Next Article in Journal
Inhibitors of DNA Glycosylases as Prospective Drugs
Previous Article in Journal
The Cannabinoid Receptor CB1 Stabilizes Sperm Chromatin Condensation Status During Epididymal Transit by Promoting Disulphide Bond Formation
Previous Article in Special Issue
A Novel Highly Sensitive Method for Measuring Inflammatory Neural-Derived APC Activity in Glial Cell Lines, Mouse Brain and Human CSF
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

TLR-4 Signaling vs. Immune Checkpoints, miRNAs Molecules, Cancer Stem Cells, and Wingless-Signaling Interplay in Glioblastoma Multiforme—Future Perspectives

1
Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, 20-954 Lublin, Poland
2
Department of Immunology, Medical University of Lublin, 20-093 Lublin, Poland
3
Department of Anatomy, Medical University of Lublin, 20-090 Lublin, Poland
4
Laboratory of Virtual Man, Department of Anatomy, Medical University of Lublin, 20-090 Lublin, Poland
5
St. John‘s Cancer Center in Lublin, 20-090 Lublin, Poland
6
MSWiA Hospital, 02-507 Warsaw, Poland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2020, 21(9), 3114; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21093114
Submission received: 20 March 2020 / Revised: 23 April 2020 / Accepted: 24 April 2020 / Published: 28 April 2020
(This article belongs to the Special Issue Neuroimmunology)

Abstract

:
Toll-like-receptor (TLR) family members were detected in the central nervous system (CNS). TLR occurrence was noticed and widely described in glioblastomamultiforme (GBM) cells. After ligand attachment, TLR-4 reorients domains and dimerizes, activates an intracellular cascade, and promotes further cytoplasmatic signaling. There is evidence pointing at a strong relation between TLR-4 signaling and micro ribonucleic acid (miRNA) expression. The TLR-4/miRNA interplay changes typical signaling and encourages them to be a target for modern immunotherapy. TLR-4 agonists initiate signaling and promote programmed death ligand-1 (PD-1L) expression. Most of those molecules are intensively expressed in the GBM microenvironment, resulting in the autocrine induction of regional immunosuppression. Another potential target for immunotreatment is connected with limited TLR-4 signaling that promotes Wnt/DKK-3/claudine-5 signaling, resulting in a limitation of GBM invasiveness. Interestingly, TLR-4 expression results in bordering proliferative trends in cancer stem cells (CSC) and GBM. All of these potential targets could bring new hope for patients suffering from this incurable disease. Clinical trials concerning TLR-4 signaling inhibition/promotion in many cancers are recruiting patients. There is still a lot to do in the field of GBM immunotherapy.

1. Introduction

Various toll-like-receptor (TLR) family members are detected in the central nervous system (CNS). They are mainly expressed in neurons and glial structures, where they play a role in recognizing unfamiliar molecules, some postapoptotic antigens consequently control repair processes and modulate inflammatory action [1].
Furthermore, TLR occurrence was noticed and widely described in glioblastoma-multiforme (GBM) mature cell fractions [2]. Studies revealed a strong expression of these receptors in cohorts of neural non-pro-oncogenic stem cells [3,4]. GBM, as the most popular and expansive glioma form, with a mean survival of 14.6 months, remains a challenge for modern therapy [5]. The characteristics of TLRs make them a promising target for GBM immunotreatment. Activated receptors stimulate the response of the immune system and control the course of many diseases, including cancers [6,7,8,9]. The necrotic ratio during glioblastoma invasion remains high. Products of cell breakdown intensively interact with the transmembrane architecture of TLRs and promote differentiation and inflammatory signaling [10]. In brain tissue, TLR-4 is detected in two main types of cells: microglial (macrophages) and macroglial cells (oligodendrocytes and astrocytes). Microglia do not intensively express TLR-4 on the surface (less than 15%), and receptors are easier to detect intracellularly. Macroglial cells superficially express TLR-4. Oligodendrocytes and astrocytes do not intracellularly express TLR-4 at all. This difference in expression could be explained by the different phagocytic functions of micro- and macroglial cells (Table 1) [11,12].
The TLR family is a group of ten receptors (TLR-1–TLR-10) characterized by the detection of a particular pattern of micro-organisms (pathogen-associated molecular patterns (PAMPs)), which are invariable for most pathogens and not present in mammalian organisms. TLR receptors are also sensitive to particles secreted during necrosis and cell death called danger-associated molecular patterns (DAMPs) [20,21,22,23]. Typically, TLRs are grouped into two major categories, endosomal (TLR-9, TLR-8, TLR-7, and TLR-3) and cell-surface-acting (TLR-10, TLR-6, TLR-5, TLR-4, TLR-2, and TLR-1) [24,25,26]. TLRs functioning endosomally are mainly activated with nucleic acids. On the other hand, a variety of molecules activate TLRs expressed on the cell surface. Most of them include lipoproteins. After ligand attachment, TLRs reorient domains and dimerize, activate intracellular cascade, and promote further cytoplasmatic signaling [27,28]. Immunotherapeutic agents aim at this activation chain, targeting immune-related disorders. Many autoregulating mechanisms control inflammation that is mediated through TLR signaling. Their activity concerns the nucleus level (countering the expression of cytokines and interleukins (TTP, ATF3, and REG-1) and the cytoplasmatic level (STAT1, AhR, Nurr1); the inhibition of adaptor complex suppressor of cytokine signaling, sterile alpha-and armadillo-motif-containing protein (SOCS1, SARM); and the cell-surface level interrupting dimerization processes: suppressor of tumorigenicity 2, single immunoglobulin IL-1R-related molecule, Rickettsia prowazekii 105 (ST2, SIGGR, and RP105) [29,30,31,32,33]. Moreover, some studies revealed microRNA molecules over activity destabilizing the mRNA of various cytokines. MiR-155-5p targets the MyD88 complex, similarly to MiR 203-5p and MiR 149-5p [30,34]. The described mechanisms of autocontrol ensure an adequate reaction to microbe-associated molecular patterns (MAMPs) and danger-associated molecular patterns (DAMPs), protecting autoimmunity and excessive response. TLR-4, after particular antigen attachment to a pathogen-associated molecular patterns (PAMP) or DAMP, is shifted from the cell surface to form the endosome during phagocytosis (Table 2).

2. TLR-4 Overview

Toll receptors (TRs) were detected in drosophila embryos by Hashimoto et al. (1988) [35,36]. Hoffmann and their study team proved that TR mutation in drosophila raised the risk of fungal infection. The above experiments indicated that the innate immune response has the ability to precisely detect the invasion of bacteria and other micro-organisms [37]. Further studies identified mammalian TR homologs that researchers called toll-like receptors (TLRs). The first, discovered by Medzhitov R. and colleagues in 1997, was named TLR-4 [38,39]. The TLR-4 gene is localized on the SSC9:119.5, 9q32-33 chromosome, structured with three exons. The receptor is built of four domains and 838 amino acids. The first, the extracellular, consists of 624 amino acid molecules; the second, the transmembrane, consists of 33 amino acids; the third, the cytoplasmatic proximal, of 159 amino acids; and the cytoplasmatic distal of 19 amino acids. The ectodomain is built of 21 leucine-rich repeat regions (LRRs), part of the extracellular domain [40,41,42]. TLR-4 presents multiple polymorphism phenomena concerning single nucleotides (SNPs). Most of them, recognized in the ectodomain, promote detrimental phenotypic outcomes [43,44].

2.1. TLR-4 Signaling

To bind lipopolysaccharides, TLR-4 requires co-operating molecule myeloid differentiation 2 molecule (MD-2), extracellularly stabilizing the ligand during activation. After antigen ligation, the receptor dimerizes and activates TIR domains; to properly signal, TLRs require MyD88, an essential adaptor activated during the first stages after pattern ligation to promote an immune response. Activation of the transmembrane receptoral structure of TLR-4 transfers signaling in two major ways, the TIRAP–MyD88-dependent pathway controlling primary nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) promotion, and the combined production of inflammatory cytokines. Activation of MyD88 results in IL-1R-associated kinase 1/TNFR-associated factor 6 (IRAK-1/TRAF-6) induction with collateral ubiquitination, and TRAF-6 promotes Transforming growth factor-beta-activated kinase 1 (TAK-1). Activation of TAK-1 results in the formation and promotion of the IKK/NF-κB complex. Mitogen-activated protein kinases-c-Jun. N-terminal kinase (MAPK-JNK) and extracellular signaling-regulated kinase (ERK1/2) kinases are also activated, which indicates activated protein-1 (Ap-1). Downsignaling promotes transcription factors, such as activated Ap1, stabilizing many genes with regulatory properties during inflammation [45,46,47].
The second toll-like receptor adaptor molecule-1-translocating chain-associating membrane (TRIF-TRAM) signaling pathway (MyD88-independent) activates interferon regulatory factor-3 (IRF-3). TRAM signaling, through TRIF activation, results in TRAF-3 and TRAF-6 promotion (Figure 1); RIP is recruited by TRAF-6, and RIP signaling is activated on TAK-1/ERK1/2/Ap-1 axis. TRAF-3 activates IKK and TBK-1 to activate IRF-3. IRF-3 as a transcription factor that upregulates genes, coding mainly the first types of IFNs and other proinflammatory cytokines. The MyD88-independent pathway stimulates TNFα secretion and production. The sequential ligation of TNFa to a proper receptor favors NF-κB promotion. Therefore, the MyD88-independent pathway induces the activation of NF-κB in a late phase via IRF-3 and the secretion of TNFα. Proinflammatory molecules, such as IFN, pro-IL-1, and pro-IL-6, are also promoted [48,49,50,51,52].

2.2. Progression of GBM

Numerous signaling pathways induce carcinogenesis, oncogenic transformation, and invasiveness of cancer cells. Interestingly, some of them promote NF-κB expression. NF-κB, as an important transcription factor, is involved in various cellular processes, including migration, proliferation, and survival. Inflammatory cytokines, PAMP and DAMP, could trigger further pro- NF-κB signaling throughout the activation of TNFRs and TLRs [53,54,55,56]. The deregulated activity of NF-κB is becoming an indicator of most neoplasm processes, including GBM. Evidence emphasizes TLRs induced carcinogenesis connected with NF-κB expression [57,58]. Kina et al. and Ferrandez et al., in studies concerning Glioma, confirmed the participation of the TLR-4/NF-κB pathway in the promotion of carcinogenesis [59,60]. Another interesting relation in glioma genesis is TLR-4/Programmed death receptor-1/Programmed death receptor-1 ligand (PD-1/PD-1L) axis interaction. Overexpression of TLR-4 results in an increase of PD-1L in GBM patients and is associated with unfavorable outcome [61]. Additionally, the increase inTLR-4 expression results in Wnt/claudine signaling forwarding to the progression of GBM and limits the effective apoptosis [62]. Progression of glioblastoma is also controlled and modulated by multiple miRNAs molecules. They control the cell circle of glioma cells on different stages of development. The invasiveness of GBM involves the change in epithelial cells polarity and loss of cell–cell adhesive properties. Down expression of E-cadherins results in modifications in the architecture of glioma cells, loss of integrity, and at last, tumor invasion. Increased expression of miR-10-b, miR-29, miR-146 accelerate invasion of GBM and predict an inauspicious outcome. Overexpression of miR-21 inhibits apoptosis and promotes GBM cell survival. miR 210-3p and miR- 93 promote angiogenesis. Moreover, miR 130b, miR 140, and miR 184 are intensively expressed in cases of histological progression of glioma [63,64,65,66,67,68,69,70]. TLR-4 signaling is modulated by multiple miRNAs molecules, and a great number of them inhibit this pathway. Xu et al. challenged the thesis that long noncoding RNA UBE2R2-AS1 targets TLR-4/miR 877-3p and promotes apoptosis and reduces invasiveness in glioma tissue. The study revealed that UBE2R2-AS1 targets miR 877-3p, inhibits its limiting effect on TLR-4, and as a result, promotes TLR-4 dependent apoptosis [71].

3. Potential Immunotherapeutic Targets

Developments made in neuroimmunology and tumor biology revealed interesting properties of TLR. Activation and TLR-4 downsignaling seem to play an interesting role in the promotion of glioma growth and invasion. On the other hand, some studies discovered that, in some special conditions, they could take part in the anticancer response. The release of HMGB1 from GBM dead cells during radio and chemotherapy stimulates the TLR4 pathway. It leads directly to Dendritic Cell maturation and efficient tumor antigen presentation leading to GBM regression in animal models [72,73,74,75,76,77]. There is adequate evidence clarifying that the modulant of TLR-4 proves efficiency and safety for GBM treatment based on previous trials concerning different neoplasms. The data imply that TLR-4 signaling has a dual disposition as a double-edged sword [61,78]. Taking control over this complex interplay could bring a satisfactory result in GBM matters. miRNAs, immune checkpoints, the Wnt axis, and glioma stem cells (SCS) could be used as a path to the complicated goal of effective glioblastoma therapy.

3.1. miRNA/TLR-4 Interplay

miRNAs represent a group of short noncoding strands of RNA with length oscillating 20–22 nucleotides each. Around 30% of all genes are controlled and regulated by miRNAs. Molecules bind to the targeted mRNA with the 3′ region called the untranslated (UTR) monitoring the expression of particular genes through degradation or translation breakdown. As a result, miRNAs are engaged in various biological processes, such as homeostasis, growth, differentiation, immune activation, apoptosis, and stress response. Otherwise, they modulate the immune response to pathogen invasion throughout the TLR signaling pathway (modulation of cytokines, transcription factors, and signaling proteins and receptors). Additionally, miRNA activity is similarly detected in normal healthy tissue in chronic diseases and cancers [79,80,81,82,83,84,85,86,87].
Levels of miRNAs are significantly higher in GBM tissue and GBM cell lines [87,88,89,90,91,92]. The impact of those molecules in carcinogenesis is still unclear, requiring further investigation [93]. There is evidence pointing at a strong relation between TLR-4 signaling and miRNA expression. TLR-4/miRNA interplay changes the typical balance between immune response and inhibition. The indirect influence of mRNAs concerning TLR downsignaling was represented by the study by Taganov et al. The study team documented lipopolysaccharide-induced expression of miR-132, miR-155, and miR 146a, regulating TLR downsignaling [94,95,96], Another study connected miR-34a expression with significant antitumor properties in glioma p-53 mutant cell line U251. Overexpression of miR-34a induces apoptosis and inhibits the growth of glioma through the activation of numerous particles. A similar effect of miR-34a was revealed by Xu et al. while investigating breast cancer. In their study, the miR-34a molecule was found to inhibit C-X-C motif ligand 10 (CXCL10). The ligand is characterized by a strong affinity to TLR, promoting cancerogenic downsignaling [97,98]. Indirect inhibition of TLR signaling results in the regression of tumor growth and expansion. Direct TLR-4 inhibition is induced by different miRNAs [99,100,101,102]. miRNA molecules directly develop inhibitory features, in particular during stages of TLR-4 activation. The MyD-88-dependent pathway IRAK1–TRAF6 complex is inhibited by miR 93-5p, miR 302b-5p, miR 124-5p, and miR 146a/b-5p. The TAK-1 complex is inhibited by miR 23b-5p, miR 142-3p, miR 155-5p, and miR 23-5p. Signaling through the Myd88-independent pathway is regulated by miR3178-5p and miR 3473-5p as inhibitors of TRAF-3, and miR 21b-3p, miR 146a-5p, and miR 302c-5p as direct inhibitors of IRF-3 [94,103,104,105,106,107,108]. TNF-alfa is inhibited by miR 19a-5p, miR125b-5p, and miR 203-5p. IL-6 is blocked by miR 9-5p, miR26a-5p, miR100-5p, and miR 365, and IL-10 is inhibited by miR-98-5p and miR-106a-5p [109,110].

3.2. Immune Checkpoints vs. TLR-4

Immune checkpoints (ICs) are molecules with the ability to modulate T cells. Coinhibitory and costimulatory properties ensure a balanced immune response. ICs protect autoimmunity and optimize the adequate response of the immune system. When carcinogenesis occurs, IC signaling becomes a route of immune escape, leading to aggressive tumor invasion. Cytotoxic T-lymphocyte antigen 4 (CTLA-4) was the first well-described IC molecule expressed on CD-8+ and CD-4+ T cells. CTLA-4 binds to the corresponding B7-1 (CD80) ligand on antigen-presenting cells (APCs). Interaction between both results in the inhibition of T cells. In GBM, CTLA-4 expression on CD8+ and CD4+ has a significant correlation with poor outcome and invasion of brain tissue. TLR signaling promotes the overexpression of CD80, enhancing the presentation of antigens. It strengthens CTLR-4-induced T-cell anergy and GBM expansion [111,112,113,114].
The PD-1/PD-1L axis is another IC pathway. PD-1 and PD-1L are the most potent immunomodulatory proteins. PD-1L was described in 1999 by the Dong study team as a B7-H1 molecule [115], which represents the B7 protein family with a homology level of around 20%. The expression of PD-1L could be divided into inducible and constitutive. The constitutive can be met on antigen-presenting cells and resting lymphocytes. Inducible expression appears during inflammation and immune response; PD-1L represents suppressive properties. Glioblastoma cells represent both constitutive and inducible expression of PD-1L [116,117]. The phenomenon of autoinduction through TLR-4 also occurs. TLR-4 activation on GBM results in signaling through the MyD-88-independent pathway, TRAF6/ERK-1/2 AP-1s, leading to the activation of the PD-1L promoter. PD-1L mRNA is translated into ribosomes, modified in the Golgi apparatus, and presented as a mature PD-1L forming on the surface of GBM cells. T cells recognize PD-1L and bind it with an adequate PD-1 receptor. The intracellular domain of the receptor consists of an (ITSM) tyrosine-based switch motif that activates an inhibitory cascade [118,119,120,121,122,123,124]. Downsignaling promotes tyrosine phosphatase 2/Zeta-chain-associated protein kinase 7 (SHP-2/Zap 70) interplay, resulting in dephosphorylation and a significant decrease in lymphocytic cytotoxicity and proliferation. Activation of the PD-1/PD-1L axis leads to T-cell anergy and increases the lymphocyte apoptotic ratio. Lipopolysaccharides (LPsS), high-mobility group box-1 proteins (HMGB1), and the heat-shock-protein (HSP) family act as TLR-4 agonists that initiate signaling promoting PD-1L expression. Most of those molecules are intensively expressed in the GBM microenvironment, resulting in the autocrine induction of regional immunosuppression controlled by the PD-1L/PD-1 axis [125,126,127].
A study performed by Beswick et al. [128] discussed the matter of TLR-4/PD1L-induced immunotolerance in colon mucosa. Additionally, Wolfle et al. described a similar inductive effect of TLR-4 activation on PD-1L expression [129]. Poor prognosis for patients with peripheral lymphomas, correlated with TLR-4 and PD-1L overexpression, presented by Zhao et al., confirmed the pro-oncogenic effect of the presented axis [130].

3.3. TLR-4/Wnt Axis and Apoptosis

Glioblastoma, as a highly aggressive neoplasm, activates or modulates many pathways promoting gliomagenesis. One of those downsignaling instances concerns wingless signaling (Wnt\claudines axis) occurring as a crucial controller of cell–cell interplay, and as a regulator of migration. Any abnormalities in Wnt lead to evolutive effects. Aberrations in Wnt balance between stimulation and inhibition usually promote carcinogenesis. The Wnt complex represents an extended network consisting of various components, crosstalk interactions, and multiple regulatory stages. Wnt usually starts with receptor and coreceptor stimulation. Receptors, such as low-density lipoprotein receptor-related protein 6 (LRP6), protein Tyr kinase 7 receptor (PTK7), Tyr kinase-like orphan receptor (ROR), skeletal muscle Tyr kinase receptor (MUSK), and Tyr kinase receptor (RYK), are treated as Wnt-related. The main regulation of receptors is intracellularly processed by phosphorylating kinases. Extracellular regulation takes place through active agonists, such as norrins, the R-spondin family, and antagonists WNT inhibitory factor (WIF), secreted frizzled-related protein (SFRP), sclerostin, and Dickkopf-related protein 1 (DKK1) [131,132,133,134,135,136,137].
Wnt pathway activation is connected with other molecules called claudins. Claudins are a family of proteins that determine tight junctions (TJs) (claudin-5, claudin-3, and claudin-1). Claudin-5 is mainly expressed on the lung epithelium and brain tissue. Claudin-5, built-in TJs, bands in CNS endothelial cells. TJs create direct intercellular adhesion reducing cell-to-cell distance. In effect, they form the real connection between particular endothelial cells, strengthen the integrity of vessels, and control the diffusion of different ions and solutions. CNS embryonal development changes the permeability ratio. An immature form of the CNS vasculature, leaky blood vessels with characteristic fenestrations, are replaced by more integrating TJs. Claudin-5 contributes to the function of the blood barrier [138,139,140,141,142]. Overexpression of claudin-5 reduced inulin diffusion through the brain-vessel endothelium in rat models [143]. On the other hand, the downexpression of claudin-5 promotes epithelial permeability and the loss of blood–brain-barrier (BBB) properties [144]. Lower levels of claudin are correlated with higher levels of TLR-4 expression [145,146,147,148].
Dickkopf-related protein-3 (DKK-3), as was mentioned above, represents a group of proteins with strong suppressive features able to halt Wnt signaling. Moreover, DKK-3 seems to be an effective inhibitor of tumor-cell growth molecules. Casili et al. performed a study revealing DKK-3 to have a regulating influence on the Wnt pathway, resulting in increased caspase-3-dependent apoptosis [62].
Caspase-3, a representative of the caspase family, is described as an effector caspase. Its properties, such as activation by many apoptotic inducers, result in rife activity leading to complete apoptosis in many cell lines. Overexpression of caspase-3 results in an apoptotic GBM phenotype, confirmed in patient samples. Studies suggested that a higher apoptotic ratio is associated with longer PFS in treating for GBM. Sensitivity to temozolomide (TMZ) treatment is also correlated with caspase-3 expression, and higher levels of caspase-3 promote TMZ efficiency. Interestingly, caspase-9, a less apoptotic molecule, is downexpressed after Dickkopf WNT Signaling Pathway Inhibitor 3 (DKK-3) upregulation, usually increasing after TLR-4 stimulation. Highly invasive cancer-cell lines express caspase-9 more intensively [62,149,150,151].
Apoptosis, as a canonical biological interaction, regulates the subsistence of organisms and limits the expansion of GBM. A study connected the expression of DKK-3 and claudine-5 in healthy brain tissue, compared with the downexpression of both in GBM patients. Significantly lower levels of DKK-3 were observed in many types of neoplasms concerning prostate cancer. The absence of TLR-4 inhibits the growth of glioblastoma lines. The expression of TLR-4 interferes with Wnt downsignaling, promoting carcinogenesis. Additionally, the lack of TLR-4 maintains claudin-5 and DKK-3 proapoptotic properties, resulting in the limitation of tumor expansion. To sum up, the downexpression of TLR-4 promotes Wnt/DKK-3/claudin-5 signaling, which limits GBM invasiveness [62,152,153].

3.4. TLR-4 Influence on Non-CSC Glioma Stem Cells

A study performed by Alvarado et al. revealed a lack of TLR-4 occurrence on cancer stem cells (CSCs), the most invasive and most aggressive subpopulations of GBM. In opposition to CSC mature GBM cells and non-CSCs, it demonstrated TLR-4 expression and a response to agonist ligation. CSCs are responsible for therapeutic ineffectiveness and tumor-mass progression. External stimulation, such as necrosis, hypoxia, uncontrolled proliferation, and acidic stress, characteristics of a tumor microenvironment, generate unfavorable conditions. Thanks to developed adaptive mechanisms, CSCs maintain the balance between self-restoration and differentiation in hostile habitats. Owing to limited TLR-4 expression, CSCs can survive in a malevolent environment. Experiments conducted by a study team explained the processes of suppressing the CSC subpopulation, in which TLR-4 signaling played a key role. The main discovery in those papers indicated intensive TLR-4 downsignaling, halting CSC expansion by reducing the activity of retinoblastoma binding protein 5 (RBBP5), which is naturally increased in this subpopulation. Another important thing to understand in the underlying processes in CSCs is that RBBP-5 promotes cardinal stem-cell transcription initiators, essential to achieve invasiveness and self-restoration. TLR-4 expression and downsignaling inhibit RBBP-5 by the phosphorylation of TBK-1. TLR signaling borders proliferative trends in CSC and GBM [154,155,156,157,158,159,160].
Recent studies concerning bladder tumors, prostate cancer, and colorectal cancer pointed at TLR signaling as a tumorigenic and tumor-progression factor [161,162,163,164]. Dapito et al. linked TLR-4 activation with the significant progression of hepatocellular carcinoma. Alvarado et al. presented the opposite effects, where the receptor limited tumor growth by the direct blockade of the self-restoration cycle. The described divergence of TLR-4 signaling could be linked with ligand properties and downsignaling; TBK-1 seems to be an explanation, which is an element of the TLR-4–MyD88-independent pathway, and activation may also be signaled through the MyD88-dependent pathway, as it occurs in hepatocellular cancer [160,165]. Further observation and experiments explaining these differences are required, which are necessary to understand the anti- and procarcinogenic features of TLR-4. Signaling through the MyD88-independent and MyD88-dependent pathway may bring totally different results. Elucidation of these opposite results is crucial for future perspectives of immunotherapy for GBM (Figure 2).

4. Trials Concerning TLR-4

4.1. In Vitro and In Vivo Agonist

Exposition of TLR-4 on LPS in in vitro studies resulted in tumor-cell-line progression. The promotion and proliferation of the U87 and U118 lines were detected, and an increase in invasion was realized in U87 [28]. Stimulation of LPS increased metalloproteinase-9 (MMP-9), a factor of invasion detected in U87 glioma cell lines. Moreover, parallel TLR-4 signaling modulated the evasion of the immune system, tumor-necrosis-factor resistance, and cell survival [166]. The spirulina complex (polysaccharide) and LPSs as TLR-4 agonists demonstrated antitumor activity in mouse models. Differences in the action of both were underlain in IL-17 interaction. IL-17, with its pleiotropic characteristic, acts as an anti- or protumor cytokine, depending on the investigated neoplasm model. An increase in the spirulina polysaccharide, in contrast to LPS, increased the levels of IL17 in mouse-model serum. The murine glioma-cell complex becomes more invasive and expands after IL-17 activation [167,168]. Moreover, Fas and TLR-4 contemporary activation results in the disappearance of TLR-4 tumor-promoting properties [169].
Prosaposin (PSAP) conserved glycoprotein acts as a neurotropic factor in the glioblastoma environment. High levels of PSAP were detected in glioblastoma patients and were associated with unfavorable outcome [27,170,171]. Jiang et al. investigated PSAP as a promotor of proliferation and tumorigenesis through TLR-4 signaling. Mice models confirmed poor prognosis related to PSAP overexpression. The study hypothesizes PSAP affinity to TLR-4. Co-localization of TLR-4 and PSAP was confirmed with immunofluorescence staining. To sum up, PSAP activates TLR-4/NF-κB signaling, induces secretion of factors responsible for inflammation and tumor growth [172]. The blockade of PSAP becomes an interesting target for improving GBM outcome.
LPS was examined as affecting GSCs, and mature glioma cells in the study performed by Han et al. rat model has proven that LPS stimulation prolonged survival time significantly in the observed glioma-cohort [173]. Interestingly some studies report an antitumoral effect induced by a bacterial infection in animal models and report some cases of GBM patients [174,175,176,177]. LPS via TLR-4 stimulation changes immuno-phenotype in GSCs and the mature form of glioma cells and induces antitumoral response [173].

4.2. Agonist and Inhibitors of TLR-4 in Oncology

TLR-4 is expressed on the surface of the cell membrane and occurs on endosomes. Moreover, the receptor has the ability to dependently signal to MyD88 and TRIF complexes. Those characteristic features are the example of evolutionary controlled carefulness in precise antigen-detection and downsignaling-promotion mechanisms. TLR-4 coreceptor myeloid differentiation factor 2 (MD-2) has an appropriate pocket that binds the corresponding ligands. Immunotherapy disrupting TLR-4 and MD-2 binding or blocking their interplay was explored [178,179,180]. Another interesting ligand inhibiting or activating TLR-4, containing a glucopyranosyl lipid adjuvant (GLA) agonist [181,182], monophosphoryl lipid A (MPLA) agonist [183,184], and lipid 4A antagonist, was discovered and explored [185]. Interestingly, TLR-4 is the most popular among the TLRs evaluated as receptors in numerous clinical trials as a potential target for immunotherapy against various pathologies concerning inflammation, viral infection, immune diseases, and cancers (Table 3) [186,187,188].

4.3. TLR-4 in Glioblastoma Multiforme—Clinical Application

The macrophage migration inhibitory factor (MIF) cytokine is treated as a potent cytokine initiating immunity and inflammation processes [189,190,191]. MIF is excreted by macrophages immediately after the ligation of bacterial products and inflammatory molecules. Studies showed a strict interplay between MIF and TLR-4 signaling. MIF knockout mice presented the downregulation of TLR-4 signaling. The response of macrophages to LPS stimulation was significantly limited [192,193,194,195]. The presented model made the basis for immunotherapy targeting MIF and TLR-4. Trial NCT03782415 contested ibudilast MN-166 in Phase 1 and compared it with TMZ combo treatment in recurrent GBM in Phase 2. A multicenter open-label study evaluated its efficacy, tolerability, and safety with a TMZ combination in 50 recurrent GBM patients. To be eligible, patients required a Karnofsky Performance Scale (KFS) > 70 (https://clinicaltrials.gov/ct2/show/NCT03782415).
Ibudilast is an agent that is able to diffuse the blood–brain barrier (BBB), developing antitumor activity by inhibiting MIF and phosphodiesterase-4 (PDE-4), also reducing TLR-4 downsignaling with pro-oncogenic properties. Action leads to excessive apoptosis and the limitation of cell proliferation in GBM [196,197,198]. Ibudilast was also observed in trials, including neuroinflammation, amyotrophic lateral sclerosis (ALS), dysphoria, treatment of alcohol-use disorders, and methamphetamine dependence (https://clinicaltrials.gov/ct2/results?cond=ibudilast&term=&cntry=&state=&city=&dist=).
Another clinical application of TLR-4 signaling concerns the antiglioma HSP vaccination. HSP, as a part of the heat shock protein peptide complex (HSSPC) (Table 4), is characterized by a strong affinity to numerous receptors on the surface of the cell. T-cell supported HSP stimulation of APCs results in downstream signaling and NF-κB activation. CD91/CD40/CD36/CD14/TLR-2 and TLR-4 seem to play crucial roles in this process. Promotion of the TLR-4/NF-κB axis in APC causes intensive secretion of chemokines and proinflammatory molecules to the tumor microenvironment. Ipso facto, HSP stimulates, indirectly, the release of inflammatory factors, such as IL-12, IL-1beta, TNF alfa, granulocyte macrophage colony-stimulating factor (GM-CSF) and inhibits GBM growth [199,200,201,202,203,204,205,206].
The study performed by Crane et al. [207] reported significant immunization after HSP administration in 11 out of 12 patients with recurrent GBM (rGBM). Median Survival time in the group of immune responders oscillated about 47 weeks after surgical resection and vaccination. Non-responding patients survived 16 weeks only. Promising results encouraged for further investigation. Bloch et al. revealed a median overall survival oscillating 42 weeks (42.6 weeks) after HSPPC-96 administration in rGBM patients. Further efforts concerning the design of Phase2 studies have been performed [208]. Clinical trials challenging the effectiveness and safety of HSPPC-96 vaccination evaluate overall survival and progression-free time. Trials also compare results with standard treatment, other immunological agents (Bevacizumab, Pembrolizumab), and the combination of drugs (https://www.clinicaltrials.gov) (Table 4).

5. Conclusions

TLR-4 signaling pathways and their wide spectrum of interactions seem to be promising targets for immunotherapy. Multimodal interplay and a variety of downsignaling encourage TLR-4 to be further considered as a potential aim for immune agents. Clinical trials contesting TLR-4 agonists and inhibitors are recruiting patients with various cancers. TLR-4-related treatment is being gradually introduced for GBM patients. There is still much to do in the field of effective immunotherapy.

Author Contributions

Each author contributed equally to this paper. Conceptualization, J.L., J.L. and J.R.; methodology, J.L. and C.G.; formal analysis, P.K. and C.G.; investigation, I.O., K.G, E.R.; resources, K.G. and J.R.; writing—original draft preparation, J.L. and J.L.; writing—review and editing, J.R., E.R. and P.K.; visualization, J.L.; supervision, J.R. and P.K.; project administration, C.G.; funding acquisition, J.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding

Conflicts of Interest

The authors declare no conflict of interest

References

  1. Lee, H.; Lee, S.; Cho, I.H.; Lee, S.J. Toll-like receptors: Sensor molecules for detecting damage to the nervous system. Curr. Protein Pept. Sci. 2013, 14, 33–42. [Google Scholar] [CrossRef] [PubMed]
  2. Tewari, R.; Choudhury, S.R.; Ghosh, S.; Mehta, V.S.; Sen, E. Involvement of TNFα-induced TLR4-NF-κB and TLR4-HIF-1α feed-forward loops in the regulation of inflammatory responses in glioma. J. Mol. Med. (Berl.) 2012, 90, 67–80. [Google Scholar] [CrossRef] [PubMed]
  3. Zeuner, M.; Bieback, K.; Widera, D. Controversial role of tolllike receptor 4 in adult stem cells. Stem Cell Rev. 2015, 11, 621–634. [Google Scholar] [CrossRef] [PubMed]
  4. Ulrich, H.; do Nascimento, I.C.; Bocsi, J.; Tárnok, A. Immunomodulation in stem cell differentiation into neurons and brain repair. Stem Cell Rev. 2014, 11, 474–486. [Google Scholar] [CrossRef]
  5. Mazurek, M.; Litak, J.; Kamieniak, P.; Kulesza, B.; Jonak, K.; Baj, J.; Grochowski, C. Metformin as Potential Therapy for High-Grade Glioma. Cancers 2020, 12, 210. [Google Scholar] [CrossRef] [Green Version]
  6. Huang, B.; Zhao, J.; Unkeless, J.C.; Feng, Z.H.; Xiong, H. TLR signaling by tumor and immune cells: A double-edged sword. Oncogene 2008, 27, 218–224. [Google Scholar] [CrossRef] [Green Version]
  7. Su, X.; Ye, J.; Hsueh, E.C.; Zhang, Y.; Hoft, D.F.; Peng, G. Tumor microenvironments direct the recruitment and expansion of human Th17 cells. J. Immunol. 2010, 184, 1630–1641. [Google Scholar] [CrossRef]
  8. Ye, J.; Ma, C.; Hsueh, E.C.; Dou, J.; Mo, W.; Liu, S.; Han, B.; Huang, Y.; Zhang, Y.; Varvares, M.A.; et al. TLR8 signaling enhances tumor immunity by preventing tumor-induced T-cell senescence. EMBO Mol. Med. 2014, 6, 1294–1311. [Google Scholar] [CrossRef]
  9. Krawczyk, C.M.; Holowka, T.; Sun, J.; Blagih, J.; Amiel, E.; DeBerardinis, R.J.; Cross, J.R.; Jung, E.; Thompson, C.B.; Jones, R.G.; et al. Toll-like receptor-induced changes in glycolytic metabolism regulate dendritic cell activation. Blood 2010, 115, 4742–4749. [Google Scholar] [CrossRef] [Green Version]
  10. Gil-Benso, R.; Monteagudo, C.; Cerdá-Nicolás, M.; Callaghan, R.C.; Pinto, S.; Martínez-Romero, A.; Pellín-Carcelén, A.; San-Miguel, T.; Cigudosa, J.C.; López-Ginés, C. Characterization of a new human melanoma cell line with CD133 expression. Hum. Cell 2012, 25, 61–67. [Google Scholar] [CrossRef] [PubMed]
  11. Jackson, M.; Hassiotou, F.; Nowak, A. Glioblastoma stem-like cells: At the root of tumor recurrence and a therapeutic target. Carcinogenesis 2015, 36, 177–185. [Google Scholar] [CrossRef] [PubMed]
  12. Nagyoszi, P.; Wilhelm, I.; Farkas, A.E.; Fazakas, C.; Dung, N.T.; Hasko, J.; Krizbai, I.A. Expression and regulation of toll-likereceptors in cerebral endothelial cells. Neurochem. Int. 2010, 57, 556–564. [Google Scholar] [CrossRef]
  13. Hanke, M.L.; Kielian, T. Toll-like receptors in health and disease in the brain: Mechanisms and therapeutic potential. Clin. Sci. 2011, 121, 367–387. [Google Scholar] [CrossRef] [Green Version]
  14. Bsibsi, M.; Ravid, R.; Gveric, D.; van Noort, J.M. Broad expression of Toll-like receptors in the human central nervous system. J. Neuropathol. Exp. Neurol. 2002, 61, 1013–1021. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Hussain, S.F.; Yang, D.; Suki, D.; Aldape, K.; Grimm, E.; Heimberger, A.B. The role of human glioma-infiltrating microglia/macrophages in mediating antitumor immune responses. Neuro-Oncol. 2006, 8, 261–279. [Google Scholar] [CrossRef] [Green Version]
  16. Bowman, C.C.; Rasley, A.; Tranguch, S.L.; Marriott, I. Cultured astrocytes express toll-like receptors for bacterial products. Glia 2003, 43, 281–291. [Google Scholar] [CrossRef]
  17. Crack, P.J.; Bray, P.J. Toll-like receptors in the brain and their potential roles in neuropathology. Immunol. Cell Biol. 2007, 85, 476–480. [Google Scholar] [CrossRef]
  18. Hu, J.; Shi, B.; Liu, X.; Jiang, M.; Yuan, C.; Jiang, B.; Song, Y.; Zeng, Y.; Wang, G. The activation of Toll-like receptor 4 reverses tumor differentiation in human glioma U251 cells via Notch pathway. Int. Immunopharmacol. 2018, 64, 33–41. [Google Scholar] [CrossRef] [PubMed]
  19. Deng, S.; Zhu, S.; Qiao, Y.; Liu, Y.J.; Chen, W.; Zhao, G.; Chen, J. Recent advances in the role of toll-like receptors and TLR agonists in immunotherapy for human glioma. Protein Cell 2014, 5, 899–911. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Kawai, T.; Akira, S. The roles of TLRs, RLRs and NLRs in pathogen recognition. Int. Immunol. 2009, 21, 317–337. [Google Scholar] [CrossRef] [Green Version]
  21. Akira, S.; Uematsu, S.; Takeuchi, O. Pathogen recognition and innate immunity. Cell 2006, 124, 783–801. [Google Scholar] [CrossRef] [Green Version]
  22. Bianchi, M.E. DAMPs, PAMPs and alarmins: All we need to know about danger. J. Leukoc. Biol. 2007, 81, 1–5. [Google Scholar] [CrossRef] [PubMed]
  23. Krysko, D.V.; Garg, A.D.; Kaczmarek, A.; Krysko, O.; Agostinis, P.; Vandenabeele, P. Immunogenic cell death and DAMPs in cancer therapy. Nat. Rev. Cancer 2012, 12, 860–875. [Google Scholar] [CrossRef] [PubMed]
  24. Meng, Y.; Kujas, M.; Marie, Y.; Paris, S.; Thillet, J.; Delattre, J.-Y.; Carpentier, A.F. Expression of TLR9 withinhuman glioblastoma. J. Neuro-Oncol. 2008, 88, 19–25. [Google Scholar] [CrossRef] [PubMed]
  25. Medvedev, A.E.; Sabroe, I.; Hasday, J.D.; Vogel, S.N. Invited review: Tolerance to microbial TLR ligands:Molecular mechanisms and relevance to disease. J. Endotoxin Res. 2006, 12, 133–150. [Google Scholar] [CrossRef] [PubMed]
  26. Aalaei-Andabili, S.H.; Rezaei, N. Toll like receptor (TLR)-induced differential expression of microRNAs(MiRs) and immune response against infection: A systematic review. J. Infect. 2013, 67, 251–264. [Google Scholar] [CrossRef] [PubMed]
  27. Han, S.; Wang, C.; Qin, X.; Xia, J.; Wu, A. LPS alters the immuno-phenotype of glioma and glioma stem-like cells and induces in vivo antitumor immunity via TLR4. J. Exp. Clin. Cancer Res. 2017, 36, 83. [Google Scholar] [CrossRef] [Green Version]
  28. Sarrazy, V.; Vedrenne, N.; Billet, F.; Bordeau, N.; Lepreux, S.; Vital, A.; Jauberteau, M.O.; Desmouliere, A. TLR4 signal transduction pathways neutralize the effect of Fas signals on glioblastoma cell proliferation and migration. Cancer Lett. 2011, 311, 195–202. [Google Scholar] [CrossRef]
  29. Han, C.; Jin, J.; Xu, S.; Liu, H.; Li, N.; Cao, X. Integrin CD11b negatively regulates TLR-triggered inflammatory responses by activating Syk and promoting degradation of MyD88 and TRIF via Cbl-b. Nat. Immunol. 2010, 11, 734–742. [Google Scholar] [CrossRef]
  30. Anwar, M.A.; Basith, S.; Choi, S. Negative regulatory approaches to the attenuation of Toll-like receptor signaling. Exp. Mol. Med. 2013, 45, e11. [Google Scholar] [CrossRef]
  31. Skaug, B.; Chen, J.; Du, F.; He, J.; Ma, A.; Chen, Z.J. Direct, noncatalytic mechanism of IKK inhibition by A20. Mol. Cell 2011, 44, 559–571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Hayden, M.S.; Ghosh, S. NF-kappaB, the first quarter-century: Remarkable progress and outstanding questions. Genes Dev. 2012, 26, 203–234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Palsson Mc Dermott, E.M.; Doyle, S.L.; McGettrick, A.F.; Hardy, M.; Husebye, H.; Banahan, K.; Gong, M.; Golenbock, D.; Espevik, T.; O’Neill, L.A.J. TAG, a splice variant of the adaptor TRAM, negatively regulates the adaptor MyD88independent TLR4 pathway. Nat. Immunol. 2009, 10, 579–586. [Google Scholar] [CrossRef] [PubMed]
  34. Spachidou, M.P.; Bourazopoulou, E.; Maratheftis, C.I.; Kapsogeorgou, E.K.; Moutsopoulos, H.M.; Tzioufas, A.G.; Manoussakis, M.N. Expression of functional Toll-like receptors by salivary gland epithelial cells: Increased mRNA expression in cells derived from patients with primary Sjogren’s syndrome. Clin. Exp. Immunol. 2007, 147, 497–503. [Google Scholar] [CrossRef]
  35. Hashimoto, C.; Hudson, K.L.; Anderson, K.V. The Toll gene of Drosophila, required for dorsal-ventral embryonic polarity, appears to encode a transmembrane protein. Cell 1988, 52, 269–279. [Google Scholar] [CrossRef]
  36. Winans, K.A.; Hashimoto, C. Ventralization of the Drosophila embryo by deletion of extracellular leucine-rich repeats in the Toll protein. Mol. Biol. Cell 1995, 6, 587–596. [Google Scholar] [CrossRef] [Green Version]
  37. Hoffmann, J.A. Innate immunity of insects. Curr. Opin. Immunol. 1995, 7, 4–10. [Google Scholar] [CrossRef]
  38. Medzhitov, R.; Preston-Hurlburt, P.; Janeway, C.A., Jr. A human homologue of the Drosophila toll protein signals activation of adaptive immunity. Nature 1997, 388, 394–397. [Google Scholar] [CrossRef]
  39. Medzhitov, R.; Janeway, C.A.J. Innate immunity: The virtues of a nonclonal system of recognition. Cell 1997, 91, 295–298. [Google Scholar] [CrossRef] [Green Version]
  40. Randow, F.; Seed, B. Endoplasmic reticulum chaperone gp96 is required for innate immunity but not cell viability. Nat. Cell Biol. 2001, 3, 891–896. [Google Scholar] [CrossRef]
  41. De Nardo, D. Toll-like receptors: Activation, signalling and transcriptionalmodulation. Cytokine 2015, 74, 181–189. [Google Scholar] [CrossRef] [PubMed]
  42. Kawai, T.; Akira, S. The role of pattern-recognition receptors in innate immunity: Update on toll-like receptors. Nat. Immunol. 2010, 11, 373–384. [Google Scholar] [CrossRef] [PubMed]
  43. Gao, D.; Li, W. Structures and recognition modes of toll-like receptors. Proteins Struct. Funct. Bioinform. 2017, 85, 3–9. [Google Scholar] [CrossRef] [PubMed]
  44. Uenishi, H.; Shinkai, H. Porcine toll-like receptors:the front line of pathogen monitoring and possible implications for disease resistance. Dev. Comp. Immunol. 2009, 33, 353–361. [Google Scholar] [CrossRef] [PubMed]
  45. Viriyakosol, S.; Tobias, P.S.; Kitchens, R.L.; Kirkland, T.N. MD-2 binds to bacterial lipopolysaccharide. J. Biol. Chem. 2001, 276, 38044–38051. [Google Scholar]
  46. Ohto, U.; Fukase, K.; Miyake, K.; Shimizu, T. Structural basis of species-specific endotoxin sensing by innate immune receptor TLR4/MD-2. Proc. Natl. Acad. Sci. USA 2012, 109, 7421–7426. [Google Scholar] [CrossRef] [Green Version]
  47. Shimazu, R.; Akashi, S.; Ogata, H.; Nagai, Y.; Fukudome, K.; Miyake, K.; Kimoto, M. MD-2, a molecule that confers lipopolysaccharide responsiveness on toll-like receptor 4. J. Exp. Med. 1999, 189, 1777–1782. [Google Scholar] [CrossRef]
  48. Kim, H.M.; Park, B.S.; Kim, J.-I.; Kim, S.E.; Lee, J.; Oh, S.C.; Enkhbayar, P.; Matsushima, M.; Lee, H.; Yoo, O.J. Crystal structure of the TLR4-MD-2 complex with bound endotoxin antagonist eritoran. Cell 2007, 130, 906–917. [Google Scholar] [CrossRef] [Green Version]
  49. O’Neill, L.A.; Bowie, A.G. The family of five: TIR-domaincontaining adaptors in Toll-like receptor signalling. Nat. Rev. Immunol. 2007, 7, 353. [Google Scholar] [CrossRef]
  50. Park, B.S.; Song, D.H.; Kim, H.M.; Choi, B.-S.; Lee, H.; Lee, J.-O. The structural basis of lipopolysaccharide recognition by the TLR4-MD-2 complex. Nature 2009, 458, 1191–1195. [Google Scholar] [CrossRef]
  51. Zeuner, M.T.; Kruger, C.L.; Volk, K.; Bieback, K.; Graeme, C.S.; Heilemann, M.; Widera, D. Biased signalling is an essential feature of TLR4 in glioma cells. Biochim. Biophys. Acta 2016, 1863, 3084–3095. [Google Scholar] [CrossRef] [PubMed]
  52. Finocchiaro, G. TLRgeting evasion of immune pathways in glioblastoma. Cell Stem Cell 2017, 20, 422–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Park, M.H.; Hong, J.T. Roles of NF-jB in cancer and inflammatory diseases and their therapeutic approaches. Cells 2016, 5, 29. [Google Scholar] [CrossRef] [PubMed]
  54. Soubannier, V.; Stifani, S. NF-κB Signalling in Glioblastoma. Biomedicines 2017, 5, 29. [Google Scholar] [CrossRef] [Green Version]
  55. Pikarsky, E.; Porat, R.M.; Stein, I.; Abramovitch, R.; Amit, S.; Kasem, S.; Gutkovich-Pyest, E.; Urieli-Shoval, S.; Galun, E.; Ben-Neriah, Y. NF-kappaB functions as a tumour promoter in inflammation-associated cancer. Nature 2004, 431, 461–466. [Google Scholar] [CrossRef]
  56. Ditsworth, D.; Zong, W.X. NF-kappaB: Key mediator of inflammationassociated cancer. Cancer Biol. Ther. 2004, 3, 1214–1216. [Google Scholar] [CrossRef] [Green Version]
  57. Kına, I.; Sultuybek, G.K.; Soydas, T.; Yenmis, G.; Biceroglu, H.; Dirican, A.; Uzan, M.; Ulutin, T. Variations in Toll-like receptor and nuclear factor-kappa B genes and the risk of glioma. Br. J. Neurosurg. 2018, 33, 165–170. [Google Scholar] [CrossRef]
  58. Hanahan, D.; Weinberg, R. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
  59. Harazono, Y.L.; Muramatsu, T.; Endo, H.; Uzawa, N.; Kawano, T.; Harada, K.; Inazawa, J.; Kozaki, K. miR- 655 Is an EMT-Suppressive MicroRNA Targeting ZEB1 and TGFBR2. PLoS ONE 2013, 8, e62757. [Google Scholar] [CrossRef] [Green Version]
  60. Gabriely, G.; Wurdinger, T.; Kesari, S.; Esau, C.C.; Burchard, J.; Linsley, P.S.; Krichevsky, A.M. MicroRNA 21 promotes glioma invasion by targeting matrix metalloproteinase regulators. Mol. Cell Biol. 2008, 28, 5369–5380. [Google Scholar] [CrossRef] [Green Version]
  61. Chaudhary, D.; Robinson, S.; Romero, D.L. Recent advances in the discovery of small molecule inhibitors of interleukin-1 receptor-associated kinase 4 (IRAK4) as a therapeutic target for inflammation and oncology disorders. J. Med. Chem. 2015, 58, 96–110. [Google Scholar] [CrossRef] [PubMed]
  62. Casili, G.; Caffo, M.; Campolo, M.; Barresi, V.; Caruso, G.; Cardali, S.M.; Lanza, M.; Mallamace, R.; Filippone, A.; Conti, A.; et al. TLR-4/Wnt modulation as new therapeutic strategy in the treatment of glioblastomas. Oncotarget 2018, 9, 37564–37580. [Google Scholar] [CrossRef] [PubMed]
  63. Li, Y.; Wang, Y.; Yu, L.; Sun, C.; Cheng, D.; Yu, S.; Wang, Q.; Yan, Y.; Kang, C.; Jin, S.; et al. miR-146b-5p inhibits glioma migration and invasion by targeting MMP16. Cancer Lett. 2013, 339, 260–269. [Google Scholar] [CrossRef] [PubMed]
  64. Cui, B.; Li, B.; Liu, Q.; Cui, Y. lncRNA CCAT1 Promotes Glioma Tumorigenesis by Sponging miR-181b. J. Cell Biochem. 2017, 118, 4548–4557. [Google Scholar] [CrossRef]
  65. Sun, L.; Yan, W.; Wang, Y.; Sun, G.; Luo, H.; Zhang, J.; Wang, X.; You, Y.; Yang, Z.; Liu, N. MicroRNA-10b induces glioma cell invasion by modulating MMP-14 and uPAR expression via HOXD10. Brain Res. 2011, 1389, 9–18. [Google Scholar] [CrossRef]
  66. Wu, D.G.; Wang, Y.Y.; Fan, L.G.; Luo, H.; Han, B.; Sun, L.H.; Wang, X.F.; Zhang, J.X.; Cao, L.; Wang, X.R.; et al. MicroRNA-7 regulates glioblastoma cell invasion via targeting focal adhesion kinase expression. Chin. Med. J. (Engl.) 2011, 124, 2616–2621. [Google Scholar]
  67. Lin, J.; Teo, S.; Lam, D.H.; Jeyaseelan, K.; Wang, S. MicroRNA-10b pleiotropically regulates invasion, angiogenicity and apoptosis of tumor cells resembling mesenchymal subtype of glioblastoma multiforme. Cell Death Dis. 2012, 3, e398. [Google Scholar] [CrossRef]
  68. Liao, H.; Bai, Y.; Qiu, S.; Zheng, L.; Huang, L.; Liu, T.; Wang, X.; Liu, Y.; Xu, N.; Yan, X.; et al. MiR-203 downregulation is responsible for chemoresistance in human glioblastoma by promoting epithelial-mesenchymal transition via SNAI2. Oncotarget 2015, 6, 8914–8928. [Google Scholar] [CrossRef]
  69. Xu, W.; Hu, G.Q.; Da Costa, C.; Tang, J.H.; Li, Q.R.; Du, L.; Pan, Y.W.; Lv, S.Q. Long noncoding RNA UBE2R2-AS1 promotes glioma cell apoptosis via targeting the miR-877-3p/TLR4 axis. Onco Targets Ther. 2019, 12, 3467–3480. [Google Scholar] [CrossRef] [Green Version]
  70. Blachere, N.E.; Li, Z.; Chandawarkar, R.Y.; Suto, R.; Jaikaria, N.S.; Basu, S.; Udono, H.; Srivastava, P.K. Heat shock protein-peptide complexes, reconstituted in vitro, elicit peptide-specific cytotoxic T lymphocyte response and tumor immunity. J. Exp. Med. 1997, 186, 1315–1322. [Google Scholar] [CrossRef]
  71. Binder, R.J.; Srivastava, P.K. Essential role of CD91 in re-presentation of gp96-chaperoned peptides. Proc. Natl. Acad. Sci. USA 2004, 101, 6128–6133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Takizawa, H.; Fritsch, K.; Kovtonyuk, L.V.; Saito, Y.; Yakkala, C.; Jacobs, K.; Ahuja, A.K.; Lopes, M.; Hausmann, A.; Hardt, W.-D. Pathogen-induced TLR4-TRIF innate immune signaling in hematopoietic stem cells promotes proliferation but reduces competitive fitness. Cell Stem Cell 2017, 21, 225–240. [Google Scholar] [CrossRef] [PubMed]
  73. Jouhi, L.; Renkonen, S.; Atula, T.; Mäkitie, A.; Haglund, C.; Hagström, J. Different toll-like receptor expression patterns in progression toward cancer. Front. Immunol 2014, 5, 638. [Google Scholar] [CrossRef] [Green Version]
  74. Gregg, K.A.; Harberts, E.; Gardner, F.M.; Pelletier, M.R.; Cayatte, C.; Yu, L.; McCarthy, M.P.; Marshall, J.D.; Ernst, R.K. Rationally designed TLR4 ligands for vaccine adjuvant discovery. MBio 2017, 8, e00492-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Shi, M.; Chen, X.; Ye, K.; Yao, Y.; Li, Y. Application potential of toll-like receptors in cancer immunotherapy: Systematic review. Medicine 2016, 95, e3951. [Google Scholar] [CrossRef] [PubMed]
  76. Apetoh, L.; Ghiringhelli, F.; Tesniere, A.; Obeid, M.; Ortiz, C.; Criollo, A.; Mignot, G.; Maiuri, M.C.; Ullrich, E.; Saulnier, P.; et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat. Med. 2007, 13, 1050–1059. [Google Scholar] [CrossRef] [PubMed]
  77. Terashita, T.; Saito, S.; Nabeka, H.; Hato, N.; Wakisaka, H.; Shimokawa, T.; Kobayashi, N.; Gyo, K.; Matsuda, S. Prosaposin-derived peptide alleviates ischaemia-induced hearing loss. Acta Otolaryngol. 2013, 133, 462–468. [Google Scholar] [CrossRef]
  78. Kuzmich, N.N.; Sivak, K.V.; Chubarev, V.N.; Porozov, Y.B.; Savateeva-Lyubimova, T.N.; Peri, F. TLR4 Signaling Pathway Modulators as Potential Therapeutics in Inflammation and Sepsis. Vaccines (Basel) 2017, 5, 34. [Google Scholar] [CrossRef] [Green Version]
  79. Ciafre, S.A.; Galardi, S.; Mangiola, A.; Ferracin, M.; Liu, C.-G.; Sabatino, G.; Negrini, M.; Maira, G.; Croce, C.M.; Farace, M.G. Extensive modulation of a set of microRNAs in primary glioblastoma. Biochem. Biophys. Res. Commun. 2005, 334, 1351–1358. [Google Scholar] [CrossRef]
  80. Shea, A.; Harish, V.; Afzal, Z.; Chijioke, J.; Kedir, H.; Dusmatova, S.; Roy, A.; Ramalinga, M.; Harris, B.; Blancato, J.; et al. MicroRNAs in glioblastoma multiforme pathogenesis and therapeutics. Cancer Med. 2016, 5, 1917–1946. [Google Scholar] [CrossRef]
  81. Møller, H.G.; Rasmussen, A.P.; Andersen, H.H.; Johnsen, K.B.; Henriksen, M.; Duroux, M. A systematic review of microRNA in glioblastoma multiforme: Micro-modulators in the mesenchymal mode of migration and invasion. Mol. Neurobiol. 2013, 47, 131–144. [Google Scholar] [CrossRef] [Green Version]
  82. Ahir, B.K.; Ozer, H.; Engelhard, H.H.; Lakka, S.S. MicroRNAs in glioblastoma pathogenesis and therapy: A comprehensive review. Crit. Rev. Oncol. Hematol. 2017, 120, 22–33. [Google Scholar] [CrossRef] [PubMed]
  83. Godlewski, J.; Nowicki, M.O.; Bronisz, A.; Williams, S.; Otsuki, A.; Nuovo, G.; Raychaudhury, A.; Newton, H.B.; Chiocca, E.A.; Lawler, S. Targeting of the Bmi-1 oncogene/stem cell renewal factor by microRNA-128 inhibits glioma proliferation and self-renewal. Cancer Res. 2008, 68, 9125–9130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Lawler, S.; Chiocca, E.A. Emerging functions of microRNAs in glioblastoma. J. Neurooncol. 2009, 92, 297–306. [Google Scholar] [CrossRef]
  85. Silber, J.; Lim, D.A.; Petritsch, C.; Persson, A.I.; Maunakea, A.K.; Yu, M.; Vandenberg, S.R.; Ginzinger, D.G.; James, C.D.; Costello, J.F.; et al. miR-124 and miR-137 inhibit proliferation of glioblastoma multiforme cells and induce differentiation of brain tumor stem cells. BMC Med. 2008, 6, 14. [Google Scholar] [CrossRef] [PubMed]
  86. Chan, J.A.; Krichevsky, A.M.; Kosik, K.S. MicroRNA-21 is an antiapoptotic factor in human glioblastoma cells. Cancer Res. 2005, 65, 6029–6033. [Google Scholar] [CrossRef] [Green Version]
  87. Yang, G.; Zhang, R.; Chen, X.; Mu, Y.; Ai, J.; Shi, C.; Liu, Y.; Shi, C.; Sun, L.; Rainov, N.G.; et al. MiR-106a inhibits glioma cell growth by targeting E2F1 independent of p53 status. J. Mol. Med. 2011, 89, 1037–1050. [Google Scholar] [CrossRef]
  88. De Smaele, E.; Ferretti, E.; Gulino, A. MicroRNAs as biomarkers for CNS cancer and other disorders. Brainres 2010, 1338, 100–111. [Google Scholar] [CrossRef]
  89. Huse, J.T.; Brennan, C.; Hambardzumyan, D.; Wee, B.; Pena, J.; Rouhanifard, S.H.; Sohn-Lee, C.; Agami, R.; Tuschl, T.; Holland, E.C.; et al. The PTEN-regulating microRNA miR-26a is amplified in high-grade gliomaand facilitates gliomagenesis in vivo. Genes Dev. 2009, 23, 1327–1337. [Google Scholar] [CrossRef] [Green Version]
  90. Corsten, M.F.; Miranda, R.; Kasmieh, R.; Krichevsky, A.M.; Weissleder, R.; Shah, K. MicroRNA-21 knockdowndisrupts glioma growthin vivoand displays synergistic cytotoxicity with neural precursor cell deliveredS-TRAIL in human gliomas. Cancer Res. 2007, 67, 8994–9000. [Google Scholar] [CrossRef] [Green Version]
  91. Wang, Q.; Li, P.; Li, A.; Jiang, W.; Wang, H.; Wang, J.; Xie, K. Plasma specifc miRNAs as predictive biomarkersfor diagnosis and prognosis of glioma. J. Exp. Clin. Cancer Res. 2019, 31, 97. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Hermansen, S.K.; Dahlrot, R.H.; Nielsen, B.S.; Hansen, S.; Kristensen, B.W. MiR-21 expression in the tumorcell compartment holds unfavourable prognostic value in gliomas. J. Neurooncol. 2013, 111, 71–81. [Google Scholar] [CrossRef]
  93. Mazurek, M.; Litak, J.; Kamieniak, P.; Osuchowska, I.; Maciejewski, R.; Roliński, J.; Grajkowska, W.; Grochowski, C. Micro RNA Molecules as Modulators of Treatment Resistance, Immune Checkpoints Controllers and Sensitive Biomarkers in Glioblastoma Multiforme. Int. J. Mol. Sci. 2020, 21, 1507. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Taganov, K.D.; Boldin, M.P.; Chang, K.J.; Baltimore, D. NF-kappaB-dependent induction of microRNA miR146, an inhibitor targeted to signaling proteins of innate immune responses. Proc. Natl. Acad. Sci. USA 2006, 103, 12481–12486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Hou, J.; Wang, P.; Lin, L.; Liu, X.; Ma, F.; An, H.; Wang, Z.; Cao, X. MicroRNA-146a feedback inhibits RIG-I-dependent type I IFN production in macrophages by targeting TRAF6, IRAK1, and IRAK2. J. Immunol. 2009, 183, 2150–2158. [Google Scholar] [CrossRef] [Green Version]
  96. Shao, M.; Rossi, S.; Chelladurai, B.; Shimizu, M.; Ntukogu, O.; Ivan, M.; Calin, G.A.; Matei, D. PDGF induced microRNA alterations in cancer cells. Nucleic Acids Res. 2011, 39, 4035–4047. [Google Scholar] [CrossRef] [Green Version]
  97. Luan, S.; Sun, L.; Huang, F. MicroRNA-34a: A novel tumor suppressor in p53-mutant glioma cell line U251. Arch. Med. Res. 2010, 41, 67–74. [Google Scholar] [CrossRef]
  98. Xu, M.; Li, D.; Yang, C.; Ji, J.S. MicroRNA-34a Inhibition of the TLR Signaling Pathway Via CXCL10 Suppresses Breast Cancer Cell Invasion and Migration. Cell Physiol. Biochem. 2018, 46, 1286–1304. [Google Scholar] [CrossRef] [Green Version]
  99. Xiang, W.; Tian, C.; Peng, S.; Zhou, L.; Pan, S.; Deng, Z. Let-7i attenuates human brain microvascular endothelial cell damage in oxygen glucose deprivation model by decreasing toll-like receptor 4 expression. Biochem. Biophys. Res. Commun. 2017, 493, 788–793. [Google Scholar] [CrossRef]
  100. Heneka, M.T.; McManus, R.M.; Latz, E. Inflammasome signalling in brain function and neurodegenerative disease. Nat. Rev. Neurosci. 2018, 19, 610–621. [Google Scholar] [CrossRef]
  101. Lippai, D.; Bala, S.; Csak, T.; Kurt-Jones, E.A.; Szabo, G. Chronic alcohol-induced microRNA-155 contributes to neuroinflammation in a TLR4-dependent manner in mice. PLoS ONE 2013, 8, e70945. [Google Scholar] [CrossRef] [Green Version]
  102. Mueller, M.; Zhou, J.; Yang, L.; Gao, Y.; Wu, F.; Schoeberlein, A.; Surbek, D.; Barnea, E.R.; Paidas, M.; Huang, Y. PreImplantation factor promotes neuroprotection by targeting microRNA let-7. Proc. Natl. Acad. Sci. USA 2014, 111, 13882–13887. [Google Scholar] [CrossRef] [Green Version]
  103. Lu, M.; Zhang, P.J.; Li, C.H.; Lv, Z.-M.; Zhang, W.-W.; Jin, C.-H. miRNA-133 augments coelomocyte phagocytosis in bacteria-challenged Apostichopus japonicus via targeting the TLR component of IRAK-1 in vitro and in vivo. Sci. Rep. 2015, 5, 12608. [Google Scholar] [CrossRef] [Green Version]
  104. Schulte, L.N.; Westermann, A.J.; Vogel, J. Differential activation and functional specialization of miR-146 and miR-155 in innate immune sensing. Nucleic Acids Res. 2013, 41, 542–553. [Google Scholar] [CrossRef]
  105. Chen, Y.; Chen, J.; Wang, H.; Shi, J.; Wu, K.; Liu, S.; Liu, Y.; Wu, J. HCV-induced miR-21 contributes to evasion of host immune system by targeting MyD88 and IRAK1. PLoS Pathog. 2013, 9, e1003248. [Google Scholar] [CrossRef]
  106. Xu, Z.; Dong, D.; Chen, X.; Huang, H.; Wen, S. MicroRNA-381 negatively regulates TLR4 signaling in A549 cells in response to LPS stimulation. Biomed. Res. Int. 2015, 2015, 849475. [Google Scholar] [CrossRef] [Green Version]
  107. Park, H.; Huang, X.; Lu, C.; Cairo, M.S.; Zhou, X. MicroRNA-146a and microRNA-146b regulate human dendritic cell apoptosis and cytokine production by targeting TRAF6 and IRAK1 proteins. J. Biol. Chem. 2015, 290, 2831–2841. [Google Scholar] [CrossRef] [Green Version]
  108. Arenas-Padilla, M.; Mata-Haro, V. Regulation of TLR signaling pathways by microRNAs: Implications in inflammatory diseases. Centr. Eur. J. Immunol. 2018, 43, 482–489. [Google Scholar] [CrossRef] [Green Version]
  109. Cherradi, N.; Feige, J.-J. Tristetraprolin (ZFP36) and TIS11B (ZFP36-L1). In Encyclopedia of Signaling Molecules; Choi, S., Ed.; Springer International Publishing: New York, NY, USA, 2018; Volume 9, pp. 5709–5718. [Google Scholar]
  110. Anwar, M.A.; Shah, M.; Kim, J.; Choi, S. Recent clinical trends in Toll-like receptor targeting therapeutics. Med. Res. Rev. 2019, 39, 1053–1090. [Google Scholar] [CrossRef] [Green Version]
  111. Mahlokozera, T.; Vellimana, A.K.; Li, T.; Mao, D.D.; Zohny, Z.S.; Kim, D.H.; Tran, D.D.; Marcus, D.S.; Fouke, S.J.; Campian, J.L.; et al. Biological and therapeutic implications of multisector sequencing in newly diagnosed glioblastomas. Neuro-Oncol. 2018, 20, 472–483. [Google Scholar] [CrossRef]
  112. Liu, F.; Huang, J.; Liu, X.; Cheng, Q.; Luo, C.; Liu, Z. CTLA-4 correlates with immune and clinical characteristics of glioma. Cancer Cell Int. 2020, 20, 7. [Google Scholar] [CrossRef]
  113. Lim, M.; Xia, Y.; Bettegowda, C.; Weller, M. Current state of immunotherapy for glioblastoma. Nat. Rev. Clin. Oncol. 2018, 15, 422–442. [Google Scholar] [CrossRef] [PubMed]
  114. Aslan, K.; Turco, V.; Blobner, J.; Sonner, J.K.; Liuzzi, A.R.; Núñez, N.G.; De Feo, D.; Kickingereder, P.; Fischer, M.; Green, E.; et al. Heterogeneity of response to immune checkpoint blockade in hypermutated experimental gliomas. Nat. Commun. 2020, 11, 931. [Google Scholar] [CrossRef] [Green Version]
  115. Dong, H.; Zhu, G.; Tamada, K.; Chen, L. B7-H1, a third member of the B7 family, costimulates T-cell proliferation and interleukin-10 secretion. Nat. Med. 1999, 5, 1365–1369. [Google Scholar] [CrossRef] [PubMed]
  116. Chen, J.; Jiang, C.C.; Jin, L.; Zhang, X.D. Regulation of PD-L1: A novel role of pro-survival signalling in cancer. Ann. Oncol. 2016, 27, 409–416. [Google Scholar] [CrossRef] [PubMed]
  117. Wang, L.L.; Li, Z.H.; Hu, X.H.; Muyayalo, K.P.; Zhang, Y.H.; Liao, A.H. The roles of the PD-1/PD-L1 pathway at immunologically privileged sites. Am. J. Reprod. Immunol. 2017, 78, e12710. [Google Scholar] [CrossRef]
  118. Sims, J.S.; Ung, T.H.; Neira, J.A.; Canoll, P.; Bruce, J.N. Biomarkers for glioma immunotherapy: The next generation. J. Neurooncol. 2015, 123, 359–372. [Google Scholar] [CrossRef]
  119. Litak, J.; Mazurek, M.; Grochowski, C.; Kamieniak, P.; Roliński, J. PD-L1/PD-1 Axis in Glioblastoma Multiforme. Int. J. Mol. Sci. 2019, 20, 5347. [Google Scholar] [CrossRef] [Green Version]
  120. Van Gool, S.W. Brain Tumor Immunotherapy: What have We Learned so Far? Front. Oncol. 2015, 5, 98. [Google Scholar] [CrossRef] [Green Version]
  121. Pfannenstiel, L.W.; McNeilly, C.; Xiang, C.; Kang, K.; Diaz-Montero, C.M.; Yu, J.S.; Gastman, B.R. Combination PD-1 blockade and irradiation of brain metastasis induces an effective abscopal effect in melanoma. Oncoimmunology 2018, 8, e1507669. [Google Scholar] [CrossRef] [Green Version]
  122. Zhang, X.; Zhu, S.; Li, T.; Liu, Y.J.; Chen, W.; Chen, J. Targeting immune checkpoints in malignant glioma. Oncotarget 2017, 8, 7157–7174. [Google Scholar] [CrossRef] [PubMed]
  123. Röver, L.K.; Gevensleben, H.; Dietrich, J.; Bootz, F.; Landsberg, J.; Goltz, D.; Dietrich, D. PD-1 (PDCD1) Promoter Methylation Is a Prognostic Factor in Patients With Diffuse Lower-Grade Gliomas Harboring Isocitrate Dehydrogenase (IDH) Mutations. EBioMedicine 2018, 28, 97–104. [Google Scholar] [CrossRef] [Green Version]
  124. Qian, J.; Wang, C.; Wang, B.; Yang, J.; Wang, Y.; Luo, F.; Xu, J.; Zhao, C.; Liu, R.; Chu, Y. The IFN-γ/PD-L1 axis between T cells and tumor microenvironment: Hints for glioma anti-PD-1/PD-L1 therapy. J. Neuroinflammation 2018, 15, 290. [Google Scholar] [CrossRef] [Green Version]
  125. Goods, B.A.; Hernandez, A.L.; Lowther, D.E.; Lucca, L.E.; Lerner, B.A.; Gunel, M.; Raddassi, K.; Coric, V.; Hafler, D.A.; Love, J.C. Functional differences between PD-1+ and PD-1-CD4+ effector T cells in healthy donors and patients with glioblastoma multiforme. PLoS ONE 2017, 12, e0181538. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Chinai, J.M.; Janakiram, M.; Chen, F.; Chen, W.; Kaplan, M.; Zang, X. New immunotherapies targeting the PD-1 pathway. Trends Pharm. Sci. 2015, 36, 587–595. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Lenting, K.; Verhaak, R.; Ter Laan, M.; Wesseling, P.; Leenders, W. Glioma: Experimental models and reality. Acta Neuropathol. 2017, 133, 263–282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Beswick, E.J.; Johnson, J.R.; Saada, J.I.; Humen, M.; House, J.; Dann, S.; Qiu, S.; Brasier, A.R.; Powell, D.W.; Reyes, V.E.; et al. TLR4 activation enhances the PD-L1-mediated tolerogenic capacity of colonic CD90+ stromal cells. J. Immunol. 2014, 193, 2218–2229. [Google Scholar] [CrossRef] [Green Version]
  129. Wolfle, S.J.; Strebovsky, J.; Bartz, H.; Sahr, A.; Arnold, C.; Kaiser, C.; Dalpke, A.H.; Heeg, K. PD-L1 expression on tolerogenic APCs is controlled by STAT-3. Eur. J. Immunol. 2011, 41, 413–424. [Google Scholar] [CrossRef]
  130. Zhao, S.; Sun, M.; Meng, H.; Ji, H.; Liu, Y.; Zhang, M.; Li, H.; Li, P.; Zhang, Y.; Zhang, Q. TLR4 expression correlated with PD-L1 expression indicates a poor prognosis in patients with peripheral T-cell lymphomas. Cancer Manag. Res. 2019, 11, 4743–4756. [Google Scholar] [CrossRef] [Green Version]
  131. He, X.; Semenov, M.; Tamai, K.; Zeng, X. LDL receptor related proteins 5 and 6 in Wnt/β-catenin signaling: Arrows point the way. Development 2004, 131, 1663–1677. [Google Scholar] [CrossRef] [Green Version]
  132. Minami, Y.; Oishi, I.; Endo, M.; Nishita, M. Ror-family receptor tyrosine kinases in noncanonical Wnt signaling: Their implications in developmental morphogenesis and human diseases. Dev. Dyn. 2010, 239, 1–15. [Google Scholar] [CrossRef] [PubMed]
  133. Peradziryi, H.; Tolwinski, N.S.; Borchers, A. The many roles of PTK7: A versatile regulator of cell–cell communication. Arch. Biochem. Biophys. 2012, 524, 71–76. [Google Scholar] [CrossRef] [PubMed]
  134. Fradkin, L.G.; Dura, J.M.; Noordermeer, J.N. Ryks: New partners for Wnts in the developing and regenerating nervous system. Trends Neurosci. 2010, 33, 84–92. [Google Scholar] [CrossRef] [PubMed]
  135. Jing, L.; Lefebvre, J.L.; Gordon, L.R.; Granato, M. Wnt signals organize synaptic prepattern and axon guidance through the zebrafish unplugged/MuSK receptor. Neuron 2009, 61, 721–733. [Google Scholar] [CrossRef] [Green Version]
  136. Kikuchi, A.; Yamamoto, H.; Sato, A.; Matsumoto, S. Discovery of MUSK as a new WNT (co-?) receptor in axon guidance. New insights into the mechanism of Wnt signaling pathway activation. Int. Rev. Cell Mol. Biol. 2011, 291, 21–71. [Google Scholar]
  137. Cruciat, C.M.; Niehrs, C. Secreted and transmembrane Wnt inhibitors and activators. Cold Spring Harb. Perspect. Biol. 2013, 5, a015081. [Google Scholar] [CrossRef] [Green Version]
  138. Abbott, N.J.; Patabendige, A.A.; Dolman, D.E.; Yusof, S.R.; Begley, D.J. Structure and function of the blood–brain barrier. Neurobiol. Dis. 2010, 37, 13–25. [Google Scholar] [CrossRef]
  139. Butt, A.M.; Jones, H.C.; Abbott, N.J. Electrical resistance across the blood–brain barrier in anaesthetized rats: A developmental study. J. Physiol. 1990, 429, 47–62. [Google Scholar] [CrossRef]
  140. Wolburg, H.; Lippoldt, A. Tight junctions of the blood–brain barrier: Development, composition and regulation. Vasc. Pharmacol. 2002, 38, 323–337. [Google Scholar] [CrossRef]
  141. Bauer, H.C.; Bauer, H.; Lametschwandtner, A.; Amberger, A.; Ruiz, P.; Steiner, M. Neovascularization and the appearance of morphological characteristics of the blood–brain barrier in the embryonic mouse central nervous system. Brain Res. Dev. Brain Res. 1993, 75, 269–278. [Google Scholar] [CrossRef]
  142. Amasheh, S.; Schmidt, T.; Mahn, M.; Florian, P.; Mankertz, J.; Tavalali, S.; Gitter, A.H.; Schulzke, J.D.; Fromm, M. Contribution of claudin-5 to barrier properties in tight junctions of epithelial cells. Cell Tissue Res. 2005, 321, 89–96. [Google Scholar] [CrossRef] [PubMed]
  143. Ohtsuki, S.; Sato, S.; Yamaguchi, H.; Kamoi, M.; Asashima, T.; Terasaki, T. Exogenous expression of claudin-5 induces barrier properties in cultured rat brain capillary endothelial cells. J. Cell Physiol. 2007, 210, 81–86. [Google Scholar] [CrossRef] [PubMed]
  144. Nitta, T.; Hata, M.; Gotoh, S.; Seo, Y.; Sasaki, H.; Hashimoto, N.; Furuse, M.; Tsukita, S. Size-selective loosening of the blood–brain barrier in claudin-5-deficient mice. J. Cell Biol. 2003, 161, 653–660. [Google Scholar] [CrossRef] [PubMed]
  145. Zuccarini, M.; Giuliani, P.; Ziberi, S.; Carluccio, M.; Iorio, P.D.; Caciagli, F.; Ciccarelli, R. The Role of Wnt Signal in Glioblastoma Development and Progression: A Possible New Pharmacological Target for the Therapy of This Tumor. Genes (Basel) 2018, 9, 105. [Google Scholar] [CrossRef] [Green Version]
  146. Rajakulendran, N.; Rowland, K.J.; Selvadurai, H.J.; Ahmadi, M.; Park, N.I.; Naumenko, S.; Dolma, S.; Ward, R.J.; So, M.; Lee, L.; et al. Wnt and Notch signaling govern self-renewal and differentiation in a subset of human glioblastoma stem cells. Genes Dev. 2019, 33, 498–510. [Google Scholar] [CrossRef]
  147. De Robertis, A.; Valensin, S.; Rossi, M.; Tunici, P.; Verani, M.; De Rosa, A.; Giordano, C.; Varrone, M.; Nencini, A.; Pratelli, C.; et al. Identification and characterization of a small-molecule inhibitor of Wnt signaling in glioblastoma cells. Mol. Cancer Ther. 2013, 12, 1180–1189. [Google Scholar] [CrossRef] [Green Version]
  148. Broekman, M.L.; Maas, S.L.N.; Abels, E.R.; Mempel, T.R.; Krichevsky, A.M.; Breakefield, X.O. Multidimensional communication in the microenvirons of glioblastoma. Nat. Rev. Neurol. 2018, 14, 482–495. [Google Scholar] [CrossRef]
  149. Zarnescu, O.; Brehar, F.M.; Chivu, M.; Ciurea, A.V. Immunohistochemical localization of caspase-3, caspase-9 and Bax in U87 glioblastoma xenografts. J. Mol. Hist. 2008, 39, 561–569. [Google Scholar] [CrossRef]
  150. Tirapelli, L.F.; Bolini, P.H.; Tirapelli, D.P.; Peria, F.M.; Becker, A.N.; Saggioro, F.P.; Carlotti, C.G., Jr. Caspase-3 and Bcl-2 expression in glioblastoma: An immunohistochemical study. Arq. Neuropsiquiatr. 2010, 68, 603–607. [Google Scholar] [CrossRef] [Green Version]
  151. Gdynia, G.; Grund, K.; Eckert, A.; Böck, B.C.; Funke, B.; Macher- Goeppinger, S.; Sieber, S.; Herold-Mende, C.; Wiestler, B.; Wiestler, O.D.; et al. Basal caspase activity promotes migration and invasiveness in glioblastoma cells. Mol. Cancer Res. 2007, 5, 1232–1240. [Google Scholar] [CrossRef] [Green Version]
  152. Jia, W.; Lu, R.; Martin, T.A.; Jiang, W.G. The role of claudin-5 in blood-brain barrier (BBB) and brain metastases (review). Mol. Med. Rep. 2014, 9, 779–785. [Google Scholar] [CrossRef] [Green Version]
  153. Hara, K.; Kageji, T.; Mizobuchi, Y.; Kitazato, K.T.; Okazaki, T.; Fujihara, T.; Nakajima, K.; Mure, H.; Kuwayama, K.; Hara, T.; et al. Blocking of the interaction between Wnt proteins and their co-receptors contributes to the anti-tumor effects of adenovirus-mediated DKK3 in glioblastoma. Cancer Lett. 2015, 356, 496–505. [Google Scholar] [CrossRef]
  154. Che, F.; Yin, J.; Quan, Y.; Xie, X.; Heng, X.; Du, Y.; Wang, L. TLR4 interaction with LPS in glioma CD133+ cancer stem cells induces cell proliferation, resistance to chemotherapy and evasion from cytotoxic T lymphocyte-induced cytolysis. Oncotarget 2017, 8, 53495–53507. [Google Scholar] [CrossRef] [Green Version]
  155. Rajesh, Y.; Pal, I.; Banik, P.; Chakraborty, S.; Borkar, S.A.; Dey, G.; Mukherjee, A.; Mandal, M. Insights into molecular therapy of glioma: Current challenges and next generation blueprint. Acta Pharm. Sin. 2017, 38, 591–613. [Google Scholar] [CrossRef] [Green Version]
  156. Dzaye, O.; Hu, F.; Derkow, K.; Haage, V.; Euskirchen, P.; Harms, C.; Lehnardt, S.; Synowitz, M.; Wolf, S.A.; Kettenmann, H. Glioma Stem Cells but Not Bulk Glioma Cells Upregulate IL-6 Secretion in Microglia/Brain Macrophages via Toll-like Receptor 4 Signaling. J. Neuropathol. Exp. Neurol. 2016, 75, 429–440. [Google Scholar] [CrossRef] [Green Version]
  157. Ma, Q.; Long, W.; Xing, C.; Chu, J.; Luo, M.; Wang, H.Y.; Liu, Q.; Wang, R.F. Cancer Stem Cells and Immunosuppressive Microenvironment in Glioma. Front. Immunol. 2018, 9, 2924. [Google Scholar] [CrossRef] [Green Version]
  158. Testa, U.; Castelli, G.; Pelosi, E. Genetic Abnormalities, Clonal Evolution, and Cancer Stem Cells of Brain Tumors. Med. Sci. (Basel) 2018, 6, 85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Widera, D.; Martínez Aguilar, R.; Cottrell, G.S. Toll-like receptor 4 and protease-activated receptor 2 in physiology and pathophysiology of the nervous system: More than just receptor cooperation? Neural Regen. Res. 2019, 14, 1196–1201. [Google Scholar] [CrossRef]
  160. Alvarado, A.G.; Thiagarajan, P.S.; Mulkearns-Hubert, E.E.; Silver, D.J.; Hale, J.S.; Alban, T.J.; Turaga, S.M.; Jarrar, A.; Reizes, O.; Longworth, M.S.; et al. Glioblastoma Cancer Stem Cells Evade Innate Immune Suppression of Self-Renewal through Reduced TLR4 Expression. Cell Stem Cell 2017, 20, 450–461. [Google Scholar] [CrossRef] [Green Version]
  161. Zhou, H.; Bao, J.; Zhu, X.; Dai, G.; Jiang, X.; Jiao, X.; Sheng, H.; Huang, J.; Yu, H. Retinoblastoma Binding Protein 5 Correlates with the Progression in Hepatocellular Carcinoma. Biomed Res. Int. 2018, 2018, 1073432. [Google Scholar] [CrossRef]
  162. Ghasemzadeh, A.; Bivalacqua, T.J.; Hahn, N.M.; Drake, C.G. New Strategies in Bladder Cancer: A Second Coming for Immunotherapy. Clin. Cancer Res. 2016, 22, 793–801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Ou, T.; Lilly, M.; Jiang, W. The Pathologic Role of Toll-Like Receptor 4 in Prostate Cancer. Front. Immunol. 2018, 9, 1188. [Google Scholar] [CrossRef] [PubMed]
  164. Li, X.X.; Sun, G.P.; Meng, J.; Li, X.; Tang, Y.X.; Li, Z.; Wang, M.F.; Liang, G.F.; Lu, X.B. Role of toll-like receptor 4 in colorectal carcinogenesis: A meta-analysis. PLoS ONE 2014, 9, e93904. [Google Scholar] [CrossRef]
  165. Dapito, D.H.; Mencin, A.; Gwak, G.Y.; Pradere, J.P.; Jang, M.K.; Mederacke, I.; Caviglia, J.M.; Khiabanian, H.; Adeyemi, A.; Bataller, R.; et al. Promotion of hepatocellular carcinoma by the intestinal microbiota and TLR4. Cancer Cell 2012, 21, 504–516. [Google Scholar] [CrossRef] [Green Version]
  166. Thuringer, D.; Hammann, A.; Benikhlef, N.; Fourmaux, E.; Bouchot, A.; Wettstein, G.; Solary, E.; Garrido, C. Transactivation of the epidermal growth factor receptor by heat shock protein 90 via Toll-like receptor 4 contributes to the migration of glioblastoma cells. J. Biol. Chem. 2011, 286, 418–3428. [Google Scholar] [CrossRef] [Green Version]
  167. Chicoine, M.R.; Zahner, M.; Won, E.K.; Kalra, R.R.; Kitamura, T.; Perry, A.; Higashikubo, R. The in vivo antitumoral effects of lipopolysaccharide against glioblastoma multiforme are mediated in part by Toll-like receptor 4. Neurosurgery 2007, 60, 372–380. [Google Scholar] [CrossRef]
  168. Kawanishi, Y.; Tominaga, A.; Okuyama, H.; Fukuoka, S.; Taguchi, T.; Kusumoto, Y.; Yawata, T.; Fujimoto, Y.; Ono, S.; Shimizu, K. Regulatory effects of Spirulina complex polysaccharides on growth of murine RSV-M glioma cells through Toll-like receptor 4. Microbiol. Immunol. 2013, 57, 63–73. [Google Scholar] [CrossRef]
  169. Shinohara, H.; Yagita, H.; Ikawa, Y.; Oyaizu, N. Fas drives cell cycle progression in glioma cells via extracellular signal-regulated kinase activation. Cancer Res. 2000, 60, 1766–1772. [Google Scholar]
  170. Wu, Y.; Sun, L.; Zou, W.; Xu, J.; Liu, H.; Wang, W.; Yun, X.; Gu, J. Prosaposin, a regulator of estrogen receptor alpha, promotes breast cancer growth. Cancer Sci. 2012, 103, 1820–1825. [Google Scholar] [CrossRef]
  171. Jiang, Y.; Zhou, J.; Luo, P.; Gao, H.; Ma, Y.; Chen, Y.S.; Li, L.; Zou, D.; Zhang, Y.; Jing, Z. Prosaposin promotes the proliferation and tumorigenesis of glioma through toll-like receptor 4 (TLR4)-mediated NF-κB signaling pathway. EBioMedicine 2018, 37, 78–79. [Google Scholar] [CrossRef] [Green Version]
  172. Bowles, A.P., Jr.; Perkins, E. Long-term remission of malignant brain tumors after intracranial infection: A report of four cases. Neurosurgery 1999, 44, 636–642. [Google Scholar] [CrossRef] [PubMed]
  173. De Bonis, P.; Albanese, A.; Lofrese, G.; de Waure, C.; Mangiola, A.; Pettorini, B.L.; Pompucci, A.; Balducci, M.; Fiorentino, A.; Lauriola, L.; et al. Postoperative infection may influence survival in patients with glioblastoma: Simply a myth. Neurosurgery 2011, 69, 864–868. [Google Scholar] [CrossRef] [PubMed]
  174. Bohman, L.E.; Gallardo, J.; Hankinson, T.C.; Waziri, A.E.; Mandigo, C.E.; McKhann, G.M., 2nd; Sisti, M.B.; Canoll, P.; Bruce, J.N. The survival impact of postoperative infection in patients with glioblastoma multiforme. Neurosurgery 2009, 64, 828–834. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Agrawal, N.; Bettegowda, C.; Cheong, I.; Geschwind, J.F.; Drake, C.G.; Hipkiss, E.L.; Tatsumi, M.; Dang, L.H.; Diaz, L.A., Jr.; Pomper, M.; et al. Bacteriolytic therapy can generate a potent immune response against experimental tumors. Proc. Natl. Acad. Sci. USA 2004, 101, 15172–15177. [Google Scholar] [CrossRef] [Green Version]
  176. Wang, H.; Cho, C.H. Effect of nf-kappab signaling on apoptosis in chronic inflammation-associated carcinogenesis. Curr. Cancer Drug Targets 2010, 10, 593–599. [Google Scholar] [CrossRef]
  177. Baldwin, A.S. Regulation of cell death and autophagy by ikk and nf-kappab: Critical mechanisms in immune function and cancer. Immunol. Rev. 2012, 246, 327–345. [Google Scholar] [CrossRef]
  178. Guo, S.; Nighot, M.; Al-Sadi, R.; Alhmoud, T.; Nighot, P.; Ma, T.Y. Lipopolysaccharide Regulation of Intestinal Tight Junction Permeability Is Mediated by TLR4 Signal Transduction Pathway Activation of FAK and MyD88. J. Immunol. 2015, 195, 4999–5010. [Google Scholar] [CrossRef]
  179. Kondo, Y.; Ikeda, K.; Tokuda, N.; Nishitani, C.; Ohto, U.; Akashi-Takamura, S.; Ito, Y.; Uchikawa, M.; Kuroki, Y.; Taguchi, R.; et al. TLR4-MD-2 complex is negatively regulated by an endogenous ligand, globotetraosylceramide. Proc. Natl. Acad. Sci. USA 2013, 110, 4714–4719. [Google Scholar] [CrossRef] [Green Version]
  180. Wang, Y.; Su, L.; Morin, M.D.; Jones, B.T.; Whitby, L.R.; Surakattula, M.M.; Huang, H.; Shi, H.; Choi, J.H.; Wang, K.W.; et al. TLR4/MD-2 activation by a synthetic agonist with no similarity to LPS. Proc. Natl. Acad. Sci. USA 2016, 113, E884–E893. [Google Scholar] [CrossRef] [Green Version]
  181. Matzner, P.; Sorski, L.; Shaashua, L.; Elbaz, E.; Lavon, H.; Melamed, R.; Rosenne, E.; Gotlieb, N.; Benbenishty, A.; Reed, S.G.; et al. Perioperative treatment with the new synthetic TLR-4 agonist GLA-SE reduces cancer metastasis without adverse effects. Int. J. Cancer 2016, 138, 1754–1764. [Google Scholar] [CrossRef] [Green Version]
  182. Coler, R.N.; Day, T.A.; Ellis, R.; Piazza, F.M.; Beckmann, A.M.; Vergara, J.; Rolf, T.; Lu, L.; Alter, G.; Hokey, D.; et al. TBVPX-113 Study Team. The TLR-4 agonist adjuvant, GLA-SE, improves magnitude and quality of immune responses elicited by the ID93 tuberculosis vaccine: First-in-human trial. NPJ Vaccines 2018, 3, 34. [Google Scholar] [CrossRef] [PubMed]
  183. Fensterheim, B.A.; Young, J.D.; Luan, L.; Kleinbard, R.R.; Stothers, C.L.; Patil, N.K.; McAtee-Pereira, A.G.; Guo, Y.; Trenary, I.; Hernandez, A.; et al. The TLR4 Agonist Monophosphoryl Lipid A Drives Broad Resistance to Infection via Dynamic Reprogramming of Macrophage Metabolism. J. Immunol. 2018, 200, 3777–3789. [Google Scholar] [CrossRef] [PubMed]
  184. Stark, R.; Choi, H.; Koch, S.; Lamb, F.; Sherwood, E. Monophosphoryl lipid A inhibits the cytokine response of endothelial cells challenged with LPS. Innate Immun. 2015, 21, 565–574. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Gao, J.; Guo, Z. Progress in the synthesis and biological evaluation of lipid A and its derivatives. Med. Res. Rev. 2018, 38, 556–601. [Google Scholar] [CrossRef]
  186. Carpentier, A.; Laigle-Donadey, F.; Zohar, S.; Capelle, L.; Behin, A.; Tibi, A.; Martin-Duverneuil, N.; Sanson, M.; Lacomblez, L.; Taillibert, S.; et al. Phase 1 trial of a CpG oligodeoxynucleotide for patients with recurrent glioblastoma. Neuro Oncol. 2006, 8, 60–66. [Google Scholar] [CrossRef]
  187. Zhou, M.; McFarland-Mancini, M.M.; Funk, H.M.; Husseinzadeh, N.; Mounajjed, T.; Drew, A.F. Toll-like receptor expression in normal ovary and ovarian tumors. Cancer Immunol. Immunother. 2009, 58, 1375–1385. [Google Scholar] [CrossRef]
  188. Andreani, V.; Gatti, G.; Simonella, L.; Rivero, V.; Maccioni, M. Activation of Toll-like receptor 4 on tumor cells in vitro inhibits subsequent tumor growth in vivo. Cancer Res. 2007, 67, 10519–10527. [Google Scholar] [CrossRef] [Green Version]
  189. Alibashe-Ahmed, M.; Roger, T.; Serre-Beinier, V.; Berishvili, E.; Reith, W.; Bosco, D.; Berney, T. Macrophage migration inhibitory factor regulates TLR4 expression and modulates TCR/CD3-mediated activation in CD4+ T lymphocytes. Sci. Rep. 2019, 9, 9380. [Google Scholar] [CrossRef] [Green Version]
  190. Kumar, R.; de Mooij, T.; Peterson, T.E.; Kaptzan, T.; Johnson, A.J.; Daniels, D.J.; Parney, I.F. Modulating glioma-mediated myeloid-derived suppressor cell development with sulforaphane. PLoS ONE 2017, 12, e0179012. [Google Scholar] [CrossRef]
  191. Li, W.; Holsinger, R.M.; Kruse, C.A.; Flügel, A.; Graeber, M.B. The potential for genetically altered microglia to influence glioma treatment. CNS Neurol. Disord. Drug Targets 2013, 12, 750–762. [Google Scholar] [CrossRef] [Green Version]
  192. Roger, T.; David, J.; Glauser, M.P.; Calandra, T. MIF regulates innate immune responses through modulation of Toll-like receptor 4. Nature 2001, 414, 920–924. [Google Scholar] [CrossRef] [PubMed]
  193. Ohta, S.; Yaguchi, T.; Okuno, H.; Chneiweiss, H.; Kawakami, Y.; Okano, H. CHD7 promotes proliferation of neural stem cells mediated by MIF. Mol. Brain 2016, 9, 96. [Google Scholar] [CrossRef] [Green Version]
  194. Roger, T.; Schneider, A.; Weier, M.; Sweep, F.C.; Le Roy, D.; Bernhagen, J.; Calandra, T.; Giannoni, E. High expression levels of macrophage migration inhibitory factor sustain the innate immune responses of neonates. Proc. Natl. Acad. Sci. USA 2016, 113, E997–E1005. [Google Scholar] [CrossRef] [Green Version]
  195. Ohkawara, T.; Takeda, H.; Nishihira, J.; Miyashita, K.; Nihiwaki, M.; Ishiguro, Y.; Takeda, K.; Akira, S.; Iwanaga, T.; Sugiyama, T.; et al. Macrophage migration inhibitory factor contributes to the development of acute dextran sulphate sodium-induced colitis in Toll-like receptor 4 knockout mice. Clin. Exp. Immunol. 2005, 141, 412–421. [Google Scholar] [CrossRef] [PubMed]
  196. Schwenkgrub, J.; Zaremba, M.; Mirowska-Guzel, D.; Kurkowska-Jastrzebska, I. Ibudilast: A nonselective phosphodiesterase inhibitor in brain disorders. Postepy Hig. Med. Dosw. 2017, 71, 137–148. [Google Scholar] [CrossRef]
  197. Ha, W.; Sevim-Nalkiran, H.; Zaman, A.M.; Matsuda, K.; Khasraw, M.; Nowak, A.K.; Chung, L.; Baxter, R.C.; McDonald, K.L. Ibudilast sensitizes glioblastoma to temozolomide by targeting Macrophage Migration Inhibitory Factor (MIF). Sci. Rep. 2019, 9, 2905. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  198. Roger, T.; Froidevaux, C.; Martin, C.; Calandra, T. Macrophage migration inhibitory factor (MIF) regulates host responses to endotoxin through modulation of Toll-like receptor 4 (TLR4). J. Endotoxin Res. 2003, 9, 119–123. [Google Scholar] [CrossRef] [PubMed]
  199. Basu, S.; Binder, R.J.; Ramalingam, T.; Srivastava, P.K. CD91 is a common receptor for heat shock proteins gp96, hsp90, hsp70, and calreticulin. Immunity 2001, 14, 303–313. [Google Scholar] [CrossRef] [Green Version]
  200. Castellino, F.; Boucher, P.E.; Eichelberg, K.; Mayhew, M.; Rothman, J.E.; Houghton, A.N.; Germain, R.N. Receptor-mediated uptake of antigen/heat shock protein complexes results in major histocompatibility complex class I antigen presentation via two distinct processing pathways. J. Exp. Med. 2000, 191, 1957–1964. [Google Scholar] [CrossRef] [Green Version]
  201. Matsutake, T.; Sawamura, T.; Srivastava, P.K. High efficiency CD91- and LOX-1-mediated representation of gp96-chaperoned peptides by MHC II molecules. Cancer Immun. 2010, 10, 7. [Google Scholar]
  202. Berwin, B.; Hart, J.P.; Pizzo, S.V.; Nicchitta, C.V. Cutting edge: CD91-independent cross-presentation of GRP94(gp96)-associated peptides. J. Immunol. 2002, 168, 4282–4286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Panjwani, N.N.; Popova, L.; Srivastava, P.K. Heat shock proteins gp96 and hsp70 activate the release of nitric oxide by APCs. J. Immunol. 2002, 168, 2997–3003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Srivastava, P. Roles of heat-shock proteins in innate and adaptive immunity. Nat. Rev. Immunol. 2002, 2, 185–194. [Google Scholar] [CrossRef]
  205. Crane, C.A.; Han, S.J.; Ahn, B.; Oehlke, J.; Kivett, V.; Fedoroff, A.; Butowski, N.; Chang, S.M.; Clarke, J.; Berger, M.S.; et al. Individual patient-specific immunity against high-grade glioma after vaccination with autologous tumor derived peptides bound to the 96 KD chaperone protein. Clin. Cancer Res. 2013, 19, 205–214. [Google Scholar] [CrossRef] [Green Version]
  206. Bloch, O.; Crane, C.A.; Fuks, Y.; Kaur, R.; Aghi, M.K.; Berger, M.S.; Butowski, N.A.; Chang, S.M.; Clarke, J.L.; McDermott, M.W.; et al. Heat-shock protein peptide complex-96 vaccination for recurrent glioblastoma: A phase II, single-arm trial. Neuro-Oncology 2014, 16, 274–279. [Google Scholar] [CrossRef]
  207. Ampie, L.; Choy, W.; Lamano, J.B.; Fakurnejad, S.; Bloch, O.; Parsa, A.T. Heat shock protein vaccines against glioblastoma: From bench to bedside. J. Neurooncol. 2015, 123, 441–448. [Google Scholar] [CrossRef] [Green Version]
  208. Khansur, E.; Shah, A.H.; Lacy, K.; Komotar, R.J. Novel Immunotherapeutics for Treatment of Glioblastoma: The Last Decade of Research. Cancer Investig. 2019, 37, 1–7. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Toll-like-receptor (TLR)-4, after activation by pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs) supported by myeloid differentiation factor-2 (MD-2), initiates downstream signaling in two major ways. The first is the MyD88-dependent pathway connected with toll-interleukin 1 receptor domain-containing adaptor protein (TIRAP) resulting in myddosome formation, a structure containing IRAK-4 and IRAK-1 kinases. Activation of IRAK-1 results in TRAF-6 induction with collateral ubiquitination and phosphorylation. TRAF-6 promotes TAK-1. Activation of TAK-1 results in IKK/NF-κB complex formation and promotion. ERK1/2 kinases are also activated, which indicates activated protein-1 (Ap-1), the factor activating the transcription process. The second pathway, MyD88 -independent, connected with translocating chain-associating membrane (TRAM), leads through toll-like receptor adaptor molecule-1 (TRIF) activation resulting in TRAF-3 and TRAF-6 promotion. RIP is recruited by TRAF-6, activating RIP signaling on the TAK-1/ERK 1/2/Ap-1 axis. TRAF-3 activates IKK and TBK-1 to activate interferon regulatory factor-3 (IRF-3). IRF-3 induces interferon expression and other proinflammatory cytokines.
Figure 1. Toll-like-receptor (TLR)-4, after activation by pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs) supported by myeloid differentiation factor-2 (MD-2), initiates downstream signaling in two major ways. The first is the MyD88-dependent pathway connected with toll-interleukin 1 receptor domain-containing adaptor protein (TIRAP) resulting in myddosome formation, a structure containing IRAK-4 and IRAK-1 kinases. Activation of IRAK-1 results in TRAF-6 induction with collateral ubiquitination and phosphorylation. TRAF-6 promotes TAK-1. Activation of TAK-1 results in IKK/NF-κB complex formation and promotion. ERK1/2 kinases are also activated, which indicates activated protein-1 (Ap-1), the factor activating the transcription process. The second pathway, MyD88 -independent, connected with translocating chain-associating membrane (TRAM), leads through toll-like receptor adaptor molecule-1 (TRIF) activation resulting in TRAF-3 and TRAF-6 promotion. RIP is recruited by TRAF-6, activating RIP signaling on the TAK-1/ERK 1/2/Ap-1 axis. TRAF-3 activates IKK and TBK-1 to activate interferon regulatory factor-3 (IRF-3). IRF-3 induces interferon expression and other proinflammatory cytokines.
Ijms 21 03114 g001
Figure 2. Potential targets for future immunotherapy concerning TLR-4 signaling in glioblastoma multiforme: Complex of miRNA molecules inhibiting TLR-4 signaling on different stages, programmed death ligand-1 (PD-1L) overexpression stimulated by TLR downsignaling, indirect retinoblastoma binding protein 5 (RBBP5) inhibition by TBK-1 phosphorylation resulting in antineoplastic activity, interplay between TLR-4 and Dickkopf-related protein 3 (DKK-3) inhibition, Wnt pathway signaling restriction, claudin-5 level reduction, and caspase-9 elevation and caspase-3 decrease as a pro-oncogenic cascade after TLR-4 activation.
Figure 2. Potential targets for future immunotherapy concerning TLR-4 signaling in glioblastoma multiforme: Complex of miRNA molecules inhibiting TLR-4 signaling on different stages, programmed death ligand-1 (PD-1L) overexpression stimulated by TLR downsignaling, indirect retinoblastoma binding protein 5 (RBBP5) inhibition by TBK-1 phosphorylation resulting in antineoplastic activity, interplay between TLR-4 and Dickkopf-related protein 3 (DKK-3) inhibition, Wnt pathway signaling restriction, claudin-5 level reduction, and caspase-9 elevation and caspase-3 decrease as a pro-oncogenic cascade after TLR-4 activation.
Ijms 21 03114 g002
Table 1. Toll-like-receptor (TLR)-4 expression in brain tissue, cancer lines, and tumors.
Table 1. Toll-like-receptor (TLR)-4 expression in brain tissue, cancer lines, and tumors.
TLR-4 ExpressionLocalizationType of Cells
mRNA/protein TissueNeurons, microglia [13,14,15]
mRNA/protein TissueAstrocytes [16,17]
mRNACell LinesGlioma (U87, SF126, U251, GI 261) [18]
mRNA/protein Tumor/Cell LinesAstrocytoma/GBM (U87MG, A172, LN229, U118) [19]
Table 2. Characteristics of TLR-4 receptor: types of cells expressing TLR-4, pathogen-associated molecular patterns (PAMPs), danger-associated molecular patterns (DAMPs), and clinical-trial agents.
Table 2. Characteristics of TLR-4 receptor: types of cells expressing TLR-4, pathogen-associated molecular patterns (PAMPs), danger-associated molecular patterns (DAMPs), and clinical-trial agents.
TLR-4
Types of CellsPAMPsDAMPsClinical-Trial Agent
Characteristics
Monocytes
Macrophages
Neutrophil
Myeloid dendritic cells
Mast cells
B cells
Intestinal epithelium
Platelets
LipopolysaccharideHSPs, heparin, fibrinogen,
fibronectin,
sulfate,
HMGB1,
ANG II
Anti-TLR-4 antibody,
lipid A derivates,
polysaccharides.
Table 3. First- and second-phase clinical trials concerning TLR-4 agonists and antagonists in the treatment of various neoplasms.
Table 3. First- and second-phase clinical trials concerning TLR-4 agonists and antagonists in the treatment of various neoplasms.
Clinical Trial NumberPhaseIndication Agonist/Antagonist of TLR-4Ligand Characteristic
NCT 023203051MelanomaAgonistMART-1
NCT 021806981Soft tissue sarcomaAgonistGLA-SE
NCT025014731,2Follicular lymphomaAgonistG100
NCT029956551Acute myeloid leukemiaAntagonistCX100
NCT020356571Merkel cell carcinomaAgonistGLA-SE
NCT022703721Breast and ovarian cancerAgonistONT-10
NCT015567891Solid tumorsAgonistONT-10
NCT026099842SarcomaAgonistCMB-305
NCT023871252Non-small lung cancerAgonistCMB-305
Table 4. Clinical trials concerning heat shock protein peptide complex (HSSPC) vaccination in glioblastoma-multiforme (GBM) patients. (https://www.clinicaltrials.gov).
Table 4. Clinical trials concerning heat shock protein peptide complex (HSSPC) vaccination in glioblastoma-multiforme (GBM) patients. (https://www.clinicaltrials.gov).
Clinical Trial NumberPhaseImmunological AgentNumber of ParticipantsIndicationAgeCountry
NCT036502572Autologous Heat Shock Protein (gp96)
Vaccine
150GBMAdultChina
NCT027225122HSPPC-96 Vaccine20hGG,
rGBM,
GBM, Ependymoma
Child/AdultUSA
NCT0301828822Pembrolizumab (anti PD-1), HSPPC-96 Vaccine108GBMAdultUSA
NCT009050602HSPPC-96 Vaccine46GBMAdultUSA
NCT002934231/2HSPPC-96 Vaccine41rGBMAdultUSA
NCT018148132HSPPC-96 Vaccine with Bevacizumab90rGBMAdultUSA

Share and Cite

MDPI and ACS Style

Litak, J.; Grochowski, C.; Litak, J.; Osuchowska, I.; Gosik, K.; Radzikowska, E.; Kamieniak, P.; Rolinski, J. TLR-4 Signaling vs. Immune Checkpoints, miRNAs Molecules, Cancer Stem Cells, and Wingless-Signaling Interplay in Glioblastoma Multiforme—Future Perspectives. Int. J. Mol. Sci. 2020, 21, 3114. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21093114

AMA Style

Litak J, Grochowski C, Litak J, Osuchowska I, Gosik K, Radzikowska E, Kamieniak P, Rolinski J. TLR-4 Signaling vs. Immune Checkpoints, miRNAs Molecules, Cancer Stem Cells, and Wingless-Signaling Interplay in Glioblastoma Multiforme—Future Perspectives. International Journal of Molecular Sciences. 2020; 21(9):3114. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21093114

Chicago/Turabian Style

Litak, Jakub, Cezary Grochowski, Joanna Litak, Ida Osuchowska, Krzysztof Gosik, Elżbieta Radzikowska, Piotr Kamieniak, and Jacek Rolinski. 2020. "TLR-4 Signaling vs. Immune Checkpoints, miRNAs Molecules, Cancer Stem Cells, and Wingless-Signaling Interplay in Glioblastoma Multiforme—Future Perspectives" International Journal of Molecular Sciences 21, no. 9: 3114. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21093114

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop