2010), Butovsky et al. (2012), Rose et al. (2012), Asai et al. (2014)
Cd11b+
Cd115+
Cd169+
Cd43+
Cd45high
Cx3cr1low
Iba1+
Ly6chigh
Ly6gneg
Patrolling Ccr2low Auffray et al. (2007), Ingersoll et al. (2010), Rose et al. (2012), Carlin et al. (2013), Michaud et al. (2013)
Cd11a+
Cd11b+
Cd115+
Cd45high
Cx3cr1high
Iba1+
Ly6clow
Cd83+
Cd16+
Cd36+
Cd274+

In vivo studies of microglia and monocyte origin revealed that microglia and monocytes are ontogenetically distinct cells. Microglia are derived from hematopoietic progenitors in the yolk sac and invade the brain parenchyma around E9.5 during the murine CNS development (Ginhoux et al. 2010). In contrast, monocytes originate from the myeloid progenitor lineage in the bone marrow (Geissmann et al. 2010), where they are generated from dendritic cell and macrophage precursor cells (Geissmann et al. 2008). In mice, monocyte subsets were first identified by differential expression of chemokine receptor, CCR2. Later, they were characterized by their expression of the inflammatory marker Ly6C (Gr1) and it is now accepted that murine monocytes are divided into two major populations: Ly6Chigh and Ly6Clow based on expression levels of Ly6C on the cell surface. In addition to Ly6C and CCR2 expression, Ly6Chigh cells were described as CD11b+/CD115+/CCR2high/CX3CR1low, whereas Ly6Clow monocytes are CD11b+/CD115+/CCR2lowCX3CR1high (Geissmann et al. 2003; Gordon and Taylor 2005; Woollard and Geissmann 2010). Inflammatory Ly6Chigh monocytes represent approximately 2–5% of circulating blood cells in the periphery, which are actively recruited to sites of infection and inflammation (Serbina et al. 2008). Ly6Clow patrolling resident monocytes are less abundant than Ly6CHi monocytes where they adhere and migrate to luminal surfaces of blood vessels in a non-inflamed state (Auffray et al. 2007). Monocyte subsets in the human blood have different repertoires of surface receptors that may dictate functional roles. Human blood monocytes consist of two primary subsets with distinct expression profiles based on the LPS co-receptor, CD14 and the low-affinity IgG receptor, CD16. CD14++/CD16 -monocytes, the most abundant in the human blood, are analogues of Ly6Chigh monocytes in mice and are referred to as “classical monocytes”. CD14++/CD16−- monocytes also express CCR2 similar to their murine counterparts (Geissmann et al. 2003). There is evidence that CD14++CD16+ monocytes equate to Ly6Clow in mice based on gene expression profiles (Ingersoll et al. 2010).

Different approaches have been established to distinguish between resident microglia and recruited-blood monocytes. Microglia distinction was previously based on the expression levels of CD45, a protein tyrosine phosphatase expressed by myeloid cells. In microglia, CD45 is expressed at lower levels compared to other macrophages that are CD45high. However, in disease, CD45 levels on microglia increase which makes them harder to distinguish from the recruited-blood monocytes (Ford et al. 1995). Recent developments in neuroimmunology, imaging, and genetics have greatly progressed our understanding of the unique nature of microglia in development, homeostasis, and disease, and have led to the identification of novel markers that are unique to resident microglia and have been used to distinguish them from bone marrow-derived macrophages (BMDM) and monocytic subsets. These markers include P2Y12, TMEM119, FCRLS, CX3CR1, Siglec-H, and olfactomedin-like 3 (Gautier et al. 2012; Chiu et al. 2013; Goldmann et al. 2013; Parkhurst et al. 2013; Yona et al. 2013; Butovsky et al. 2014; Bennett et al. 2016). However, similar to CD45, some of these markers may be modulated by activation and/or expressed in other cell types outside of the CNS and may not be suitable for unequivocally identifying or targeting tissue-resident microglia.

In addition to these novel markers, other methods have been developed to specifically target and manipulate microglia or macrophages. Initially, bone marrow chimeras were used to distinguish the two cell types. In this mouse model, fluorescently labeled bone marrow cells replace the original bone marrow cells after irradiation. This technique allows for tracking the origin and fate of infiltrating cells. This approach presents with some limitations, since the irradiation itself induces an opening of the BBB and the recruitment of monocytes to the CNS. It was demonstrated that CNS infiltration of peripheral monocytes is minimal if a head shield is used during irradiation (Ajami et al. 2007) or blood circulations are shared between GFP-positive and GFP-negative mice (Mildner et al. 2007). The latter method is termed ‘parabiosis’ and can also be used to distinguish microglia from monocytes. By combining mice that express different alleles of the CD45 gene (CD45.1 and CD45.2), Wang et al. (2016) analyzed infiltration of peripheral monocytes in mouse models of AD. Another important tool to study microglia vs. monocytes was the development of the CX3CR1-CreERT mouse (Jung et al. 2000), which takes advantage of the longevity of microglia compared to peripheral monocytes, that also express CX3CR1, but renew within a few weeks. This model is now used routinely to specifically target microglia.

With this in mind, it will be crucial to take advantage of and combine microglia-specific tools, such as transcriptomic profiles, proteomic signatures, and surface markers, to reduce the observation of artifacts arising from other myeloid cells (Gautier et al. 2012; Chiu et al. 2013; Goldmann et al. 2013; Parkhurst et al. 2013; Yona et al. 2013; Butovsky et al. 2014; Bennett et al. 2016).

Microglia vs. monocytes in amyotrophic lateral sclerosis

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the loss of both upper and lower motor neurons leading to progressive skeletal muscle weakness and paralysis. Growing evidence has proven that non-neuronal cells directly contribute to motor neuron damage in ALS (Musaro 2010). Several studies in genetic models have well established that toxicity to motor neurons requires damage from mutant SOD1 acting within non-neuronal cells. Indeed, restricted expression of SOD1 mutations to neurons is not sufficient to induce ALS pathology (Pramatarova et al. 2001; Lino et al. 2002); however, wild-type neurons acquire an ALS phenotype when surrounded by glial cells carrying the SOD1 mutation (Clement et al. 2003).

In both animal models and ALS patients, neuronal loss is accompanied by a well-characterized neuroinflammatory reaction (McGeer et al. 1991; Kawamata et al. 1992; Engelhardt et al. 1993; Hall et al. 1998; Alexianu et al. 2001; McGeer and McGeer 2002), which includes proliferation of resident microglia (Henkel et al. 2004; Turner et al. 2004; Solomon et al. 2006; Appel et al. 2011). In ALS, microglia activation involves changes in morphology, gene expression, and surface receptors (Shibata et al. 2009; Butovsky et al. 2012, 2015). Microglia from mutant SOD1 mice (referred to hereafter as SOD1) display a phenotype that differs from both activation with LPS and the M1/M2 dichotomy (Chiu et al. 2013; Krasemann et al. 2017). Microglial activation in the spinal cord is an early event (Sanagi et al. 2010) that precedes clinical onset (muscle weakness), evidence of significant motor neuron loss, astrocyte reactivity (Alexianu et al. 2001), and increases with disease progression (Hall et al. 1998; Elliott 2001; Olsen et al. 2001). Several studies have demonstrated that modulation of microglial response in SOD1 mice alters disease progression. SOD1-expressing microglia adopt a pro-inflammatory phenotype (Kobayashi et al. 2013), produce excessive reactive oxidative species (Harraz et al. 2008), and exhibit enhanced neurotoxicity after LPS treatment (Xiao et al. 2007) and in late phases of the disease (Liao et al. 2012), supporting the concept that microgliosis contributes to motor neuron degeneration. Consistent with these findings, inhibition of NF-κB suppresses the neuroinflammatory component of microglial toxicity in co-cultured motor neurons (Frakes et al. 2014), and deletion of galectin-3 exacerbates microglial activation and accelerates disease progression in SOD1 mice (Lerman et al. 2012). Another study demonstrated that a lack of IL-1β expression or treatment with IL-1β receptor antagonist extended the lifespan of SOD1 mice and attenuated the inflammatory pathology (Meissner et al. 2010). Moreover, deletion of SOD1 transgene within CD11b+ myeloid cells (Boillee et al. 2006; Wang et al. 2009) improves survival in SOD1 mice. Others studies have shown the importance of CSF1R signaling in microglia and its contribution to neurodegeneration in SOD1 models (Gowing et al. 2009; Martinez-Muriana et al. 2016). However, it has also been proposed that microglia at pre-onset stage in SOD1 mice exhibit an anti-inflammatory phenotype (enriched for BDNF and CD163) and attenuate innate immune response compared with microglia at symptomatic stage (Liao et al. 2012; Gravel et al. 2016), supporting a trophic and neuroprotective role of microglia at the early phase of disease and a subsequent transformation with disease progression.

Studies in ALS patients have found that peripheral monocytes are functionally altered (Zhang et al. 2005, 2006; Nardo et al. 2011), skewed towards a pro-inflammatory state (Butovsky et al. 2015; Zhao et al. 2017), and can invade the CNS (Zondler 2016). Specifically, CD14+/CD16 inflammatory monocyte levels in blood correlate with functional score in ALS (Murdock et al. 2016). Monocytes from rapidly progressive patients have a more inflammatory profile than monocytes from slower progressing patients (Zhao et al. 2017). Analogous changes are seen in SOD1 mice, even before the symptomatic onset of disease, defined by the appearance of tremors and the hind limb splay defect, and their degree of activation correlates with disease progression (Butovsky et al. 2012). Furthermore, myeloid cells are increased in ALS and SOD1 spinal cords (Engelhardt and Appel 1990; Appel et al. 1993; Obal et al. 2001; McGeer and McGeer 2002; Wilms et al. 2003; Henkel et al. 2004; Chiu et al. 2009; Butovsky et al. 2012). One hypothesis is that disruptions in the BBB (Evans et al. 2013) might facilitate the migration of peripheral monocytes to the CNS (Butovsky et al. 2012; Zondler 2016). In human patients, the levels of CD14+ monocytes in the blood are decreased during early disease, indicating potential recruitment to the CNS (Mantovani et al. 2009). Several groups also described abnormal levels of pro-inflammatory cytokines in cerebral spinal fluid (CSF) and sera in patients with ALS (Simpson et al. 2004; Mitchell et al. 2009). Since microglia express high levels of CCL2 (Butovsky et al. 2012), which is also elevated in CSF of ALS patients (Nagata et al. 2007), and blood monocytes express higher levels of its receptor (Butovsky et al. 2012), a chemoattraction through the CCL2–CCR2 axis has been suggested to be responsible for recruiting inflammatory Ly6C+ monocytes into the CNS (Mildner et al. 2007), possibly aggravating motor neuron injury. Importantly, blocking these infiltrating monocytes pharmacologically results in delayed disease symptoms and extended survival in SOD1 mice (Butovsky et al. 2012). However, Zondler (2016) found that the presence of peripheral myeloid cells (CD169+) in the spinal cord correlates with the number of surviving motor neurons in SOD1 transgenic mice, suggesting that peripheral macrophages may exert neuroprotection under certain conditions. Others have sustained that circulating monocytes do not contribute to spinal cord inflammation in ALS and have found that monocyte infiltration is limited to the peripheral nerves (Chiu et al. 2009; Graber et al. 2010; Lincecum et al. 2010). One justification for these inconsistent results is that the experimental paradigm of irradiation and bone marrow transplantation may introduce confounding variables that modify the natural course of the disease. Irradiation itself can provoke the entrance of cells in the CNS, most likely by altering the BBB tightness (Mildner et al. 2007), and introduces progenitor populations that are not in the blood under physiological circumstances (Ajami et al. 2007). Parabiosis experiments (surgically joining the circulatory systems of two organisms) between SOD1G93A and control mice expressing GFP in bone marrow cells and showed no evidence of infiltration into the CNS from periphery in parabiotic chimeras (Ajami et al. 2007). The limitation of this elegant approach, though, is that the donor cells were from naı¨ve wild-type mice. Hence, this paradigm does not account for the disease-associated dysregulation of peripheral inflammatory monocytes. These controversial results regarding the role of microglia and monocytes suggest their complex role in ALS and can be due to different experimental models (bone marrow transplantation studies, gene excision strategies, and pharmacological targeting of molecules) that are not easily comparable.

C9ORF72 expansion (DeJesus-Hernandez et al. 2011) is currently the most commonly demonstrated mutation related to ALS. Although its function is poorly understood, three independent studies shed light on its critical role in the immune system: C9orf72 expression was highest in myeloid cells, and its loss led to lysosomal accumulation, myeloid expansion, altered immune responses in both macrophages and microglia, with age-related neuroinflammation (Burberry et al. 2016; O’Rourke et al. 2016), and signs of severe autoimmunity (Atanasio et al. 2016). Though the absence of neurodegeneration in these C9orf72-deficient mice provides an argument against the loss of C9orf72 function as the sole driver of disease, these findings of altered myeloid cell function in C9orf72-deficient mice might support the possibility that abnormal immune responses play a fundamental role in ALS.

Facing such conflicting data on the effects of inflammation, between neuroprotection and neurotoxicity in ALS, a recent hypothesis suggests that the immune response in ALS occurs in two separate phases: protective in earlier stages and toxic in late stages of the disease (Appel et al. 2011; Hooten et al. 2015). This could explain the discrepancies that have been previously reported. The potential for different mechanisms underlying the immune response in disease initiation and progression has recently been proposed (Murdock et al. 2015). In SOD1 mice, a decrease in microglia number in the entire spinal cord was observed in the pre-symptomatic stage, suggesting a distinct microglial involvement in different stages of the disease (Gerber et al. 2012). During periods of slow disease progression, an anti-inflammatory process governs neuroinflammation, while in later phases of rapid progression, neuroinflammation is directed towards a strong inflammatory state (Liao et al. 2012). Most likely, microglia are the fulcrum of this complex immune response and drive the polarization from one phase to the other during the course of the disease. Therefore, modulation of immune pathways may be more complex than anticipated. It remains for translational research studies of human ALS to document that both protective and toxic mechanisms are involved in the pathogenesis of the disease, and to employ such processes for therapeutic benefit.

Microglia vs. monocytes in Alzheimer’s disease

Dementias affect approximately 47 million people worldwide, with Alzheimer’s disease (AD) being the most common form of dementia (Prince et al. 2016). One of the major hallmarks of the disease is the extracellular accumulation of amyloid-beta (Aβ) protein as well as the development of intracellular neurofibrillary tangles (NFTs) comprised of hyperphosphorylated tau protein. Furthermore, AD pathology is accompanied by neuroinflammation, which also contributes to disease pathogenesis (Selkoe 2002; Zhang et al. 2013). Changes in cytokine levels are found in the CSF of individuals with mild cognitive impairment—amnestic type (MCI), who have an increased risk to develop AD, suggesting that this neuroinflammation occurs early in the disease development and might trigger AD (Tarkowski et al. 2003; Brosseron et al. 2014).

Recent GWAS studies identified variants of the myeloid cell genes Triggering receptor expressed on myeloid cells 2 (TREM2), CD33, and complement receptor 1 (CR1) to be associated with an increased risk to develop AD (Bradshaw et al. 2013a, b; Guerreiro et al. 2013; Jonsson et al. 2013), shifting the focus on resident microglia and peripheral myeloid cells. In addition, reactive myeloid cells accumulate specifically around Aβ-plaques in the brains of AD mouse models and humans. These findings are based on the usage of the common myeloid marker Iba-1, which raised the question whether these cells are actually resident microglia or peripheral myeloid cells that infiltrated the brain parenchyma during the development of the disease. Several studies suggested that there is a recruitment of peripheral myeloid cells to the AD brain. However, these studies have been conducted using bone marrow chimeras. Another study described the peripheral origin of plaque associated myeloid cells based on the microglial marker P2Y12 (Jay et al. 2015). However, this molecule is downregulated by reactive microglia as they can be found around amyloid plaques (Haynes et al. 2006; Krasemann et al. 2017). A study by Michaud et al. (2013) has suggested that peripheral myeloid cells do not actually infiltrate the brain parenchyma, but that their role is the removal of vascular Aβ from within the lumen of veins. As discussed above, Wang et al. (2016) distinguished microglia and monocytes by parabiosis of mice expressing either CD45.1 or CD45.2. Their study showed that there is no significant recruitment of peripheral monocytes to Aβ-plaques in the analyzed mouse models. This controversy emphasizes the need for a marker that can distinguish resident microglia from peripheral myeloid cells, regardless of the activation status. It remains a possibility that peripheral myeloid cells can be guided to the brain to help the resident microglia clear up amyloid plagues, yet conflicting reports exist as to whether they are capable of this. The approach of a transient depletion of resident microglia using an inducible suicide gene and their subsequent replacement with peripheral myeloid cells did not have any impact on Aβ plaque burden or APP processing (Prokop et al. 2015; Varvel et al. 2015). Conversely, intravenous infusion of monocytes from young mice at least transiently improved memory function and reduced Aβ plaque burden (Hohsfield and Humpel 2015). Another group did not see an effect of injected monocytes alone, but when these monocytes were genetically engineered to express neprilysin, an Ab degrading enzyme, amyloid deposition was arrested (Lebson 2010).

Similar to the conflicting evidence of the role of monocytes in AD pathology, the question also remains as to the degree of involvement of resident microglia cells in the emergence and progression of AD. As previously discussed, variants of microglial genes like TREM2, CD33, and CR1 are risk factors for AD and have become the focus of many studies. TREM2 is a transmembrane glycoprotein that binds phospholipids present in the brain due to neuronal cell death or myelin damage (Wang et al. 2015b) and has been shown to bind ApoE (Apolipoprotein E) (Atagi et al. 2015), whose isoform ε4 is a major risk factor for AD (Corder et al. 1993). We recently identified a TREM2-dependent pathway, which induces ApoE signaling in microglia. The TREM2-APOE pathway suppresses the homeostatic microglial signature and induces a transcriptional signature that is common in neurodegenerative diseases (Krasemann et al. 2017). This microglia phenotype switch is triggered by phagocytosis of dying neurons and, therefore, is specifically associated with neuritic Aβ-plaques in mouse and human AD. Trem2 deficiency in APP-PS1 mice restored the microglial homeostatic signature, decreased clustering of microglia around Aβ-plaques, and reduced Aβ-plaque burden at early stages of the disease (Jay et al. 2015; Krasemann et al. 2017). However, at later stages, Trem2 deficiency exacerbated the pathology (Wang et al. 2015b; Jay et al. 2017), indicating that TREM2 might have contrasting roles during different stages of the disease. Yuan et al. (2016) dissected the effect of Trem2 deficiency in AD mouse models and TREM2 variants in human cases on the structure of Aβ-plaques and axonal dystrophy, and found that both in mice and humans, plaques are more filamentous and less compact, which leads to increased axonal dystrophy. A recent study analyzing soluble TREM2 (sTREM2) in the CSF of patients with different disease stages of AD, from preclinical to dementia stages, revealed that sTREM2 is already upregulated in the early stages of the disease and is associated with neuronal injury markers (Suarez-Calvet et al. 2016). This may reflect an early response of microglia to neuronal injury. TREM2 has also been shown to be modulated by the CD33 AD risk allele, another risk factor for late-onset AD (Bradshaw et al. 2013a, b; Chan et al. 2015). This study found that TREM2 expression is increased in monocytes from carriers with the CD33 risk allele and that CD33 expression itself is increased on microglia in human AD brains, and elevated levels of CD33 inhibited microglial uptake and phagocytosis of Aβ (Bradshaw et al. 2013a, b; Griciuc et al. 2013).

The complement system has also been demonstrated to play an important role in microglia response to Aβ. Microglia are responsible for complement-mediated synaptic loss (Hong et al. 2016), which can be rescued by deleting complement factors C1q, C3, or CR3 in a mouse model of AD (Maier et al. 2008; Shi et al. 2017). In spite of this specific response to amyloid, microglia are not able to halt disease progression. This might be because they are impaired in their phagocytic function by the presence of Aβ, chronic inflammation, or an age-linked dysfunction (Streit et al. 2004; Hickman et al. 2013; Krabbe et al. 2013). This functional impairment as well as their secretion of various pro-inflammatory cytokines in response to Ab (Vom Berg et al. 2012; Heppner et al. 2015; Wang et al. 2015a) led to the assumption that inhibition of the microglial reaction could ameliorate the pathology. Two different approaches were used to test whether complete depletion of microglia would impact disease progression. Firstly, Grathwohl et al. (2009) demonstrated that depletion of microglia using the herpes simplex virus thymidine kinase (HSVTK) suicide gene had no effect on plaque pathology or neuritic dystrophy in APP-PS1 and APP23 mice. Secondly, treatment of AD mice with a CSF1-receptor antagonist to deplete microglia (PLX3397 or GW2580) reduced neuronal loss and improved cognition, but did not affect amyloid-plaque burden (Dagher et al. 2015; Olmos-Alonso et al. 2016; Spangenberg et al. 2016).

Multiple studies have aimed at modulating the cytokine profile in the brains of AD mice. Among them, Heneka et al. (2013) described that reduction of IL-1β through deletion of the NLRP3 inflammasome decreased Aβ and had a protective effect on memory. Inhibiting the IL-12/23 pathway similarly reduced AD-like pathology (Vom Berg et al. 2012). On the other hand, overexpression of IL-6 in AD mice attenuated plaque deposition and enhanced microglia-mediated phagocytosis of Aβ in vitro (Chakrabarty et al. 2010). Deficiency in the anti-inflammatory cytokine IL-10 increased microglial phagocytosis and reduced Aβ-plaque pathology (Guillot-Sestier et al. 2015). Taken together, it should be acknowledged that the response of microglia in AD is complex and graded (Raivich et al. 1999) and not solely ‘good’ or ‘bad’.

Microglia vs. monocytes in Parkinson’s disease

Parkinson’s disease (PD) is the second most common neurodegenerative disease in the aging population (Pringsheim et al. 2014). It is a chronic progressive disease characterized by cytoplasmic aggregates of Lewy bodies comprised of α-synuclein and ubiquitin. Progressive degeneration and ultimately the loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) lead to a decrease in dopamine levels which manifest as the motor symptoms associated with PD such as tremors, rigidity, gait dysfunction, and bradykinesia (Hornykiewicz and Kish 1987). In addition to progressive neurodegeneration, inflammation plays a central role in the pathophysiology of the disease. As with many neurodegenerative diseases, the current underlying pathological mechanisms that lead to neuronal cell loss/death, neuroinflammation, and motor impairment remain elusive. Insight in the pathological mechanisms of PD has largely stemmed from animal models incorporating toxins such as neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) in mice and 6-hydroxydopamine (6-OHDA) in rats/monkeys or genetic-based models using α-synuclein or LRRK2, and PARKIN gene modifications (Dauer and Przedborski 2003; Blesa and Przedborski 2014).

Immune system involvement in PD was demonstrated by the observation of elevated pro-inflammatory mediators (TNFα, IL1β, and IL6) in the CSF and brains of PD patients (Mogi et al. 1994a, b). McGeer et al. (1988) showed for the first time that reactive microglia are evident in the SNpc of human post-mortem tissue, suggesting that neuroinflammation is an additional hallmark of PD. In addition, positron emission tomography (PET) studies have shown that microgliosis is a prominent feature in PD brains (Gerhard et al. 2006; Bartels et al. 2010). The role of microglia in PD was further strengthened by a GWAS study identifying a single nucleotide polymorphism (SNP) in the human leukocyte antigen gene (HLA-DRA), which is expressed by microglia, as a genetic risk factor for late-onset PD (Hamza et al. 2010). Furthermore, the distribution of microglia, with the greatest density in the SN and striatum, leads to the assumption that microglia are involved in the disease development or progression or possibly both. In line with this, peripheral injections of LPS induced microgliosis and reduced tyrosine hydroxylase-positive neurons in the SN, illustrating that activation of microglia can indeed induce neurotoxicity in dopaminergic neurons (Qin et al. 2007; Bodea et al. 2014).

Reactive microglia have also been detected in the SN in toxin-induced as well as transgenic mouse models for PD (Czlonkowska et al. 1996; Sanchez-Guajardo et al. 2010; Hoenen et al. 2016). Furthermore, mice lacking the fractalkine receptor (CX3CR1), which is expressed by microglia and is important for microglia–neuron communication, show extensive neuronal loss in the toxin-induced MPTP PD mouse model (Cardona et al. 2006).

In addition to investigating the role of microglia, there have also been several efforts to study the peripheral immune system in PD. Elevated serum levels of cytokines (IL-6, IL-10, TNFα, and IFNγ) have been reported in peripheral blood from PD patients (Brodacki et al. 2008; Reale et al. 2009). A study by Lampe et al. showed higher expression levels of polymorphic major histocompatibility complex class II molecules, HLA-DR and HLA-DQ, expressed by monocytes in the CSF and peripheral blood of PD patients compared with healthy subjects, indicating that the peripheral immune system is activated in PD (Lampe et al. 2003).

Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene are the most common cause of familial PD and a risk factor for sporadic PD. Lrrk2 has been shown to be implicated in peripheral inflammation and is seen to be relatively abundant in mononuclear cells and macrophages in the periphery (Hakimi et al. 2011). GWAS studies revealed that LRRK2 modifications are associated with autoimmune diseases such as Crohn’s disease, ulcerative colitis, and inflammatory bowel disease (Umeno et al. 2011). Likewise, LRRK2 expression is seen to be increased by pro-inflammatory mediators like IL-1β, TNFα, and IFN-γ (Gardet et al. 2010; Hongge et al. 2015). A study investigating the role of LRRK2 in monocyte dysregulation in PD reported elevated LRRK2 levels in both (CD14dim/CD16+ and CD14++/CD16) monocyte subsets from PD patients in comparison to healthy controls (Bliederhaeuser et al. 2016). The increased LRRK2 expression level positively correlates with the frequency of inflammatory monocytes from PD patients (Cook et al. 2017). In addition, monocytes from PD patients display a distinct transcriptome signature, have an elevated inflammatory profile, and are “hyper-responsive” to LPS stimulation in comparison to healthy controls (Grozdanov et al. 2014). Thus, indicating that there is an underlying inflammatory reaction in the disease.

Similar to the situation in AD, it is still being debated whether these inflammatory monocytes can infiltrate the CNS in the context of PD. A study by Rodriguez et al. (2007) using a chronic toxin-induced MPTP PD model in GFP bone marrow chimeric mice suggests that BMDM enter the brain in their treatment paradigm. Using the same model with only a subchronic dose of MPTP, Depboylu et al. (2012) showed that these infiltrating cells are not involved in the disease development, since resident microglia are the major phagocytes of neuronal cell debris. Another study suggested that deletion of CCR2+ monocytes in the MPTP model does not impact dopamine levels in the striatum (Kalkonde et al. 2007). Parillaud et al. also demonstrated that infiltrating CCR2+ monocytes in the acute MPTP model did not contribute to the dopaminergic neuronal loss (Parillaud et al. 2017). However, it was also demonstrated that BMDM, which were genetically engineered to over-express the neuroprotective glial cell line-derived neurotrophic factor (GDNF) were able to ameliorate MPTP-induced neurodegeneration (Biju et al. 2010), emphasizing the complexity of the immune and CNS involvement.

Taken together, these studies illustrate that microglia and peripheral myeloid cells are implicated in PD pathogenesis. Whether or not blood-borne monocytes infiltrate the CNS in this disease remains to be determined. Considering the evidence that circulating monocytes exhibit a hyper-activate inflammatory phenotype in PD, they may present as an attractive therapeutic target to attenuate the peripheral immune dysregulation in PD progression.

Microglia vs. monocytes in multiple sclerosis

Multiple sclerosis is a complex inflammatory autoimmune disease accompanied by focal plaques and demyelination in white matter brain regions and spinal cord (Symonds 1975; Compston and Coles 2002; Lassmann et al. 2007). MS is also considered in part to be a neurodegenerative disorder as axonal damage, cell loss, and degeneration are common pathological features, especially at late stages of the disease (Haines et al. 2011). The mechanism of MS pathology is elusive but is known to involve complex interactions between systems and cell types including neurons, glia, and immune cells accompanied by permeability of the BBB (Bruck et al. 1995). Supported by the main animal model of MS, the experimental autoimmune encephalomyelitis (EAE), MS was traditionally thought of as a predominantly T-cell-mediated autoimmune disease. Extrapolation of data from EAE studies translating to MS clinical trials has only been partially successful (Gold et al. 2006, Aharoni 2013), which could be in part to the inflammatory process in MS being driven by other immune cells (Cardona et al. 2008, Lassmann et al. 2012, Croxford et al. 2015).

A central role for inflammatory monocytes in disease progression has been demonstrated in EAE mouse models. Myeloid cells, particularly monocytes, infiltrate the CNS in the early stages of EAE development and contribute to the inflammatory response and clinical disease (Ajami et al. 2011; Mikita et al. 2011). Chemokine receptor CCR2 is critical for the accumulation of Ly6Chigh/CCR2high monocytes in the CNS. CCR2 and its ligand CCL2 play an important role in regulating Ly6Chigh/CCR2+ Monocyte infiltration to the CNS and facilitate tissue damage in MS and EAE (Mildner et al. 2007, 2009; King et al. 2009). In EAE, monocytes that are recruited to the CNS are CCR2high/CX3CR1low (Saederup et al. 2010; Garcia et al. 2013), further distinguishing them from resident microglia. Patients with MS display high levels of monocyte-secreted inflammatory molecules in serum compared to healthy individuals, demonstrating a role for peripheral monocytes in disease progression. For example, increased levels of serum TNFα and α have been reported with the onset of MS relapse (Rudick and Ransohoff 1992; Imamura et al. 1993; Huang et al. 2004). The inflammatory profile of monocytes from patients with MS can vary greatly between MS type, disease severity, and gender (Kallaur et al. 2013). Infiltrating monocytes are abundant in active MS lesions and are associated with demyelination (Bruck et al. 1995; Lucchinetti et al. 2000).

This active demyelination and neurodegeneration is also associated with reactive microgliosis (Lassmann 2014). Due to the difficulty in distinguishing monocytes and microglia both morphologically and functionally, the complex roles of microglia vs. monocytes in the inflammatory/degenerative cascade in MS have yet to be fully revealed. However, with the advancement of specific markers and identification of molecular signatures of these cells, significant advancements are being made in this field (Chiu et al. 2013; Hickman et al. 2013; Butovsky et al. 2014). Microglia and macrophages accumulate in active sites of demyelination and neurodegeneration in MS. A recent comprehensive study of microglia and macrophage phenotypic differences used novel cell-specific markers to investigate these phagocytes over different stages of MS (Zrzavy et al. 2017). On one hand, they demonstrate that the homeostatic microglia marker P2Y12 is lost in active MS lesions, but reappears in inactive lesions. On the other hand, they used the marker TMEM119, which is expressed on microglia, but not on infiltrating monocytes, to distinguish phagocytic cells within the active lesion (Butovsky et al. 2014; Bennett et al. 2016; Satoh et al. 2016). With the use of this marker, they demonstrated that the majority of cells associated with active demyelination originate from resident microglia, indicating that microglia play a central role in MS disease progression (Zrzavy et al. 2017).

Earlier studies have described that reactive microglia are present at early and late stages of disease (van der Valk and Amor 2009; Singh et al. 2013). They are present in demyelinating lesions (Ponomarev et al. 2005) and correlate with the axonal and oligodendrocyte pathology (Trapp et al. 1998; Henderson et al. 2009). Experiments using bone marrow chimeric mice showed that microglial activation even occurs before the onset of EAE (Ponomarev et al. 2005). Once microglia are activated, they can damage other CNS cells, in particular oligodendrocytes and neurons (Butovsky et al. 2006a, b; Peferoen et al. 2014). Microglia in MS contain myelin and axonal debris, present higher expression of MHC class II and co-stimulatory molecules, and secrete different inflammatory and neurotoxic mediators in lesions (Boven et al. 2006; Butovsky et al. 2006a, b; Huizinga et al. 2012; Vogel et al. 2013). Depletion of microglia using the herpes simplex virus thymidine kinase (HSVTK) suicide gene attenuated disease severity and demyelination in EAE (Heppner et al. 2005). Similarly, microglia-specific deletion of transforming growth factor (TGF)-β-activated kinase 1 (TAK) with the use of the CX3CR1-CreERT2 mouse model reduced CNS inflammation and myelin damage (Heppner et al. 2005; Ponomarev et al. 2005; Goldmann et al. 2013).

These studies show that microglia can promote disease progression in EAE and MS. Pre-active lesions, which are clusters of activated microglia in the white matter of MS patients, develop without demyelination and peripheral cell infiltration (De Groot et al. 2001) and thus may represent the early stage of inflammatory lesions (van Noort et al. 2011). However, microglia also can be neuroprotective and promote CNS repair by the clearance of inhibitory myelin debris (Miron et al. 2013; Lampron et al. 2015). Furthermore, IL-4- or IFNγ-stimulated microglia promote oligodendrogenesis and neurogenesis (Butovsky et al. 2006a, b). These studies demonstrate the fine balance between neuroprotection and neurotoxicity effects of microglia in EAE.

Homozygous mutations in TREM2, a myeloid cell-surface receptor which is also implicated in AD, lead to demyelination and early onset dementia in humans (Paloneva et al. 2002). In the cuprizone-induced mouse model of demyelination, Trem2 deficiency rendered microglia defective in clearing myelin debris, which resulted in a persistent demyelination (Cantoni et al. 2015; Poliani et al. 2015). Thus, microglial reaction after inflammatory and neurodegenerative signals depends on a fine-tuning of immunomodulatory mediators and microglia phenotypes.

A study using the CCR2RFP–CX3CR1GFP mouse model demonstrated distinct roles for microglia and infiltrating monocytes during EAE. In this model, recruited monocytes are associated with nodes of Ranvier and initiate demyelination, whereas microglia appear to clear debris (Yamasaki et al. 2014). The mechanism of recruitment remains relatively elusive with some studies demonstrating that it is the “activation” of resident microglia that may represent one of the initial steps in EAE pathogenesis, preceding and possibly triggering the infiltration of bloodderived cells (Zrzavy et al. 2017). Yamasaki et al. (2014) present divergent molecular and functional signatures of both cell types, which are maintained during neuroinflammation. These characteristic signatures reflect ontogenic differences between microglia and blood-borne monocytes and strongly argue for cell-specific neurotoxic and protective properties in neuroinflammatory disorders.

Conclusion

Microglia and monocytes have recently been in the spotlight when discussing CNS pathologies. In the healthy body, these cells exist in parallel systems; however, during disease, their paths might cross (King et al. 2009; Shi and Pamer 2011). As described here, both central and peripheral immune responses contribute to the development and progression of many neurodegenerative diseases. As emphasized throughout this review, the complex roles of microglia vs. monocytes in these diseases have yet to be fully revealed due to the difficulty of distinguishing these cell types both morphologically and functionally.

Over the past decade, several tools have been developed to distinguish microglia and peripheral monocytes. As described in the introduction, one widely used tool has been bone marrow chimeras. However, the irradiation needed to generate these mice leads to BBB damage and subsequent monocytes infiltration into the CNS, which would otherwise not occur. This method was improved by introducing head shields to protect the BBB. As an alternative to bone marrow chimeras, parabiosis, which allows for a shared blood circulation between two mice, can be used to distinguish resident microglia and peripheral monocytes (Hashimoto et al. 2013; Wang et al. 2016). Additional tools have been developed to specifically target microglia, such as the Cx3cr1-Cre/ERT mouse model. With these models, however, it has to be kept in mind that peripheral monocytes will be affected too and only the Cre-inducible model, not the constitutive model, can target microglia alone. Recently, the Sall1-Cre mouse has been proposed as an alternative to Cx3cr1-Cre with the advantage of targeting only resident microglia and not peripheral monocytes (Buttgereit et al. 2016).

The description of specific markers and identification of molecular signatures of these cells has helped to generate significant advancements in this field (Chiu et al. 2013; Hickman et al. 2013; Butovsky et al. 2014). Transcriptomic gene assays and computational biology, along with proteomic and epigenomic analysis, have provided unbiased insights into the contextual factors driving authentic, in vivo microglial reaction states (Gosselin et al. 2014; Matcovitch-Natan 2016; Gosselin et al. 2017). These new tools will also be helpful in answering the question of whether peripheral monocytes migrate to the CNS during certain neurodegenerative diseases and to further distinguish the distinct roles of microglia and monocytes. In this context, the fate of infiltrating monocytes also has to be taken into consideration. Either their appearance in the CNS is just transient (Ajami et al. 2011) or they could be reprogrammed by their new environment and become more like microglia, making them harder to distinguish (Gosselin et al. 2014; Lavin et al. 2014; Varvel et al. 2015). Either way, their role in neurodegenerative diseases might be restricted to the short time that they are present in the CNS as a distinct cell type.

Approaches using genetically engineered cells to deliver neuroprotective molecules as demonstrated in a mouse model of PD might be an interesting therapeutic option. Specific targeting and modulation of resident microglia and peripheral myeloid cells in combinatorial manner could potentially alleviate disease progression. Assuming that immune cells infiltrate the CNS, diminishing the immune response in the periphery may have beneficial outcomes and specific targeting of the peripheral immune system may lead to new avenues of therapeutics to rescue CNS homeostasis, restore function, and reduce inflammatory burden. The recent development of cell-specific markers in combination with the modern imaging advances such as PET to distinguish between functionally different resident and infiltrating cells could prove to be highly beneficial. It is important to keep in mind, however, that it is not fully understood whether the immune responses in CNS diseases are detrimental or beneficial. As we have discussed in this review, the microglia and monocyte response could be both neuroprotective and neurotoxic depending on the stage and progression of the disease. There is a fine balancing act between both states, and a microglia and/or monocyte reaction in neurodegenerative diseases may “tip the balance” toward a ‘good’, ‘bad’ or ‘neither’ response. Therefore, modulation of the immune pathways may be more complex than anticipated and timing of therapeutic intervention may be pivotal.

References

  1. Aharoni R (2013) New findings and old controversies in the research of multiple sclerosis and its model experimental autoimmune encephalomyelitis. Expert Rev Clin Immunol 9(5):423–440 [DOI] [PubMed] [Google Scholar]
  2. Ajami B, Bennett JL, Krieger C, Tetzlaff W, Rossi FM (2007) Local self-renewal can sustain CNS microglia maintenance and function throughout adult life. Nat Neurosci 10(12):1538–1543 [DOI] [PubMed] [Google Scholar]
  3. Ajami B, Bennett JL, Krieger C, McNagny KM, Rossi FM (2011) Infiltrating monocytes trigger EAE progression, but do not contribute to the resident microglia pool. Nat Neurosci 14(9):1142–1149 [DOI] [PubMed] [Google Scholar]
  4. Alexianu ME, Kozovska M, Appel SH (2001) Immune reactivity in a mouse model of familial ALS correlates with disease progression. Neurology 57(7):1282–1289 [DOI] [PubMed] [Google Scholar]
  5. Appel SH, Smith RG, Engelhardt JI, Stefani E (1993) Evidence for autoimmunity in amyotrophic lateral sclerosis. J Neurol Sci 118(2):169–174 [DOI] [PubMed] [Google Scholar]
  6. Appel SH, Zhao W, Beers DR, Henkel JS (2011) The microglialmotoneuron dialogue in ALS. Acta Myol 30(1):4–8 [PMC free article] [PubMed] [Google Scholar]
  7. Asai H, Ikezu S, Woodbury ME, Yonemoto GM, Cui L, Ikezu T (2014) Accelerated neurodegeneration and neuroinflammation in transgenic mice expressing P301L tau mutant and tau-tubulin kinase 1. Am J Pathol 184(3):808–818 [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Asai H, Ikezu S, Tsunoda S, Medalla M, Luebke J, Haydar T, Wolozin B, Butovsky O, Kugler S, Ikezu T (2015) Depletion of microglia and inhibition of exosome synthesis halt tau propagation. Nat Neurosci 18(11):1584–1593 [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Atagi Y, Liu CC, Painter MM, Chen XF, Verbeeck C, Zheng H, Li X, Rademakers R, Kang SS, Xu H, Younkin S, Das P, Fryer JD, Bu G (2015) Apolipoprotein E is a ligand for triggering receptor expressed on myeloid cells 2 (TREM2). J Biol Chem 290(43):26043–26050 [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Atanasio A, Decman V, White D, Ramos M, Ikiz B, Lee HC, Siao CJ, Brydges S, LaRosa E, Bai Y, Fury W, Burfeind P, Zamfirova R, Warshaw G, Orengo J, Oyejide A, Fralish M, Auerbach W, Poueymirou W, Freudenberg J, Gong G, Zambrowicz B, Valenzuela D, Yancopoulos G, Murphy A, Thurston G, Lai KM (2016) C9orf72 ablation causes immune dysregulation characterized by leukocyte expansion, autoantibody production, and glomerulonephropathy in mice. Sci Rep 6:23204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Auffray C, Fogg D, Garfa M, Elain G, Join-Lambert O, Kayal S, Sarnacki S, Cumano A, Lauvau G, Geissmann F (2007) Monitoring of blood vessels and tissues by a population of monocytes with patrolling behavior. Science 317(5838):666–670 [DOI] [PubMed] [Google Scholar]
  12. Bartels AL, Willemsen AT, Doorduin J, de Vries EF, Dierckx RA, Leenders KL (2010) [11C]-PK11195 PET: quantification of neuroinflammation and a monitor of anti-inflammatory treatment in Parkinson’s disease? Parkinsonism Relat Disord 16(1):57–59 [DOI] [PubMed] [Google Scholar]
  13. Bennett ML, Bennett FC, Liddelow SA, Ajami B, Zamanian JL, Fernhoff NB, Mulinyawe SB, Bohlen CJ, Adil A, Tucker A, Weissman IL, Chang EF, Li G, Grant GA, Hayden Gephart MG, Barres BA (2016) New tools for studying microglia in the mouse and human CNS. Proc Natl Acad Sci USA 113(12):E1738–1746 [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Biju K, Zhou Q, Li G, Imam SZ, Roberts JL, Morgan WW, Clark RA, Li S (2010) Macrophage-mediated GDNF delivery protects against dopaminergic neurodegeneration: a therapeutic strategy for Parkinson’s disease. Mol Ther 18(8):1536–1544 [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Blesa J, Przedborski S (2014) Parkinson’s disease: animal models and dopaminergic cell vulnerability. Front Neuroanat 8:155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Bliederhaeuser C, Zondler L, Grozdanov V, Ruf WP, Brenner D, Melrose HL, Bauer P, Ludolph AC, Gillardon F, Kassubek J, Weishaupt JH, Danzer KM (2016) LRRK2 contributes to monocyte dysregulation in Parkinson’s disease. Acta Neuropathol Commun 4(1):123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Bodea LG, Wang Y, Linnartz-Gerlach B, Kopatz J, Sinkkonen L, Musgrove R, Kaoma T, Muller A, Vallar L, Di Monte DA, Balling R, Neumann H (2014) Neurodegeneration by activation of the microglial complement-phagosome pathway. J Neurosci 34(25):8546–8556 [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Boillee S, Yamanaka K, Lobsiger CS, Copeland NG, Jenkins NA, Kassiotis G, Kollias G, Cleveland DW (2006) Onset and progression in inherited ALS determined by motor neurons and microglia. Science 312(5778):1389–1392 [DOI] [PubMed] [Google Scholar]
  19. Boven LA, Van Meurs M, Van Zwam M, Wierenga-Wolf A, Hintzen RQ, Boot RG, Aerts JM, Amor S, Nieuwenhuis EE, Laman JD (2006) Myelin-laden macrophages are anti-inflammatory, consistent with foam cells in multiple sclerosis. Brain 129(Pt2):517–526 [DOI] [PubMed] [Google Scholar]
  20. Bradshaw EM, Chibnik LB, Keenan BT, Ottoboni L, Raj T, Tang A, Rosenkrantz LL, Imboywa S, Lee M, Von Korff A, I. Alzheimer Disease Neuroimaging, Morris MC, Evans DA, Johnson K, Sperling RA, Schneider JA, Bennett DA, De Jager PL (2013a) CD33 Alzheimer’s disease locus: altered monocyte function and amyloid biology. Nat Neurosci 16(7):848–850 [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Bradshaw EM, Chibnik LB, Keenan BT, Ottoboni L, Raj T, Tang A, Rosenkrantz LL, Imboywa S, Lee M, Von Korff A, Morris MC, Evans DA, Johnson K, Sperling RA, Schneider JA, Bennett DA, De Jager PL, Jager PLD (2013b) CD33 Alzheimer’s disease locus: altered monocyte function and amyloid biology. Nat Neurosci 16:848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Brodacki B, Staszewski J, Toczylowska B, Kozlowska E, Drela N, Chalimoniuk M, Stepien A (2008) Serum interleukin (IL-2, IL-10, IL-6, IL-4), TNFalpha, and INFgamma concentrations are elevated in patients with atypical and idiopathic parkinsonism. Neurosci Lett 441(2):158–162 [DOI] [PubMed] [Google Scholar]
  23. Brosseron F, Krauthausen M, Kummer M, Heneka MT (2014) Body fluid cytokine levels in mild cognitive impairment and Alzheimer’s disease: a comparative overview. Mol Neurobiol 50:534–544 [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Bruck W, Porada P, Poser S, Rieckmann P, Hanefeld F, Kretzschmar HA, Lassmann H (1995) Monocyte/macrophage differentiation in early multiple sclerosis lesions. Ann Neurol 38(5):788–796 [DOI] [PubMed] [Google Scholar]
  25. Burberry A, Suzuki N, Wang JY, Moccia R, Mordes DA, Stewart MH, Suzuki-Uematsu S, Ghosh S, Singh A, Merkle FT, Koszka K, Li QZ, Zon L, Rossi DJ, Trowbridge JJ, Notarangelo LD, Eggan K (2016) Loss-of-function mutations in the C9ORF72 mouse ortholog cause fatal autoimmune disease. Sci Transl Med 8(347):347ra393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Butovsky O, Landa G, Kunis G, Ziv Y, Avidan H, Greenberg N, Schwartz A, Smirnov I, Pollack A, Jung S, Schwartz M (2006a) Induction and blockage of oligodendrogenesis by differently activated microglia in an animal model of multiple sclerosis. J Clin Investig 116(4):905–915 [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Butovsky O, Ziv Y, Schwartz A, Landa G, Talpalar AE, Pluchino S, Martino G, Schwartz M (2006b) Microglia activated by IL-4 or IFN-gamma differentially induce neurogenesis and oligodendrogenesis from adult stem/progenitor cells. Mol Cell Neurosci 31(1):149–160 [DOI] [PubMed] [Google Scholar]
  28. Butovsky O, Siddiqui S, Gabriely G, Lanser AJ, Dake B, Murugaiyan G, Doykan CE, Wu PM, Gali RR, Iyer LK, Lawson R, Berry J, Krichevsky AM, Cudkowicz ME, Weiner HL (2012) Modulating inflammatory monocytes with a unique microRNA gene signature ameliorates murine ALS. J Clin Investig 122(9):3063–3087 [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Butovsky O, Jedrychowski MP, Moore CS, Cialic R, Lanser AJ, Gabriely G, Koeglsperger T, Dake B, Wu PM, Doykan CE, Fanek Z, Liu L, Chen Z, Rothstein JD, Ransohoff RM, Gygi SP, Antel JP, Weiner HL (2014) Identification of a unique TGF-betadependent molecular and functional signature in microglia. Nat Neurosci 17(1):131–143 [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Butovsky O, Jedrychowski MP, Cialic R, Krasemann S, Murugaiyan G, Fanek Z, Greco DJ, Wu PM, Doykan CE, Kiner O, Lawson RJ, Frosch MP, Pochet N, Fatimy RE, Krichevsky AM, Gygi SP, Lassmann H, Berry J, Cudkowicz ME, Weiner HL (2015) Targeting miR-155 restores abnormal microglia and attenuates disease in SOD1 mice. Ann Neurol 77(1):75–99 [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Buttgereit A, Lelios I, Yu X, Vrohlings M, Krakoski NR, Gautier EL, Nishinakamura R, Becher B, Greter M (2016) Sall1 is a transcriptional regulator defining microglia identity and function. Nat Immunol 17(12):1397–1406 [DOI] [PubMed] [Google Scholar]
  32. Cantoni C, Bollman B, Licastro D, Xie M, Mikesell R, Schmidt R, Yuede CM, Galimberti D, Olivecrona G, Klein RS, Cross AH, Otero K, Piccio L (2015) TREM2 regulates microglial cell activation in response to demyelination in vivo. Acta Neuropathol 129(3):429–447 [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Cardona AE, Pioro EP, Sasse ME, Kostenko V, Cardona SM, Dijkstra IM, Huang D, Kidd G, Dombrowski S, Dutta R, Lee JC, Cook DN, Jung S, Lira SA, Littman DR, Ransohoff RM (2006) Control of microglial neurotoxicity by the fractalkine receptor. Nat Neurosci 9(7):917–924 [DOI] [PubMed] [Google Scholar]
  34. Cardona AE, Sasse ME, Liu L, Cardona SM, Mizutani M, Savarin C, Hu T, Ransohoff RM (2008) Scavenging roles of chemokine receptors: chemokine receptor deficiency is associated with increased levels of ligand in circulation and tissues. Blood 112(2):256–263 [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Carlin LM, Stamatiades EG, Auffray C, Hanna RN, Glover L, Vizcay-Barrena G, Hedrick CC, Cook HT, Diebold S, Geissmann F (2013) Nr4a1-dependent Ly6C(low) monocytes monitor endothelial cells and orchestrate their disposal. Cell 153(2):362–375 [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Carvey PM, Hendey B, Monahan AJ (2009) The blood–brain barrier in neurodegenerative disease: a rhetorical perspective. J Neurochem 111(2):291–314 [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Chakrabarty P, Jansen-West K, Beccard A, Ceballos-Diaz C, Levites Y, Verbeeck C, Zubair AC, Dickson D, Golde TE, Das P (2010) Massive gliosis induced by interleukin-6 suppresses Abeta deposition in vivo: evidence against inflammation as a driving force for amyloid deposition. FASEB J 24(2):548–559 [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Chan G, White CC, Winn PA, Cimpean M, Replogle JM, Glick LR, Cuerdon NE, Ryan KJ, Johnson KA, Schneider JA, Bennett DA, Chibnik LB, Sperling RA, Bradshaw EM, De Jager PL (2015) CD33 modulates TREM2: convergence of Alzheimer loci. Nat Neurosci 18(11):1556–1558 [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Chaudhuri A (2013) Multiple sclerosis is primarily a neurodegenerative disease. J Neural Transm (Vienna) 120(10):1463–1466 [DOI] [PubMed] [Google Scholar]
  40. Chiu IM, Phatnani H, Kuligowski M, Tapia JC, Carrasco MA, Zhang M, Maniatis T, Carroll MC (2009) Activation of innate and humoral immunity in the peripheral nervous system of ALS transgenic mice. Proc Natl Acad Sci USA 106(49):20960–20965 [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Chiu IM, Morimoto ET, Goodarzi H, Liao JT, O’Keeffe S, Phatnani HP, Muratet M, Carroll MC, Levy S, Tavazoie S, Myers RM, Maniatis T (2013) A neurodegeneration-specific gene-expression signature of acutely isolated microglia from an amyotrophic lateral sclerosis mouse model. Cell Rep 4(2):385–401 [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Clement AM, Nguyen MD, Roberts EA, Garcia ML, Boillee S, Rule M, McMahon AP, Doucette W, Siwek D, Ferrante RJ, Brown RH Jr, Julien JP, Goldstein LS, Cleveland DW (2003) Wild-type nonneuronal cells extend survival of SOD1 mutant motor neurons in ALS mice. Science 302(5642):113–117 [DOI] [PubMed] [Google Scholar]
  43. Compston A, Coles A (2002) Multiple sclerosis. Lancet 359(9313):1221–1231 [DOI] [PubMed] [Google Scholar]
  44. Cook DA, Kannarkat GT, Cintron AF, Butkovich LM, Fraser KB, Chang J, Grigoryan N, Factor SA, West AB, Boss JM, Tansey MG (2017) LRRK2 levels in immune cells are increased in Parkinson’s disease. NPJ Parkinsons Dis 3:11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Corder EH, Saunders AM, Strittmatter WJ, Schmechel DE, Gaskell PC, Small GW, Roses AD, Haines JL, Pericak-Vance MA (1993) Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset families. Science 261(5123):921–923 [DOI] [PubMed] [Google Scholar]
  46. Croxford AL, Lanzinger M, Hartmann FJ, Schreiner B, Mair F, Pelczar P, Clausen BE, Jung S, Greter M, Becher B (2015) The cytokine GM-CSF drives the inflammatory signature of CCR2 + monocytes and licenses autoimmunity. Immunity 43(3):502–514 [DOI] [PubMed] [Google Scholar]
  47. Czlonkowska A, Kohutnicka M, Kurkowska-Jastrzebska I, Czlonkowski A (1996) Microglial reaction in MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) induced Parkinson’s disease mice model. Neurodegeneration 5(2):137–143 [DOI] [PubMed] [Google Scholar]
  48. Dagher NN, Najafi AR, Kayala KM, Elmore MR, White TE, Medeiros R, West BL, Green KN (2015) Colony-stimulating factor 1 receptor inhibition prevents microglial plaque association and improves cognition in 3xTg-AD mice. J Neuroinflammation 12:139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Dauer W, Przedborski S (2003) Parkinson’s disease: mechanisms and models. Neuron 39(6):889–909 [DOI] [PubMed] [Google Scholar]
  50. De Groot CJ, Bergers E, Kamphorst W, Ravid R, Polman CH, Barkhof F, van der Valk P (2001) Post-mortem MRI-guided sampling of multiple sclerosis brain lesions: increased yield of active demyelinating and (p)reactive lesions. Brain 124(Pt8):1635–1645 [DOI] [PubMed] [Google Scholar]
  51. DeJesus-Hernandez M, Mackenzie IR, Boeve BF, Boxer AL, Baker M, Rutherford NJ, Nicholson AM, Finch NA, Flynn H, Adamson J, Kouri N, Wojtas A, Sengdy P, Hsiung GY, Karydas A, Seeley WW, Josephs KA, Coppola G, Geschwind DH, Wszolek ZK, Feldman H, Knopman DS, Petersen RC, Miller BL, Dickson DW, Boylan KB, Graff-Radford NR, Rademakers R (2011) Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron 72(2):245–256 [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Depboylu C, Stricker S, Ghobril JP, Oertel WH, Priller J, Hoglinger GU (2012) Brain-resident microglia predominate over infiltrating myeloid cells in activation, phagocytosis and interaction with T-lymphocytes in the MPTP mouse model of Parkinson disease. Exp Neurol 238(2):183–191 [DOI] [PubMed] [Google Scholar]
  53. Elliott JL (2001) Cytokine upregulation in a murine model of familial amyotrophic lateral sclerosis. Brain Res Mol Brain Res 95(1–2):172–178 [DOI] [PubMed] [Google Scholar]
  54. Engelhardt JI, Appel SH (1990) IgG reactivity in the spinal cord and motor cortex in amyotrophic lateral sclerosis. Arch Neurol 47(11):1210–1216 [DOI] [PubMed] [Google Scholar]
  55. Engelhardt JI, Tajti J, Appel SH (1993) Lymphocytic infiltrates in the spinal cord in amyotrophic lateral sclerosis. Arch Neurol 50(1):30–36 [DOI] [PubMed] [Google Scholar]
  56. Evans MC, Couch Y, Sibson N, Turner MR (2013) Inflammation and neurovascular changes in amyotrophic lateral sclerosis. Mol Cell Neurosci 53:34–41 [DOI] [PubMed] [Google Scholar]
  57. Ferguson B, Matyszak MK, Esiri MM, Perry VH (1997) Axonal damage in acute multiple sclerosis lesions. Brain 120(Pt3):393–399 [DOI] [PubMed] [Google Scholar]
  58. Ford AL, Goodsall AL, Hickey WF, Sedgwick JD (1995) Normal adult ramified microglia separated from other central nervous system macrophages by flow cytometric sorting. Phenotypic differences defined and direct ex vivo antigen presentation to myelin basic protein-reactive CD4+ T cells compared. J Immunol 154(9):4309–4321 [PubMed] [Google Scholar]
  59. Frakes AE, Ferraiuolo L, Haidet-Phillips AM, Schmelzer L, Braun L, Miranda CJ, Ladner KJ, Bevan AK, Foust KD, Godbout JP, Popovich PG, Guttridge DC, Kaspar BK (2014) Microglia induce motor neuron death via the classical NF-kappaB pathway in amyotrophic lateral sclerosis. Neuron 81(5):1009–1023 [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Garcia JA, Pino PA, Mizutani M, Cardona SM, Charo IF, Ransohoff RM, Forsthuber TG, Cardona AE (2013) Regulation of adaptive immunity by the fractalkine receptor during autoimmune inflammation. J Immunol 191(3):1063–1072 [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Gardet A, Benita Y, Li C, Sands BE, Ballester I, Stevens C, Korzenik JR, Rioux JD, Daly MJ, Xavier RJ, Podolsky DK (2010) LRRK2 is involved in the IFN-gamma response and host response to pathogens. J Immunol 185(9):5577–5585 [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Gautier EL, Shay T, Miller J, Greter M, Jakubzick C, Ivanov S, Helft J, Chow A, Elpek KG, Gordonov S, Mazloom AR, Ma’ayan A, Chua WJ, Hansen TH, Turley SJ, Merad M, Randolph GJ, Immunological Genome C (2012) Gene-expression profiles and transcriptional regulatory pathways that underlie the identity and diversity of mouse tissue macrophages. Nat Immunol 13(11):1118–1128 [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Geissmann F, Jung S, Littman DR (2003) Blood monocytes consist of two principal subsets with distinct migratory properties. Immunity 19(1):71–82 [DOI] [PubMed] [Google Scholar]
  64. Geissmann F, Auffray C, Palframan R, Wirrig C, Ciocca A, Campisi L, Narni-Mancinelli E, Lauvau G (2008) Blood monocytes: distinct subsets, how they relate to dendritic cells, and their possible roles in the regulation of T-cell responses. Immunol Cell Biol 86(5):398–408 [DOI] [PubMed] [Google Scholar]
  65. Geissmann F, Manz MG, Jung S, Sieweke MH, Merad M, Ley K (2010) Development of monocytes, macrophages, and dendritic cells. Science 327(5966):656–661 [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Gerber YN, Sabourin JC, Rabano M, Vivanco M, Perrin FE (2012) Early functional deficit and microglial disturbances in a mouse model of amyotrophic lateral sclerosis. PLoS One 7(4):e36000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Gerhard A, Pavese N, Hotton G, Turkheimer F, Es M, Hammers A, Eggert K, Oertel W, Banati RB, Brooks DJ (2006) In vivo imaging of microglial activation with [11C](R)-PK11195 PET in idiopathic Parkinson’s disease. Neurobiol Dis 21(2):404–412 [DOI] [PubMed] [Google Scholar]
  68. Ginhoux F, Greter M, Leboeuf M, Nandi S, See P, Gokhan S, Mehler MF, Conway SJ, Ng LG, Stanley ER, Samokhvalov IM, Merad M (2010) Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science 330(6005):841–845 [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Glass CK, Saijo K, Winner B, Marchetto MC, Gage FH (2010) Mechanisms underlying inflammation in neurodegeneration. Cell 140(6):918–934 [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Goate A, Chartier-Harlin MC, Mullan M, Brown J, Crawford F, Fidani L, Giuffra L, Haynes A, Irving N, James L et al. (1991) Segregation of a missense mutation in the amyloid precursor protein gene with familial Alzheimer’s disease. Nature 349(6311):704–706 [DOI] [PubMed] [Google Scholar]
  71. Gold R, Linington C, Lassmann H (2006) Understanding pathogenesis and therapy of multiple sclerosis via animal models: 70 years of merits and culprits in experimental autoimmune encephalomyelitis research. Brain 129(Pt 8):1953–1971 [DOI] [PubMed] [Google Scholar]
  72. Goldmann T, Wieghofer P, Muller PF, Wolf Y, Varol D, Yona S, Brendecke SM, Kierdorf K, Staszewski O, Datta M, Luedde T, Heikenwalder M, Jung S, Prinz M (2013) A new type of microglia gene targeting shows TAK1 to be pivotal in CNS autoimmune inflammation. Nat Neurosci 16(11):1618–1626 [DOI] [PubMed] [Google Scholar]
  73. Gordon S, Taylor PR (2005) Monocyte and macrophage heterogeneity. Nat Rev Immunol 5(12):953–964 [DOI] [PubMed] [Google Scholar]
  74. Gosselin D, Link VM, Romanoski CE, Fonseca GJ, Eichenfield DZ, Spann NJ, Stender JD, Chun HB, Garner H, Geissmann F, Glass CK (2014) Environment drives selection and function of enhancers controlling tissue-specific macrophage identities. Cell 159(6):1327–1340 [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Gosselin D, Skola D, Coufal NG, Holtman IR, Schlachetzki JCM, Sajti E, Jaeger BN, O’Connor C, Fitzpatrick C, Pasillas MP, Pena M, Adair A, Gonda DD, Levy ML, Ransohoff RM, Gage FH, Glass CK (2017) An environment-dependent transcriptional network specifies human microglia identity. Science 356(6344):eaal3222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Gowing G, Lalancette-Hebert M, Audet JN, Dequen F, Julien JP (2009) Macrophage colony stimulating factor (M-CSF) exacerbates ALS disease in a mouse model through altered responses of microglia expressing mutant superoxide dismutase. Exp Neurol 220(2):267–275 [DOI] [PubMed] [Google Scholar]
  77. Graber DJ, Hickey WF, Harris BT (2010) Progressive changes in microglia and macrophages in spinal cord and peripheral nerve in the transgenic rat model of amyotrophic lateral sclerosis. J Neuroinflammation 7:8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Grathwohl SA, Kalin RE, Bolmont T, Prokop S, Winkelmann G, Kaeser SA, Odenthal J, Radde R, Eldh T, Gandy S, Aguzzi A, Staufenbiel M, Mathews PM, Wolburg H, Heppner FL, Jucker M (2009) Formation and maintenance of Alzheimer’s disease betaamyloid plaques in the absence of microglia. Nat Neurosci 12(11):1361–1363 [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Gravel M, Beland LC, Soucy G, Abdelhamid E, Rahimian R, Gravel C, Kriz J (2016) IL-10 controls early microglial phenotypes and disease onset in ALS caused by misfolded superoxide dismutase1. J Neurosci 36(3):1031–1048 [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Greenhalgh AD, Passos Dos Santos R, Zarruk JG, Salmon CK, Kroner A, David S (2016) Arginase-1 is expressed exclusively by infiltrating myeloid cells in CNS injury and disease. Brain Behav Immun 56:61–67 [DOI] [PubMed] [Google Scholar]
  81. Greter M, Lelios I, Croxford AL (2015) Microglia versus myeloid cell nomenclature during brain inflammation. Front Immunol 6:249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Griciuc A, Serrano-Pozo A, Parrado AR, Lesinski AN, Asselin CN, Mullin K, Hooli B, Choi SH, Hyman BT, Tanzi RE (2013) Alzheimer’s disease risk gene CD33 inhibits microglial uptake of amyloid beta. Neuron 78(4):631–643 [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Grozdanov V, Bliederhaeuser C, Ruf WP, Roth V, Fundel-Clemens K, Zondler L, Brenner D, Martin-Villalba A, Hengerer B, Kassubek J, Ludolph AC, Weishaupt JH, Danzer KM (2014) Inflammatory dysregulation of blood monocytes in Parkinson’s disease patients. Acta Neuropathol 128(5):651–663 [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Guerreiro R, Wojtas A, Bras J, Carrasquillo M, Rogaeva E, Majounie E, Cruchaga C, Sassi C, Kauwe JSK, Younkin S, Hazrati L, Collinge J, Pocock J, Lashley T, Williams J, Lambert Amouyel P, Goate A, Rademakers R, Morgan K, Powell J, St George-Hyslop Singleton A, Hardy J (2013) TREM2 variants Alzheimer’s disease. N Engl J Med 368:117–127 [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Guillot-Sestier MV, Doty KR, Gate D, Rodriguez J Jr, Leung RezaiZadeh K, Town T (2015) Il10 deficiency rebalances innate immunity to mitigate Alzheimer-like pathology. Neuron 85(3):534–548 [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Haines JD, Inglese M, Casaccia P (2011) Axonal damage in multiple sclerosis. Mt Sinai J Med 78(2):231–243 [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Hakimi M, Selvanantham T, Swinton E, Padmore RF, Tong Kabbach G, Venderova K, Girardin SE, Bulman DE, Scherzer CR, LaVoie MJ, Gris D, Park DS, Angel JB, Shen J, Philpott Schlossmacher MG (2011) Parkinson’s disease-linked LRRK2 expressed in circulating and tissue immune cells and upregulated following recognition of microbial structures. J Neural Transm (Vienna) 118(5):795–808 [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Hall ED, Oostveen JA, Gurney ME (1998) Relationship of microglial and astrocytic activation to disease onset and progression in transgenic model of familial ALS. Glia 23(3):249–256 [DOI] [PubMed] [Google Scholar]
  89. Hamza TH, Zabetian CP, Tenesa A, Laederach A, Montimurro Yearout D, Kay DM, Doheny KF, Paschall J, Pugh E, Kusel Collura R, Roberts J, Griffith A, Samii A, Scott WK, Nutt Factor SA, Payami H (2010) Common genetic variation in HLA region is associated with late-onset sporadic Parkinson’s disease. Nat Genet 42(9):781–785 [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Harraz MM, Marden JJ, Zhou W, Zhang Y, Williams A, Sharov Nelson K, Luo M, Paulson H, Schoneich C, Engelhardt(2008) SOD1 mutations disrupt redox-sensitive Rac regulation of NADPH oxidase in a familial ALS model. J Clin Investig 118(2):659–670 [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Hashimoto D, Chow A, Noizat C, Teo P, Beasley MB, Leboeuf Becker CD, See P, Price J, Lucas D, Greter M, Mortha A, Boyer SW, Forsberg EC, Tanaka M, van Rooijen N, Garcia-Sastre Stanley ER, Ginhoux F, Frenette PS, Merad M (2013) Tissue-resident macrophages self-maintain locally throughout adult with minimal contribution from circulating monocytes. Immunity 38(4):792–804 [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Haynes SE, Hollopeter G, Yang G, Kurpius D, Dailey ME, Gan Julius D (2006) The P2Y12 receptor regulates microglial activation by extracellular nucleotides. Nat Neurosci 9(12):1512–1519 [DOI] [PubMed] [Google Scholar]
  93. Henderson AP, Barnett MH, Parratt JD, Prineas JW (2009) Multiple sclerosis: distribution of inflammatory cells in newly forming lesions. Ann Neurol 66(6):739–753 [DOI] [PubMed] [Google Scholar]
  94. Heneka MT, Kummer MP, Stutz A, Delekate A, Schwartz S, Vieira-Saecker A, Griep A, Axt D, Remus A, Tzeng TC, Gelpi E, A, Korte M, Latz E, Golenbock DT (2013) NLRP3 is activated in Alzheimer’s disease and contributes to pathology in APP/PS1 mice. Nature 493(7434):674–678 [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Henkel JS, Engelhardt JI, Siklos L, Simpson EP, Kim SH, Pan Goodman JC, Siddique T, Beers DR, Appel SH (2004) Presence of dendritic cells, MCP-1, and activated microglia/macrophages in amyotrophic lateral sclerosis spinal cord tissue. Ann Neurol 55(2):221–235 [DOI] [PubMed] [Google Scholar]
  96. Heppner FL, Greter M, Marino D, Falsig J, Raivich G, Hovelmeyer N, Waisman A, Rulicke T, Prinz M, Priller J, Becher B, Aguzzi A (2005) Experimental autoimmune encephalomyelitis repressed by microglial paralysis. Nat Med 11(2):146–152 [DOI] [PubMed] [Google Scholar]
  97. Heppner FL, Ransohoff RM, Becher B (2015) Immune attack: role of inflammation in Alzheimer disease. Nat Rev Neurosci 16(6):358–372 [DOI] [PubMed] [Google Scholar]
  98. Hickman SE, Kingery ND, Ohsumi TK, Borowsky ML, Wang Means TK, El Khoury J (2013) The microglial sensome revealed by direct RNA sequencing. Nat Neurosci 16(12):1896–1905 [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Hoenen C, Gustin A, Birck C, Kirchmeyer M, Beaume N, Felten P, Grandbarbe L, Heuschling P, Heurtaux T (2016) Alphasynuclein proteins promote pro-inflammatory cascades in microglia: stronger effects of the A53T mutant. PLoS One 11(9):e0162717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Hohsfield LA, Humpel C (2015) Intravenous infusion of monocytes isolated from 2-week-old mice enhances clearance of betaamyloid plaques in an Alzheimer mouse model. PLoS One 10:1–18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Holtman IR, Raj DD, Miller JA, Schaafsma W, Yin Z, Brouwer N, Wes PD, Moller T, Orre M, Kamphuis W, Hol EM, Boddeke EW, Eggen BJ (2015) Induction of a common microglia gene expression signature by aging and neurodegenerative conditions: a co-expression meta-analysis. Acta Neuropathol Commun 3:31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Hong S, Beja-Glasser VF, Nfonoyim BM, Frouin A, Li S, Ramakrishnan S, Merry KM, Shi Q, Rosenthal A, Barres BA, Lemere CA, Selkoe DJ, Stevens B (2016) Complement and microglia mediate early synapse loss in Alzheimer mouse models. Science 352(6286):712–716 [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Hongge L, Kexin G, Xiaojie M, Nian X, Jinsha H (2015) The role of LRRK2 in the regulation of monocyte adhesion to endothelial cells. J Mol Neurosci 55(1):233–239 [DOI] [PubMed] [Google Scholar]
  104. Hooten KG, Beers DR, Zhao W, Appel SH (2015) Protective and toxic neuroinflammation in amyotrophic lateral sclerosis. Neurotherapeutics 12(2):364–375 [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Hornykiewicz O, Kish SJ (1987) Biochemical pathophysiology of Parkinson’s disease. Adv Neurol 45:19–34 [PubMed] [Google Scholar]
  106. Huang WX, Huang P, Hillert J (2004) Increased expression of caspase-1 and interleukin-18 in peripheral blood mononuclear cells in patients with multiple sclerosis. Mult Scler 10(5):482–487 [DOI] [PubMed] [Google Scholar]
  107. Huizinga R, van der Star BJ, Kipp M, Jong R, Gerritsen W, Clarner T, Puentes F, Dijkstra CD, van der Valk P, Amor S (2012) Phagocytosis of neuronal debris by microglia is associated with neuronal damage in multiple sclerosis. Glia 60(3):422–431 [DOI] [PubMed] [Google Scholar]
  108. Imamura K, Suzumura A, Hayashi F, Marunouchi T (1993) Cytokine production by peripheral blood monocytes/macrophages in multiple sclerosis patients. Acta Neurol Scand 87(4):281–285 [DOI] [PubMed] [Google Scholar]
  109. Ingersoll MA, Spanbroek R, Lottaz C, Gautier EL, Frankenberger M, Hoffmann R, Lang R, Haniffa M, Collin M, Tacke F, Habenicht AJ, Ziegler-Heitbrock L, Randolph GJ (2010) Comparison of gene expression profiles between human and mouse monocyte subsets. Blood 115(3):e10–19 [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Jay TR, Miller CM, Cheng PJ, Graham LC, Bemiller S, Broihier ML, Xu G, Margevicius D, Karlo JC, Sousa GL, Cotleur AC, Butovsky O, Bekris L, Staugaitis SM, Leverenz JB, Pimplikar SW, Landreth GE, Howell GR, Ransohoff RM, Lamb BT (2015) TREM2 deficiency eliminates TREM2+ inflammatory macrophages and ameliorates pathology in Alzheimer’s disease mouse models. J Exp Med 212(3):287–295 [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Jay TR, Hirsch AM, Broihier ML, Miller CM, Neilson LE, Ransohoff RM, Lamb BT, Landreth GE (2017) Disease progressiondependent effects of TREM2 deficiency in a mouse model of Alzheimer’s disease. J Neurosci 37(3):637–647 [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Jonsson T, Stefansson H, Steinberg S, Jonsdottir I, Jonsson PV, Snaedal J, Bjornsson S, Huttenlocher J, Levey AI, Lah JJ, Rujescu D, Hampel H, Giegling I, Andreassen OA, Engedal K, Ulstein I, Djurovic S, Ibrahim-Verbaas C, Hofman A, Ikram MA, van Duijn CM, Thorsteinsdottir U, Kong A, Stefansson K (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]
  113. Jung S, Aliberti J, Graemmel P, Sunshine MJ, Kreutzberg GW, Sher A, Littman DR (2000) Analysis of fractalkine receptor CX(3)CR1 function by targeted deletion and green fluorescent protein reporter gene insertion. Mol Cell Biol 20(11):4106–4114 [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Kalkonde YV, Morgan WW, Sigala J, Maffi SK, Condello C, Kuziel W, Ahuja SS, Ahuja SK (2007) Chemokines in the MPTP model of Parkinson’s disease: absence of CCL2 and its receptor CCR2 does not protect against striatal neurodegeneration. Brain Res 1128(1):1–11 [DOI] [PubMed] [Google Scholar]
  115. Kallaur AP, Oliveira SR, Colado Simao AN, Delicatode Almeida ER, Kaminami Morimoto H, Lopes J, de Carvalho Jennings Pereira WL, Marques Andrade R, Muliterno Pelegrino L, Donizete Borelli S, Kaimen-Maciel DR, Reiche EM (2013) Cytokine profile in relapsing-remitting multiple sclerosis patients and the association between progression and activity of the disease. Mol Med Rep 7(3):1010–1020 [DOI] [PubMed] [Google Scholar]
  116. Kawachi I, Lassmann H (2017) Neurodegeneration in multiple sclerosis and neuromyelitis optica. J Neurol Neurosurg Psychiatry 88(2):137–145 [DOI] [PubMed] [Google Scholar]
  117. Kawamata T, Akiyama H, Yamada T, McGeer PL (1992) Immunologic reactions in amyotrophic lateral sclerosis brain and spinal cord tissue. Am J Pathol 140(3):691–707 [PMC free article] [PubMed] [Google Scholar]
  118. Keren-Shaul H, Spinrad A, Weiner A, Matcovitch-Natan O, Dvir-Szternfeld R, Ulland TK, David E, Baruch K, Lara-Astaiso D, Toth B, Itzkovitz S, Colonna M, Schwartz M, Amit I (2017) A unique microglia type associated with restricting development of Alzheimer’s disease. Cell 169(7):1276–1290 e1217 [DOI] [PubMed] [Google Scholar]
  119. King IL, Dickendesher TL, Segal BM (2009) Circulating Ly-6C+ myeloid precursors migrate to the CNS and play a pathogenic role during autoimmune demyelinating disease. Blood 113(14):3190–3197 [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Kobayashi K, Imagama S, Ohgomori T, Hirano K, Uchimura K, Sakamoto K, Hirakawa A, Takeuchi H, Suzumura A, Ishiguro N, Kadomatsu K (2013) Minocycline selectively inhibits M1 polarization of microglia. Cell Death Dis 4:e525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Krabbe G, Halle A, Matyash V, Rinnenthal JL, Eom GD, Bernhardt U, Miller KR, Prokop S, Kettenmann H, Heppner FL (2013) Functional impairment of microglia coincides with Beta-amyloid deposition in mice with Alzheimer-like pathology. PLoS One 8(4):e60921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Krasemann S, Madore C, Cialic R, Baufeld C, Calcagno N, El Fatimy R, O’Loughlin E, Xu Y, Fanek Z, Greco DJ, Smith ST, Tweet G, Conde-Sanroman P, Garcias M, Weng Z, Chen H, Tjon E, Mazaheri F, Zrzavy T, Hartman K, Glatzel M, Worthmann A, Lemere CA, Ikezu T, Heppner FL, Lassmann H, Weiner HL, Litvak V, Ochando J, Haass C, Butovsky O (2017) The TREM2-APOE pathway drives the transcriptional phenotype of dysfunctional microglia in neurodegenerative diseases. Immunity 47:566–581 [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Lampe JB, Gossrau G, Kempe A, Fussel M, Schwurack K, Schroder R, Krause S, Kohnen R, Walter MC, Reichmann H, Lochmuller H (2003) Analysis of HLA class I and II alleles in sporadic inclusion-body myositis. J Neurol 250(11):1313–1317 [DOI] [PubMed] [Google Scholar]
  124. Lampron A, Larochelle A, Laflamme N, Prefontaine P, Plante MM, Sanchez MG, Yong VW, Stys PK, Tremblay ME, Rivest S (2015) Inefficient clearance of myelin debris by microglia impairs remyelinating processes. J Exp Med 212(4):481–495 [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Lassmann H (2014) Mechanisms of white matter damage in multiple sclerosis. Glia 62(11):1816–1830 [DOI] [PubMed] [Google Scholar]
  126. Lassmann H, Bruck W, Lucchinetti CF (2007) The immunopathology of multiple sclerosis: an overview. Brain Pathol 17(2):210–218 [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Lassmann H, van Horssen J, Mahad D (2012) Progressive multiple sclerosis: pathology and pathogenesis. Nat Rev Neurol 8(11):647–656 [DOI] [PubMed] [Google Scholar]
  128. Lavin Y, Winter D, Blecher-Gonen R, David E, Keren-Shaul H, Merad M, Jung S, Amit I (2014) Tissue-resident macrophage enhancer landscapes are shaped by the local microenvironment. Cell 159(6):1312–1326 [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Lebson L, Nash K, Kamath S, Herber D, Carty N, Lee DC, Li Q, Szekeres K, Jinwal U, Koren J, Dickey CA, Gottschall PE, Morgan D, Gordon MN (2010) Trafficking CD11b-positive blood cells deliver therapeutic genes to the brain of amyloiddepositing transgenic mice. J Neurosci 30:9651–9658 [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Lerman BJ, Hoffman EP, Sutherland ML, Bouri K, Hsu DK, Liu FT, Rothstein JD, Knoblach SM (2012) Deletion of galectin-3 exacerbates microglial activation and accelerates disease progression and demise in a SOD1(G93A) mouse model of amyotrophic lateral sclerosis. Brain Behav 2(5):563–575 [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Liao B, Zhao W, Beers DR, Henkel JS, Appel SH (2012) Transformation from a neuroprotective to a neurotoxic microglial phenotype in a mouse model of ALS. Exp Neurol 237(1):147–152 [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Lincecum JM, Vieira FG, Wang MZ, Thompson K, De Zutter GS, Kidd J, Moreno A, Sanchez R, Carrion IJ, Levine BA, AlNakhala BM, Sullivan SM, Gill A, Perrin S (2010) From transcriptome analysis to therapeutic anti-CD40L treatment in the SOD1 model of amyotrophic lateral sclerosis. Nat Genet 42(5):392–399 [DOI] [PubMed] [Google Scholar]
  133. Lino MM, Schneider C, Caroni P (2002) Accumulation of SOD1 mutants in postnatal motoneurons does not cause motoneuron pathology or motoneuron disease. J Neurosci 22(12):4825–4832 [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Lucchinetti C, Bruck W, Parisi J, Scheithauer B, Rodriguez M, Lassmann H (2000) Heterogeneity of multiple sclerosis lesions: implications for the pathogenesis of demyelination. Ann Neurol 47(6):707–717 [DOI] [PubMed] [Google Scholar]
  135. Maier M, Peng Y, Jiang L, Seabrook TJ, Carroll MC, Lemere CA (2008) Complement C3 deficiency leads to accelerated amyloid beta plaque deposition and neurodegeneration and modulation of the microglia/macrophage phenotype in amyloid precursor protein transgenic mice. J Neurosci 28(25):6333–6341 [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Mantovani S, Garbelli S, Pasini A, Alimonti D, Perotti C, Melazzini M, Bendotti C, Mora G (2009) Immune system alterations in sporadic amyotrophic lateral sclerosis patients suggest an ongoing neuroinflammatory process. J Neuroimmunol 210(1–2):73–79 [DOI] [PubMed] [Google Scholar]
  137. Martinez-Muriana A, Mancuso R, Francos-Quijorna I, Olmos-Alonso A, Osta R, Perry VH, Navarro X, Gomez-Nicola D, 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]
  138. Matcovitch-Natan O, Winter DR, Giladi A, Vargas Aguilar S, Spinrad A, Sarrazin S, Ben-Yehuda H, David E, Zelada Gonzalez F, Perrin P, Keren-Shaul H, Gury M, Lara-Astaiso D, Thaiss CA, Cohen M, Bahar Halpern K, Baruch K, Deczkowska A, LorenzoVivas E, Itzkovitz S, Elinav E, Sieweke MH, Schwartz M, Amit I (2016) Microglia development follows a stepwise program to regulate brain homeostasis. Science 353(6301):aad8670. [DOI] [PubMed] [Google Scholar]
  139. McGeer PL, McGeer EG (2002) Inflammatory processes in amyotrophic lateral sclerosis. Muscle Nerve 26(4):459–470 [DOI] [PubMed] [Google Scholar]
  140. McGeer PL, Itagaki S, Boyes BE, McGeer EG (1988) Reactive microglia are positive for HLA-DR in the substantia nigra of Parkinson’s and Alzheimer’s disease brains. Neurology 38(8):1285–1291 [DOI] [PubMed] [Google Scholar]
  141. McGeer PL, McGeer EG, Kawamata T, Yamada T, Akiyama H (1991) Reactions of the immune system in chronic degenerative neurological diseases. Can J Neurol Sci 18(3 Suppl):376–379 [DOI] [PubMed] [Google Scholar]
  142. Meissner F, Molawi K, Zychlinsky A (2010) Mutant superoxide dismutase 1-induced IL-1beta accelerates ALS pathogenesis. Proc Natl Acad Sci USA 107(29):13046–13050 [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Michaud JP, Bellavance MA, Préfontaine P, Rivest S (2013) Realtime in vivo imaging reveals the ability of monocytes to clear vascular amyloid beta. Cell Rep 5:646–653 [DOI] [PubMed] [Google Scholar]
  144. Mikita J, Dubourdieu-Cassagno N, Deloire MS, Vekris A, Biran M, Raffard G, Brochet B, Canron MH, Franconi JM, Boiziau C, Petry KG (2011) Altered M1/M2 activation patterns of monocytes in severe relapsing experimental rat model of multiple sclerosis. Amelioration of clinical status by M2 activated monocyte administration. Mult Scler 17(1):2–15 [DOI] [PubMed] [Google Scholar]
  145. Mildner A, Schmidt H, Nitsche M, Merkler D, Hanisch UK, Mack M, Heikenwalder M, Bruck W, Priller J, Prinz M (2007) Microglia in the adult brain arise from Ly-6ChiCCR2+ monocytes only under defined host conditions. Nat Neurosci 10(12):1544–1553 [DOI] [PubMed] [Google Scholar]
  146. Mildner A, Mack M, Schmidt H, Bruck W, Djukic M, Zabel MD, Hille A, Priller J, Prinz M (2009) CCR2+ Ly-6Chi monocytes are crucial for the effector phase of autoimmunity in the central nervous system. Brain 132(Pt 9):2487–2500 [DOI] [PubMed] [Google Scholar]
  147. Miron VE, Boyd A, Zhao JW, Yuen TJ, Ruckh JM, Shadrach JL, van Wijngaarden P, Wagers AJ, Williams A, Franklin RJM, Ffrench Constant C (2013) M2 microglia and macrophages drive oligodendrocyte differentiation during CNS remyelination. Nat Neurosci 16(9):1211–1218 [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Mitchell RM, Freeman WM, Randazzo WT, Stephens HE, Beard JL, Simmons Z, Connor JR (2009) A CSF biomarker panel for identification of patients with amyotrophic lateral sclerosis. Neurology 72(1):14–19 [DOI] [PubMed] [Google Scholar]
  149. Mogi M, Harada M, Kondo T, Riederer P, Inagaki H, Minami M, Nagatsu T (1994a) Interleukin-1 beta, interleukin-6, epidermal growth factor and transforming growth factor-alpha are elevated in the brain from parkinsonian patients. Neurosci Lett 180(2):147–150 [DOI] [PubMed] [Google Scholar]
  150. Mogi M, Harada M, Riederer P, Narabayashi H, Fujita K, Nagatsu T (1994b) Tumor necrosis factor-alpha (TNF-alpha) increases both in the brain and in the cerebrospinal fluid from parkinsonian patients. Neurosci Lett 165(1–2):208–210 [DOI] [PubMed] [Google Scholar]
  151. Murdock BJ, Bender DE, Segal BM, Feldman EL (2015) The dual roles of immunity in ALS: injury overrides protection. Neurobiol Dis 77:1–12 [DOI] [PubMed] [Google Scholar]
  152. Murdock BJ, Bender DE, Kashlan SR, Figueroa-Romero C, Backus C, Callaghan BC, Goutman SA, Feldman EL (2016) Increased ratio of circulating neutrophils to monocytes in amyotrophic lateral sclerosis. Neurology Neuroimmunol Neuroinflammation 3(4):e242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Musaro A (2010) State of the art and the dark side of amyotrophic lateral sclerosis. World J Biol Chem 1(5):62–68 [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Nagata T, Nagano I, Shiote M, Narai H, Murakami T, Hayashi T, Shoji M, Abe K (2007) Elevation of MCP-1 and MCP-1/VEGF ratio in cerebrospinal fluid of amyotrophic lateral sclerosis patients. Neurol Res 29(8):772–776 [DOI] [PubMed] [Google Scholar]
  155. Nardo G, Pozzi S, Pignataro M, Lauranzano E, Spano G, Garbelli S, Mantovani S, Marinou K, Papetti L, Monteforte M, Torri V, Paris L, Bazzoni G, Lunetta C, Corbo M, Mora G, Bendotti C, Bonetto V (2011) Amyotrophic lateral sclerosis multiprotein biomarkers in peripheral blood mononuclear cells. PLoS One 6(10):e25545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Obal I, Jakab JS, Siklos L, Engelhardt JI (2001) Recruitment of activated microglia cells in the spinal cord of mice by ALS IgG. NeuroReport 12(11):2449–2452 [DOI] [PubMed] [Google Scholar]
  157. Olmos-Alonso A, Schetters ST, Sri S, Askew K, Mancuso R, Vargas-Caballero M, Holscher C, Perry VH, Gomez-Nicola D (2016) Pharmacological targeting of CSF1R inhibits microglial proliferation and prevents the progression of Alzheimer’s-like pathology. Brain 139(Pt 3):891–907 [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Olsen MK, Roberds SL, Ellerbrock BR, Fleck TJ, McKinley DK, Gurney ME (2001) Disease mechanisms revealed by transcription profiling in SOD1-G93A transgenic mouse spinal cord. Ann Neurol 50(6):730–740 [DOI] [PubMed] [Google Scholar]
  159. O’Rourke JG, Bogdanik L, Yanez A, Lall D, Wolf AJ, Muhammad AK, Ho R, Carmona S, Vit JP, Zarrow J, Kim KJ, Bell S, Harms MB, Miller TM, Dangler CA, Underhill DM, Goodridge HS, Lutz CM, Baloh RH (2016) C9orf72 is required for proper macrophage and microglial function in mice. Science 351(6279):1324–1329 [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Paloneva J, Manninen T, Christman G, Hovanes K, Mandelin J, Adolfsson R, Bianchin M, Bird T, Miranda R, Salmaggi A, Tranebjaerg L, Konttinen Y, Peltonen L (2002) Mutations in two genes encoding different subunits of a receptor signaling complex result in an identical disease phenotype. Am J Hum Genet 71(3):656–662 [DOI] [PMC free article] [PubMed] [Google Scholar]
  161. Parillaud VR, Lornet G, Monnet Y, Privat AL, Haddad AT, Brochard V, Bekaert A, de Chanville CB, Hirsch EC, Combadiere C, Hunot S, Lobsiger CS (2017) Analysis of monocyte infiltration in MPTP mice reveals that microglial CX3CR1 protects against neurotoxic over-induction of monocyte-attracting CCL2 by astrocytes. J Neuroinflammation 14(1):60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. Parkhurst CN, Yang G, Ninan I, Savas JN, Yates JR 3rd, Lafaille JJ, Hempstead BL, Littman DR, Gan WB (2013) Microglia promote learning-dependent synapse formation through brain-derived neurotrophic factor. Cell 155(7):1596–1609 [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Peferoen L, Kipp M, van der Valk P, van Noort JM, Amor S (2014) Oligodendrocyte-microglia cross-talk in the central nervous system. Immunology 141(3):302–313 [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Poliani PL, Wang Y, Fontana E, Robinette ML, Yamanishi Y, Gilfillan S, Colonna M (2015) TREM2 sustains microglial expansion during aging and response to demyelination. J Clin Investig 125(5):2161–2170 [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Polymeropoulos MH, Lavedan C, Leroy E, Ide SE, Dehejia A, Dutra A, Pike B, Root H, Rubenstein J, Boyer R, Stenroos ES, Chandrasekharappa S, Athanassiadou A, Papapetropoulos T, Johnson WG, Lazzarini AM, Duvoisin RC, Di Iorio G, Golbe LI, Nussbaum RL (1997) Mutation in the alpha-synuclein gene identified in families with Parkinson’s disease. Science 276(5321):2045–2047 [DOI] [PubMed] [Google Scholar]
  166. Ponomarev ED, Shriver LP, Maresz K, Dittel BN (2005) Microglial cell activation and proliferation precedes the onset of CNS autoimmunity. J Neurosci Res 81(3):374–389 [DOI] [PubMed] [Google Scholar]
  167. Pramatarova A, Laganiere J, Roussel J, Brisebois K, Rouleau GA (2001) Neuron-specific expression of mutant superoxide dismutase 1 in transgenic mice does not lead to motor impairment. J Neurosci 21(10):3369–3374 [DOI] [PMC free article] [PubMed] [Google Scholar]
  168. Prince M, Ali GC, Guerchet M, Prina AM, Albanese E, Wu YT (2016) Recent global trends in the prevalence and incidence of dementia, and survival with dementia. Alzheimers Res Ther 8(1):23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Pringsheim T, Jette N, Frolkis A, Steeves TD (2014) The prevalence of Parkinson’s disease: a systematic review and meta-analysis. Mov Disord 29(13):1583–1590 [DOI] [PubMed] [Google Scholar]
  170. Prinz M, Priller J, Sisodia SS, Ransohoff RM (2011) Heterogeneity of CNS myeloid cells and their roles in neurodegeneration. Nat Neurosci 14(10):1227–1235 [DOI] [PubMed] [Google Scholar]
  171. Prokop S, Miller KR, Drost N, Handrick S, Mathur V, Luo J, Wegner A, Wyss-Coray T, Heppner FL (2015) Impact of peripheral myeloid cells on amyloidpathology in Alzheimer’s disease-like mice. J Exp Med 212:1811–1818 [DOI] [PMC free article] [PubMed] [Google Scholar]
  172. Qin L, Wu X, Block ML, Liu Y, Breese GR, Hong JS, Knapp DJ, Crews FT (2007) Systemic LPS causes chronic neuroinflammation and progressive neurodegeneration. Glia 55(5):453–462 [DOI] [PMC free article] [PubMed] [Google Scholar]
  173. Raivich G, Bohatschek M, Kloss CU, Werner A, Jones LL, Kreutzberg GW (1999) Neuroglial activation repertoire in the injured brain: graded response, molecular mechanisms and cues to physiological function. Brain Res Brain Res Rev 30(1):77–105 [DOI] [PubMed] [Google Scholar]
  174. Reale M, Greig NH, Kamal MA (2009) Peripheral chemo-cytokine profiles in Alzheimer’s and Parkinson’s diseases. Mini Rev Med Chem 9(10):1229–1241 [DOI] [PMC free article] [PubMed] [Google Scholar]
  175. Rodriguez M, Alvarez-Erviti L, Blesa FJ, Rodriguez-Oroz MC, Arina A, Melero I, Ramos LI, Obeso JA (2007) Bone-marrow-derived cell differentiation into microglia: a study in a progressive mouse model of Parkinson’s disease. Neurobiol Dis 28(3):316–325 [DOI] [PubMed] [Google Scholar]
  176. Rose S, Misharin A, Perlman H (2012) A novel Ly6C/Ly6G-based strategy to analyze the mouse splenic myeloid compartment. Cytometry A 81(4):343–350 [DOI] [PMC free article] [PubMed] [Google Scholar]
  177. Rosen DR, Siddique T, Patterson D, Figlewicz DA, Sapp P, Hentati A, Donaldson D, Goto J, O’Regan JP, Deng HX et al. (1993) Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature 362(6415):59–62 [DOI] [PubMed] [Google Scholar]
  178. Rudick RA, Ransohoff RM (1992) Cytokine secretion by multiple sclerosis monocytes. Relationship to disease activity. Arch Neurol 49(3):265–270 [DOI] [PubMed] [Google Scholar]
  179. Saederup N, Cardona AE, Croft K, Mizutani M, Cotleur AC, Tsou CL, Ransohoff RM, Charo IF (2010) Selective chemokine receptor usage by central nervous system myeloid cells in CCR2-red fluorescent protein knock-in mice. PLoS One 5(10):e13693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  180. Sanagi T, Yuasa S, Nakamura Y, Suzuki E, Aoki M, Warita H, Itoyama Y, Uchino S, Kohsaka S, Ohsawa K (2010) Appearance of phagocytic microglia adjacent to motoneurons in spinal cord tissue from a presymptomatic transgenic rat model of amyotrophic lateral sclerosis. J Neurosci Res 88(12):2736–2746 [DOI] [PubMed] [Google Scholar]
  181. Sanchez-Guajardo V, Febbraro F, Kirik D, Romero-Ramos M (2010) Microglia acquire distinct activation profiles depending on the degree of alpha-synuclein neuropathology in a rAAV based model of Parkinson’s disease. PLoS One 5(1):e8784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  182. Satoh J, Kino Y, Asahina N, Takitani M, Miyoshi J, Ishida T, Saito Y (2016) TMEM119 marks a subset of microglia in the human brain. Neuropathology 36(1):39–49 [DOI] [PubMed] [Google Scholar]
  183. Selkoe DJ (2002) Alzheimer’s disease is a synaptic failure. Science 298(5594):789–791 [DOI] [PubMed] [Google Scholar]
  184. Serbina NV, Jia T, Hohl TM, Pamer EG (2008) Monocyte-mediated defense against microbial pathogens. Annu Rev Immunol 26:421–452 [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. Shi C, Pamer EG (2011) Monocyte recruitment during infection and inflammation. Nat Rev Immunol 11(11):762–774 [DOI] [PMC free article] [PubMed] [Google Scholar]
  186. Shi Q, Chowdhury S, Ma R, Le KX, Hong S, Caldarone BJ, Stevens B, Lemere CA (2017) Complement C3 deficiency protects against neurodegeneration in aged plaque-rich APP/PS1 mice. Sci Transl Med 9(392):eaaf6295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  187. Shibata N, Kakita A, Takahashi H, Ihara Y, Nobukuni K, Fujimura H, Sakoda S, Sasaki S, Iwata M, Morikawa S, Hirano A, Kobayashi M (2009) Activation of signal transducer and activator of transcription-3 in the spinal cord of sporadic amyotrophic lateral sclerosis patients. Neurodegener Dis 6(3):118–126 [DOI] [PubMed] [Google Scholar]
  188. Simpson EP, Henry YK, Henkel JS, Smith RG, Appel SH (2004) Increased lipid peroxidation in sera of ALS patients: a potential biomarker of disease burden. Neurology 62(10):1758–1765 [DOI] [PubMed] [Google Scholar]
  189. Singh S, Metz I, Amor S, van der Valk P, Stadelmann C, Bruck W (2013) Microglial nodules in early multiple sclerosis white matter are associated with degenerating axons. Acta Neuropathol 125(4):595–608 [DOI] [PMC free article] [PubMed] [Google Scholar]
  190. Solomon JN, Lewis CA, Ajami B, Corbel SY, Rossi FM, Krieger C (2006) Origin and distribution of bone marrow-derived cells in the central nervous system in a mouse model of amyotrophic lateral sclerosis. Glia 53(7):744–753 [DOI] [PubMed] [Google Scholar]
  191. Spangenberg EE, Lee RJ, Najafi AR, Rice RA, Elmore MR, Blurton-Jones M, West BL, Green KN (2016) Eliminating microglia in Alzheimer’s mice prevents neuronal loss without modulating amyloid-beta pathology. Brain 139(Pt 4):1265–1281 [DOI] [PMC free article] [PubMed] [Google Scholar]
  192. Streit WJ, Sammons NW, Kuhns AJ, Sparks DL (2004) Dystrophic microglia in the aging human brain. Glia 45(2):208–212 [DOI] [PubMed] [Google Scholar]
  193. Suarez-Calvet M, Araque Caballero MA, Kleinberger G, Bateman RJ, Fagan AM, Morris JC, Levin J, Danek A, Ewers M, Haass C, Dominantly Inherited Alzheimer Network (2016) Early changes in CSF sTREM2 in dominantly inherited Alzheimer’s disease occur after amyloid deposition and neuronal injury. Sci Transl Med 8(369):369ra178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  194. Symonds CP (1975) Multiple sclerosis and the swayback story. Lancet 1(7899):155–156 [DOI] [PubMed] [Google Scholar]
  195. Tarkowski E, Andreasen N, Tarkowski A, Blennow K (2003) Intrathecal inflammation precedes development of Alzheimer’s disease. J Neurol Neurosurg Psychiatry 74:1200–1205 [DOI] [PMC free article] [PubMed] [Google Scholar]
  196. Trapp BD, Peterson J, Ransohoff RM, Rudick R, Mork S, Bo L (1998) Axonal transection in the lesions of multiple sclerosis. N Engl J Med 338(5):278–285 [DOI] [PubMed] [Google Scholar]
  197. Turner MR, Cagnin A, Turkheimer FE, Miller CC, Shaw CE, Brooks DJ, Leigh PN, Banati RB (2004) Evidence of widespread cerebral microglial activation in amyotrophic lateral sclerosis: an [11C](R)-PK11195 positron emission tomography study. Neurobiol Dis 15(3):601–609 [DOI] [PubMed] [Google Scholar]
  198. Umeno J, Asano K, Matsushita T, Matsumoto T, Kiyohara Y, Iida M, Nakamura Y, Kamatani N, Kubo M (2011) Meta-analysis of published studies identified eight additional common susceptibility loci for Crohn’s disease and ulcerative colitis. Inflamm Bowel Dis 17(12):2407–2415 [DOI] [PubMed] [Google Scholar]
  199. van der Valk P, Amor S (2009) Preactive lesions in multiple sclerosis. Curr Opin Neurol 22(3):207–213 [DOI] [PubMed] [Google Scholar]
  200. van Noort JM, van den Elsen PJ, van Horssen J, Geurts JJ, van der Valk P, Amor S (2011) Preactive multiple sclerosis lesions offer novel clues for neuroprotective therapeutic strategies. CNS Neurol Disord Drug Targets 10(1):68–81 [DOI] [PubMed] [Google Scholar]
  201. Varvel NH, Grathwohl SA, Degenhardt K, Resch C, Bosch A, Jucker M, Neher JJ (2015) Replacement of brain-resident myeloid cells does not alter cerebral amyloid-beta deposition in mouse models of Alzheimer’s disease. J Exp Med 212(11):1803–1809 [DOI] [PMC free article] [PubMed] [Google Scholar]
  202. Vogel DY, Vereyken EJ, Glim JE, Heijnen PD, Moeton M, van der Valk P, Amor S, Teunissen CE, van Horssen J, Dijkstra CD (2013) Macrophages in inflammatory multiple sclerosis lesions have an intermediate activation status. J Neuroinflammation 10:35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  203. Vom Berg J, Prokop S, Miller KR, Obst J, Kalin RE, Lopategui-Cabezas I, Wegner A, Mair F, Schipke CG, Peters O, Winter Y, Becher B, Heppner FL (2012) Inhibition of IL-12/IL-23 signaling reduces Alzheimer’s disease-like pathology and cognitive decline. Nat Med 18(12):1812–1819 [DOI] [PubMed] [Google Scholar]
  204. Wang L, Sharma K, Grisotti G, Roos RP (2009) The effect of mutant SOD1 dismutase activity on non-cell autonomous degeneration in familial amyotrophic lateral sclerosis. Neurobiol Dis 35(2):234–240 [DOI] [PMC free article] [PubMed] [Google Scholar]
  205. Wang WY, Tan MS, Yu JT, Tan L (2015a) Role of pro-inflammatory cytokines released from microglia in Alzheimer’s disease. Ann Transl Med 3(10):136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  206. Wang Y, Cella M, Mallinson K, Ulrich JD, Young KL, Robinette ML, Gilfillan S, Krishnan GM, Sudhakar S, Zinselmeyer BH, Holtzman DM, Cirrito JR, Colonna M (2015b) TREM2 lipid sensing sustains the microglial response in an Alzheimer’s disease model. Cell 160(6):1061–1071 [DOI] [PMC free article] [PubMed] [Google Scholar]
  207. Wang Y, Ulland TK, Ulrich JD, Song W, Tzaferis JA, Hole JT, Yuan P, Mahan TE, Shi Y, Gilfillan S, Cella M, Grutzendler J, DeMattos RB, Cirrito JR, Holtzman DM, Colonna M (2016) TREM2-mediated early microglial response limits diffusion and toxicity of amyloid plaques. J Exp Med 213(5):667–675 [DOI] [PMC free article] [PubMed] [Google Scholar]
  208. Wilms H, Sievers J, Dengler R, Bufler J, Deuschl G, Lucius R (2003) Intrathecal synthesis of monocyte chemoattractant protein-1 (MCP-1) in amyotrophic lateral sclerosis: further evidence for microglial activation in neurodegeneration. J Neuroimmunol 144(1–2):139–142 [DOI] [PubMed] [Google Scholar]
  209. Woollard KJ, Geissmann F (2010) Monocytes in atherosclerosis: subsets and functions. Nat Rev Cardiol 7(2):77–86 [DOI] [PMC free article] [PubMed] [Google Scholar]
  210. Wyss-Coray T, Mucke L (2002) Inflammation in neurodegenerative disease—a double-edged sword. Neuron 35(3):419–432 [DOI] [PubMed] [Google Scholar]
  211. Xiao Q, Zhao W, Beers DR, Yen AA, Xie W, Henkel JS, Appel SH (2007) Mutant SOD1(G93A) microglia are more neurotoxic relative to wild-type microglia. J Neurochem 102(6):2008–2019 [DOI] [PubMed] [Google Scholar]
  212. Yamasaki R, Lu H, Butovsky O, Ohno N, Rietsch AM, Cialic R, Wu PM, Doykan CE, Lin J, Cotleur AC, Kidd G, Zorlu MM, Sun N, Hu W, Liu L, Lee JC, Taylor SE, Uehlein L, Dixon D, Gu J, Floruta CM, Zhu M, Charo IF, Weiner HL, Ransohoff RM (2014) Differential roles of microglia and monocytes in the inflamed central nervous system. J Exp Med 211(8):1533–1549 [DOI] [PMC free article] [PubMed] [Google Scholar]
  213. Yona S, Kim KW, Wolf Y, Mildner A, Varol D, Breker M, Strauss-Ayali D, Viukov S, Guilliams M, Misharin A, Hume DA, Perlman H, Malissen B, Zelzer E, Jung S (2013) Fate mapping reveals origins and dynamics of monocytes and tissue macrophages under homeostasis. Immunity 38(1):79–91 [DOI] [PMC free article] [PubMed] [Google Scholar]
  214. Yuan P, Condello C, Keene CD, Wang Y, Bird TD, Paul SM, Luo W, Colonna M, Baddeley D, Grutzendler J (2016) TREM2 haplodeficiency in mice and humans impairs the microglia barrier function leading to decreased amyloid compaction and severe axonal dystrophy. Neuron 90(4):724–739 [DOI] [PMC free article] [PubMed] [Google Scholar]
  215. Zhang R, Gascon R, Miller RG, Gelinas DF, Mass J, Hadlock K, Jin X, Reis J, Narvaez A, McGrath MS (2005) Evidence for systemic immune system alterations in sporadic amyotrophic lateral sclerosis (sALS). J Neuroimmunol 159(1–2):215–224 [DOI] [PubMed] [Google Scholar]
  216. Zhang R, Gascon R, Miller RG, Gelinas DF, Mass J, Lancero M, Narvaez A, McGrath MS (2006) MCP-1 chemokine receptor CCR2 is decreased on circulating monocytes in sporadic amyotrophic lateral sclerosis (sALS). J Neuroimmunol 179(1–2):87–93 [DOI] [PubMed] [Google Scholar]
  217. Zhang B, Gaiteri C, Bodea L-G, Wang Z, McElwee J, Podtelezhnikov Alexei A, Zhang C, Xie T, Tran L, Dobrin R, Fluder E, Clurman B, Melquist S, Narayanan M, Suver C, Shah H, Mahajan M, Gillis T, Mysore J, MacDonald Marcy E, Lamb John R, Bennett David A, Molony C, Stone David J, Gudnason V, Myers Amanda J, Schadt Eric E, Neumann H, Zhu J, Emilsson V (2013) Integrated systems approach identifies genetic nodes and networks in late-onset Alzheimer’s disease. Cell 153:707–720 [DOI] [PMC free article] [PubMed] [Google Scholar]
  218. Zhao W, Beers DR, Hooten KG, Sieglaff DH, Zhang A, Kalyana-Sundaram S, Traini CM, Halsey WS, Hughes AM, Sathe GM, Livi GP, Fan GH, Appel SH (2017) Characterization of gene expression phenotype in amyotrophic lateral sclerosis monocytes. JAMA Neurol 74(6):677–685 [DOI] [PMC free article] [PubMed] [Google Scholar]
  219. Zondler L, Muller K, Khalaji S, Bliederhauser C, Ruf WP, Grozdanov V, Thiemann M, Fundel-Clemes K, Freischmidt A, Holzmann K, Strobel B, Weydt P, Witting A, Tha l, Helferich AM, Hengerer B, Gottschalk KE, Hill O, Kluge M, Ludolph AC, Danzer KM, Weishaupt JH (2016) Peripheral monocytes are functionally altered and invade the CNS in ALS patients. Acta Neuropathol 132:391–411 [DOI] [PubMed] [Google Scholar]
  220. Zrzavy T, Hametner S, Wimmer I, Butovsky O, Weiner HL, Lassmann H (2017) Loss of ‘homeostatic’ microglia and patterns of their activation in active multiple sclerosis. Brain 140:1900–1913 [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES