Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 1260 word(s) 1260 2022-01-12 05:17:35 |
2 format is correct Meta information modification 1260 2022-01-25 10:19:15 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Kögel, D. STAT3 Enhances Sensitivity of Glioblastoma to Cell Death. Encyclopedia. Available online: https://encyclopedia.pub/entry/18754 (accessed on 12 May 2024).
Kögel D. STAT3 Enhances Sensitivity of Glioblastoma to Cell Death. Encyclopedia. Available at: https://encyclopedia.pub/entry/18754. Accessed May 12, 2024.
Kögel, Donat. "STAT3 Enhances Sensitivity of Glioblastoma to Cell Death" Encyclopedia, https://encyclopedia.pub/entry/18754 (accessed May 12, 2024).
Kögel, D. (2022, January 25). STAT3 Enhances Sensitivity of Glioblastoma to Cell Death. In Encyclopedia. https://encyclopedia.pub/entry/18754
Kögel, Donat. "STAT3 Enhances Sensitivity of Glioblastoma to Cell Death." Encyclopedia. Web. 25 January, 2022.
STAT3 Enhances Sensitivity of Glioblastoma to Cell Death
Edit

Glioblastoma is the most common primary brain cancer in adults. One reason for the development and malignancy of this tumor is the misregulation of certain cellular proteins. The oncoprotein Signal Transducer and Activator of Transcription (STAT3) that is frequently overactive in glioblastoma cells is associated with more aggressive disease and decreased patient survival. Autophagy is a form of cellular self digestion that normally maintains cell integrity and provides nutrients and basic building blocks required for growth. While glioblastoma is known to be particularly resistant to conventional therapies, recent research has suggested that these tumors are more sensitive to excessive overactivation of autophagy, leading to autophagy-dependent tumor cell death.

STAT3 glioblastoma autophagy lysosome

1. Introduction

Glioblastoma (GBM; grade IV glioma, WHO) is a primary brain tumor that mainly occurs in adults. GBM-patients have a very dismal prognosis with a median survival of ~15 months despite aggressive radiochemotherapy after maximally possible surgery [1]. Since GBMs are characterized by their diffuse infiltrative growth, a complete surgical resection is nearly impossible and the tumors quickly recur, often with an even more aggressive growth [2]. Different molecular subtypes of GBM have been described based on the genetic profiles of the tumors. Although their nomenclature is not uniformly used, the general consensus agrees on three GBM subtypes: proneural, classical and mesenchymal [3][4], of which the latter one is the most aggressive. Signal Transducer and Activator of Transcription (STAT3) is a molecular hub-like oncoprotein that is highly overactivated in GBM and is associated with the mesenchymal subtype [5][6]. STAT3 is known to regulate multiple hallmarks of cancer such as proliferation, cell survival, angiogenesis and immune evasion (reviewed in [7][8]). The activation of STAT3 is mediated by multiple upstream kinases [9] which phosphorylate the protein at two different sites (reviewed in [10], Tyrosine705 (pY705Stat3) and Serine727 (pS727Stat3). Upon activation, STAT3 forms homodimers or heterodimers with other proteins of the STAT-family, translocates into the nucleus and induces pronounced changes in gene expression. Known targets include genes responsible for various hallmarks of cancer like migration/invasion (MMP2, MMP9; [11][12]) or EMT-like features and immune evasion/suppression (e.g., SNAI1; [13][14]). Targeting STAT3 by upstream signaling inhibitors of the STAT3 pathway or by genetic depletion decreases glioma cell proliferation and migration in vitro and prolongs overall survival of tumor-bearing mice in vivo [15][16][17][18][19][20][21].
An underappreciated function of STAT3 is its involvement in autophagy and lysosomal function. Recent studies could already show that STAT3 can transcriptionally regulate several genes relevant for autophagy [22][23][24][25][26][27]. In addition, unphosphorylated, cytoplasmic STAT3 can have direct, transcription-independent effects on the autophagy pathway [28]. Furthermore, upon translocation to the mitochondria STAT3 was found to decrease mitochondrial reactive oxygen species (ROS) that in turn may trigger autophagy [22][29][30][31][32]. Notably, STAT3 has also been reported to be important for regulating lysosomal membrane permeabilization (LMP) and lysosome-dependent cell death (LDCD) during post-lactational regression of the mammary gland epithelium [33][34] and in breast cancer cells [35]. Curiously, under these circumstances STAT3 rather promotes cell death, a function that is in stark contrast to its canonical anti-apoptotic pro-survival role.
The lysosomes have been fittingly described as the “suicide bags” of the cells, because of their low pH and content of acidic hydrolases which are able to degrade macromolecules [36]. Damage to the lysosomal membrane, for example through cationic amphiphilic drugs [37][38], can trigger lysosomal membrane permeabilization (LMP), causing lysosomal rupture and release of its content into the cytosol [39]. The concomitant release of lysosomal cathepsin proteases and hydrolytic enzymes can lead to a specific type of cell death termed lysosome-dependent cell death (LDCD), that depending on the extent of LMP can have necrotic, apoptotic or apoptosis-like features [39]. LDCD has been observed in several pathophysiological conditions such as inflammation, aging, neurodegeneration and cardiovascular disorders [37][39][40][41]. It has been reported that cancer cells hijack lysosomes to remodel the extracellular matrix in order to increase tissue invasion and metastasis, while at the same time destabilizing them [42]. Accordingly, an increased expression, activity and secretion of cathepsins, as well as changes in lysosomal volume, composition and cellular distribution have been observed in different tumors [43]. Based on the findings suggesting both an increased lysosomal activity and concomitant lysosomal destabilization in certain tumors, these organelles might represent a double-edged sword and render cancer cells more prone to lysosome-targeting agents and LDCD via excessive activation of LMP [42].

2. Research Findings

The oncogenic transcription factor STAT3 acts as a signaling hub molecule involved in regulation of most, if not all hallmarks of cancer, including proliferation, tumor invasion, altered cellular metabolism, angiogenesis, immune evasion and cell survival. Overactivation of STAT3 is frequently observed in malignant gliomas and correlated to the mesenchymal subtype of GBM, a particularly aggressive and treatment-resistant molecular subgroup of these tumors [9][44][45]. Therefore, STAT3 represents a very interesting target for GBM therapy. However, STAT3 and other oncogenic transcription factors are a unique class of targets that are very difficult to drug. Therefore, many of the current efforts are focusing on the pharmacological inhibition of upstream JAK kinases in the JAK/Stat pathway, including a phase I clinical trial with the JAK inhibitor WP1066 (NCT01904123), although it should be noted that non-receptor tyrosine kinases such as bone marrow and X-linked (BMX) and members of the Src family can bypass JAK-dependent activation of STAT3 in GBM [9][46]. An alternative approach to target STAT3-driven GBMs is to identify and exploit particular vulnerabilities of these tumors. Extensive and prolonged overactivation of autophagy and the concomitant induction of ACD represents a particular sensitivity of GBM cells and an interesting new therapeutic approach [47][48][49][50][51]. While STAT3 has been found to be important for the (positive and negative) regulation of autophagy [32], the exact role of STAT3 in autophagy is highly dependent on the subcellular localization of STAT3 and the cellular context [32].
Mechanistically, this death-sensitizing role of STAT3 appears to be independent from alterations in the early steps of the autophagy pathway, because STAT3-deficient and proficient cell lines displayed a very similar extent of LC-3-conversion and an unchanged autophagic flux in the experiments. These findings indicate that STAT3 may rather interfere at the later steps of the autophagosomal/lysosomal pathway, in particular at the lysosomal level. Consistent with this hypothesis, an LMP-promoting role of STAT3 has been documented in several previous studies, for example during mammary gland involution [33][34] as well as in breast cancer cells treated with a derivative of the natural molecule riccardin D [35]. As already outlined above, IM, TIC and Pimo are FIASMAs that accumulate within the lysosomes and induce the detachment and inactivation of acid sphingomyelinase from the lysosomal membrane [52], thereby enhancing deregulation of lipid homeostasis. This event has in turn been correlated to lysosomal stress and LMP [53][54][55]
In addition to enhanced lysosomal activation, the demand for cholesterol may be a second vulnerability of (STAT3-driven) tumors, in particular GBMs that are incapable of de novo cholesterol synthesis and rely on exogenous cholesterol [56][57][58]. Hence, the combined targeting of cholesterol trafficking and lysosomal function, as demonstrated to occur with the drugs used in this entry [38], appears to be an interesting strategy for the treatment of GBM.

3. Conclusions

Here, the entry presents a novel pro-death function of the oncoprotein STAT3, which is frequently overactivated in multiple human cancers including glioblastoma and is associated with the most aggressive mesenchymal subtype. Using isogenic Crispr/Cas9-Knockouts and stable shRNA-mediated STAT3-KD cells the loss of STAT3 leads to reduced sensitivity to known ACD-inducers was shown. It is shown further that this is accompanied by reduced lysosomal lipid accumulation in STAT3-deficient cells and therefore less leakage of lysosomal contents in the cytosol, which causes LDCD in STAT3-proficient cells. In conclusion, these findings offer new research directions to develop targeted therapies of STAT3-activated, autophagy-proficient tumors such as glioblastoma.

References

  1. Louis, D.N.; Perry, A.; Reifenberger, G.; von Deimling, A.; Figarella-Branger, D.; Cavenee, W.K.; Ohgaki, H.; Wiestler, O.D.; Kleihues, P.; Ellison, D.W. The 2016 World Health Organization classification of tumors of the central nervous system: A summary. Acta Neuropathol. 2016, 131, 803–820.
  2. Becker, K.P.; Yu, J. Status quo—standard-of-care medical and radiation therapy for glioblastoma. Cancer J. 2012, 18, 12–19.
  3. Verhaak, R.G.; Hoadley, K.A.; Purdom, E.; Wang, V.; Qi, Y.; Wilkerson, M.D.; Miller, C.R.; Ding, L.; Golub, T.; Mesirov, J.P.; et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 2010, 17, 98–110.
  4. Olar, A.; Aldape, K.D. Using the molecular classification of glioblastoma to inform personalized treatment. J. Pathol. 2014, 232, 165–177.
  5. Carro, M.S.; Lim, W.K.; Alvarez, M.J.; Bollo, R.J.; Zhao, X.; Snyder, E.Y.; Sulman, E.P.; Anne, S.L.; Doetsch, F.; Colman, H.; et al. The transcriptional network for mesenchymal transformation of brain tumours. Nature 2010, 463, 318–325.
  6. Hossain, A.; Gumin, J.; Gao, F.; Figueroa, J.; Shinojima, N.; Takezaki, T.; Priebe, W.; Villarreal, D.; Kang, S.G.; Joyce, C.; et al. Mesenchymal stem cells isolated from human gliomas increase proliferation and maintain stemness of glioma stem cells through the IL-6/gp130/STAT3 pathway. Stem Cells 2015, 33, 2400–2415.
  7. Yu, H.; Lee, H.; Herrmann, A.; Buettner, R.; Jove, R. Revisiting STAT3 signalling in cancer: New and unexpected biological functions. Nat. Rev. Cancer 2014, 14, 736–746.
  8. Galoczova, M.; Coates, P.; Vojtesek, B. STAT3, stem cells, cancer stem cells and p63. Cell. Mol. Biol. Lett. 2018, 23, 12.
  9. Kim, J.E.; Patel, M.; Ruzevick, J.; Jackson, C.M.; Lim, M. STAT3 Activation in glioblastoma: Biochemical and therapeutic implications. Cancers 2014, 6, 376–395.
  10. Brantley, E.C.; Benveniste, E.N. Signal transducer and activator of transcription-3: A molecular hub for signaling pathways in gliomas. Mol. Cancer Res. 2008, 6, 675–684.
  11. Luwor, R.B.; Baradaran, B.; Taylor, L.E.; Iaria, J.; Nheu, T.V.; Amiry, N.; Hovens, C.M.; Wang, B.; Kaye, A.H.; Zhu, H.J. Targeting Stat3 and Smad7 to restore TGF-beta cytostatic regulation of tumor cells in vitro and in vivo. Oncogene 2013, 32, 2433–2441.
  12. Westermarck, J.; Kahari, V.M. Regulation of matrix metalloproteinase expression in tumor invasion. FASEB J. 1999, 13, 781–792.
  13. Wendt, M.K.; Balanis, N.; Carlin, C.R.; Schiemann, W.P. STAT3 and epithelial-mesenchymal transitions in carcinomas. JAK-STAT 2014, 3, e28975.
  14. Kudo-Saito, C.; Shirako, H.; Takeuchi, T.; Kawakami, Y. Cancer metastasis is accelerated through immunosuppression during Snail-induced EMT of cancer cells. Cancer Cell 2009, 15, 195–206.
  15. Esposito, C.L.; Nuzzo, S.; Catuogno, S.; Romano, S.; de Nigris, F.; de Franciscis, V. STAT3 gene silencing by aptamer-siRNA chimera as selective therapeutic for glioblastoma. Mol. Ther. Nucleic Acids 2018, 10, 398–411.
  16. Kiprianova, I.; Remy, J.; Milosch, N.; Mohrenz, I.V.; Seifert, V.; Aigner, A.; Kogel, D. Sorafenib sensitizes glioma cells to the BH3 mimetic ABT-737 by targeting MCL1 in a STAT3-dependent manner. Neoplasia 2015, 17, 564–573.
  17. Priester, M.; Copanaki, E.; Vafaizadeh, V.; Hensel, S.; Bernreuther, C.; Glatzel, M.; Seifert, V.; Groner, B.; Kogel, D.; Weissenberger, J. STAT3 silencing inhibits glioma single cell infiltration and tumor growth. Neuro-Oncology 2013, 15, 840–852.
  18. Senft, C.; Priester, M.; Polacin, M.; Schroder, K.; Seifert, V.; Kogel, D.; Weissenberger, J. Inhibition of the JAK-2/STAT3 signaling pathway impedes the migratory and invasive potential of human glioblastoma cells. J. Neuro-Oncol. 2011, 101, 393–403.
  19. Sherry, M.M.; Reeves, A.; Wu, J.L.K.; Cochran, B.H. STAT3 is required for proliferation and maintenance of multipotency in glioblastoma stem cells. Stem Cells 2009, 27, 2383–2392.
  20. Takeda, K.; Kaisho, T.; Yoshida, N.; Takeda, J.; Kishimoto, T.; Akira, S. Stat3 activation is responsible for IL-6-dependent T cell proliferation through preventing apoptosis: Generation and characterization of T cell-specific Stat3-deficient mice. J. Immunol. 1998, 161, 4652–4660.
  21. Linder, B.; Weirauch, U.; Ewe, A.; Uhmann, A.; Seifert, V.; Mittelbronn, M.; Harter, P.N.; Aigner, A.; Kogel, D. Therapeutic targeting of Stat3 using lipopolyplex nanoparticle-formulated siRNA in a syngeneic orthotopic mouse glioma model. Cancers 2019, 11, 333.
  22. Gong, J.; Munoz, A.R.; Chan, D.; Ghosh, R.; Kumar, A.P. STAT3 down regulates LC3 to inhibit autophagy and pancreatic cancer cell growth. Oncotarget 2014, 5, 2529–2541.
  23. Sun, L.; Hu, L.; Cogdell, D.; Lu, L.; Gao, C.; Tian, W.; Zhang, Z.; Kang, Y.; Fleming, J.B.; Zhang, W. MIR506 induces autophagy-related cell death in pancreatic cancer cells by targeting the STAT3 pathway. Autophagy 2017, 13, 703–714.
  24. Jung, J.E.; Lee, H.G.; Cho, I.H.; Chung, D.H.; Yoon, S.H.; Yang, Y.M.; Lee, J.W.; Choi, S.; Park, J.W.; Ye, S.K.; et al. STAT3 is a potential modulator of HIF-1-mediated VEGF expression in human renal carcinoma cells. FASEB J. 2005, 19, 1296–1298.
  25. Jung, J.E.; Kim, H.S.; Lee, C.S.; Shin, Y.J.; Kim, Y.N.; Kang, G.H.; Kim, T.Y.; Juhnn, Y.S.; Kim, S.J.; Park, J.W.; et al. STAT3 inhibits the degradation of HIF-1alpha by pVHL-mediated ubiquitination. Exp. Mol. Med. 2008, 40, 479–485.
  26. Pratt, J.; Annabi, B. Induction of autophagy biomarker BNIP3 requires a JAK2/STAT3 and MT1-MMP signaling interplay in Concanavalin-A-activated U87 glioblastoma cells. Cell. Signal. 2014, 26, 917–924.
  27. Pensa, S.; Lloyd-Lewis, B.; Sargeant, T.J.; Resemann, H.K.; Kahn, C.R.; Watson, C.J. Signal transducer and activator of transcription 3 and the phosphatidylinositol 3-kinase regulatory subunits p55alpha and p50alpha regulate autophagy in vivo. FEBS J. 2014, 281, 4557–4567.
  28. Shen, S.; Niso-Santano, M.; Adjemian, S.; Takehara, T.; Malik, S.A.; Minoux, H.; Souquere, S.; Marino, G.; Lachkar, S.; Senovilla, L.; et al. Cytoplasmic STAT3 represses autophagy by inhibiting PKR activity. Mol. Cell 2012, 48, 667–680.
  29. Szczepanek, K.; Chen, Q.; Larner, A.C.; Lesnefsky, E.J. Cytoprotection by the modulation of mitochondrial electron transport chain: The emerging role of mitochondrial STAT3. Mitochondrion 2012, 12, 180–189.
  30. Szczepanek, K.; Lesnefsky, E.J.; Larner, A.C. Multi-tasking: Nuclear transcription factors with novel roles in the mitochondria. Trends Cell Biol. 2012, 22, 429–437.
  31. Wegrzyn, J.; Potla, R.; Chwae, Y.J.; Sepuri, N.B.; Zhang, Q.; Koeck, T.; Derecka, M.; Szczepanek, K.; Szelag, M.; Gornicka, A.; et al. Function of mitochondrial Stat3 in cellular respiration. Science 2009, 323, 793–797.
  32. You, L.; Wang, Z.; Li, H.; Shou, J.; Jing, Z.; Xie, J.; Sui, X.; Pan, H.; Han, W. The role of STAT3 in autophagy. Autophagy 2015, 11, 729–739.
  33. Sargeant, T.J.; Lloyd-Lewis, B.; Resemann, H.K.; Ramos-Montoya, A.; Skepper, J.; Watson, C.J. Stat3 controls cell death during mammary gland involution by regulating uptake of milk fat globules and lysosomal membrane permeabilization. Nat. Cell Biol. 2014, 16, 1057–1068.
  34. Kreuzaler, P.A.; Staniszewska, A.D.; Li, W.; Omidvar, N.; Kedjouar, B.; Turkson, J.; Poli, V.; Flavell, R.A.; Clarkson, R.W.; Watson, C.J. Stat3 controls lysosomal-mediated cell death in vivo. Nat. Cell Biol. 2011, 13, 303–309.
  35. Li, L.; Sun, B.; Gao, Y.; Niu, H.; Yuan, H.; Lou, H. STAT3 contributes to lysosomal-mediated cell death in a novel derivative of riccardin D-treated breast cancer cells in association with TFEB. Biochem. Pharmacol. 2018, 150, 267–279.
  36. de Duve, C. Lysosomes revisited. Eur. J. Biochem. 1983, 137, 391–397.
  37. Serrano-Puebla, A.; Boya, P. Lysosomal membrane permeabilization in cell death: New evidence and implications for health and disease. Ann. N. Y. Acad. Sci. 2016, 1371, 30–44.
  38. Meyer, N.; Henkel, L.; Linder, B.; Zielke, S.; Tascher, G.; Trautmann, S.; Geisslinger, G.; Münch, C.; Fulda, S.; Tegeder, I.; et al. Autophagy activation, lipotoxicity and lysosomal membrane permeabilization synergize to promote pimozide- and loperamide-induced glioma cell death. Autophagy 2020, 17, 3424–3443.
  39. Aits, S.; Jaattela, M. Lysosomal cell death at a glance. J. Cell Sci. 2013, 126, 1905–1912.
  40. Gomez-Sintes, R.; Ledesma, M.D.; Boya, P. Lysosomal cell death mechanisms in aging. Ageing Res. Rev. 2016, 32, 150–168.
  41. Stoka, V.; Turk, V.; Turk, B. Lysosomal cathepsins and their regulation in aging and neurodegeneration. Ageing Res. Rev. 2016, 32, 22–37.
  42. Kallunki, T.; Olsen, O.D.; Jaattela, M. Cancer-associated lysosomal changes: Friends or foes? Oncogene 2013, 32, 1995–2004.
  43. Piao, S.; Amaravadi, R.K. Targeting the lysosome in cancer. Ann. N. Y. Acad. Sci. 2016, 1371, 45–54.
  44. Levy, D.E.; Lee, C.K. What does Stat3 do? J. Clin. Investig. 2002, 109, 1143–1148.
  45. Liu, Y.; Li, C.; Lin, J. STAT3 as a therapeutic target for glioblastoma. Anti-Cancer Agents Med. Chem. 2010, 10, 512–519.
  46. Shi, Y.; Guryanova, O.A.; Zhou, W.; Liu, C.; Huang, Z.; Fang, X.; Wang, X.; Chen, C.; Wu, Q.; He, Z.; et al. Ibrutinib inactivates BMX-STAT3 in glioma stem cells to impair malignant growth and radioresistance. Sci. Transl. Med. 2018, 10, 600.
  47. Shchors, K.; Massaras, A.; Hanahan, D. Dual targeting of the autophagic regulatory circuitry in gliomas with repurposed drugs elicits cell-lethal autophagy and therapeutic benefit. Cancer Cell 2015, 28, 456–471.
  48. Meyer, N.; Zielke, S.; Michaelis, J.B.; Linder, B.; Warnsmann, V.; Rakel, S.; Osiewacz, H.D.; Fulda, S.; Mittelbronn, M.; Munch, C.; et al. AT 101 induces early mitochondrial dysfunction and HMOX1 (heme oxygenase 1) to trigger mitophagic cell death in glioma cells. Autophagy 2018, 14, 1693–1709.
  49. Voss, V.; Senft, C.; Lang, V.; Ronellenfitsch, M.W.; Steinbach, J.P.; Seifert, V.; Kogel, D. The pan-Bcl-2 inhibitor (−)-gossypol triggers autophagic cell death in malignant glioma. Mol. Cancer Res. 2010, 8, 1002–1016.
  50. Hernandez-Tiedra, S.; Fabrias, G.; Davila, D.; Salanueva, I.J.; Casas, J.; Montes, L.R.; Anton, Z.; Garcia-Taboada, E.; Salazar-Roa, M.; Lorente, M.; et al. Dihydroceramide accumulation mediates cytotoxic autophagy of cancer cells via autolysosome destabilization. Autophagy 2016, 12, 2213–2229.
  51. Salazar, M.; Carracedo, A.; Salanueva, I.J.; Hernandez-Tiedra, S.; Lorente, M.; Egia, A.; Vazquez, P.; Blazquez, C.; Torres, S.; Garcia, S.; et al. Cannabinoid action induces autophagy-mediated cell death through stimulation of ER stress in human glioma cells. J. Clin. Invest. 2009, 119, 1359–1372.
  52. Kornhuber, J.; Tripal, P.; Reichel, M.; Muhle, C.; Rhein, C.; Muehlbacher, M.; Groemer, T.W.; Gulbins, E. Functional inhibitors of acid sphingomyelinase (FIASMAs): A novel pharmacological group of drugs with broad clinical applications. Cell. Physiol. Biochem. 2010, 26, 9–20.
  53. Petersen, N.H.; Olsen, O.D.; Groth-Pedersen, L.; Ellegaard, A.M.; Bilgin, M.; Redmer, S.; Ostenfeld, M.S.; Ulanet, D.; Dovmark, T.H.; Lønborg, A.; et al. Transformation-associated changes in sphingolipid metabolism sensitize cells to lysosomal cell death induced by inhibitors of acid sphingomyelinase. Cancer Cell 2013, 24, 379–393.
  54. Boya, P.; Kroemer, G. Lysosomal membrane permeabilization in cell death. Oncogene 2008, 27, 6434–6451.
  55. Gabande-Rodriguez, E.; Boya, P.; Labrador, V.; Dotti, C.G.; Ledesma, M.D. High sphingomyelin levels induce lysosomal damage and autophagy dysfunction in Niemann Pick disease type A. Cell Death Differ. 2014, 21, 864–875.
  56. Wiklund, E.D.; Catts, V.S.; Catts, S.V.; Ng, T.F.; Whitaker, N.J.; Brown, A.J.; Lutze-Mann, L.H. Cytotoxic effects of antipsychotic drugs implicate cholesterol homeostasis as a novel chemotherapeutic target. Int. J. Cancer 2010, 126, 28–40.
  57. Kuzu, O.F.; Gowda, R.; Noory, M.A.; Robertson, G.P. Modulating cancer cell survival by targeting intracellular cholesterol transport. Br. J. Cancer 2017, 117, 513–524.
  58. Villa, G.R.; Hulce, J.J.; Zanca, C.; Bi, J.; Ikegami, S.; Cahill, G.L.; Gu, Y.; Lum, K.M.; Masui, K.; Yang, H.; et al. An LXR-cholesterol axis creates a metabolic co-dependency for brain cancers. Cancer Cell 2016, 30, 683–693.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 294
Revisions: 2 times (View History)
Update Date: 26 Jan 2022
1000/1000