Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Neuron

Review

Microglia/Brain Macrophages as Central


Drivers of Brain Tumor Pathobiology
David H. Gutmann1 and Helmut Kettenmann2,*
1Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
2CellularNeurosciences, Max-Delbru €ck Center for Molecular Medicine in the Helmholtz Association, Berlin 13125, Germany
*Correspondence: kettenmann@mdc-berlin.de
https://doi.org/10.1016/j.neuron.2019.08.028

One of the most common brain tumors in children and adults is glioma or astrocytoma. There are few effective
therapies for these cancers, and patients with malignant glioma fare poorly, even after aggressive surgery,
chemotherapy, and radiation. Over the past decade, it is now appreciated that these tumors are composed
of numerous distinct neoplastic and non-neoplastic cell populations, which could each influence overall
tumor biology and response to therapy. Among these noncancerous cell types, monocytes (microglia and
macrophages) predominate. In this Review, we discuss the complex interactions involving microglia and
macrophages relevant to glioma formation, progression, and response to therapy.

Introduction relevant to patient outcome. In one study, there was a positive


The most common brain tumor arising in children and adults is association between the numbers of CD68+, CD163+, and
the glioma (or astrocytoma). These tumors are classified by the CD206+ GAMs in the vital tumor core and prolonged overall
World Health Organization according to histologic features, survival of patients with IDH1R132H-non-mutant glioblastoma
running the spectrum from low-grade (I and II) to high-grade multiforme (GBM; Zeiner et al., 2019). Similarly, others found
(III and IV) gliomas. With the advent of molecular diagnostics, that microglia density, as determined by Iba1 labeling, corre-
the nosology of adult glioma has changed (Louis et al., 2016), lated with improved outcomes for patients with gliomas,
now incorporating the presence or absence of specific genetic whereas CD204 expression in GAMs was associated with
(e.g., IDH1 and IDH2 mutations) and genomic (e.g., chromosome poorer survival (Sørensen et al., 2018). In contrast, the expres-
1p/19q co-deletions) alterations. Similarly, pediatric diffuse gli- sion of microglia or macrophage-related genes in adult and pe-
omas are molecularly defined by mutations in histone H3 genes diatric malignant gliomas of the mesenchymal subtype revealed
(H3-K27M mutant diffuse midline gliomas). a negative correlation between GAMs accumulation and patient
Glioma tissue is not only composed of cancer cells, but also survival in adults, but not in children (Engler et al., 2012). While
contains nontransformed cells, predominantly resident microglia GAMs enrichment is higher in the mesenchymal subtype of
from the brain and circulating blood monocytes (macrophages), high-grade glioma relative to proneural or classical gliomas
which comprise 30%–50% of the cellular content of these tu- (Kaffes et al., 2019), the mechanisms underlying GAMs-medi-
mors (Simmons et al., 2011). Microglia, the brain resident im- ated patient survival remain to be determined.
mune cells, were first described by Pio del Rio Hortega in 1919
(Sierra et al., 2016) and later reported by Wilder Penfield to infil- GAMs Regulate Glioma Formation and Progression
trate glioma tissue as cells with an amoeboid morphology, Numerous studies have revealed that GAMs are critical for both
similar to microglia found in multiple sclerosis or Alzheimer dis- low-grade and high-grade gliomagenesis and continued tumor
ease tissue (Penfield, 1925). growth. In experimental malignant (high-grade) glioma models,
Microglia develop from primitive yolk sac myeloid precursors microglia depletion reduces glioma growth (Hambardzumyan
and enter the brain during embryogenesis (Ginhoux et al., et al., 2016; Figure 1). While the mechanisms underlying this
2010) to mature into distinct populations of CNS monocytes. pro-tumoral effect likely vary from tumor to tumor, numerous po-
Additionally, there may be other ontologies for brain microglia tential etiologies have been identified. In these studies, trans-
that reflect different waves of yolk sac hematopoiesis (De forming growth factor b (TGF-b), stress-inducible protein 1
et al., 2018). In contrast to resident microglia, macrophages (STI-1), interleukin-6 (IL-6), IL-1b, and epidermal growth factor
most typically enter the brain in the context of pathology, either (EGF) are factors released from GAMs, which can each promote
from the blood (bone-marrow-derived macrophages) or through tumor growth (Hambardzumyan et al., 2016). Moreover, micro-
direct vascular channels that connect the skull bone marrow to glia, but not bone-marrow-derived macrophages, induce
the brain (Herisson et al., 2018). platelet-derived growth factor receptor expression in a subset
Over the last decade, it has become clear that brain microglia of tumor cells of mouse and human low and high-grade glioma.
and infiltrating peripheral macrophages (collectively termed This receptor expression stimulates the migratory capacity of gli-
GAMs) are not merely bystander or ‘‘reactive’’ immune-sys- oma cells, which accelerates tumor progression (Wallmann
tem-like cells and interact with numerous other cell types to et al., 2018). Similarly, GAMs are the predominant source of
actively influence brain tumor biology. However, there is still the secreted form of osteopontin (also known as SPP1), which
no consensus on the clinical significance of GAM infiltration suppresses glioma growth in some contexts (Szulzewsky et al.,

442 Neuron 104, November 6, 2019 ª 2019 Elsevier Inc.


Neuron

Review

gliomas gain some measure of stromal independence, malignant


gliomas harboring NF1 mutations express CCL5, establishing an
autocrine loop for mesenchymal glioblastoma survival (Pan et al.,
2017a). The requirement for microglia and microglia-produced
Ccl5 is further underscored by studies in which murine Nf1 optic
glioma stem cells are implanted into mice. While wild-type mice
support the formation of glioma-like lesions following transplan-
tation (Chen et al., 2015), no tumors form in mice lacking the two
chemokine receptors that direct monocyte migration (Cx3cr1
and Ccr2) or in mice deficient in Ccl5 expression (Pan et al.,
2018; Guo et al., 2019). While less well studied, KIAA1549:
BRAF-expressing low-grade gliomas are also dependent on
monocytes in the tumor microenvironment, such that gliomas
fail to form following the transplantation of murine KIAA1549:
BRAF-expressing cerebellar neural stem cells in Ccr2-deficient
mice (Chen et al., 2019a).

Figure 1. GAMs Increase Glioma Growth through the Release of Glioma Cells Attract or Recruit GAMs
Mitogens and Invasion-Promoting Factors
There are a variety of glioma-derived factors that can function as
Brain microglia and infiltrating peripheral macrophages (GAMs) become re-
programmed to produce growth factors that increase glioma cell proliferation, chemoattractants for GAMs, including CCL2, CX3CL1, SDF-1,
attenuate glioma cell apoptosis, and promote tumor cell migration. In this CSF-1, granulocyte/macrophage-colony stimulating factor
fashion, GAMs secrete factors (IL-10, IL-6, IL-1b, EGF, and CCL5) that enhance (GM-CSF), lysine oxidase (LOX), and potentially EGF (Roesch
glioma cell growth through binding to their cognate receptors (right). In addition,
SPP1 has been reported to both increase (‘‘+’’) and inhibit (‘‘ ’’) glioma cell et al., 2018). This directed monocyte recruitment occurs through
growth. Moreover, GAMs elaborate other factors that increase extracellular the establishment of chemokine gradients that attract mono-
matrix (ECM) degradation and promote glioma cell invasion and motility (left). cytes to the evolving tumor bed (Figure 2). Studies employing
Ccr2 and Cx3cr1 reporter mice have revealed that two of the
2018) and reduces glioma cell apoptosis in other experimental most important chemoattractants are Ccl2 and Cx3cl1, which
settings (Chen et al., 2019b). traditionally have been thought to drive directional migration of
In contrast to their malignant counterparts, low-grade gliomas macrophages and microglia, respectively. In this regard, Ccl2
predominate in children and harbor a paucity of genetic muta- is produced by low-grade glioma stem cells expressing the
tions. These tumors arise either sporadically, caused frequently KIAA1549:BRAF genomic alteration, leading to monocyte
by genomic alterations involving the BRAF kinase gene attraction (Chen et al., 2019a), whereas NF1 mutation in both
(KIAA1549:BRAF fusion) (Pfister et al., 2008; Yu et al., 2009), or low-grade and high-grade gliomas attracts microglia through
in the setting of the Neurofibromatosis type 1 (NF1) cancer pre- Cx3cl1 (Chen et al., 2017; Guo et al., 2019). Similarly, in experi-
disposition syndrome (Listernick et al., 1989). Importantly, Nf1 mental glioblastoma models, Ccl2 is produced by the tumor
loss or BRAF fusion is usually not sufficient for glioma formation cells, which attract macrophages (Platten et al., 2003), such
in rodents, unless coupled with supportive growth factors from that Ccl2/Ccr2 inhibition prolongs mouse survival (Chen et al.,
the tumor microenvironment. In this regard, Nf1 loss (Larribere 2017; Hutter et al., 2019). While there are few studies that specif-
et al., 2015) or KIAA1549:BRAF expression (Jacob et al., 2011) ically focused on chemokine networks responsible for monocyte
in numerous cell types induces cellular senescence, which re- attraction, other molecules are also likely involved in recruiting
quires stromal signals to overcome and lead to neoplasia. monocytes to the tumor (Dijksterhuis et al., 2015; Brandenburg
To explore the role of these permissive stromal signals, studies et al., 2016).
using Nf1 genetically engineered mouse strains that develop op- Moreover, emerging evidence supports the view that microglia
tic glioma, the signature brain tumor seen in children with NF1, and macrophages colonize different regions of malignant gli-
have uncovered essential roles for microglia in glioma formation omas, such that macrophages appear to be recruited early dur-
and maintenance. First, optic glioma formation is delayed in mice ing tumorigenesis and occupy perivascular regions (Chen et al.,
with reduced expression of the key chemokine receptor involved 2017). However, conflicting data exist regarding the major
in directed microglia migration (CX3CR1) (Pong et al., 2013a). monocyte population in these tumors, with some studies report-
Second, optic glioma growth in vivo is attenuated following treat- ing a microglia predominance (Hutter et al., 2019) and others
ment with either minocycline (crude microglia inhibitor; Dagina- demonstrating that infiltrating macrophages represent the ma-
katte and Gutmann, 2007; Pan et al., 2017b) or JNK inhibitors jority of the GAMs population (Chen et al., 2017; Yu et al.,
(targeting the signaling pathway hyperactivated in Nf1 mutant 2019). These differences could reflect the specific experimental
microglia) (Daginakatte et al., 2008). Third, RNA sequencing of mouse model systems (chicken retroviral receptor [RCAS] model
murine Nf1 optic glioma-associated GAMs demonstrated that versus GL261 or T387 cell lines) used in each of these studies,
these cells secrete CCL5, which is a potent mitogen for glioma suggesting that variations in GAMs populations may be differen-
cells (Solga et al., 2015). Treatment of Nf1 optic glioma-bearing tially dictated by the molecular properties of the glioma.
mice with a neutralizing CCL5 antibody dramatically attenuated Support for potential mutation-specific effects on GAMs
tumor growth in vivo. Consistent with the idea that high-grade recruitment comes from two studies: First, murine high-grade

Neuron 104, November 6, 2019 443


Neuron

Review

Work in many laboratories using different microglia isolation


methods and RNA analysis platforms has identified other
GAMs-specific markers; however, no marker has been discov-
ered to date that uniquely distinguishes GAMs from other micro-
glia populations.
While microglia are the professional phagocytes of the CNS, in
the setting of glioma, they do not attack or phagocytose glioma
cells. This may reflect increased expression of the anti-phago-
cytic (‘‘don’t eat me’’) surface protein CD47 that binds to its
cognate receptor SIRPa on phagocytic cells to inhibit its phago-
cytic activity. In this regard, anti-CD47 antibody treatment
induced microglial tumor phagocytosis in a glioblastoma xeno-
graft model and reduced tumor expansion (Hutter et al., 2019).
A similar effect of a humanized anti-CD47 antibody was observed
using five aggressive and etiologically distinct pediatric brain
tumors: group 3 medulloblastoma (primary and metastatic), atyp-
ical teratoid rhabdoid tumor, primitive neuroectodermal tumor,
Figure 2. Glioma Cells Recruit and Reprogram GAMs through the
pediatric glioblastoma, and diffuse intrinsic pontine glioma (Gho-
Elaboration of Chemokines and Other Soluble Factors
Chemokines produced by glioma cells actively recruit resident microglia from lamin et al., 2017). Moreover, in glioma patients, CD47 expression
the brain, as well as macrophages from the blood, through binding to their inversely correlated with histopathologic grading (low-grade
cognate receptors on GAMs (right). In addition, glioma cells produce proteins versus high-grade glioma), and high levels of CD47 were associ-
that increase cytokine release (EGF, Let7, and tenascin-C [TNC]), phagocy-
tosis (ATP), and matrix metalloprotease (MMP9 and MMP14) expression ated with lower overall survival rates (Li et al., 2017).
(versican) (left). How microglia become reprogrammed in the setting of glioma
remains to be fully elucidated. One family of receptors expressed
gliomas harboring a mutant IDH1 allele exhibit reduced macro- on microglia may play an instructive role in the establishment of
phage and microglia infiltration, which correlated with lower these new functional states. Toll-like receptors (TLR) were initially
levels of chemokine expression (e.g., CCL2 and CXCL2) (Aman- recognized as pathogen sensors, but it is now appreciated that
kulor et al., 2017). Moreover, the IDH1/2-mediated accumulation they can also be activated by endogenous ligands. TLR2 expres-
of the oncometabolite 2-hydroxyglutarate reduced the expres- sion is increased in GAMs (Hu et al., 2015), such that mouse
sion of pro-inflammatory chemokines (Han et al., 2019). Second, GL261 glioma cells implanted into the brains of Tlr2 knockout
differences in microglia recruitment were observed in mice mice have smaller tumors and enhanced survival. This TLR2 de-
genetically engineered to develop low-grade gliomas with pendency is partially mediated by increased production of a
distinct patient-derived germline NF1 gene mutations and coop- membrane-residing protease (MMP14) essential for activating
erating genetic alterations (e.g., heterozygous Pten loss) through MMP2 and for promoting malignant glioma invasion (Vinnakota
the elaboration of specific chemokines (Guo et al., 2019). These et al., 2013). In addition to MMP14, TLR2 signaling also triggers
findings suggest that variations in the GAMs composition of gli- the release of MMP9 from microglia, which serves as an addi-
omas could be dictated by the particular immune chemoattrac- tional factor for degrading the extracellular matrix to promote gli-
tant molecules produced by cancer cells with different oma invasion and growth (Hu et al., 2014). TLR2 signaling is also
mutations. involved in the interaction of GAMs with T cells. As such, TLR2-
mediated growth glioma is reduced using TLR2 blocking anti-
GAMs Acquire Unique Phenotypes in the Setting of bodies (Hu et al., 2015). Beyond its role in glioma growth, TLR2
Glioma activation may also function to facilitate glioma immune evasion
Once GAMs are recruited to the glioma milieu, they adopt new through reduced MHC class II molecule expression and reduced
cellular and molecular identities that reflect the enactment of CD4+ T cells activation (Qian et al., 2018).
epigenetic or transcriptional programs that create monocyte In addition to TLR2, TLR4 is also important for regulating mi-
populations critical for glioma homeostasis and progression. croglial IL-6 secretion, a mitogen for glioma stem cells. Analysis
To identify these GAM transcriptional programs, two mouse gli- of human glioma tissue confirmed that GAMs are the major
oma models, inoculated GL261 cells and the RCAS transgenic source of IL-6 in gliomas (Dzaye et al., 2016). One of the endog-
system, have been used to compare the expression profiles of enous ligands for TLR4 is the cell-adhesion molecule tenascin C.
glioma-associated microglia or macrophages and naive control In tumors with genetically reduced tenascin C expression, micro-
cells. Not surprisingly, these profiles do not correspond to the glia acquire a more amoeboid morphology and increased
M1/M2 classification scheme often used to describe macro- expression of major histocompatibility complex (MHC) class II
phages/microglia in homeostatic or pro-inflammatory settings. molecules (Xia et al., 2016), similar to that observed with TLR2
Two genes were increased in GAMs, Gpnmb and Spp1, which silencing.
have also been described in other specialized microglia popula- Finally, monocytes likely receive instructive cues from other
tions (e.g., disease-associated microglia [DAM] and prolifera- cell types. One of these cell types is the T lymphocyte. Using a
tive-region-associated microglia [PAM]) and were associated low-grade glioma stem cell transplantation system, glioma-like
with poor prognosis in human GBM (Szulzewsky et al., 2015). lesions did not form in mice with impaired T cell function

444 Neuron 104, November 6, 2019


Neuron

Review

In vitro culture assays have been employed for many years,


typically involving established glioma cell lines (e.g., mouse
GL261 or human U87 high-grade glioma cells) and freshly iso-
lated microglia or the BV2 microglia-like cell line (Mu €ller et al.,
2017; Gu et al., 2017). These platforms in vitro are well suited
to determine the functions of defined cell types, such as quanti-
fying chemoattraction by tumor cells or microglia-induced gli-
oma growth and migratory properties, but have only limited value
in determining the interactions of these cell types in the natural
context of a glioma.
An intermediate model between in vivo and cell culture sys-
tems is represented by the organotypic brain slice model, in
which rodent glioma cells are introduced into rodent brain slices.
This allows for direct visualization of the interactions between
microglia and tumor cells using fluorescent protein-expressing
tumor cells and transgenic mice with microglia-restricted
expression of different fluorescent proteins (Bayerl et al., 2016;
Ghoochani et al., 2016; Resende et al., 2016). Further refine-
ments of these platforms will enable the analysis of microglia
and tumor motility using induced models of glioma, rather than
using established cell lines (Juliano et al., 2018).
Figure 3. Microglia Are Reprogrammed by Numerous Cell Types and A newer experimental platform is the zebrafish larval brain. The
Conditions in the Context of Glioma optically transparent zebrafish larva allows for real-time visuali-
As highly adaptive cells, GAMs gene expression and function can be changed
by glioma treatment (chemotherapy and radiation therapy), environmental zations of the interactions between implanted mammalian
stresses (hypoxia), interactions with other non-neoplastic cell types (e.g., as- (even human) glioma cells and genetically labeled GAMs (Astell
trocytes), and signals from the glioma cells themselves. Each of these modi- and Sieger, 2017; Yan et al., 2019). Using the Irf8 / zebrafish
fications could alter the properties of GAM in ways that either promote or
inhibit continued tumor growth or invasion. mutant, which lacks microglia, the contribution of microglia to tu-
mor growth has been explored (Hamilton et al., 2016). While the
current xenotransplantation models have employed established
(Pan et al., 2018). These T cells, found in small abundance in both human glioblastoma cell lines, future modifications may incorpo-
human and mouse NF1 mutant tumors, secrete paracrine factors rate reporter fish in which gliomas are induced by specific can-
important for inducing microglia to express CCL5 and drive gli- cer-causing genetic alterations (Chia et al., 2018; Jung et al.,
oma cell growth. Understanding how these T cells are recruited 2013), thus providing a more natural physiologic context in which
to the tumor, how they are activated, and the mechanisms that to examine the glioma ecosystem. This model has the potential
underlie their reprogramming of microglia represent opportu- to serve as a drug screening platform, as well as facilitate the
nities to dissect the immunologic determinants that underlie gli- development of future immunotherapeutic approaches that
oma formation and maintenance. target microglia.
While these vertebrate models have been very instructive, they
Experimental Model Systems to Study Glioma-Microglia do not fully recapitulate their human counterparts. In addition to
Interactions structural and lifespan differences between rodents and people,
Current available platforms to examine the interactions between the human brain contains more white matter relative to the
monocytes and glioma cells involve co-culture systems and or- mouse brain. For this reason, the generation of human cerebral
ganotypic systems, as well as small- and large-animal models. organoids from induced pluripotent stem cells (iPSCs) has
In addition to several genetically engineered mouse models (for recently been developed as a proxy for the human brain (Qian
review, see Gargiulo, 2018), large-animal platforms involving et al., 2019), allowing for the implantation of patient-derived gli-
pigs and dogs have been utilized. Spontaneously occurring oma stem cells (Linkous et al., 2019; Hubert et al., 2016). Alterna-
low- and high-grade gliomas have been observed in dogs, but tively, brain tumors can be generated by genomic editing. For
a systematic study of GAMs in these tumors has not been per- example, CRISPR/CAS9 insertion of an oncogenic RAS allele
formed (Bentley et al., 2017). Similarly, swine engineered with a into the TP53 locus in human iPSCs was used to generate
patient germline NF1 gene mutation develop low-grade optic gli- high-grade gliomas following transplantation into immunodefi-
omas, as observed in their human counterparts; however, little is cient animals (Ogawa et al., 2018). These humanized systems
known about the role of GAMs populations in these tumors (Isak- facilitate a close examination of the interactions between glioma
son et al., 2018). Moreover, human glioma cell lines can be in- cells and the non-neoplastic cells in the human brain.
jected into the brains of immunosuppressed pigs as xenograft
models (Selek et al., 2014; Khoshnevis et al., 2017), but the Outlook
use of immunocompromised animals limits a full analysis of Unfortunately, to date, the clinical application of these basic sci-
GAM contributions due to secondary defects in microglia func- ence advances has been modest. In this regard, numerous stra-
tion (Pan et al., 2018). tegies aimed at silencing GAMs function has not translated well

Neuron 104, November 6, 2019 445


Neuron

Review

to human clinical trials. For example, the tetracycline analog min- macrophages often express markers traditionally associated
ocycline, which blocks microglia activation and reduces glioma with microglia following integration into the cancer ecosystem,
expansion in experimental glioma mouse models of both high- including downregulation of CD45 and upregulation of CX3CR1
grade (Markovic et al., 2011) and low-grade (Daginakatte and expression (Pong et al., 2013b; Chen et al., 2017). These adap-
Gutmann, 2007; Toonen et al., 2017) glioma, has led to three tations make it difficult to confidently ascribe unique functions
clinical trials with no clear clinical benefit (NCT01580969, to distinct monocyte populations (Mu €ller et al., 2017). However,
NCT02272270, and NCT02770378). Similarly, PLX3397-induced with the availability of more stably expressed markers and the
depletion of microglia attenuates malignant glioma growth in use of spatial proteomics and single-cell RNA-sequencing stra-
mice (Pyonteck et al., 2013); however, no efficacy was observed tegies (Li et al., 2019; Keren-Shaul et al., 2017; Masuda et al.,
in human clinical trials (NCT01349036), despite good patient 2019), it should become possible to define the individual contri-
tolerability and adequate blood-brain barrier penetration (Bu- butions of these monocyte populations to overall glioma biology
towski et al., 2016). Finally, antibody targeting of microglial (Haage et al., 2019; Chen et al., 2017; Bowman et al., 2016;
TLRs reduces high-grade glioma growth in a mouse brain tumor Mu€ller et al., 2017). Understanding how distinct subpopulations
slice model (Hu et al., 2015) and exhibits good safety and toler- of microglia or macrophages contribute to glioma pathobiology
ability profiles in healthy subjects (Reilly et al., 2013) but has not may also help to resolve conflicting reports in the literature
progressed further in clinical trials for glioma. regarding monocyte content and overall patient survival with gli-
While targeting microglia and macrophages represents a oma. Importantly, defining these different GAM species is neces-
novel and potentially efficacious therapeutic approach, it is sary in order to design effective therapeutic approaches that
possible that adaptive changes in the tumor ecosystem will selectively impair the monocyte populations most critical for tu-
occur, creating a treatment-induced whack-a-mole scenario. mor maintenance and progression.
In this regard, microglia can be reprogrammed by radiation
(Monje et al., 2002; Allen et al., 2014; Monje et al., 2003) and/or ACKNOWLEDGMENTS
chemotherapy (Gibson et al., 2019) to create new functional
states, with different abilities to promote tumor growth. Similarly, We appreciate the support from a Berlin Institute of Health/Einstein fellowship
grant to D.H.G. and H.K., an Alexander von Humboldt Foundation grant to
hypoxia in necrotic areas of high-grade gliomas harbor high D.H.G., and the Helmholtz-Gemeinschaft, Zukunftsthema ‘‘Immunology and
levels of extracellular ATP, which could act on microglia express- Inflammation’’ grant to H.K. (ZT-0027).
ing purinergic receptors to increase cell motility, phagocytic ac-
tivity, and cytokine release (Kettenmann et al., 2011). Moreover, REFERENCES
it is possible that immunotherapies, like chimeric antigen recep-
Allen, A.R., Eilertson, K., Chakraborti, A., Sharma, S., Baure, J., Habdank-Ko-
tor T cell therapy, will similarly alter microglia dynamics to estab- laczkowski, J., Allen, B., Rosi, S., Raber, J., and Fike, J.R. (2014). Radiation
lish new functional states. For this reason, it becomes exceeding exposure prior to traumatic brain injury induces responses that differ as a func-
important to understand the complex relationships established tion of animal age. Int. J. Radiat. Biol. 90, 214–223.
between GAMs and the other cell types in the tumor in order to Amankulor, N.M., Kim, Y., Arora, S., Kargl, J., Szulzewsky, F., Hanke, M., Mar-
develop treatments that disrupt these interactions that support gineantu, D.H., Rao, A., Bolouri, H., Delrow, J., et al. (2017). Mutant IDH1 reg-
ulates the tumor-associated immune system in gliomas. Genes Dev. 31,
glioma maintenance (Figure 3). 774–786.
Additionally, it is also vital to consider other factors that could
Astell, K.R., and Sieger, D. (2017). Investigating microglia-brain tumor cell in-
influence microglia function, including brain location and patient
teractions in vivo in the larval zebrafish brain. Methods Cell Biol. 138, 593–626.
sex. To this end, the incidence of malignant glioma is higher in
males (Ostrom et al., 2018), but females generally have better Badie, B., and Schartner, J.M. (2000). Flow cytometric characterization of tu-
mor-associated macrophages in experimental gliomas. Neurosurgery 46,
overall survival (Gittleman et al., 2018). While these sexually 957–961, discussion 961–962.
dimorphic differences may reflect direct effects on cancer cells,
Bayerl, S.H., Niesner, R., Cseresnyes, Z., Radbruch, H., Pohlan, J., Branden-
it is equally possible that they act at the level of GAMs. Emerging burg, S., Czabanka, M.A., and Vajkoczy, P. (2016). Time lapse in vivo
evidence from numerous laboratories has revealed differences in microscopy reveals distinct dynamics of microglia-tumor environment interac-
brain microglia gene expression and function in normal male and tions-a new role for the tumor perivascular space as highway for trafficking mi-
croglia. Glia 64, 1210–1226.
female mice (Guneykaya et al., 2018; Villa et al., 2018; Thion
et al., 2018), as well as between male and female mice in the Bentley, R.T., Ahmed, A.U., Yanke, A.B., Cohen-Gadol, A.A., and Dey, M.
(2017). Dogs are man’s best friend: in sickness and in health. Neuro-oncol.
setting of a diverse number of different brain diseases, including 19, 312–322.
murine Nf1 optic glioma microglia-induced neuronal damage
and vision loss (Toonen et al., 2017) Bowman, R.L., Klemm, F., Akkari, L., Pyonteck, S.M., Sevenich, L., Quail, D.F.,
Dhara, S., Simpson, K., Gardner, E.E., Iacobuzio-Donahue, C.A., et al. (2016).
Finally, the lack of robust GAM-specific markers has limited macrophage ontogeny underlies differences in tumor-specific education in
our ability to define the distinct contributions of microglia and brain malignancies. Cell Rep. 17, 2445–2459.
macrophages to glioma biology. In most studies, CD45 expres- Brandenburg, S., Mu €ller, A., Turkowski, K., Radev, Y.T., Rot, S., Schmidt, C.,
sion by fluorescence-activated cell sorting (FACS) is used to Bungert, A.D., Acker, G., Schorr, A., Hippe, A., et al. (2016). Resident microglia
distinguish microglia (CD45low) from peripheral macrophages rather than peripheral macrophages promote vascularization in brain tumors
and are source of alternative pro-angiogenic factors. Acta Neuropathol. 131,
(CD45high) (Badie and Schartner, 2000), leading to conflicting 365–378.
conclusions regarding the distribution of microglia and macro-
Butowski, N., Colman, H., De Groot, J.F., Omuro, A.M., Nayak, L., Wen, P.Y.,
phages within gliomas. Moreover, microglia increase CD45 Cloughesy, T.F., Marimuthu, A., Haidar, S., Perry, A., et al. (2016).
expression in the context of glioma (Mu € ller et al., 2015), whereas Orally administered colony stimulating factor 1 receptor inhibitor PLX3397 in

446 Neuron 104, November 6, 2019


Neuron

Review
recurrent glioblastoma: an Ivy Foundation Early Phase Clinical Trials Con- other central nervous system tumor from 2000-2014. Neuro-oncol 20 (suppl
sortium phase II study. Neuro-oncol. 18, 557–564. 7), vii6–vii16.

Chen, Y.H., McGowan, L.D., Cimino, P.J., Dahiya, S., Leonard, J.R., Lee, D.Y., Gu, R., Zhang, X., Zhang, G., Tao, T., Yu, H., Liu, L., Dou, Y., Li, A., and Qin, J.
and Gutmann, D.H. (2015). Mouse low-grade gliomas contain cancer stem (2017). Probing the bi-directional interaction between microglia and gliomas in
cells with unique molecular and functional properties. Cell Rep. 10, a tumor microenvironment on a microdevice. Neurochem. Res. 42,
1899–1912. 1478–1487.

Chen, Z., Feng, X., Herting, C.J., Garcia, V.A., Nie, K., Pong, W.W., Rasmus- Guneykaya, D., Ivanov, A., Hernandez, D.P., Haage, V., Wojtas, B., Meyer, N.,
sen, R., Dwivedi, B., Seby, S., Wolf, S.A., et al. (2017). Cellular and molecular Maricos, M., Jordan, Buonfiglioli, A., Gielniewski, B., et al. (2018). Transcrip-
identity of tumor-associated macrophages in glioblastoma. Cancer Res. 77, tional and translational differences of microglia from male and female brains.
2266–2278. Cell Rep. 24, 2773–2783.e2776.

Chen, R., Keoni, C., Waker, C.A., Lober, R.M., Chen, Y.H., and Gutmann, D.H. Guo, X., Pan, Y., and Gutmann, D.H. (2019). Genetic and genomic alterations
(2019a). KIAA1549-BRAF expression establishes a permissive tumor microen- differentially dictate low-grade glioma growth through cancer stem cell-spe-
vironment through NFkB-mediated CCL2 production. Neoplasia 21, 52–60. cific chemokine recruitment of T cells and microglia. Neuro-oncol. noz080.

Chen, P., Zhao, D., Li, J., Liang, X., Li, J., Chang, A., Henry, V.K., Lan, Z., Haage, V., Semtner, M., Vidal, R.O., Hernandez, D.P., Pong, W.W., Chen, Z.,
Spring, D.J., Rao, G., Wang, Y.A., and DePinho, R.A. (2019b). Symbiotic Hambardzumyan, D., Magrini, V., Ly, A., Walker, J., et al. (2019). Comprehen-
macrophage-glioma cell interactions reveal synthetic lethality in PTEN-null gli- sive gene expression meta-analysis identifies signature genes that distinguish
oma. Cancer Cell 35, 868–884.e866. microglia from peripheral monocytes/macrophages in health and glioma. Acta
Neuropathol. Commun. 7, 20.
Chia, K., Mazzolini, J., Mione, M., and Sieger, D. (2018). Tumor initiating cells
induce Cxcr4-mediated infiltration of pro-tumoral macrophages into the brain. Hambardzumyan, D., Gutmann, D.H., and Kettenmann, H. (2016). The role of
eLife 7, e31918. microglia and macrophages in glioma maintenance and progression. Nat.
Neurosci. 19, 20–27.
Daginakatte, G.C., and Gutmann, D.H. (2007). Neurofibromatosis-1 (Nf1) het-
erozygous brain microglia elaborate paracrine factors that promote Nf1-defi- Hamilton, L., Astell, K.R., Velikova, G., and Sieger, D. (2016). A zebrafish live
cient astrocyte and glioma growth. Hum. Mol. Genet. 16, 1098–1112. imaging model reveals differential responses of microglia toward glioblastoma
cells in vivo. Zebrafish 13, 523–534.
Daginakatte, G.C., Gianino, S.M., Zhao, N.W., Parsadanian, A.S., and Gut-
Han, C.J., Zheng, J.Y., Sun, L., Yang, H.C., Cao, Z.Q., Zhang, X.H., Zheng,
mann, D.H. (2008). Increased c-Jun-NH2-kinase signaling in neurofibroma-
L.T., and Zhen, X.C. (2019). The oncometabolite 2-hydroxyglutarate inhibits
tosis-1 heterozygous microglia drives microglia activation and promotes optic
microglial activation via the AMPK/mTOR/NF-kB pathway. Acta Pharmacol.
glioma proliferation. Cancer Res. 68, 10358–10366.
Sin. Published online April 23, 2019. https://doi.org/10.1038/s41401-019-
De, S., Van Deren, D., Peden, E., Hockin, M., Boulet, A., Titen, S., and Capec- 0225-9.
chi, M.R. (2018). Two distinct ontogenies confer heterogeneity to mouse brain
Herisson, F., Frodermann, V., Courties, G., Rohde, D., Sun, Y., Vandoorne, K.,
microglia. Development 145, dev152306.
Wojtkiewicz, G.R., Masson, G.S., Vinegoni, C., Kim, J., et al. (2018). Direct
vascular channels connect skull bone marrow and the brain surface enabling
Dijksterhuis, J.P., Arthofer, E., Marinescu, V.D., Nelander, S., Uhlén, M.,
myeloid cell migration. Nat. Neurosci. 21, 1209–1217.
Pontén, F., Mulder, J., and Schulte, G. (2015). High levels of WNT-5A in human
glioma correlate with increased presence of tumor-associated microglia/ Hu, F., Ku, M.C., Markovic, D., Dzaye, O., Lehnardt, S., Synowitz, M., Wolf,
monocytes. Exp. Cell Res. 339, 280–288. S.A., and Kettenmann, H. (2014). Glioma-associated microglial MMP9 expres-
sion is upregulated by TLR2 signaling and sensitive to minocycline. Int. J. Can-
Dzaye, O., Hu, F., Derkow, K., Haage, V., Euskirchen, P., Harms, C., Lehnardt,
cer 135, 2569–2578.
S., Synowitz, M., Wolf, S.A., and Kettenmann, H. (2016). Glioma stem cells but
not bulk glioma cells upregulate IL-6 secretion in microglia/brain macrophages Hu, F., Dzaye, O., Hahn, A., Yu, Y., Scavetta, R.J., Dittmar, G., Kaczmarek,
via Toll-like receptor 4 signaling. J. Neuropathol. Exp. Neurol. 75, 429–440. A.K., Dunning, K.R., Ricciardelli, C., Rinnenthal, J.L., et al. (2015). Glioma-
derived versican promotes tumor expansion via glioma-associated micro-
Engler, J.R., Robinson, A.E., Smirnov, I., Hodgson, J.G., Berger, M.S., Gupta, glial/macrophages Toll-like receptor 2 signaling. Neuro-oncol. 17, 200–210.
N., James, C.D., Molinaro, A., and Phillips, J.J. (2012). Increased microglia/
macrophage gene expression in a subset of adult and pediatric astrocytomas. Hubert, C.G., Rivera, M., Spangler, L.C., Wu, Q., Mack, S.C., Prager, B.C.,
PLoS ONE 7, e43339. Couce, M., McLendon, R.E., Sloan, A.E., and Rich, J.N. (2016). A three-dimen-
sional organoid culture system derived from human glioblastomas recapitu-
Gargiulo, G. (2018). Next-generation in vivo modeling of human cancers. Front. lates the hypoxic gradients and cancer stem cell heterogeneity of tumors
Oncol. 8, 429. found in vivo. Cancer Res. 76, 2465–2477.
Gholamin, S., Mitra, S.S., Feroze, A.H., Liu, J., Kahn, S.A., Zhang, M., Esparza, Hutter, G., Theruvath, J., Graef, C.M., Zhang, M., Schoen, M.K., Manz, E.M.,
R., Richard, C., Ramaswamy, V., Remke, M., et al. (2017). Disrupting the Bennett, M.L., Olson, A., Azad, T.D., Sinha, R., et al. (2019). Microglia are
CD47-SIRPa anti-phagocytic axis by a humanized anti-CD47 antibody is an effector cells of CD47-SIRPa antiphagocytic axis disruption against glioblas-
efficacious treatment for malignant pediatric brain tumors. Sci. Transl. Med. toma. Proc. Natl. Acad. Sci. USA 116, 997–1006.
9, eaaf2968.
Isakson, S.H., Rizzardi, A.E., Coutts, A.W., Carlson, D.F., Kirstein, M.N.,
Ghoochani, A., Yakubov, E., Sehm, T., Fan, Z., Hock, S., Buchfelder, M., Fisher, J., Vitte, J., Williams, K.B., Pluhar, G.E., Dahiya, S., et al. (2018). Genet-
€poglu, I.Y., and Savaskan, N.E. (2016). A versatile ex vivo technique for as-
Eyu ically engineered minipigs model the major clinical features of human neurofi-
saying tumor angiogenesis and microglia in the brain. Oncotarget 7, bromatosis type 1. Commun. Biol. 1, 158.
1838–1853.
Jacob, K., Quang-Khuong, D.A., Jones, D.T., Witt, H., Lambert, S., Albrecht,
Gibson, E.M., Nagaraja, S., Ocampo, A., Tam, L.T., Wood, L.S., Pallegar, P.N., S., Witt, O., Vezina, C., Shirinian, M., Faury, D., et al. (2011). Genetic aberra-
Greene, J.J., Geraghty, A.C., Goldstein, A.K., Ni, L., et al. (2019). Methotrexate tions leading to MAPK pathway activation mediate oncogene-induced senes-
chemotherapy induces persistent tri-glial dysregulation that underlies chemo- cence in sporadic pilocytic astrocytomas. Clin. Cancer Res. 17, 4650–4660.
therapy-related cognitive impairment. Cell 176, 43–55.e13.
Juliano, J., Gil, O., Hawkins-Daarud, A., Noticewala, S., Rockne, R.C., Gal-
Ginhoux, F., Greter, M., Leboeuf, M., Nandi, S., See, P., Gokhan, S., Mehler, laher, J., Massey, S.C., Sims, P.A., Anderson, A.R.A., Swanson, K.R., and
M.F., Conway, S.J., Ng, L.G., Stanley, E.R., et al. (2010). Fate mapping analysis Canoll, P. (2018). Comparative dynamics of microglial and glioma cell motility
reveals that adult microglia derive from primitive macrophages. Science 330, at the infiltrative margin of brain tumours. J. R. Soc. Interface 15, 20170582.
841–845.
Jung, I.H., Leem, G.L., Jung, D.E., Kim, M.H., Kim, E.Y., Kim, S.H., Park, H.C.,
Gittleman, H., Boscia, A., Ostrom, Q.T., Truitt, G., Fritz, Y., Kruchko, C., and and Park, S.W. (2013). Glioma is formed by active Akt1 alone and promoted by
Barnholtz-Sloan, J.S. (2018). Survivorship in adults with malignant brain and active Rac1 in transgenic zebrafish. Neuro-oncol. 15, 290–304.

Neuron 104, November 6, 2019 447


Neuron

Review
Kaffes, I., Szulzewsky, F., Chen, Z., Herting, C.J., Gabanic, B., Velázquez nervous system tumors diagnosed in the United States in 2011-2015.
Vega, J.E., Shelton, J., Switchenko, J.M., Ross, J.L., McSwain, L.F., et al. Neuro-oncol 20 (suppl 4), iv1–iv86.
(2019). Human Mesenchymal glioblastomas are characterized by an increased
immune cell presence compared to proneural and classical tumors. OncoIm- Pan, Y., Smithson, L.J., Ma, Y., Hambardzumyan, D., and Gutmann, D.H.
munology. Published online August 22, 2019. https://doi.org/10.1080/ (2017a). Ccl5 establishes an autocrine high-grade glioma growth regulatory
2162402X.2019.1655360. circuit critical for mesenchymal glioblastoma survival. Oncotarget 8,
32977–32989.
Keren-Shaul, H., Spinrad, A., Weiner, A., Matcovitch-Natan, O., Dvir-Sztern-
feld, R., Ulland, T.K., David, E., Baruch, K., Lara-Astaiso, D., Toth, B., et al. Pan, Y., Bush, E.C., Toonen, J.A., Ma, Y., Solga, A.C., Sims, P.A., and Gut-
(2017). A unique microglia type associated with restricting development of Alz- mann, D.H. (2017b). Whole tumor RNA-sequencing and deconvolution reveal
heimer’s disease. Cell 169, 1276–1290.e1217. a clinically-prognostic PTEN/PI3K-regulated glioma transcriptional signature.
Oncotarget 8, 52474–52487.
Kettenmann, H., Hanisch, U.K., Noda, M., and Verkhratsky, A. (2011). Physi-
ology of microglia. Physiol. Rev. 91, 461–553. Pan, Y., Xiong, M., Chen, R., Ma, Y., Corman, C., Maricos, M., Kindler, U.,
Semtner, M., Chen, Y.H., Dahiya, S., and Gutmann, D.H. (2018). Athymic
Khoshnevis, M., Carozzo, C., Bonnefont-Rebeix, C., Belluco, S., Leveneur, O., mice reveal a requirement for T-cell-microglia interactions in establishing a
Chuzel, T., Pillet-Michelland, E., Dreyfus, M., Roger, T., Berger, F., and Ponce, microenvironment supportive of Nf1 low-grade glioma growth. Genes Dev.
F. (2017). Development of induced glioblastoma by implantation of a human 32, 491–496.
xenograft in Yucatan minipig as a large animal model. J. Neurosci. Methods
282, 61–68. Penfield, W. (1925). Microglia and the process of phagocytosis in glioma. Am.
J. Pathol. 1, 77–90, 15.
Larribere, L., Wu, H., Novak, D., Galach, M., Bernhardt, M., Orouji, E., Weina,
K., Knappe, N., Sachpekidis, C., Umansky, L., et al. (2015). NF1 loss induces Pfister, S., Janzarik, W.G., Remke, M., Ernst, A., Werft, W., Becker, N., Toedt,
senescence during human melanocyte differentiation in an iPSC-based G., Wittmann, A., Kratz, C., Olbrich, H., et al. (2008). BRAF gene duplication
model. Pigment Cell Melanoma Res. 28, 407–416. constitutes a mechanism of MAPK pathway activation in low-grade astrocy-
tomas. J. Clin. Invest. 118, 1739–1749.
Li, F., Lv, B., Liu, Y., Hua, T., Han, J., Sun, C., Xu, L., Zhang, Z., Feng, Z., Cai,
Y., et al. (2017). Blocking the CD47-SIRPa axis by delivery of anti-CD47 anti- Platten, M., Kretz, A., Naumann, U., Aulwurm, S., Egashira, K., Isenmann, S.,
body induces antitumor effects in glioma and glioma stem cells. OncoImmu- and Weller, M. (2003). Monocyte chemoattractant protein-1 increases micro-
nology 7, e1391973. glial infiltration and aggressiveness of gliomas. Ann. Neurol. 54, 388–392.

Li, Q., Cheng, Z., Zhou, L., Darmanis, S., Neff, N.F., Okamoto, J., Gulati, G., Pong, W.W., Higer, S.B., Gianino, S.M., Emnett, R.J., and Gutmann, D.H.
Bennett, M.L., Sun, L.O., Clarke, L.E., et al. (2019). Developmental heteroge- (2013a). Reduced microglial CX3CR1 expression delays neurofibromatosis-1
neity of microglia and brain myeloid cells revealed by deep single-cell RNA glioma formation. Ann. Neurol. 73, 303–308.
sequencing. Neuron 101, 207–223.e210.
Pong, W.W., Walker, J., Wylie, T., Magrini, V., Luo, J., Emnett, R.J., Choi, J.,
Linkous, A., Balamatsias, D., Snuderl, M., Edwards, L., Miyaguchi, K., Milner, Cooper, M.L., Griffith, M., Griffith, O.L., et al. (2013b). F11R is a novel mono-
T., Reich, B., Cohen-Gould, L., Storaska, A., Nakayama, Y., et al. (2019). cyte prognostic biomarker for malignant glioma. PLoS ONE 8, e77571.
Modeling patient-derived glioblastoma with cerebral organoids. Cell Rep.
26, 3203–3211.e3205. Pyonteck, S.M., Akkari, L., Schuhmacher, A.J., Bowman, R.L., Sevenich, L.,
Quail, D.F., Olson, O.C., Quick, M.L., Huse, J.T., Teijeiro, V., et al. (2013).
Listernick, R., Charrow, J., Greenwald, M.J., and Esterly, N.B. (1989). Optic gli- CSF-1R inhibition alters macrophage polarization and blocks glioma progres-
omas in children with neurofibromatosis type 1. J. Pediatr. 114, 788–792. sion. Nat. Med. 19, 1264–1272.

Louis, D.N., Perry, A., Reifenberger, G., von Deimling, A., Figarella-Branger, D., Qian, J., Luo, F., Yang, J., Liu, J., Liu, R., Wang, L., Wang, C., Deng, Y., Lu, Z.,
Cavenee, W.K., Ohgaki, H., Wiestler, O.D., Kleihues, P., and Ellison, D.W. Wang, Y., et al. (2018). TLR2 promotes glioma immune evasion by downregu-
(2016). The 2016 World Health Organization Classification of Tumors of the lating MHC class II molecules in microglia. Cancer Immunol. Res. 6,
Central Nervous System: a summary. Acta Neuropathol. 131, 803–820. 1220–1233.

Markovic, D.S., Vinnakota, K., van Rooijen, N., Kiwit, J., Synowitz, M., Glass, Qian, X., Song, H., and Ming, G.L. (2019). Brain organoids: advances, applica-
R., and Kettenmann, H. (2011). Minocycline reduces glioma expansion and in- tions and challenges. Development 146, dev166074.
vasion by attenuating microglial MT1-MMP expression. Brain Behav. Immun.
25, 624–628. Reilly, M., Miller, R.M., Thomson, M.H., Patris, V., Ryle, P., McLoughlin, L.,
Mutch, P., Gilboy, P., Miller, C., Broekema, M., et al. (2013). Randomized, dou-
Masuda, T., Sankowski, R., Staszewski, O., Böttcher, C., Amann, L., Sagar, ble-blind, placebo-controlled, dose-escalating phase I, healthy subjects study
Scheiwe, C., Nessler, S., Kunz, P., van Loo, G., et al. (2019). Spatial and tem- of intravenous OPN-305, a humanized anti-TLR2 antibody. Clin. Pharmacol.
poral heterogeneity of mouse and human microglia at single-cell resolution. Ther. 94, 593–600.
Nature 566, 388–392.
Resende, F.F., Bai, X., Del Bel, E.A., Kirchhoff, F., Scheller, A., and Titze-de-Al-
Monje, M.L., Mizumatsu, S., Fike, J.R., and Palmer, T.D. (2002). Irradiation in- meida, R. (2016). Evaluation of TgH(CX3CR1-EGFP) mice implanted with
duces neural precursor-cell dysfunction. Nat. Med. 8, 955–962. mCherry-GL261 cells as an in vivo model for morphometrical analysis of gli-
oma-microglia interaction. BMC Cancer 16, 72.
Monje, M.L., Toda, H., and Palmer, T.D. (2003). Inflammatory blockade re-
stores adult hippocampal neurogenesis. Science 302, 1760–1765. Roesch, S., Rapp, C., Dettling, S., and Herold-Mende, C. (2018). When im-
mune cells turn bad-tumor-associated microglia/macrophages in glioma. Int.
Mu€ller, A., Brandenburg, S., Turkowski, K., Mu
€ller, S., and Vajkoczy, P. (2015). J. Mol. Sci. 19, E436.
Resident microglia, and not peripheral macrophages, are the main source of
brain tumor mononuclear cells. Int. J. Cancer 137, 278–288. Selek, L., Seigneuret, E., Nugue, G., Wion, D., Nissou, M.F., Salon, C., Seurin,
M.J., Carozzo, C., Ponce, F., Roger, T., and Berger, F. (2014). Imaging and his-
Mu€ller, S., Kohanbash, G., Liu, S.J., Alvarado, B., Carrera, D., Bhaduri, A., tological characterization of a human brain xenograft in pig: the first induced
Watchmaker, P.B., Yagnik, G., Di Lullo, E., Malatesta, M., et al. (2017). Sin- glioma model in a large animal. J. Neurosci. Methods 221, 159–165.
gle-cell profiling of human gliomas reveals macrophage ontogeny as a basis
for regional differences in macrophage activation in the tumor microenviron- Sierra, A., de Castro, F., Del Rı́o-Hortega, J., Rafael Iglesias-Rozas, J., Gar-
ment. Genome Biol. 18, 234. rosa, M., and Kettenmann, H. (2016). The ‘‘Big-Bang’’ for modern glial biology:
translation and comments on Pı́o del Rı́o-Hortega 1919 series of papers on
Ogawa, J., Pao, G.M., Shokhirev, M.N., and Verma, I.M. (2018). Glioblastoma microglia. Glia 64, 1801–1840.
model using human cerebral organoids. Cell Rep. 23, 1220–1229.
Simmons, G.W., Pong, W.W., Emnett, R.J., White, C.R., Gianino, S.M., Rodri-
Ostrom, Q.T., Gittleman, H., Truitt, G., Boscia, A., Kruchko, C., and Barnholtz- guez, F.J., and Gutmann, D.H. (2011). Neurofibromatosis-1 heterozygosity in-
Sloan, J.S. (2018). CBTRUS statistical report: primary brain and other central creases microglia in a spatially and temporally restricted pattern relevant to

448 Neuron 104, November 6, 2019


Neuron

Review
mouse optic glioma formation and growth. J. Neuropathol. Exp. Neurol. Vinnakota, K., Hu, F., Ku, M.C., Georgieva, P.B., Szulzewsky, F., Pohlmann, A.,
70, 51–62. Waiczies, S., Waiczies, H., Niendorf, T., Lehnardt, S., et al. (2013). Toll-like re-
ceptor 2 mediates microglia/brain macrophage MT1-MMP expression and gli-
Solga, A.C., Pong, W.W., Kim, K.Y., Cimino, P.J., Toonen, J.A., Walker, J., Wy- oma expansion. Neuro-oncol. 15, 1457–1468.
lie, T., Magrini, V., Griffith, M., Griffith, O.L., et al. (2015). RNA sequencing of
tumor-associated microglia reveals Ccl5 as a stromal chemokine critical for
Wallmann, T., Zhang, X.M., Wallerius, M., Bolin, S., Joly, A.L., Sobocki, C.,
neurofibromatosis-1 glioma growth. Neoplasia 17, 776–788.
Leiss, L., Jiang, Y., Bergh, J., Holland, E.C., et al. (2018). Microglia induce
Sørensen, M.D., Dahlrot, R.H., Boldt, H.B., Hansen, S., and Kristensen, B.W. PDGFRB expression in glioma cells to enhance their migratory capacity.
(2018). Tumour-associated microglia/macrophages predict poor prognosis iScience 9, 71–83.
in high-grade gliomas and correlate with an aggressive tumour subtype. Neu-
ropathol. Appl. Neurobiol. 44, 185–206. Xia, S., Lal, B., Tung, B., Wang, S., Goodwin, C.R., and Laterra, J. (2016). Tu-
mor microenvironment tenascin-C promotes glioblastoma invasion and nega-
Szulzewsky, F., Pelz, A., Feng, X., Synowitz, M., Markovic, D., Langmann, T., tively regulates tumor proliferation. Neuro-oncol. 18, 507–517.
Holtman, I.R., Wang, X., Eggen, B.J., Boddeke, H.W., et al. (2015). Glioma-
associated microglia/macrophages display an expression profile different
Yan, C., Brunson, D.C., Tang, Q., Do, D., Iftimia, N.A., Moore, J.C., Hayes,
from M1 and M2 polarization and highly express Gpnmb and Spp1. PLoS M.N., Welker, A.M., Garcia, E.G., Dubash, T.D., et al. (2019). ). Visualizing en-
ONE 10, e0116644.
grafted human cancer and therapy responses in immunodeficient zebrafish.
€neykaya, D., Cimino, P.J., Hambard- Cell 177, 1903–1914.e1914.
Szulzewsky, F., Schwendinger, N., Gu
zumyan, D., Synowitz, M., Holland, E.C., and Kettenmann, H. (2018). Loss of
host-derived osteopontin creates a glioblastoma-promoting microenviron- Yu, J., Deshmukh, H., Gutmann, R.J., Emnett, R.J., Rodriguez, F.J., Watson,
ment. Neuro-oncol. 20, 355–366. M.A., Nagarajan, R., and Gutmann, D.H. (2009). Alterations of BRAF and
HIPK2 loci predominate in sporadic pilocytic astrocytoma. Neurology 73,
Thion, M.S., Low, D., Silvin, A., Chen, J., Grisel, P., Schulte-Schrepping, J., 1526–1531.
Blecher, R., Ulas, T., Squarzoni, P., Hoeffel, G., et al. (2018). Microbiome
influences prenatal and adult microglia in a sex-specific manner. Cell 172, Yu, K., Youshani, A.S., Wilkinson, F.L., O’Leary, C., Cook, P., Laaniste, L., Liao,
500–516.e516. A., Mosses, D., Waugh, C., Shorrock, H., et al. (2019). A nonmyeloablative
chimeric mouse model accurately defines microglia and macrophage contri-
Toonen, J.A., Solga, A.C., Ma, Y., and Gutmann, D.H. (2017). Estrogen activa-
bution in glioma. Neuropathol. Appl. Neurobiol. 45, 119–140.
tion of microglia underlies the sexually dimorphic differences in Nf1 optic
glioma-induced retinal pathology. J. Exp. Med. 214, 17–25.
Zeiner, P.S., Preusse, C., Golebiewska, A., Zinke, J., Iriondo, A., Muller, A.,
Villa, A., Gelosa, P., Castiglioni, L., Cimino, M., Rizzi, N., Pepe, G., Lolli, F., €ller-Eschner, M., Bernatz, S., et al. (2019). Distribu-
Kaoma, T., Filipski, K., Mu
Marcello, E., Sironi, L., Vegeto, E., and Maggi, A. (2018). Sex-specific features tion and prognostic impact of microglia/macrophage subpopulations in gli-
of microglia from adult mice. Cell Rep. 23, 3501–3511. omas. Brain Pathol. 29, 513–529.

Neuron 104, November 6, 2019 449

You might also like