2,956
Views
6
CrossRef citations to date
0
Altmetric
Research Paper

Identification and validation of an autophagy-related signature for predicting survival in lower-grade glioma

, , , , &
Pages 9692-9708 | Received 24 Jul 2021, Accepted 22 Sep 2021, Published online: 02 Dec 2021

References

  • Lopes MBS. The 2017 World Health Organization classification of tumors of the pituitary gland: a summary. Acta Neuropathol. 2017;134:521–535.
  • Brat DJ, Verhaak RG, Aldape KD, et al. Comprehensive, integrative genomic analysis of diffuse lower-grade gliomas. N Engl J Med. 2015;372:2481–2498.
  • Ostrom QT, Gittleman H, Xu J, et al. CBTRUS statistical report: primary brain and other central nervous system tumors diagnosed in the United States in 2009–2013. Neuro Oncol. 2016;18(suppl_5):v1–v75.
  • Claus EB, Walsh KM, Wiencke JK, et al. Survival and low-grade glioma: the emergence of genetic information. Neurosurg Focus. 2015;38:E6.
  • Weller M, van den Bent M, Tonn JC, et al. European Association for Neuro-Oncology (EANO) guideline on the diagnosis and treatment of adult astrocytic and oligodendroglial gliomas. Lancet Oncol. 2017;18:e315–e329.
  • Wongsurawat T, Jenjaroenpun P, De Loose A, et al. A novel Cas9-targeted long-read assay for simultaneous detection of IDH1/2 mutations and clinically relevant MGMT methylation in fresh biopsies of diffuse glioma. Acta Neuropathol Commun. 2020;8:87.
  • Paech D, Windschuh J, Oberhollenzer J, et al. Assessing the predictability of IDH mutation and MGMT methylation status in glioma patients using relaxation-compensated multipool CEST MRI at 7.0 T. Neuro Oncol. 2018;20:1661–1671.
  • Brigliadori G, Foca F, Dall’Agata M, et al. Defining the cutoff value of MGMT gene promoter methylation and its predictive capacity in glioblastoma. J Neurooncol. 2016;128:333–339.
  • Ceccarelli M, Barthel FP, Malta TM, et al. Molecular profiling reveals biologically discrete subsets and pathways of progression in diffuse glioma. Cell. 2016;164:550–563.
  • Lyamzaev KG, Tokarchuk AV, Panteleeva AA, et al. Induction of autophagy by depolarization of mitochondria. Autophagy. 2018;14:921–924.
  • Giampieri F, Afrin S, Forbes-Hernandez TY, et al. Autophagy in human health and disease: novel therapeutic opportunities. Antioxid Redox Signal. 2019;30:577–634.
  • Singh SS, Vats S, Chia AY, et al. Dual role of autophagy in hallmarks of cancer. Oncogene. 2018;37:1142–1158.
  • Sun Z, Jing C, Xiao C, et al. An autophagy-related long non-coding RNA prognostic signature accurately predicts survival outcomes in bladder urothelial carcinoma patients. Aging (Albany NY). 2020;12:15624–15637.
  • Levine B, Kroemer G. Biological functions of autophagy genes: a disease perspective. Cell. 2019;176:11–42.
  • Ariosa AR, Lahiri V, Lei Y, et al. A perspective on the role of autophagy in cancer. Biochim Biophys Acta Mol Basis Dis. 2021;166262.
  • White E. The role for autophagy in cancer. J Clin Invest. 2015;125:42–46.
  • Ulasov IV, Lenz G, Lesniak MS. Autophagy in glioma cells: an identity crisis with a clinical perspective. Cancer Lett. 2018;428:139–146.
  • Baumert BG, Hegi ME, van den Bent MJ, et al. Temozolomide chemotherapy versus radiotherapy in high-risk low-grade glioma (EORTC 22033-26033): a randomised, open-label, phase 3 intergroup study. Lancet Oncol. 2016;17:1521–1532.
  • How CW, Ong YS, Low SS, et al. How far have we explored fungi to fight cancer? Semin Cancer Biol. 2021. DOI:10.1016/j.semcancer.2021.03.009
  • Tomczak K, Czerwińska P, Wiznerowicz M. The Cancer Genome Atlas (TCGA): an immeasurable source of knowledge. Contemp Oncol (Pozn). 2015;19:A68–77.
  • GTEx Consortium. The GTEx Consortium atlas of genetic regulatory effects across human tissues. Science. 2020;369:1318–1330.
  • Ritchie ME, Phipson B, Wu D, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43:e47.
  • Tu Z, Wu L, Wang P, et al. N6-methylandenosine-related lncRNAs are potential biomarkers for predicting the overall survival of lower-grade glioma patients. Front Cell Dev Biol. 2020;8:642.
  • Galluzzi L, Bravo-San Pedro JM, Levine B, et al. Pharmacological modulation of autophagy: therapeutic potential and persisting obstacles. Nat Rev Drug Discov. 2017;16:487–511.
  • Hazlewood JE, Dumenil T, Le TT, et al. Injection site vaccinology of a recombinant vaccinia-based vector reveals diverse innate immune signatures. PLoS Pathog. 2021;17:e1009215–e1009215.
  • Seiler M, Huang CC, Szalma S, et al. ConsensusCluster: a software tool for unsupervised cluster discovery in numerical data. Omics. 2010;14:109–113.
  • Yang L, Liu L, Gao H, et al. The stem cell factor SALL4 is an essential transcriptional regulator in mixed lineage leukemia-rearranged leukemogenesis. J Hematol Oncol. 2017;10:159.
  • Szklarczyk D, Gable AL, Lyon D, et al. STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 2019;47:D607–d613.
  • Zhu X, Luo H, Xu Y. Transcriptome analysis reveals an important candidate gene involved in both nodal metastasis and prognosis in lung adenocarcinoma. Cell Biosci. 2019;9:92.
  • Best MG, Sol N, Vancura A, et al. Swarm intelligence-enhanced detection of non-small-cell lung cancer using tumor-educated platelets. Cancer Cell. 2017;32:238–252.e239.
  • Rybinska I, Sandri M, Bianchi F, et al. Extracellular matrix features discriminate aggressive HER2-positive breast cancer patients who benefit from trastuzumab treatment. Cells. 2020;9:434.
  • Pontén F, Jirström K, Uhlen M. The human protein atlas–a tool for pathology. J Pathol. 2008;216:387–393.
  • Liu B, Liu J, Liu K, et al. A prognostic signature of five pseudogenes for predicting lower-grade gliomas. Biomed Pharmacother. 2019;117:109116.
  • Yu F, Asghar S, Zhang M, et al. Local strategies and delivery systems for the treatment of malignant gliomas. J Drug Target. 2019;27:367–378.
  • Amaravadi R, Kimmelman AC, White E. Recent insights into the function of autophagy in cancer. Genes Dev. 2016;30:1913–1930.
  • Amaravadi RK, Kimmelman AC, Debnath J. Targeting autophagy in cancer: recent advances and future directions. Cancer Discov. 2019;9:1167–1181.
  • Zheng J, Wang B, Zheng R, et al. Linc-RA1 inhibits autophagy and promotes radioresistance by preventing H2Bub1/USP44 combination in glioma cells. Cell Death Dis. 2020;11:758.
  • Song S, Long M, Yu G, et al. Urinary exosome miR-30c-5p as a biomarker of clear cell renal cell carcinoma that inhibits progression by targeting HSPA5. J Cell Mol Med. 2019;23:6755–6765.
  • Care C, Sornjai W, Jaratsittisin J, et al. Discordant activity of kaempferol towards dengue virus and japanese encephalitis virus. Molecules. 2020;25:1246.
  • Jia Z-Y, Shen T-Y, Jiang W, et al. Identification of molecular mechanisms of glutamine in pancreatic cancer. Oncol Lett. 2017;14:6395–6402.
  • Kim SY, Kim HJ, Kim HJ, et al. HSPA5 negatively regulates lysosomal activity through ubiquitination of MUL1 in head and neck cancer. Autophagy. 2018;14:385–403.
  • Cerezo M, Rocchi S. New anti-cancer molecules targeting HSPA5/BIP to induce endoplasmic reticulum stress, autophagy and apoptosis. Autophagy. 2017;13:216–217.
  • Zhang J, Jiang Y, Jia Z, et al. Association of elevated GRP78 expression with increased lymph node metastasis and poor prognosis in patients with gastric cancer. Clin Exp Metastasis. 2006;23:401–410.
  • Wei PL, Kuo LJ, Wang W, et al. Silencing of glucose-regulated protein 78 (GRP78) enhances cell migration through the upregulation of vimentin in hepatocellular carcinoma cells. Ann Surg Oncol. 2012;19(Suppl 3):S572–579.
  • Raiter A, Vilkin A, Gingold R, et al. The presence of anti-GRP78 antibodies in the serum of patients with colorectal carcinoma: a potential biomarker for early cancer detection. Int J Biol Markers. 2014;29:e431–435.
  • Birts CN, Banerjee A, Darley M, et al. p53 is regulated by aerobic glycolysis in cancer cells by the CtBP family of NADH-dependent transcriptional regulators. Sci Signal. 2020;13:eaau9529.
  • Cordani M, Butera G, Pacchiana R, et al. Molecular interplay between mutant p53 proteins and autophagy in cancer cells. Biochim Biophys Acta Rev Cancer. 2017;1867:19–28.
  • Yang A, Rajeshkumar NV, Wang X, et al. Autophagy is critical for pancreatic tumor growth and progression in tumors with p53 alterations. Cancer Discov. 2014;4:905–913.
  • Mori T, Kitani Y, Hatakeyama D, et al. Predation threats for a 24-h period activated the extension of axons in the brains of Xenopus tadpoles. Sci Rep. 2020;10:11737.
  • Knight JRP, Garland G, Pöyry T, et al. Control of translation elongation in health and disease. Dis Model Mech. 2020;13.
  • De Gassart A, Demaria O, Panes R, et al. Pharmacological eEF2K activation promotes cell death and inhibits cancer progression. EMBO Rep. 2016;17:1471–1484.
  • Ye J, Zou G, Zhu R, et al. Structural basis of GABARAP-mediated GABA(A) receptor trafficking and functions on GABAergic synaptic transmission. Nat Commun. 2021;12:297.
  • Joachim J, Jefferies HB, Razi M, et al. Activation of ULK kinase and autophagy by GABARAP trafficking from the centrosome is regulated by WAC and GM130. Mol Cell. 2015;60:899–913.
  • Klebig C, Seitz S, Arnold W, et al. Characterization of {gamma}-aminobutyric acid type A receptor-associated protein, a novel tumor suppressor, showing reduced expression in breast cancer. Cancer Res. 2005;65:394–400.
  • Greig FH, Nixon GF. Phosphoprotein enriched in astrocytes (PEA)-15: a potential therapeutic target in multiple disease states. Pharmacol Ther. 2014;143:265–274.
  • Bartholomeusz C, Rosen D, Wei C, et al. PEA-15 induces autophagy in human ovarian cancer cells and is associated with prolonged overall survival. Cancer Res. 2008;68:9302–9310.
  • McCullough J, Clippinger AK, Talledge N, et al. Structure and membrane remodeling activity of ESCRT-III helical polymers. Science. 2015;350:1548–1551.
  • Zheng K, He Z, Kitazato K, et al. Selective autophagy regulates cell cycle in cancer therapy. Theranostics. 2019;9:104–125.
  • Groner B, von Manstein V. Jak Stat signaling and cancer: opportunities, benefits and side effects of targeted inhibition. Mol Cell Endocrinol. 2017;451:1–14.
  • Wagner EF, Nebreda AR. Signal integration by JNK and p38 MAPK pathways in cancer development. Nat Rev Cancer. 2009;9:537–549.