Page 338 - Read Online
P. 338

Page 10 of 14                Choi et al. Neuroimmunol Neuroinflammation 2018;5:42  I  http://dx.doi.org/10.20517/2347-8659.2018.47


               in peri-tumoral areas and instead resulted in a significant increase in macrophage recruitment from the
               periphery and subsequent infiltration. Of key importance is that deletion of Cx3cr1 in mice saw an increase
               in IL-1β production from both microglia and macrophages, implicating this receptor in the suppression of
                             [79]
               IL-1β production .
               The importance of IL-1β lies in the IL-1β/CCL2/IL-6 interaction between microglia and glioma cells [Figure 2A-C].
               Specifically, IL-1β released from both microglia and macrophages activates the p38 mitogen-activated protein
               kinase (MAPK) pathway in glioma cells, which in turn results in increased expression of CCL2-the agonist
                                          [80]
               for CCR2 on microglia [Table 1] . This results in an increase in microglial production of IL-6 and eventually
               MMP-2, which facilitates tumor migration, invasion, and gliomagenesis [Figure 2C] [81,82] . The CX3CR1/IL-1β/
               CCL2 pathway continues to be an area of active interest, particularly in regard to reduction of IL-6 pro-tumor
                                                                                  [80]
               signaling and inhibition of MMP-based pathways in microglia [Figure 2C and D] .
               Similarly, studies have examined the p38 MAPK pathway in microglia and its potential for anti-tumor ther-
               apy. Minocycline, a tetracycline that inhibits the p38 MAPK pathway, appears to counteract the pro-tumor
               phenotype of microglia and reduce tumor growth in vitro and in vivo by inhibiting downstream microglial
               MT1-MMP expression in mouse models. Decrease in MT1-MMP expression is in turn associated with de-
               creased MMP-2 activity, which follows a similar treatment schema as mentioned above in the IL-1β/CCL2/
               IL-6 pathway [Figure 2C] [83,84] .

               Also stimulating secretion of CCL2, IL-6, IL-1, and NO is TNFα, which is readily produced by glioma-
               associated microglia [79,85] . TNF receptor 1 (TNFR1) activation leads to the degradation of IκBa, an inhibitor
               of NFκB signaling. This degradation leads to a positive feedback loop with p65/p50 nuclear translocation
               and subsequent transcriptional activation of TNFα [86,87] . Meanwhile, NFκB activation also activates pro-
               migratory genes that contribute to tumor invasiveness involving several pro-tumor chemokines and MMP
                       [88]
               pathways . As such, there is ongoing interest in targeting TNFR1 and the related NFκB pathway in
                       [86]
               microglia .
               Moreover, GBM cells induce TLR2/6 activation in both macrophages and microglia via the myeloid
               differentiation primary response 88/TLR8 signaling pathway, which in turn leads to an increase in
                                                                                                  [89]
               metalloproteinases like MMP-9 that facilitate tumor invasion and angiogenesis [Figure 2C and D] . TLR2
               on microglia is also directly involved with promoting tumor invasion with downstream production of MT1-
                    [90]
               MMP . Studies done with murine GL261 glioma cells injected into TLR2 KO mice resulted in smaller
                                                                               [90]
               tumor burden and reduced MT1-MMP levels in glioma-associated microglia .

               Contributing further empiric credence to the idea that macrophages and microglia are distinct populations
                                    [91]
               in the TME, Jacobs et al.  found that propentofylline, a methylxanthine, directly acts only on microglia-and
               not on macrophages-through tumor necrosis factor receptor superfamily, member 19 (TNFRSF19)/TROY
               inhibition. In a rat glioma model, they demonstrated that TROY is upregulated in infiltrating microglia, with
               downstream expression of pro-tumor genes Pyk2, Rac1, and pJNK. The potential effectiveness of targeting
               TROY was shown through a series of siRNA experiments that resulted in inhibition of microglial migration
               towards glioma cells and, as a result, decreased pro-tumor activity [Figure 2C].

               Another important pathway involves signal transducer and activator of transcription 3 (STAT3), which
               is upregulated in both GBM and glioma-associated microglia and is associated with GBM pathogenesis,
                                                                    [95]
               progression, and immune evasion [92-94] . A study by Lisi et al.  in 2014 examined the use of rapamycin
               to inhibit mTOR in glioma; the result was the reversal of pro-tumor functions in microglia with glioma-
               specific cytotoxic behavior. Normally, mTOR leads to an increase in STAT3 expression, which in turn results
               in increased production of pro-tumor cytokines IL-6, IL-10, CXCL2, and HIF-1α [Figure 2C-E]. With mTOR
   333   334   335   336   337   338   339   340   341   342   343