Role of Inflammatory Mediators, Macrophages, and Neutrophils in Glioma Maintenance and Progression: Mechanistic Understanding and Potential Therapeutic Applications
Abstract
:Simple Summary
Abstract
1. Introduction
2. Glioma Microenvironment
3. Molecular Mechanism of Neuroinflammation
4. Tumorigenic Role of Inflammatory Molecules in Glioma
4.1. Role of Tumor-Associated Macrophages (TAMs) in Glioma Growth and Progression
4.2. Relationship between Inflammatory Mediators and TAMs
4.3. Role of Tumor-Associated Neutrophils in Glioma Growth and Progression
5. Signal Transduction Pathways Involved in Glioma Progression
5.1. NF-κB Signaling Pathway
5.2. JAK/STAT Signaling Pathway
5.3. MAP Kinase Signaling Pathway
5.4. PI3K/Akt/mTOR Signaling Pathway
5.5. TLRs Signaling Pathway
5.5.1. TLR Expression in the Brain
5.5.2. Role of TLR Axis in Glioma Development and Associated Neuroinflammation
5.5.3. Canonical (MyD88)-Dependent TLR Signaling Pathway
5.5.4. Non-Canonical (TRIF)-Dependent TLR Signaling Pathway
5.5.5. TLR Expression and Glioma Progression
6. Therapeutic Applications
6.1. Therapeutic Potential of Targeting Glioma-Derived Inflammatory Mediators
6.2. Therapeutic Potential of Targeting TAMs and TANs in Glioma
6.3. Therapeutic Potential of Targeting Signaling Pathways in Glioma
6.3.1. NF-κB Pathway
6.3.2. JAK/STAT Pathway
6.3.3. MAP Kinase Pathway
6.3.4. PI3K/Akt/mTOR Pathway
6.3.5. TLR Pathway
6.4. Therapeutic Potential of Non-Steroidal Anti-Inflammatory Drugs against Glioma Progression
7. Conclusions and Future Possibilities
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Perry, A.; Wesseling, P. Histologic classification of gliomas. Handb. Clin. Neurol. 2016, 134, 71–95. [Google Scholar] [CrossRef]
- Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 coun-tries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Molinaro, A.M.; Taylor, J.W.; Wiencke, J.K.; Wrensch, M.R. Genetic and molecular epidemiology of adult diffuse glioma. Nat. Rev. Neurol. 2019, 15, 405–417. [Google Scholar] [CrossRef] [PubMed]
- Rinaldi, M.; Caffo, M.; Minutoli, L.; Marini, H.; Abbritti, R.V.; Squadrito, F.; Trichilo, V.; Valenti, A.; Barresi, V.; Altavilla, D.; et al. ROS and Brain Gliomas: An Overview of Potential and Innovative Therapeutic Strategies. Int. J. Mol. Sci. 2016, 17, 984. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- McNeill, K.A. Epidemiology of Brain Tumors. Neurol. Clin. 2016, 34, 981–998. [Google Scholar] [CrossRef]
- Korniluk, A.; Koper, O.; Kemona, H.; Dymicka-Piekarska, V. From inflammation to cancer. Ir. J. Med. Sci. 2017, 186, 57–62. [Google Scholar] [CrossRef] [Green Version]
- Greten, F.R.; Grivennikov, S.I. Inflammation and Cancer: Triggers, Mechanisms, and Consequences. Immunity 2019, 51, 27–41. [Google Scholar] [CrossRef]
- Yeung, Y.T.; McDonald, K.L.; Grewal, T.; Munoz, L. Interleukins in glioblastoma pathophysiology: Implications for therapy. Br. J. Pharmacol. 2013, 168, 591–606. [Google Scholar] [CrossRef] [Green Version]
- Solinas, G.; Marchesi, F.; Garlanda, C.; Mantovani, A.; Allavena, P. Inflammation-mediated promotion of invasion and metastasis. Cancer Metastasis Rev. 2010, 29, 243–248. [Google Scholar] [CrossRef]
- Iwami, K.; Natsume, A.; Wakabayashi, T. Cytokine networks in glioma. Neurosurg. Rev. 2011, 34, 253–264. [Google Scholar] [CrossRef]
- Hanahan, D.; Coussens, L.M. Accessories to the Crime: Functions of Cells Recruited to the Tumor Microenvironment. Cancer Cell 2012, 21, 309–322. [Google Scholar] [CrossRef] [Green Version]
- Gajewski, T.F.; Schreiber, H.; Fu, Y.-X. Innate and adaptive immune cells in the tumor microenvironment. Nat. Immunol. 2013, 14, 1014–1022. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Marvel, D.; Gabrilovich, D.I. Myeloid-derived suppressor cells in the tumor microenvironment: Expect the unexpected. J. Clin. Investig. 2015, 125, 3356–3364. [Google Scholar] [CrossRef]
- Arneth, B. Tumor Microenvironment. Medicina 2019, 56, 15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Olson, O.; Joyce, J.A. Cysteine cathepsin proteases: Regulators of cancer progression and therapeutic response. Nat. Rev. Cancer 2015, 15, 712–729. [Google Scholar] [CrossRef]
- Oelschlaegel, D.; Sadan, T.W.; Salpeter, S.; Krug, S.; Blum, G.; Schmitz, W.; Schulze, A.; Michl, P. Cathepsin Inhibition Modulates Metabolism and Polarization of Tumor-Associated Macrophages. Cancers 2020, 12, 2579. [Google Scholar] [CrossRef] [PubMed]
- Gonzalez, H.; Hagerling, C.; Werb, Z. Roles of the immune system in cancer: From tumor initiation to metastatic progression. Genes Dev. 2018, 32, 1267–1284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Michelson, N.; Rincon-Torroella, J.; Quiñones-Hinojosa, A.; Greenfield, J.P. Exploring the role of inflammation in the malignant transformation of low-grade gliomas. J. Neuroimmunol. 2016, 297, 132–140. [Google Scholar] [CrossRef] [PubMed]
- Shabab, T.; Khanabdali, R.; Moghadamtousi, S.Z.; Kadir, H.A.; Mohan, G. Neuroinflammation pathways: A general review. Int. J. Neurosci. 2016, 127, 624–633. [Google Scholar] [CrossRef]
- Galdiero, M.R.; Marone, G.; Mantovani, A. Cancer inflammation and cytokines. Cold Spring Harb. Perspect. Biol. 2018, 10, a028662. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shastri, A.; Bonifati, D.M.; Kishore, U. Innate Immunity and Neuroinflammation. Mediat. Inflamm. 2013, 2013, 1–19. [Google Scholar] [CrossRef]
- Coussens, L.M.; Zitvogel, L.; Palucka, A.K. Neutralizing tumor-promoting chronic inflammation: A magic bullet? Science 2013, 339, 286–291. [Google Scholar] [CrossRef] [Green Version]
- Woo, S.-R.; Corrales, L.; Gajewski, T.F. Innate Immune Recognition of Cancer. Annu. Rev. Immunol. 2015, 33, 445–474. [Google Scholar] [CrossRef] [PubMed]
- Obermeier, B.; Daneman, R.; Ransohoff, R.M. Development, maintenance and disruption of the blood-brain barrier. Nat. Med. 2013, 19, 1584–1596. [Google Scholar] [CrossRef]
- Wang, Y.; Xiang, Y.; Xin, V.W.; Wang, X.-W.; Peng, X.-C.; Liu, X.-Q.; Wang, D.; Li, N.; Cheng, J.-T.; Lyv, Y.-N.; et al. Dendritic cell biology and its role in tumor immunotherapy. J. Hematol. Oncol. 2020, 13, 1–18. [Google Scholar] [CrossRef] [PubMed]
- Lan, T.; Chen, L.; Wei, X. Inflammatory Cytokines in Cancer: Comprehensive Understanding and Clinical Progress in Gene Therapy. Cells 2021, 10, 100. [Google Scholar] [CrossRef]
- Ha, E.T.; Antonios, J.P.; Soto, H.; Prins, R.M.; Yang, I.; Kasahara, N.; Liau, L.M.; Kruse, C.A. Chronic inflammation drives glioma growth: Cellular and molecular factors responsible for an immunosuppressive mi-croenvironment. Neuroimmunol. Neuroinflammation 2014, 1, 66–76. [Google Scholar]
- Bonnin, D.A.A.; Havrda, M.C.; Israel, M.A. Glioma Cell Secretion: A Driver of Tumor Progression and a Potential Therapeutic Target. Cancer Res. 2018, 78, 6031–6039. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hasan, T.; Caragher, S.P.; Shireman, J.M.; Park, C.H.; Atashi, F.; Baisiwala, S.; Lee, G.; Guo, D.; Wang, J.Y.; Dey, M.; et al. Interleukin-8/CXCR2 signaling regulates therapy-induced plasticity and enhances tumorigenicity in glioblastoma. Cell. Death Dis. 2019, 10, 1–17. [Google Scholar] [CrossRef] [Green Version]
- Lu, T.; Tian, L.; Han, L.; Vogelbaum, M.; Stark, G.R. Dose-dependent cross-talk between the transforming growth factor-β and interleukin-1 signaling pathways. Proc. Natl. Acad. Sci. USA 2007, 104, 4365–4370. [Google Scholar] [CrossRef] [Green Version]
- Taniura, S.; Kamitani, H.; Watanabe, T.; Eling, T.E. Induction of cyclooxygenase-2 expression by interleukin-1β in human glioma cell line, U87MG. Neurol. Med. Chir. 2008, 48, 500–505. [Google Scholar] [CrossRef] [Green Version]
- Qiu, J.; Li, Q.; Bell, K.A.; Yao, X.; Du, Y.; Zhang, E.; Yu, J.J.; Yu, Y.; Shi, Z.; Jiang, J. Small-molecule inhibition of prostaglandin E receptor 2 impairs cyclooxygenase-associated malignant glioma growth. Br. J. Pharmacol. 2019, 176, 1680–1699. [Google Scholar] [CrossRef] [PubMed]
- Palumbo, P.; Lombardi, F.; Augello, F.R.; Giusti, I.; Dolo, V.; Leocata, P.; Cifone, M.G.; Cinque, B. Biological effects of selective COX-2 inhibitor NS398 on human glioblastoma cell lines. Cancer Cell Int. 2020, 20, 1–17. [Google Scholar] [CrossRef] [PubMed]
- Griffin, B.D.; Moynagh, P. Persistent Interleukin-1β Signaling Causes Long Term Activation of NFκB in a Promoter-specific Manner in Human Glial Cells. J. Biol. Chem. 2006, 281, 10316–10326. [Google Scholar] [CrossRef] [Green Version]
- Liu, Q.; Li, G.; Li, R.; Shen, J.; He, Q.; Deng, L.; Zhang, C.; Zhang, J. IL-6 promotion of glioblastoma cell invasion and angiogenesis in U251 and T98G cell lines. J. Neuro-Oncol. 2010, 100, 165–176. [Google Scholar] [CrossRef] [PubMed]
- Munoz, L.; Yeung, Y.T.; Grewal, T. Oncogenic Ras modulates p38 MAPK-mediated inflammatory cytokine production in glio-blastoma cells. Cancer Biol. Ther. 2016, 17, 355–363. [Google Scholar] [CrossRef] [Green Version]
- Kosmopoulos, M.; Christofides, A.; Drekolias, D.; Zavras, P.; Gargalionis, A.N.; Piperi, C. Critical Role of IL-8 Targeting in Gliomas. Curr. Med. Chem. 2018, 25, 1954–1967. [Google Scholar] [CrossRef] [PubMed]
- Ping, Y.-F.; Yao, X.-H.; Jiang, J.-Y.; Zhao, L.-T.; Yu, S.-C.; Jiang, T.; Lin, M.C.M.; Chen, J.-H.; Wang, B.; Zhang, R.; et al. The chemokine CXCL12 and its receptor CXCR4 promote glioma stem cell-mediated VEGF production and tumour angiogenesis via PI3K/AKT signalling. J. Pathol. 2011, 224, 344–354. [Google Scholar] [CrossRef]
- Lokker, N.A.; Sullivan, C.M.; Hollenbach, S.J.; Israel, M.A.; Giese, N.A. Platelet-derived growth factor (PDGF) autocrine signaling regulates survival and mitogenic pathways in glio-blastoma cells: Evidence that the novel PDGF-C and PDGF-D ligands may play a role in the development of brain tumors. Cancer Res. 2002, 62, 3729–3735. [Google Scholar] [PubMed]
- Shih, A.H.; Holland, E.C. Platelet-derived growth factor (PDGF) and glial tumorigenesis. Cancer Lett. 2006, 232, 139–147. [Google Scholar] [CrossRef] [PubMed]
- Paulsson, J.; Lindh, M.B.; Jarvius, M.; Puputti, M.; Nister, M.; Nupponen, N.N.; Paulus, W.; Soderberg, N.; Dresemann, G.; von Deimling, A.; et al. Prognostic but not predictive role of platelet-derived growth factor receptors in patients with recurrent glioblasto-ma. Int. J. Cancer 2011, 128, 1981–1988. [Google Scholar] [CrossRef] [Green Version]
- Smith, D.; Shimamura, T.; Barbera, S.; Bejcek, B.E. NF-κB controls growth of glioblastomas/astrocytomas. Mol. Cell. Biochem. 2008, 307, 141–147. [Google Scholar] [CrossRef] [PubMed]
- Shapouri-Moghaddam, A.; Mohammadian, S.; Vazini, H.; Taghadosi, M.; Esmaeili, S.-A.; Mardani, F.; Seifi, B.; Mohammadi, A.; Afshari, J.T.; Sahebkar, A. Macrophage plasticity, polarization, and function in health and disease. J. Cell. Physiol. 2018, 233, 6425–6440. [Google Scholar] [CrossRef]
- Watters, J.J.; Schartner, J.M.; Badie, B. Microglia function in brain tumors. J. Neurosci. Res. 2005, 81, 447–455. [Google Scholar] [CrossRef] [PubMed]
- Schiffer, D.; Mellai, M.; Bovio, E.; Annovazzi, L. The neuropathological basis to the functional role of microglia/macrophages in gliomas. Neurol. Sci. 2017, 38, 1571–1577. [Google Scholar] [CrossRef] [PubMed]
- Lisi, L.; Stigliano, E.; Lauriola, L.; Navarra, P.; Russo, C.D. Proinflammatory-activated glioma cells induce a switch in microglial polarization and activation status, from a predominant M2b phenotype to a mixture of M1 and M2a/B polarized cells. ASN Neuro 2014, 6, 171–183. [Google Scholar] [CrossRef] [Green Version]
- Mantovani, A.; Sica, A.; Sozzani, S.; Allavena, P.; Vecchi, A.; Locati, M. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 2004, 25, 677–686. [Google Scholar] [CrossRef]
- Jackaman, C.; Tomay, F.; Duong, L.; Razak, N.B.A.; Pixley, F.; Metharom, P.; Nelson, D. Aging and cancer: The role of macrophages and neutrophils. Ageing Res. Rev. 2017, 36, 105–116. [Google Scholar] [CrossRef]
- Szebeni, G.J.; Vizler, C.; Kitajka, K.; Puskas, L.G. Inflammation and cancer: Extra-and intracellular determinants of tumor-associated macrophages as tumor pro-moters. Mediat. Inflamm. 2017, 2017, 9294018. [Google Scholar] [CrossRef]
- Lee, H.L.; Jang, J.W.; Lee, S.W.; Yoo, S.W.; Kwon, J.H.; Nam, S.W.; Bae, S.H.; Choi, J.Y.; Han, N.I.; Yoon, S.K. Inflammatory cytokines and change of Th1/Th2 balance as prognostic indicators for hepatocellular carcinoma in pa-tients treated with transarterial chemoembolization. Sci. Rep. 2019, 9, 3260. [Google Scholar] [CrossRef] [Green Version]
- Duluc, D.; Delneste, Y.; Tan, F.; Moles, M.-P.; Grimaud, L.; Lenoir, J.; Preisser, L.; Anegon, I.; Catala, L.; Ifrah, N.; et al. Tumor-associated leukemia inhibitory factor and IL-6 skew monocyte differentiation into tumor-associated macro-phage-like cells. Blood 2007, 110, 4319–4330. [Google Scholar] [CrossRef]
- Hambardzumyan, D.; Gutmann, D.; Kettenmann, H. The role of microglia and macrophages in glioma maintenance and progression. Nat. Neurosci. 2015, 19, 20–27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gadani, S.P.; Cronk, J.C.; Norris, G.T.; Kipnis, J. IL-4 in the Brain: A Cytokine to Remember. J. Immunol. 2012, 189, 4213–4219. [Google Scholar] [CrossRef] [PubMed]
- Rolle, C.E.; Sengupta, S.; Lesniak, M.S. Mechanisms of Immune Evasion by Gliomas. Adv. Exp. Med. Biol. 2012, 746, 53–76. [Google Scholar] [CrossRef] [PubMed]
- Hori, T.; Sasayama, T.; Tanaka, K.; Koma, Y.-I.; Nishihara, M.; Tanaka, H.; Nakamizo, S.; Nagashima, H.; Maeyama, M.; Fujita, Y.; et al. Tumor-associated macrophage related interleukin-6 in cerebrospinal fluid as a prognostic marker for glioblastoma. J. Clin. Neurosci. 2019, 68, 281–289. [Google Scholar] [CrossRef]
- Deshmane, S.L.; Kremlev, S.; Amini, S.; Sawaya, B.E. Monocyte Chemoattractant Protein-1 (MCP-1): An Overview. J. Interf. Cytokine Res. 2009, 29, 313–326. [Google Scholar] [CrossRef]
- Platten, M.; Kretz, A.; Naumann, U.; Aulwurm, S.; Egashira, K.; Isenmann, S.; Weller, M. Monocyte chemoattractant protein-1 increases microglial infiltration and aggressiveness of gliomas. Ann. Neurol. 2003, 54, 388–392. [Google Scholar] [CrossRef]
- Saio, M.; Okada, M.; Kito, Y.; Ohe, N.; Yano, H.; Yoshimura, S.; Iwama, T.; Takami, T. Tumor-associated macrophage/microglia infiltration in human gliomas is correlated with MCP-3, but not MCP-1. Int. J. Oncol. 2009, 34, 1621–1627. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lin, L.; Doherty, D.; Lile, J.; Bektesh, S.; Collins, F. GDNF: A glial cell line-derived neurotrophic factor for midbrain dopaminergic neurons. Science 1993, 260, 1130–1132. [Google Scholar] [CrossRef] [PubMed]
- Wiesenhofer, B.; Stockhammer, G.; Kostron, H.; Maier, H.; Hinterhuber, H.; Humpel, C. Glial cell line-derived neurotrophic factor (GDNF) and its receptor (GFR-alpha 1) are strongly expressed in human gliomas. Acta Neuropathol. 2000, 99, 131–137. [Google Scholar] [CrossRef] [PubMed]
- Ku, M.-C.; Wolf, S.A.; Respondek, D.; Matyash, V.; Pohlmann, A.; Waiczies, S.; Waiczies, H.; Niendorf, T.; Synowitz, M.; Glass, R.; et al. GDNF mediates glioblastoma-induced microglia attraction but not astrogliosis. Acta Neuropathol. 2013, 125, 609–620. [Google Scholar] [CrossRef]
- Sielska, M.; Przanowski, P.; Pasierbińska, M.; Wojnicki, K.; Poleszak, K.; Wojtas, B.; Grzeganek, D.; Ellert-Miklaszewska, A.; Ku, M.-C.; Kettenmann, H.; et al. Tumour-derived CSF2/granulocyte macrophage colony stimulating factor controls myeloid cell accumulation and progression of gliomas. Br. J. Cancer 2020, 123, 438–448. [Google Scholar] [CrossRef] [PubMed]
- Hamilton, J.A. Colony-stimulating factors in inflammation and autoimmunity. Nat. Rev. Immunol. 2008, 8, 533–544. [Google Scholar] [CrossRef]
- Nijaguna, M.B.; Patil, V.; Urbach, S.; Shwetha, S.D.; Sravani, K.; Hegde, A.S.; Chandramouli, B.A.; Arivazhagan, A.; Marin, P.; Santosh, V.; et al. Glioblastoma-derived Macrophage Colony-stimulating Factor (MCSF) Induces Microglial Release of Insulin-like Growth Factor-binding Protein 1 (IGFBP1) to Promote Angiogenesis. J. Biol. Chem. 2015, 290, 23401–23415. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Revoltella, R.P.; Menicagli, M.; Campani, D. Granulocyte–macrophage colony-stimulating factor as an autocrine survival-growth factor in human gliomas. Cytokine 2012, 57, 347–359. [Google Scholar] [CrossRef]
- Bettinger, I.; Thanos, S.; Paulus, W. Microglia promote glioma migration. Acta Neuropathol. 2002, 103, 351–355. [Google Scholar] [CrossRef]
- Hwang, J.-S.; Jung, E.-H.; Kwon, M.-Y.; Han, I.-O. Glioma-secreted soluble factors stimulate microglial activation: The role of interleukin-1β and tumor necrosis factor-α. J. Neuroimmunol. 2016, 298, 165–171. [Google Scholar] [CrossRef] [PubMed]
- da Fonseca, A.C.C.; Wang, H.; Fan, H.; Chen, X.; Zhang, I.; Zhang, L.; Lima, F.R.S.; Badie, B. Increased expression of stress inducible protein 1 in glioma-associated microglia/macrophages. J. Neuroimmunol. 2014, 274, 71–77. [Google Scholar] [CrossRef] [Green Version]
- Zheng, Y.; Yang, W.; Aldape, K.; He, J.; Lu, Z. Epidermal growth factor (EGF)-enhanced vascular cell adhesion molecule-1 (VCAM-1) expression promotes macro-phage and glioblastoma cell interaction and tumor cell invasion. J. Biol. Chem. 2013, 288, 31488–31495. [Google Scholar] [CrossRef] [Green Version]
- Takenaka, M.C.; Gabriely, G.; Rothhammer, V.; Mascanfroni, I.D.; Wheeler, M.A.; Chao, C.-C.; Gutiérrez-Vázquez, C.; Kenison, J.; Tjon, E.C.; Barroso, A.; et al. Control of tumor-associated macrophages and T cells in glioblastoma via AHR and CD39. Nat. Neurosci. 2019, 22, 729–740. [Google Scholar] [CrossRef]
- Joseph, J.V.; Balasubramaniyan, V.; Walenkamp, A.; Kruyt, F.A. TGF-β as a therapeutic target in high grade gliomas–Promises and challenges. Biochem. Pharmacol. 2013, 85, 478–485. [Google Scholar] [CrossRef]
- Wesolowska, A.; Kwiatkowska, A.; Slomnicki, L.; Dembinski, M.; Master, A.; Sliwa, M.; Franciszkiewicz, K.; Chouaib, S.; Kaminska, B. Microglia-derived TGF-beta as an important regulator of glioblastoma invasion--an inhibition of TGF-beta-dependent effects by shRNA against human TGF-beta type II receptor. Oncogene 2008, 27, 918–930. [Google Scholar] [CrossRef] [Green Version]
- Wang, L.; Wang, L.; Liu, Z.; Balivada, S.; Shrestha, T.; Bossmann, S.; Pyle, M.; Pappan, L.; Shi, J.; Troyer, D. Interleukin-1β and transforming growth factor-β cooperate to induce neurosphere formation and increase tumorigen-icity of adherent LN-229 glioma cells. Stem Cell Res. Ther. 2012, 3, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, X.-J.; Chen, G.-L.; Yu, S.-C.; Xu, C.; Xin, Y.-H.; Li, T.-T.; Shi, Y.; Gu, A.; Duan, J.-J.; Qian, C.; et al. TGF-β1 enhances tumor-induced angiogenesis via JNK pathway and macrophage infiltration in an improved zebrafish embryo/xenograft glioma model. Int. Immunopharmacol. 2012, 15, 191–198. [Google Scholar] [CrossRef]
- Zaynagetdinov, R.; Sherrill, T.P.; Polosukhin, V.V.; Han, W.; Ausborn, J.A.; McLoed, A.G.; McMahon, F.B.; Gleaves, L.A.; Degryse, A.L.; Stathopoulos, G.T.; et al. A Critical Role for Macrophages in Promotion of Urethane-Induced Lung Carcinogenesis. J. Immunol. 2011, 187, 5703–5711. [Google Scholar] [CrossRef]
- Grabowski, M.M.; Sankey, E.W.; Ryan, K.J.; Chongsathidkiet, P.; Lorrey, S.J.; Wilkinson, D.; Fecci, P.E. Immune suppression in gliomas. J. Neuro-Oncol. 2020, 151, 3–12. [Google Scholar] [CrossRef] [PubMed]
- Prosniak, M.; Harshyne, L.A.; Andrews, D.W.; Kenyon, L.; Bedelbaeva, K.; Apanasovich, T.V.; Heber-Katz, E.; Curtis, M.T.; Cotzia, P.; Hooper, D. Glioma Grade Is Associated with the Accumulation and Activity of Cells Bearing M2 Monocyte Markers. Clin. Cancer Res. 2013, 19, 3776–3786. [Google Scholar] [CrossRef] [Green Version]
- Lu, J.; Xu, Z.; Duan, H.; Ji, H.; Zhen, Z.; Li, B.; Wang, H.; Tang, H.; Zhou, J.; Guo, T.; et al. Tumor-associated macrophage interleukin-β promotes glycerol-3-phosphate dehydrogenase activation, glycolysis and tumorigenesis in glioma cells. Cancer Sci. 2020, 111, 1979–1990. [Google Scholar] [CrossRef] [PubMed]
- Grégoire, H.; Roncali, L.; Rousseau, A.; Chérel, M.; Delneste, Y.; Jeannin, P.; Hindré, F.; Garcion, E. Targeting Tumor Associated Macrophages to Overcome Conventional Treatment Resistance in Glioblastoma. Front. Pharmacol. 2020, 11, 368. [Google Scholar] [CrossRef] [Green Version]
- Kohanbash, G.; McKaveney, K.; Sakaki, M.; Ueda, R.; Mintz, A.H.; Amankulor, N.; Fujita, M.; Ohlfest, J.R.; Okada, H. GM-CSF Promotes the Immunosuppressive Activity of Glioma-Infiltrating Myeloid Cells through Interleukin-4 Receptor-α. Cancer Res. 2013, 73, 6413–6423. [Google Scholar] [CrossRef] [Green Version]
- Soehnlein, O.; Lindbom, L. Phagocyte partnership during the onset and resolution of inflammation. Nat. Rev. Immunol. 2010, 10, 427–439. [Google Scholar] [CrossRef]
- Khan, S.; Mittal, S.; McGee, K.; Alfaro, K.; Majd, N.; Balasubramaniyan, V.; De Groot, J.F. Role of Neutrophils and Myeloid-Derived Suppressor Cells in Glioma Progression and Treatment Resistance. Int. J. Mol. Sci. 2020, 21, 1954. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Uribe-Querol, E.; Rosales, C. Neutrophils in Cancer: Two Sides of the Same Coin. J. Immunol. Res. 2015, 2015, 983698. [Google Scholar] [CrossRef] [Green Version]
- Hurt, B.; Schulick, R.; Edil, B.; El Kasmi, K.C.; Barnett, C. Cancer-promoting mechanisms of tumor-associated neutrophils. Am. J. Surg. 2017, 214, 938–944. [Google Scholar] [CrossRef]
- Kusumanto, Y.H.; Dam, W.A.; Hospers, G.A.; Meijer, C.; Mulder, N.H. Platelets and granulocytes, in particular the neutrophils, form important compartments for circulating vas-cular endothelial growth factor. Angiogenesis 2003, 6, 283–287. [Google Scholar] [CrossRef]
- Mishalian, I.; Bayuh, R.; Levy, L.; Zolotarov, L.; Michaeli, J.; Fridlender, Z.G. Tumor-associated neutrophils (TAN) develop pro-tumorigenic properties during tumor progression. Cancer Immunol. Immunother. 2013, 62, 1745–1756. [Google Scholar] [CrossRef]
- Fossati, G.; Ricevuti, G.; Edwards, S.W.; Walker, C.; Dalton, A.; Rossi, M.L. Neutrophil infiltration into human gliomas. Acta Neuropathol. 1999, 98, 349–354. [Google Scholar] [CrossRef] [PubMed]
- Wu, L.; Awaji, M.; Saxena, S.; Varney, M.L.; Sharma, B. IL-17-CXC Chemokine Receptor 2 Axis Facilitates Breast Cancer Progression by Up-Regulating Neutrophil Recruitment. Am. J. Pathol. 2020, 190, 222–233. [Google Scholar] [CrossRef] [PubMed]
- Rahbar, A.; Cederarv, M.; Wolmer-Solberg, N.; Tammik, C.; Stragliotto, G.; Peredo, I.; Fornara, O.; Xu, X.; Dzabic, M.; Taher, C.; et al. Enhanced neutrophil activity is associated with shorter time to tumor progression in glioblastoma patients. OncoImmunology 2015, 5, e1075693. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sippel, T.R.; White, J.; Nag, K.; Tsvankin, V.; Klaassen, M.; Kleinschmidt-DeMasters, B.; Waziri, A. Neutrophil Degranulation and Immunosuppression in Patients with GBM: Restoration of Cellular Immune Function by Targeting Arginase I. Clin. Cancer Res. 2011, 17, 6992–7002. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Christofides, A.; Kosmopoulos, M.; Piperi, C. Pathophysiological mechanisms regulated by cytokines in gliomas. Cytokine 2015, 71, 377–384. [Google Scholar] [CrossRef] [PubMed]
- Sen, E. Targeting inflammation-induced transcription factor activation: An open frontier for glioma therapy. Drug Discov. Today 2011, 16, 1044–1051. [Google Scholar] [CrossRef]
- 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] [Green Version]
- Cianciulli, A.; Porro, C.; Calvello, R.; Trotta, T.; Lofrumento, D.D.; Panaro, M.A. Microglia Mediated Neuroinflammation: Focus on PI3K Modulation. Biomolecules 2020, 10, 137. [Google Scholar] [CrossRef] [Green Version]
- Wong, S.; Kamarudin, M.; Naidu, R. Anticancer Mechanism of Curcumin on Human Glioblastoma. Nutrients 2021, 13, 950. [Google Scholar] [CrossRef]
- Sen, R.; Baltimore, D. Multiple nuclear factors interact with the immunoglobulin enhancer sequences. Cell 1986, 46, 705–716. [Google Scholar] [CrossRef]
- Hayden, M.S.; Ghosh, S. Shared principles in NF-kappaB signaling. Cell 2008, 132, 344–362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Taniguchi, K.; Karin, M. NF-κB, inflammation, immunity and cancer: Coming of age. Nat. Rev. Immunol. 2018, 18, 309–324. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Q.; Lenardo, M.J.; Baltimore, D. 30 Years of NF-κB: A Blossoming of Relevance to Human Pathobiology. Cell 2017, 168, 37–57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Olivera, A.; Moore, T.W.; Hu, F.; Brown, A.P.; Sun, A.; Liotta, D.C.; Snyder, J.P.; Yoon, Y.; Shim, H.; Marcus, A.I.; et al. Inhibition of the NF-κB signaling pathway by the curcumin analog, 3,5-Bis(2-pyridinylmethylidene)-4-piperidone (EF31): Anti-inflammatory and anti-cancer properties. Int. Immunopharmacol. 2012, 12, 368–377. [Google Scholar] [CrossRef] [Green Version]
- Cooks, T.; Pateras, I.S.; Tarcic, O.; Solomon, H.; Schetter, A.J.; Wilder, S.; Lozano, G.; Pikarsky, E.; Forshew, T.; Rosenfeld, N.; et al. Mutant p53 prolongs NF-κB activation and promotes chronic inflammation and inflammation-associated colorectal cancer. Cancer Cell 2013, 23, 634–646. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Atkinson, G.P.; Nozell, S.E.; Benveniste, E.T. NF-kappaB and STAT3 signaling in glioma: Targets for future therapies. Expert Rev. Neurother. 2010, 10, 575–586. [Google Scholar] [CrossRef] [Green Version]
- Song, L.; Liu, L.; Wu, Z.; Li, Y.; Ying, Z.; Lin, C.; Wu, J.; Hu, B.; Cheng, S.Y.; Li, M.; et al. TGF-β induces miR-182 to sustain NF-κB activation in glioma subsets. J. Clin. Investig. 2012, 122, 3563–3578. [Google Scholar] [CrossRef] [PubMed]
- Brat, D.J.; Bellail, A.C.; Van Meir, E.G. The role of interleukin-8 and its receptors in gliomagenesis and tumoral angiogenesis. Neuro-Oncology 2005, 7, 122–133. [Google Scholar] [CrossRef] [PubMed]
- Esencay, M.; Newcomb, E.W.; Zagzag, D. HGF upregulates CXCR4 expression in gliomas via NF-kappaB: Implications for glioma cell migration. J. Neurooncol. 2010, 99, 33–40. [Google Scholar] [CrossRef] [Green Version]
- McFarland, B.C.; Hong, S.W.; Rajbhandari, R.; Twitty, G.B., Jr.; Gray, G.K.; Yu, H.; Benveniste, E.N.; Nozell, S.E. NF-κB-induced IL-6 ensures STAT3 activation and tumor aggressiveness in glioblastoma. PLoS ONE 2013, 8, e78728. [Google Scholar] [CrossRef]
- Piperi, C.; Papavassiliou, K.A.; Papavassiliou, A.G. Pivotal Role of STAT3 in Shaping Glioblastoma Immune Microenvironment. Cells 2019, 8, 1398. [Google Scholar] [CrossRef] [Green Version]
- Bose, S.; Banerjee, S.; Mondal, A.; Chakraborty, U.; Pumarol, J.; Croley, C.; Bishayee, A. Targeting the JAK/STAT Signaling Pathway Using Phytocompounds for Cancer Prevention and Therapy. Cells 2020, 9, 1451. [Google Scholar] [CrossRef]
- Stark, G.R.; Darnell, J.E. The JAK-STAT Pathway at Twenty. Immunity 2012, 36, 503–514. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kruczyk, M.; Przanowski, P.; Dabrowski, M.; Swiatek-Machado, K.; Mieczkowski, J.; Wallerman, O.; Ronowicz, A.; Piotrowski, A.; Wadelius, C.; Kaminska, B.; et al. Integration of genome-wide of Stat3 binding and epigenetic modification mapping with transcriptome reveals novel Stat3 target genes in glioma cells. Biochim. Biophys. Acta (BBA)-Bioenerg. 2014, 1839, 1341–1350. [Google Scholar] [CrossRef]
- Banerjee, S.; Biehl, A.; Gadina, M.; Hasni, S.; Schwartz, D. JAK–STAT Signaling as a Target for Inflammatory and Autoimmune Diseases: Current and Future Prospects. Drugs 2017, 77, 521–546. [Google Scholar] [CrossRef]
- West, A.J.; Tsui, V.; Stylli, S.S.; Nguyen, H.; Morokoff, A.P.; Kaye, A.H.; Luwor, R.B. The role of interleukin-6-STAT3 signalling in glioblastoma. Oncol. Lett. 2018, 16, 4095–4104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, X.; Nie, X.; Mao, J.; Zhang, Y.; Yin, K.; Jiang, S. Perfluorooctanesulfonate induces neuroinflammation through the secretion of TNF-α mediated by the JAK2/STAT3 pathway. NeuroToxicology 2018, 66, 32–42. [Google Scholar] [CrossRef]
- Ding, Z.; Kloss, J.M.; Tuncali, S.; Tran, N.L.; Loftus, J.C. TROY signals through JAK1-STAT3 to promote glioblastoma cell migration and resistance. Neoplasia 2020, 22, 352–364. [Google Scholar] [CrossRef] [PubMed]
- Liu, X.; Yang, J.; Deng, W. The inflammatory cytokine IL-22 promotes murine gliomas via proliferation. Exp. Ther. Med. 2017, 13, 1087–1092. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rahaman, S.O.; Vogelbaum, M.A.; Haque, S.J. Aberrant Stat3 signaling by interleukin-4 in malignant glioma cells: Involve-ment of IL-13Ralpha2. Cancer Res. 2005, 65, 2956–2963. [Google Scholar] [CrossRef] [Green Version]
- Lo, H.W.; Cao, X.; Zhu, H.; Ali-Osman, F. Cyclooxygenase-2 is a novel transcriptional target of the nuclear EGFR-STAT3 and EGFRvIII-STAT3 signaling axes. Mol. Cancer Res. 2010, 8, 232–245. [Google Scholar] [CrossRef] [Green Version]
- Cho, H.J.; Park, J.H.; Nam, J.H.; Chang, Y.C.; Park, B.; Hoe, H.S. Ascochlorin Suppresses MMP-2-Mediated Migration and Invasion by Targeting FAK and JAK-STAT Signaling Cas-cades. J. Cell. Biochem. 2018, 119, 300–313. [Google Scholar] [CrossRef]
- Kim, E.K.; Choi, E.-J. Pathological roles of MAPK signaling pathways in human diseases. Biochim. Biophys. Acta (BBA)-Mol. Basis Dis. 2010, 1802, 396–405. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Robinson, M.J.; Cobb, M.H. Mitogen-activated protein kinase pathways. Curr. Opin. Cell. Biol. 1997, 9, 180–186. [Google Scholar] [CrossRef]
- Arnold, A.; Papanikolaou, A. Cyclin D1 in Breast Cancer Pathogenesis. J. Clin. Oncol. 2005, 23, 4215–4224. [Google Scholar] [CrossRef] [PubMed]
- Meng, Q.; Xia, Y. c-Jun, at the crossroad of the signaling network. Protein Cell 2011, 2, 889–898. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Miller, D.M.; Thomas, S.D.; Islam, A.; Muench, D.; Sedoris, K. c-Myc and Cancer Metabolism. Clin. Cancer Res. 2012, 18, 5546–5553. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Burotto, M.; Chiou, V.L.; Lee, J.-M.; Kohn, E.C. The MAPK pathway across different malignancies: A new perspective. Cancer 2014, 120, 3446–3456. [Google Scholar] [CrossRef] [Green Version]
- Kim, E.K.; Choi, E.-J. Compromised MAPK signaling in human diseases: An update. Arch. Toxicol. 2015, 89, 867–882. [Google Scholar] [CrossRef]
- Perini, G.F.; Ribeiro, G.N.; Neto, J.V.P.; Campos, L.T.; Hamerschlak, N. BCL-2 as therapeutic target for hematological malignancies. J. Hematol. Oncol. 2018, 11, 1–15. [Google Scholar] [CrossRef]
- Jo, S.H.; Kim, M.E.; Cho, J.H.; Lee, Y.; Lee, J.; Park, Y.-D.; Lee, J.S. Hesperetin inhibits neuroinflammation on microglia by suppressing inflammatory cytokines and MAPK pathways. Arch. Pharmacal Res. 2019, 42, 695–703. [Google Scholar] [CrossRef] [PubMed]
- Nickl-Jockschat, T.; Arslan, F.; Doerfelt, A.; Bogdahn, U.; Bosserhoff, A.; Hau, P. An imbalance between Smad and MAPK pathways is responsible for TGF-beta tumor promoting effects in high-grade gliomas. Int. J. Oncol. 2007, 30, 499–507. [Google Scholar] [PubMed]
- Yoshino, Y.; Aoyagi, M.; Tamaki, M.; Duan, L.; Morimoto, T.; Ohno, K. Activation of p38 MAPK and/or JNK contributes to increased levels of VEGF secretion in human malignant glioma cells. Int. J. Oncol. 2006, 29, 981–987. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Anand, M.; Van Meter, T.E.; Fillmore, H. Epidermal growth factor induces matrix metalloproteinase-1 (MMP-1) expression and invasion in glioma cell lines via the MAPK pathway. J. Neuro-Oncol. 2011, 104, 679–687. [Google Scholar] [CrossRef]
- Jin, T.; Li, N.; Yang, T.; Liu, F.; Kong, J.; Zhou, Y. PTPN1 promotes the progression of glioma by activating the MAPK/ERK and PI3K/AKT pathways and is associated with poor patient survival. Oncol. Rep. 2019, 42, 717–725. [Google Scholar] [CrossRef]
- Song, H.; Moon, A. Glial cell-derived neurotrophic factor (GDNF) promotes low-grade Hs683 glioma cell migration through JNK, ERK-1/2 and p38 MAPK signaling pathways. Neurosci. Res. 2006, 56, 29–38. [Google Scholar] [CrossRef]
- Karar, J.; Maity, A. PI3K/AKT/mTOR Pathway in Angiogenesis. Front. Mol. Neurosci. 2011, 4, 51. [Google Scholar] [CrossRef] [Green Version]
- Hawkins, P.T.; Stephens, L.R. Emerging evidence of signalling roles for PI(3,4)P2 in Class I and II PI3K-regulated pathways. Biochem. Soc. Trans. 2016, 44, 307–314. [Google Scholar] [CrossRef] [PubMed]
- Zhang, J.; Wang, L.; Wang, H.; Su, Z.; Pang, X. Neuroinflammation and central PI3K/Akt/mTOR signal pathway contribute to bone cancer pain. Mol. Pain 2019, 15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Porta, C.; Paglino, C.; Mosca, A. Targeting PI3K/Akt/mTOR Signaling in Cancer. Front. Oncol. 2014, 4, 64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Burris, H.A. Overcoming acquired resistance to anticancer therapy: Focus on the PI3K/AKT/mTOR pathway. Cancer Chemother. Pharmacol. 2013, 71, 829–842. [Google Scholar] [CrossRef] [PubMed]
- Escobedo, J.A.; Navankasattusas, S.; Kavanaugh, W.M.; Milfay, D.; Fried, V.A.; Williams, L.T. cDNA cloning of a novel 85 kd protein that has SH2 domains and regulates binding of PI3-kinase to the PDGF beta-receptor. Cell 1991, 65, 75–82. [Google Scholar] [CrossRef]
- Otsu, M.; Hiles, I.; Gout, I.; Fry, M.; Larrea, F.R.; Panayotou, G.; Thompson, A.; Dhand, R.; Hsuan, J.; Totty, N.; et al. Characterization of two 85 kd proteins that associate with receptor tyrosine kinases, middle-T/pp60c-src complexes, and PI3-kinase. Cell 1991, 65, 91–104. [Google Scholar] [CrossRef]
- Hiles, I.D.; Otsu, M.; Volinia, S.; Fry, M.; Gout, I.; Dhand, R.; Panayotou, G.; Larrea, F.R.; Thompson, A.; Totty, N.F.; et al. Phosphatidylinositol 3-kinase: Structure and expression of the 110 kd catalytic subunit. Cell 1992, 70, 419–429. [Google Scholar] [CrossRef]
- Stephens, L.R.; Jackson, T.R.; Hawkins, P.T. Agonist-stimulated synthesis of phosphatidylinositol(3,4,5)-trisphosphate: A new intracellular signalling system? Biochim. Biophys. Acta 1993, 1179, 27–75. [Google Scholar] [CrossRef]
- Li, X.; Wu, C.; Chen, N.; Gu, H.; Yen, A.; Cao, L.; Wang, E.; Wang, L. PI3K/Akt/mTOR signaling pathway and targeted therapy for glioblastoma. Oncotarget 2016, 7, 33440–33450. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Haddadi, N.; Lin, Y.; Travis, G.; Simpson, A.M.; Nassif, N.T.; McGowan, E.M. PTEN/PTENP1: ’Regulating the regulator of RTK-dependent PI3K/Akt signalling’, new targets for cancer thera-py. Mol. Cancer 2018, 17, 37. [Google Scholar] [CrossRef]
- Efeyan, A.; Sabatini, D.M. mTOR and cancer: Many loops in one pathway. Curr. Opin. Cell Biol. 2010, 22, 169–176. [Google Scholar] [CrossRef] [Green Version]
- Brown, E.J.; Albers, M.W.; Shin, T.B.; Ichikawa, K.; Keith, C.T.; Lane, W.S.; Schreiber, S.L. A mammalian protein targeted by G1-arresting rapamycin–receptor complex. Nat. Cell Biol. 1994, 369, 756–758. [Google Scholar] [CrossRef]
- Sabatini, D.M.; Erdjument-Bromage, H.; Lui, M.; Tempst, P.; Snyder, S.H. RAFT1: A mammalian protein that binds to FKBP12 in a rapamycin-dependent fashion and is homologous to yeast TORs. Cell 1994, 78, 35–43. [Google Scholar] [CrossRef]
- Sabers, C.J.; Martin, M.M.; Brunn, G.J.; Williams, J.M.; Dumont, F.J.; Wiederrecht, G.; Abraham, R.T. Isolation of a Protein Target of the FKBP12-Rapamycin Complex in Mammalian Cells. J. Biol. Chem. 1995, 270, 815–822. [Google Scholar] [CrossRef] [Green Version]
- Ma, X.M.; Blenis, J. Molecular mechanisms of mTOR-mediated translational control. Nat. Rev. Mol. Cell. Biol. 2009, 10, 307–318. [Google Scholar] [CrossRef]
- Moschetta, M.; Reale, A.; Marasco, C.; Vacca, A.; Carratù, M.R. Therapeutic targeting of the mTOR-signalling pathway in cancer: Benefits and limitations. Br. J. Pharmacol. 2014, 171, 3801–3813. [Google Scholar] [CrossRef] [Green Version]
- Cargnello, M.; Tcherkezian, J.; Roux, P.P. The expanding role of mTOR in cancer cell growth and proliferation. Mutagenesis 2015, 30, 169–176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, L.C.; Cook, R.S.; Chen, J. mTORC1 and mTORC2 in cancer and the tumor microenvironment. Oncogene 2016, 36, 2191–2201. [Google Scholar] [CrossRef] [Green Version]
- Tao, F.; Zhu, J.; Duan, L.; Wu, J.; Zhang, J.; Yao, K.; Bo, J.; Zu, H. Anti-inflammatory effects of doxepin hydrochloride against LPS-induced C6-glioma cell inflammatory reaction by PI3K-mediated Akt signaling. J. Biochem. Mol. Toxicol. 2020, 34, e22424. [Google Scholar] [CrossRef] [PubMed]
- Castellino, R.C.; Durden, D.L. Mechanisms of Disease: The PI3K–Akt–PTEN signaling node—an intercept point for the control of angiogenesis in brain tumors. Nat. Clin. Pract. Neurol. 2007, 3, 682–693. [Google Scholar] [CrossRef] [PubMed]
- Koul, D. PTEN Signaling pathways in glioblastoma. Cancer Biol. Ther. 2008, 7, 1321–1325. [Google Scholar] [CrossRef] [PubMed]
- Lisi, L.; Ciotti, G.M.P.; Chiavari, M.; Pizzoferrato, M.; Mangiola, A.; Kalinin, S.; Feinstein, D.L.; Navarra, P. Phospho-mTOR expression in human glioblastoma microglia-macrophage cells. Neurochem. Int. 2019, 129, 104485. [Google Scholar] [CrossRef]
- Ye, X.; Zhu, M.; Che, X.; Wang, H.; Liang, X.-J.; Wu, C.; Xue, X.; Yang, J. Lipopolysaccharide induces neuroinflammation in microglia by activating the MTOR pathway and downregulating Vps34 to inhibit autophagosome formation. J. Neuroinflammation 2020, 17, 1–17. [Google Scholar] [CrossRef] [Green Version]
- Shen, X.; Zhang, J.; Zhang, X.; Wang, Y.; Hu, Y.; Guo, J. Retinoic Acid-Induced Protein 14 (RAI14) Promotes mTOR-Mediated Inflammation Under Inflammatory Stress and Chemical Hypoxia in a U87 Glioblastoma Cell Line. Cell. Mol. Neurobiol. 2019, 39, 241–254. [Google Scholar] [CrossRef] [PubMed]
- Cui, L.; Ren, T.; Zhao, H.; Chen, S.; Zheng, M.; Gao, X.; Feng, D.; Yang, L.; Jin, X.; Zhuo, R. Suppression of PTTG1 inhibits cell angiogenesis, migration and invasion in glioma cells. Med. Oncol. 2020, 37, 1–11. [Google Scholar] [CrossRef]
- Huang, W.; Ding, X.; Ye, H.; Wang, J.; Shao, J.; Huang, T. Hypoxia enhances the migration and invasion of human glioblastoma U87 cells through PI3K/Akt/mTOR/HIF-1α pathway. NeuroReport 2018, 29, 1578–1585. [Google Scholar] [CrossRef]
- Akil, H.; Abbaci, A.; Lalloué, F.; Bessette, B.; Costes, L.M.M.; Domballe, L.; Charreau, S.; Guilloteau, K.; Karayan-Tapon, L.; Bernard, F.-X.; et al. IL22/IL-22R Pathway Induces Cell Survival in Human Glioblastoma Cells. PLoS ONE 2015, 10, e0119872. [Google Scholar] [CrossRef] [Green Version]
- Tu, M.; Wange, W.; Cai, L.; Zhu, P.; Gao, Z.; Zheng, W. IL-13 receptor α2 stimulates human glioma cell growth and metastasis through the Src/PI3K/Akt/mTOR signaling pathway. Tumor Biol. 2016, 37, 14701–14709. [Google Scholar] [CrossRef]
- Chen, H.-Y.; Lin, L.-T.; Wang, M.-L.; Lee, S.-H.; Tsai, M.-L.; Tsai, C.-C.; Liu, W.-H.; Chen, T.-C.; Yang, Y.-P.; Lee, Y.-Y.; et al. Musashi-1 regulates AKT-derived IL-6 autocrinal/paracrinal malignancy and chemoresistance in glioblastoma. Oncotarget 2016, 7, 42485–42501. [Google Scholar] [CrossRef] [Green Version]
- Bai, D.; Zhao, Y.; Zhu, Q.; Zhou, Y.; Zhao, Y.; Zhang, T.; Guo, Q.; Lu, N. LZ205, a newly synthesized flavonoid compound, exerts anti-inflammatory effect by inhibiting M1 macrophage polari-zation through regulating PI3K/AKT/mTOR signaling pathway. Exp. Cell. Res. 2018, 364, 84–94. [Google Scholar] [CrossRef] [PubMed]
- Alzahrani, A.S. PI3K/Akt/mTOR inhibitors in cancer: At the bench and bedside. Semin. Cancer Biol. 2019, 59, 125–132. [Google Scholar] [CrossRef] [PubMed]
- Janeway, C.A., Jr. Approaching the asymptote? Evolution and revolution in immunology. Cold. Spring Harb. Symp. Quant. Biol. 1989, 54, 1–13. [Google Scholar] [CrossRef]
- Kumar, V. Toll-like receptors in the pathogenesis of neuroinflammation. J. Neuroimmunol. 2019, 332, 16–30. [Google Scholar] [CrossRef] [PubMed]
- Stephenson, J.; Nutma, E.; Van Der Valk, P.; Amor, S. Inflammation in CNS neurodegenerative diseases. Immunology 2018, 154, 204–219. [Google Scholar] [CrossRef] [Green Version]
- Li, J.-K.; Balic, J.J.; Yu, L.; Jenkins, B. TLR Agonists as Adjuvants for Cancer Vaccines. Adv. Exp. Med. Biol. 2017, 1024, 195–212. [Google Scholar] [CrossRef]
- Smith, M.; García-Martínez, E.; Pitter, M.R.; Fucikova, J.; Spisek, R.; Zitvogel, L.; Kroemer, G.; Galluzzi, L. Trial Watch: Toll-like receptor agonists in cancer immunotherapy. OncoImmunology 2018, 7, e1526250. [Google Scholar] [CrossRef]
- Takeuchi, O.; Akira, S. Pattern Recognition Receptors and Inflammation. Cell 2010, 140, 805–820. [Google Scholar] [CrossRef] [Green Version]
- Kirkland, T.N.; Virca, G.D.; Kuus-Reichel, T.; Multer, F.K.; Kim, S.Y.; Ulevitch, R.J.; Tobias, P.S. Identification of lipopolysaccharide-binding proteins in 70Z/3 cells by photoaffinity cross-linking. J. Biol. Chem. 1990, 265. [Google Scholar] [CrossRef]
- 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] [PubMed]
- Wright, S.; Ramos, R.; Tobias, P.; Ulevitch, R.; Mathison, J. CD14, a receptor for complexes of lipopolysaccharide (LPS) and LPS binding protein. Science 1990, 249, 1431–1433. [Google Scholar] [CrossRef] [PubMed]
- Schumann, R.; Leong, S.R.; Flaggs, G.; Gray, P.; Wright, S.; Mathison, J.; Tobias, P.; Ulevitch, R. Structure and function of lipopolysaccharide binding protein. Science 1990, 249, 1429–1431. [Google Scholar] [CrossRef]
- Wesche, H.; Henzel, W.; Shillinglaw, W.; Li, S.; Cao, Z. MyD88: An Adapter That Recruits IRAK to the IL-1 Receptor Complex. Immunity 1997, 7, 837–847. [Google Scholar] [CrossRef] [Green Version]
- Muzio, M.; Ni, J.; Feng, P.; Dixit, V.M. IRAK (Pelle) Family Member IRAK-2 and MyD88 as Proximal Mediators of IL-1 Signaling. Science 1997, 278, 1612–1615. [Google Scholar] [CrossRef] [PubMed]
- Yamamoto, M.; Sato, S.; Mori, K.; Hoshino, K.; Takeuchi, O.; Takeda, K.; Akira, S. Cutting edge: A novel Toll/IL-1 receptor domain-containing adapter that preferentially activates the IFN-beta promoter in the Toll-like receptor signaling. J. Immunol. 2002, 169, 6668–6672. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tartey, S.; Takeuchi, O. Pathogen recognition and Toll-like receptor targeted therapeutics in innate immune cells. Int. Rev. Immunol. 2017, 36, 57–73. [Google Scholar] [CrossRef]
- Fitzgerald, K.; Palsson-McDermott, E.M.; Bowie, A.; Jefferies, C.A.; Mansell, A.; Brady, G.; Brint, E.; Dunne, A.; Gray, P.; Harte, M.T.; et al. Mal (MyD88-adapter-like) is required for Toll-like receptor-4 signal transduction. Nat. Cell Biol. 2001, 413, 78–83. [Google Scholar] [CrossRef]
- Horng, T.; Barton, G.; Medzhitov, R. TIRAP: An adapter molecule in the Toll signaling pathway. Nat. Immunol. 2001, 2, 835–841. [Google Scholar] [CrossRef]
- Meylan, E.; Burns, K.; Hofmann, K.; Blancheteau, V.; Martinon, F.; Kelliher, M.; Tschopp, J. RIP1 is an essential mediator of Toll-like receptor 3-induced NF-kappa B activation. Nat. Immunol. 2004, 5, 503–507. [Google Scholar] [CrossRef] [PubMed]
- Chakraborty, S.; Li, L.; Tang, H.; Xie, Y.; Puliyappadamba, V.T.; Raisanen, J.; Burma, S.; Boothman, A.D.; Cochran, B.; Wu, J.; et al. Cytoplasmic TRADD Confers a Worse Prognosis in Glioblastoma. Neoplasia 2013, 15, 888–897. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ambrosini, E.; Aloisi, F. Chemokines and Glial Cells: A Complex Network in the Central Nervous System. Neurochem. Res. 2004, 29, 1017–1038. [Google Scholar] [CrossRef]
- Siddiqui, A.; Akhtar, S.; Shah, Z.; Othman, I.; Kumari, Y. Inflammation Drives Alzheimer’s Disease: Emphasis on 5-lipoxygenase Pathways. Curr. Neuropharmacol. 2021, 19, 885–895. [Google Scholar] [CrossRef] [PubMed]
- Jack, C.S.; Arbour, N.; Manusow, J.; Montgrain, V.; Blain, M.; McCrea, E.; Shapiro, A.; Antel, J. TLR Signaling Tailors Innate Immune Responses in Human Microglia and Astrocytes. J. Immunol. 2005, 175, 4320–4330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nakajima, K.; Tohyama, Y.; Maeda, S.; Kohsaka, S.; Kurihara, T. Neuronal regulation by which microglia enhance the production of neurotrophic factors for GABAergic, catecholaminergic, and cholinergic neurons. Neurochem. Int. 2007, 50, 807–820. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Glumac, P.M.; Lebeau, A.M. The role of CD133 in cancer: A concise review. Clin. Transl. Med. 2018, 7, 18. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed] [Green Version]
- 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]
- Gupta, P.; Ghosh, S.; Nagarajan, A.; Mehta, V.S.; Sen, E. β-defensin-3 negatively regulates TLR4-HMGB1 axis mediated HLA-G expression in IL-1β treated glioma cells. Cell Signal 2013, 25, 682–689. [Google Scholar] [CrossRef] [PubMed]
- Haghparast, A.; Kharaji, M.H.; Malvandi, A.M. Down-regulation of CD14 transcripts in human glioblastoma cell line U87 MG. Iran. J. Immunol. 2011, 8, 111–119. [Google Scholar]
- Echigo, R.; Sugimoto, N.; Yachie, A.; Ohno-Shosaku, T. Cannabinoids inhibit peptidoglycan-induced phosphorylation of NF-κB and cell growth in U87MG human malig-nant glioma cells. Oncol. Rep. 2012, 28, 1176–1180. [Google Scholar] [CrossRef] [Green Version]
- Hu, F.; Ku, M.-C.; Markovic, D.; Dzaye, A.O.D.; Lehnardt, S.; Synowitz, M.; Wolf, S.; Kettenmann, H. Glioma-associated microglial MMP9 expression is upregulated by TLR2 signaling and sensitive to minocycline. Int. J. Cancer 2014, 135, 2569–2578. [Google Scholar] [CrossRef]
- Wang, C.; Cao, S.; Yan, Y.; Ying, Q.; Jiang, T.; Xu, K.; Wu, A. TLR9 expression in glioma tissues correlated to glioma progression and the prognosis of GBM patients. BMC Cancer 2010, 10, 415. [Google Scholar] [CrossRef] [Green Version]
- Miyar, A.; Habibi, I.; Ebrahimi, A.; Mansourpour, D.; Mokarizadeh, A.; Rajabi, A.; Farshgar, R.; Eshaghzadeh, M.; Zamani-Ahmadmahmudi, M.; Nodushan, S.M.H.T. Predictive and prognostic value of TLR9 and NFKBIA gene expression as potential biomarkers for human glioma diagnosis. J. Neurol. Sci. 2016, 368, 314–317. [Google Scholar] [CrossRef]
- Herrmann, A.; Cherryholmes, G.; Schroeder, A.; Phallen, J.; Alizadeh, D.; Xin, H.; Wang, T.; Lee, H.; Lahtz, C.; Swiderski, P.; et al. TLR9 Is Critical for Glioma Stem Cell Maintenance and Targeting. Cancer Res. 2014, 74, 5218–5228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sinha, S.; Koul, N.; Dixit, D.; Sharma, V.; Sen, E. IGF-1 induced HIF-1α-TLR9 cross talk regulates inflammatory responses in glioma. Cell. Signal. 2011, 23, 1869–1875. [Google Scholar] [CrossRef] [PubMed]
- Kudo, M.; Jono, H.; Shinriki, S.; Yano, S.; Nakamura, H.; Makino, K.; Hide, T.; Muta, D.; Ueda, M.; Ota, K.; et al. Antitumor effect of humanized anti–interleukin-6 receptor antibody (tocilizumab) on glioma cell proliferation. J. Neurosurg. 2009, 111, 219–225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zanotto-Filho, A.; Gonçalves, R.M.; Klafke, K.; de Souza, P.; Dillenburg, F.C.; Carro, L.; Gelain, D.P.; Moreira, J.C.F. Inflammatory landscape of human brain tumors reveals an NFκB dependent cytokine pathway associated with mesenchymal glioblastoma. Cancer Lett. 2017, 390, 176–187. [Google Scholar] [CrossRef] [PubMed]
- Camorani, S.; Esposito, C.; Rienzo, A.; Catuogno, S.; Iaboni, M.; Condorelli, G.; de Franciscis, V.; Cerchia, L. Inhibition of Receptor Signaling and of Glioblastoma-derived Tumor Growth by a Novel PDGFRβ Aptamer. Mol. Ther. 2014, 22, 828–841. [Google Scholar] [CrossRef] [Green Version]
- Huang, H.C.; Huang, C.Y.; Lin-Shiau, S.Y.; Lin, J.K. Ursolic acid inhibits IL-1beta or TNF-alpha-induced C6 glioma invasion through suppressing the association ZIP/p62 with PKC-zeta and downregulating the MMP-9 expression. Mol. Carcinog. 2009, 48, 517–531. [Google Scholar] [CrossRef] [PubMed]
- Roy, L.-O.; Poirier, M.-B.; Fortin, D. Chloroquine inhibits the malignant phenotype of glioblastoma partially by suppressing TGF-beta. Investig. New Drugs 2015, 33, 1020–1031. [Google Scholar] [CrossRef]
- Gravina, G.L.; Mancini, A.; Colapietro, A.; Vitale, F.; Vetuschi, A.; Pompili, S.; Rossi, G.; Marampon, F.; Richardson, P.J.; Patient, L. The novel CXCR4 antagonist, PRX177561, reduces tumor cell proliferation and accelerates cancer stem cell dif-ferentiation in glioblastoma preclinical models. Tumour Biol. 2017, 39, 1010428317695528. [Google Scholar] [CrossRef] [Green Version]
- Gartung, A.; Yang, J.; Sukhatme, V.P.; Bielenberg, D.R.; Fernandes, D.; Chang, J.; Schmidt, B.A.; Hwang, S.H.; Zurakowski, D.; Huang, S.; et al. Suppression of chemotherapy-induced cytokine/lipid mediator surge and ovarian cancer by a dual COX-2/sEH inhibitor. Proc. Natl. Acad. Sci. USA 2019, 116, 1698–1703. [Google Scholar] [CrossRef] [Green Version]
- Massara, M.; Persico, P.; Bonavita, O.; Poeta, V.M.; Locati, M.; Simonelli, M.; Bonecchi, R. Neutrophils in Gliomas. Front. Immunol. 2017, 8, 1349. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Beltraminelli, T.; De Palma, M. Biology and therapeutic targeting of tumour-associated macrophages. J. Pathol. 2020, 250, 573–592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gao, H.; Zhang, I.Y.; Zhang, L.; Song, Y.; Liu, S.; Ren, H.; Liu, H.; Zhou, H.; Su, Y.; Yang, Y.; et al. S100B suppression alters polarization of infiltrating myeloid-derived cells in gliomas and inhibits tumor growth. Cancer Lett. 2018, 439, 91–100. [Google Scholar] [CrossRef] [PubMed]
- Laudati, E.; Curro, D.; Navarra, P.; Lisi, L. Blockade of CCR5 receptor prevents M2 microglia phenotype in a microglia-glioma paradigm. Neurochem. Int. 2017, 108, 100–108. [Google Scholar] [CrossRef]
- Mukherjee, S.; Fried, A.; Hussaini, R.; White, R.; Baidoo, J.; Yalamanchi, S.; Banerjee, P. Phytosomal curcumin causes natural killer cell-dependent repolarization of glioblastoma (GBM) tumor-associated microglia/macrophages and elimination of GBM and GBM stem cells. J. Exp. Clin. Cancer Res. 2018, 37, 168. [Google Scholar] [CrossRef] [Green Version]
- Yang, F.; He, Z.; Duan, H.; Zhang, D.; Li, J.; Yang, H.; Dorsey, J.F.; Zou, W.; Nabavizadeh, S.A.; Bagley, S.J.; et al. Synergistic immunotherapy of glioblastoma by dual targeting of IL-6 and CD40. Nat. Commun. 2021, 12, 1–15. [Google Scholar] [CrossRef]
- Coniglio, S.J.; Eugenin, E.; Dobrenis, K.; Stanley, E.R.; West, B.; Symons, M.H.; Segall, E.J. Microglial Stimulation of Glioblastoma Invasion Involves Epidermal Growth Factor Receptor (EGFR) and Colony Stimulating Factor 1 Receptor (CSF-1R) Signaling. Mol. Med. 2012, 18, 519–527. [Google Scholar] [CrossRef] [PubMed]
- Pyonteck, S.; Akkari, L.; Schuhmacher, A.J.; Bowman, R.L.; Sevenich, L.; Quail, D.F.; Olson, O.; Quick, M.L.; Huse, J.T.; Teijeiro, V.; et al. CSF-1R inhibition alters macrophage polarization and blocks glioma progression. Nat. Med. 2013, 19, 1264–1272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Drill, M.; Powell, K.L.; Kan, L.K.; Jones, N.C.; O’Brien, T.J.; Hamilton, J.A.; Monif, M. Inhibition of purinergic P2X receptor 7 (P2X7R) decreases granulocyte-macrophage colony-stimulating factor (GM-CSF) expression in U251 glioblastoma cells. Sci. Rep. 2020, 10, 14844. [Google Scholar] [CrossRef]
- Kast, R.E.; Scheuerle, A.; Wirtz, C.R.; Karpel-Massler, G.; Halatsch, M.-E. The rationale of targeting neutrophils with dapsone during glioblastoma treatment. Anti-Cancer Agents Med. Chem. 2011, 11, 756–761. [Google Scholar] [CrossRef]
- Karpel-Massler, G.; Kast, R.E.; Siegelin, M.D.; Dwucet, A.; Schneider, E.; Westhoff, M.-A.; Wirtz, C.R.; Chen, X.Y.; Halatsch, M.-E.; Bolm, C. Anti-glioma Activity of Dapsone and Its Enhancement by Synthetic Chemical Modification. Neurochem. Res. 2017, 42, 3382–3389. [Google Scholar] [CrossRef]
- Xue, J.; Zhao, Z.; Zhang, L.; Xue, L.; Shen, S.; Wen, Y.; Wei, Z.; Wang, L.; Kong, L.; Sun, H.; et al. Neutrophil-mediated anticancer drug delivery for suppression of postoperative malignant glioma recurrence. Nat. Nanotechnol. 2017, 12, 692–700. [Google Scholar] [CrossRef]
- Avci, N.G.; Ebrahimzadeh-Pustchi, S.; Akay, Y.M.; Esquenazi, Y.; Tandon, N.; Zhu, J.-J.; Akay, M. NF-κB inhibitor with Temozolomide results in significant apoptosis in glioblastoma via the NF-κB(p65) and actin cytoskeleton regulatory pathways. Sci. Rep. 2020, 10, 13352. [Google Scholar] [CrossRef]
- Geeviman, K.; Babu, D.; Prakash Babu, P. Pantoprazole Induces Mitochondrial Apoptosis and Attenuates NF-κB Signaling in Glioma Cells. Cell. Mol. Neurobiol. 2018, 38, 1491–1504. [Google Scholar] [CrossRef]
- Park, S.Y.; Lim, S.L.; Jang, H.J.; Lee, J.H.; Um, J.Y.; Kim, S.H.; Ahn, K.S.; Lee, S.G. Embelin induces apoptosis in human glioma cells through inactivating NF-κB. J. Pharmacol. Sci. 2013, 121, 192–199. [Google Scholar] [CrossRef] [Green Version]
- Jumnongprakhon, P.; Govitrapong, P.; Tocharus, C.; Pinkaew, D.; Tocharus, J. Melatonin Protects Methamphetamine-Induced Neuroinflammation Through NF-κB and Nrf2 Pathways in Glioma Cell Line. Neurochem. Res. 2015, 40, 1448–1456. [Google Scholar] [CrossRef]
- Mediesse, F.K.; Boudjeko, T.; Hasitha, A.; Gangadhar, M.; Mbacham, W.F.; Yogeeswari, P. Inhibition of lipopolysaccharide (LPS)-induced neuroinflammatory response by polysaccharide fractions of Kha-ya grandifoliola (C.D.C.) stem bark, Cryptolepis sanguinolenta (Lindl.) Schltr and Cymbopogon citratus Stapf leaves in raw 264.7 macrophages and U87 glioblastoma cells. BMC Complement. Altern. Med. 2018, 18, 86. [Google Scholar]
- Xie, T.X.; Xia, Z.; Zhang, N.; Gong, W.; Huang, S. Constitutive NF-kappaB activity regulates the expression of VEGF and IL-8 and tumor angiogenesis of human glio-blastoma. Oncol. Rep. 2010, 23, 725–732. [Google Scholar]
- Barberi, T.; Martin, A.; Suresh, R.; Barakat, D.J.; Harris-Bookman, S.; Drake, C.G.; Lim, M.; Friedman, A.D. Absence of host NF-κB p50 induces murine glioblastoma tumor regression, increases survival, and decreases T-cell induction of tumor-associated macrophage M2 polarization. Cancer Immunol. Immunother. 2018, 67, 1491–1503. [Google Scholar] [CrossRef]
- Park, J.-H.; Seo, Y.H.; Jang, J.-H.; Jeong, C.-H.; Lee, S.; Park, B. Asiatic acid attenuates methamphetamine-induced neuroinflammation and neurotoxicity through blocking of NF-kB/STAT3/ERK and mitochondria-mediated apoptosis pathway. J. Neuroinflammation 2017, 14, 240. [Google Scholar] [CrossRef]
- Porro, C.; Cianciulli, A.; Trotta, T.; Lofrumento, D.D.; Panaro, M.A. Curcumin Regulates Anti-Inflammatory Responses by JAK/STAT/SOCS Signaling Pathway in BV-2 Microglial Cells. Biology 2019, 8, 51. [Google Scholar] [CrossRef] [Green Version]
- Jensen, K.; Cseh, O.; Aman, A.; Weiss, S.; Luchman, H.A. The JAK2/STAT3 inhibitor pacritinib effectively inhibits patient-derived GBM brain tumor initiating cells in vitro and when used in combination with temozolomide increases survival in an orthotopic xenograft model. PLoS ONE 2017, 12, e0189670. [Google Scholar] [CrossRef] [Green Version]
- Kiprianova, I.; Remy, J.; Milosch, N.; Mohrenz, I.V.; Seifert, V.; Aigner, A.; Kögel, D. Sorafenib Sensitizes Glioma Cells to the BH3 Mimetic ABT-737 by Targeting MCL1 in a STAT3-Dependent Manner. Neoplasia 2015, 17, 564–573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Komohara, Y.; Fujiwara, Y.; Kudo, R.; Tsurushima, K.; Ohnishi, K.; Ikeda, T.; Takeya, M. Oleanolic acid inhibits macrophage differentiation into the M2 phenotype and glioblastoma cell proliferation by suppressing the activation of STAT3. Oncol. Rep. 2011, 26, 1533–1537. [Google Scholar] [CrossRef] [PubMed]
- Hussain, S.F.; Kong, L.-Y.; Jordan, J.; Conrad, C.; Madden, T.; Fokt, I.; Priebe, W.; Heimberger, A.B. A Novel Small Molecule Inhibitor of Signal Transducers and Activators of Transcription 3 Reverses Immune Tolerance in Malignant Glioma Patients. Cancer Res. 2007, 67, 9630–9636. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Michaud-Levesque, J.; Bousquet-Gagnon, N.; Béliveau, R. Quercetin abrogates IL-6/STAT3 signaling and inhibits glioblastoma cell line growth and migration. Exp. Cell Res. 2012, 318, 925–935. [Google Scholar] [CrossRef]
- Ding, D.; Wei, S.; Song, Y.; Li, L.; Du, G.; Zhan, H.; Cao, Y. Osthole Exhibits Anti-Cancer Property in Rat Glioma Cells Through Inhibiting PI3K/Akt and MAPK Signaling Pathways. Cell. Physiol. Biochem. 2013, 32, 1751–1760. [Google Scholar] [CrossRef] [PubMed]
- Zeng, K.-W.; Wang, S.; Dong, X.; Jiang, Y.; Tu, P.-F. Sesquiterpene dimer (DSF-52) from Artemisia argyi inhibits microglia-mediated neuroinflammation via suppres-sion of NF-κB, JNK/p38 MAPKs and Jak2/Stat3 signaling pathways. Phytomedicine 2014, 21, 298–306. [Google Scholar] [CrossRef]
- Goldsmith, C.S.; Kim, S.M.; Karunarathna, N.; Neuendorff, N.; Toussaint, L.G.; Earnest, D.J.; Bell-Pedersen, D. Inhibition of p38 MAPK activity leads to cell type-specific effects on the molecular circadian clock and time-dependent reduction of glioma cell invasiveness. BMC Cancer 2018, 18, 43. [Google Scholar] [CrossRef] [PubMed]
- Lien, L.-M.; Wang, M.-J.; Chen, R.-J.; Chiu, H.-C.; Wu, J.-L.; Shen, M.-Y.; Chou, D.-S.; Sheu, J.-R.; Lin, K.-H.; Lu, W.-J. Nobiletin, a Polymethoxylated Flavone, Inhibits Glioma Cell Growth and Migration via Arresting Cell Cycle and Suppressing MAPK and Akt Pathways. Phytotherapy Res. 2015, 30, 214–221. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Zou, S.; Lan, Y.-L.; Xing, J.-S.; Lan, X.-Q.; Zhang, B. Solasonine inhibits glioma growth through anti-inflammatory pathways. Am. J. Transl. Res. 2017, 9, 3977–3989. [Google Scholar]
- Yeung, Y.T.; Bryce, N.S.; Adams, S.; Braidy, N.; Konayagi, M.; McDonald, K.L.; Teo, C.; Guillemin, G.J.; Grewal, T.; Munoz, L. p38 MAPK inhibitors attenuate pro-inflammatory cytokine production and the invasiveness of human U251 glio-blastoma cells. J. Neurooncol. 2012, 109, 35–44. [Google Scholar] [CrossRef]
- Zhu, Y.; Liu, X.; Zhao, P.; Zhao, H.; Gao, W.; Wang, L. Celastrol Suppresses Glioma Vasculogenic Mimicry Formation and Angiogenesis by Blocking the PI3K/Akt/mTOR Signaling Pathway. Front. Pharmacol. 2020, 11, 25. [Google Scholar] [CrossRef] [Green Version]
- Speranza, M.C.; Nowicki, M.; Behera, P.; Cho, C.-F.; Chiocca, E.A.; Lawler, S.E. BKM-120 (Buparlisib): A Phosphatidyl-Inositol-3 Kinase Inhibitor with Anti-Invasive Properties in Glioblas-toma. Sci. Rep. 2016, 6, 20189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cheng, Y.; Zhang, Y.; Zhang, L.; Ren, X.; Huber-Keener, K.; Liu, X.; Zhou, L.; Liao, J.; Keihack, H.; Yan, L.; et al. MK-2206, a Novel Allosteric Inhibitor of Akt, Synergizes with Gefitinib against Malignant Glioma via Modulating Both Autophagy and Apoptosis. Mol. Cancer Ther. 2011, 11, 154–164. [Google Scholar] [CrossRef] [Green Version]
- Crane, C.; Panner, A.; Pieper, R.O.; Arbiser, J.; Parsa, A.T. Honokiol-mediated inhibition of PI3K/mTOR pathway: A potential strategy to overcome immunoresistance in glioma, breast, and prostate carcinoma without impacting T cell function. J. Immunother. 2009, 32, 585–592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kaneda, M.M.; Messer, K.S.; Ralainirina, N.; Li, H.; Leem, C.J.; Gorjestani, S.; Woo, G.; Nguyen, A.V.; Figueiredo, C.C.; Foubert, P.; et al. PI3Kγ is a molecular switch that controls immune suppression. Nat. Cell Biol. 2016, 539, 437–442. [Google Scholar] [CrossRef] [Green Version]
- Wang, W.; Xia, X.; Mao, L.; Wang, S. The CCAAT/Enhancer-Binding Protein Family: Its Roles in MDSC Expansion and Function. Front. Immunol. 2019, 10, 1804. [Google Scholar] [CrossRef] [PubMed]
- Chen, G.; Yue, Y.; Qin, J.; Xiao, X.; Ren, Q.; Xiao, B. Plumbagin suppresses the migration and invasion of glioma cells via downregulation of MMP-2/9 expression and inaction of PI3K/Akt signaling pathway in vitro. J. Pharmacol. Sci. 2017, 134, 59–67. [Google Scholar] [CrossRef]
- Lisi, L.; Laudati, E.; Navarra, P.; Russo, C.D. The mTOR kinase inhibitors polarize glioma-activated microglia to express a M1 phenotype. J. Neuroinflammation 2014, 11, 125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chandrika, G.; Natesh, K.; Ranade, D.; Chugh, A.; Shastry, P. Suppression of the invasive potential of Glioblastoma cells by mTOR inhibitors involves modulation of NFκB and PKC-α signaling. Sci. Rep. 2016, 6, 22455. [Google Scholar] [CrossRef] [Green Version]
- El Andaloussi, A.; Sonabend, A.M.; Han, Y.; Lesniak, M.S. Stimulation of TLR9 with CpG ODN enhances apoptosis of glioma and prolongs the survival of mice with experimental brain tumors. Glia 2006, 54, 526–535. [Google Scholar] [CrossRef]
- Huang, Y.; Zhang, Q.; Lubas, M.; Yuan, Y.; Yalcin, F.; Efe, I.E.; Xia, P.; Motta, E.; Buonfigioli, A.; Lehnardt, S.; et al. Synergistic Toll-like Receptor 3/9 Signaling Affects Properties and Impairs Glioma-Promoting Activity of Microglia. J. Neurosci. 2020, 40, 6428–6443. [Google Scholar] [CrossRef]
- Jordan, M.; Waxman, D.J. CpG-1826 immunotherapy potentiates chemotherapeutic and anti-tumor immune responses to metro-nomic cyclophosphamide in a preclinical glioma model. Cancer Lett. 2016, 373, 88–96. [Google Scholar] [CrossRef] [Green Version]
- Wanderley, C.W.; Colon, D.F.; Luiz, J.P.M.; Oliveira, F.F.; Viacava, P.R.; Leite, A.C.; Pereira, A.J.; Silva, C.M.; Silva, C.R.; Silva, R.L.; et al. Paclitaxel reduces tumor growth by reprogramming tumor-associated macrophages to an M1- profile in a TLR4-dependent manner. Cancer Res. 2018, 78, 5891–5900. [Google Scholar] [CrossRef] [Green Version]
- Abarca-Merlin, D.M.; Maldonado-Bernal, C.; Alvarez-Arellano, L. Toll-Like Receptors as Therapeutic Targets in Central Nervous System Tumors. BioMed Res. Int. 2019, 2019, 5286358–5286359. [Google Scholar] [CrossRef]
- Triller, P.; Bachorz, J.; Synowitz, M.; Kettenmann, H.; Markovic, D. O-Vanillin Attenuates the TLR2 Mediated Tumor-Promoting Phenotype of Microglia. Int. J. Mol. Sci. 2020, 21, 2959. [Google Scholar] [CrossRef] [Green Version]
- Vinnakota, K.; Hu, F.; Ku, M.-C.; Georgieva, P.B.; Szulzewsky, F.; Pohlmann, A.; Waiczies, S.; Waiczies, H.; Niendorf, T.; Lehnardt, S.; et al. Toll-like receptor 2 mediates microglia/brain macrophage MT1-MMP expression and glioma expansion. Neuro-Oncology 2013, 15, 1457–1468. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, Y.; Ju, Y.; Liu, J.; Chen, Y.; Huo, X.; Liu, L. Inhibition of proliferation and migration and induction of apoptosis in glioma cells by silencing TLR4 expression levels via RNA interference. Oncol. Lett. 2020, 21, 13. [Google Scholar] [CrossRef] [PubMed]
- Badri, W.; Miladi, K.; Nazari, Q.A.; Greige-Gerges, H.; Fessi, H.; Elaissari, A. Encapsulation of NSAIDs for inflammation management: Overview, progress, challenges and prospects. Int. J. Pharm. 2016, 515, 757–773. [Google Scholar] [CrossRef] [PubMed]
- Lee, M.; Sparatore, A.; Del Soldato, P.; Mcgeer, E.; McGeer, P.L. Hydrogen sulfide-releasing NSAIDs attenuate neuroinflammation induced by microglial and astrocytic activation. Glia 2009, 58, 103–113. [Google Scholar] [CrossRef] [PubMed]
- Deardorff, W.J.; Grossberg, G.T. Targeting neuroinflammation in Alzheimer’s disease: Evidence for NSAIDs and novel therapeu-tics. Expert Rev. Neurother. 2017, 17, 17–32. [Google Scholar] [CrossRef] [PubMed]
- Wong, R.S.Y. Role of Nonsteroidal Anti-Inflammatory Drugs (NSAIDs) in Cancer Prevention and Cancer Promotion. Adv. Pharmacol. Sci. 2019, 2019, 3418975. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Amirian, E.S.; Ostrom, Q.T.; Armstrong, G.N.; Lai, R.K.; Gu, X.; Jacobs, I.D.; Jalali, A.; Claus, E.B.; Barnholtz-Sloan, J.S.; Il’Yasova, D.; et al. Aspirin, Non-Steroidal Anti-Inflammatory Drugs (NSAIDs), and Glioma Risk: Original Data from the Glioma International Case-Control Study and a Meta-Analysis. Cancer Epidemiol. Biomark. Prev. 2018, 28, 555–562. [Google Scholar] [CrossRef] [Green Version]
- Fujita, M.; Kohanbash, G.; Fellows-Mayle, W.; Hamilton, R.L.; Komohara, Y.; Decker, S.A.; Ohlfest, J.R.; Okada, H. COX-2 Blockade Suppresses Gliomagenesis by Inhibiting Myeloid-Derived Suppressor Cells. Cancer Res. 2011, 71, 2664–2674. [Google Scholar] [CrossRef] [Green Version]
- Leidgens, V.; Seliger, C.; Jachnik, B.; Welz, T.; Leukel, P.; Vollmann-Zwerenz, A.; Bogdahn, U.; Kreutz, M.; Grauer, O.M.; Hau, P. Ibuprofen and Diclofenac Restrict Migration and Proliferation of Human Glioma Cells by Distinct Molecular Mechanisms. PLoS ONE 2015, 10, e0140613. [Google Scholar] [CrossRef] [Green Version]
- Gerthofer, V.; Kreutz, M.; Renner, K.; Jachnik, B.; Dettmer, K.; Oefner, P.; Riemenschneider, M.J.; Proescholdt, M.; Vollmann-Zwerenz, A.; Hau, P.; et al. Combined Modulation of Tumor Metabolism by Metformin and Diclofenac in Glioma. Int. J. Mol. Sci. 2018, 19, 2586. [Google Scholar] [CrossRef] [Green Version]
- Sareddy, G.R.; Geeviman, K.; Ramulu, C.; Babu, P.P. The nonsteroidal anti-inflammatory drug celecoxib suppresses the growth and induces apoptosis of human glio-blastoma cells via the NF-κB pathway. J. Neurooncol. 2012, 106, 99–109. [Google Scholar] [CrossRef]
- Goswami, S.; Walle, T.; Cornish, A.E.; Basu, S.; Anandhan, S.; Fernandez, I.; Vence, L.; Blando, J.; Zhao, H.; Yadav, S.S.; et al. Immune profiling of human tumors identifies CD73 as a combinatorial target in glioblastoma. Nat. Med. 2019, 26, 39–46. [Google Scholar] [CrossRef] [PubMed]
- Miyazaki, T.; Ishikawa, E.; Sugii, N.; Matsuda, M. Therapeutic Strategies for Overcoming Immunotherapy Resistance Mediated by Immunosuppressive Factors of the Glioblastoma Microenvironment. Cancers 2020, 12, 1960. [Google Scholar] [CrossRef] [PubMed]
- Engel, A.L.; Holt, E.G.; Lu, H. The pharmacokinetics of Toll-like receptor agonists and the impact on the immune system. Expert Rev. Clin. Pharmacol. 2011, 4, 275–289. [Google Scholar] [CrossRef] [Green Version]
- Keshavarz, A.; Pourbagheri-Sigaroodi, A.; Zafari, P.; Bagheri, N.; Ghaffari, S.H.; Bashash, D. Toll-like receptors (TLRs) in cancer; with an extensive focus on TLR agonists and antagonists. IUBMB Life 2020, 73, 10–25. [Google Scholar] [CrossRef]
- Luchner, M.; Reinke, S.; Milicic, A. TLR Agonists as Vaccine Adjuvants Targeting Cancer and Infectious Diseases. Pharmaceutics 2021, 13, 142. [Google Scholar] [CrossRef]
- Wang, Y.; Tu, Q.; Yan, W.; Xiao, D.; Zeng, Z.; Ouyang, Y.; Huang, L.; Cai, J.; Zeng, X.; Chen, Y.J.; et al. CXC195 suppresses proliferation and inflammatory response in LPS-induced human hepatocellular carcinoma cells via regulating TLR4-MyD88-TAK1-mediated NF-κB and MAPK pathway. Biochem. Biophys. Res. Commun. 2015, 456, 373–379. [Google Scholar] [CrossRef] [PubMed]
- Kashani, B.; Zandi, Z.; Bashash, D.; Zaghal, A.; Momeny, M.; Poursani, E.M.; Pourbagheri-Sigaroodi, A.; Mousavi, S.A.; Ghaffari, S.H. Small molecule inhibitor of TLR4 inhibits ovarian cancer cell proliferation: New insight into the anticancer effect of TAK-242 (Resatorvid). Cancer Chemother. Pharmacol. 2019, 85, 47–59. [Google Scholar] [CrossRef] [PubMed]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Basheer, A.S.; Abas, F.; Othman, I.; Naidu, R. Role of Inflammatory Mediators, Macrophages, and Neutrophils in Glioma Maintenance and Progression: Mechanistic Understanding and Potential Therapeutic Applications. Cancers 2021, 13, 4226. https://doi.org/10.3390/cancers13164226
Basheer AS, Abas F, Othman I, Naidu R. Role of Inflammatory Mediators, Macrophages, and Neutrophils in Glioma Maintenance and Progression: Mechanistic Understanding and Potential Therapeutic Applications. Cancers. 2021; 13(16):4226. https://doi.org/10.3390/cancers13164226
Chicago/Turabian StyleBasheer, Abdul Samad, Faridah Abas, Iekhsan Othman, and Rakesh Naidu. 2021. "Role of Inflammatory Mediators, Macrophages, and Neutrophils in Glioma Maintenance and Progression: Mechanistic Understanding and Potential Therapeutic Applications" Cancers 13, no. 16: 4226. https://doi.org/10.3390/cancers13164226
APA StyleBasheer, A. S., Abas, F., Othman, I., & Naidu, R. (2021). Role of Inflammatory Mediators, Macrophages, and Neutrophils in Glioma Maintenance and Progression: Mechanistic Understanding and Potential Therapeutic Applications. Cancers, 13(16), 4226. https://doi.org/10.3390/cancers13164226