The Potential Links between lncRNAs and Drug Tolerance in Lung Adenocarcinoma
Abstract
:1. Introduction
2. Long Noncoding RNAs
3. Drug Tolerance
3.1. Epigenetic Reprogramming
3.2. Slow Cycling Phenotype
3.3. Regulation of a SASP-like Phenotype
3.4. Stem and Developmental Cell-like Features in DTPs
Developmental Lung Genes Co-Ordinated by lncRNAs
3.5. Metabolic Reprogramming
3.6. Adaptive Mutability
4. Therapeutic Challenges and Potential
5. Conclusions
Author Contributions
Funding
Data Availability Statement
Conflicts of Interest
References
- Rotow, J.; Bivona, T.G. Understanding and targeting resistance mechanisms in NSCLC. Nat. Rev. Cancer 2017, 17, 637–658. [Google Scholar] [CrossRef] [PubMed]
- Pu, Y.; Li, L.; Peng, H.; Liu, L.; Heymann, D.; Robert, C.; Vallette, F.; Shen, S. Drug-tolerant persister cells in cancer: The cutting edges and future directions. Nat. Rev. Clin. Oncol. 2023, 20, 799–813. [Google Scholar] [CrossRef] [PubMed]
- Sharma, S.V.; Lee, D.Y.; Li, B.; Quinlan, M.P.; Takahashi, F.; Maheswaran, S.; McDermott, U.; Azizian, N.; Zou, L.; Fischbach, M.A.; et al. A chromatin-mediated reversible drug-tolerant state in cancer cell subpopulations. Cell 2010, 141, 69–80. [Google Scholar] [CrossRef] [PubMed]
- Madhusoodanan, J. How persister bacteria evade antibiotics, prolong infections. Proc. Natl. Acad. Sci. USA 2022, 119, e2215617119. [Google Scholar] [CrossRef] [PubMed]
- Oren, Y.; Tsabar, M.; Cuoco, M.S.; Amir-Zilberstein, L.; Cabanos, H.F.; Hütter, J.-C.; Hu, B.; Thakore, P.I.; Tabaka, M.; Fulco, C.P.; et al. Cycling cancer persister cells arise from lineages with distinct programs. Nature 2021, 596, 576–582. [Google Scholar] [CrossRef] [PubMed]
- Russo, M.; Crisafulli, G.; Sogari, A.; Reilly, N.M.; Arena, S.; Lamba, S.; Bartolini, A.; Amodio, V.; Magrì, A.; Novara, L.; et al. Adaptive mutability of colorectal cancers in response to targeted therapies. Science 2019, 366, 1473–1480. [Google Scholar] [CrossRef]
- Noronha, A.; Nataraj, N.B.; Lee, J.S.; Zhitomirsky, B.; Oren, Y.; Oster, S.; Lindzen, M.; Mukherjee, S.; Will, R.; Ghosh, S.; et al. AXL and error-prone DNA replication confer drug resistance and offer strategies to treat EGFR-mutant lung cancer. Cancer Discov. 2022, 12, 2666–2683. [Google Scholar] [CrossRef] [PubMed]
- Rambow, F.; Marine, J.C.; Goding, C.R. Melanoma plasticity and phenotypic diversity: Therapeutic barriers and opportunities. Genes Dev. 2019, 33, 1295–1318. [Google Scholar] [CrossRef] [PubMed]
- Chang, C.A.; Jen, J.; Jiang, S.; Sayad, A.; Mer, A.S.; Brown, K.R.; Nixon, A.M.; Dhabaria, A.; Tang, K.H.; Venet, D.; et al. Ontogeny and Vulnerabilities of Drug-Tolerant Persisters in HER2+ Breast Cancer. Cancer Discov. 2022, 12, 1022–1045. [Google Scholar] [CrossRef]
- Liau, B.B.; Sievers, C.; Donohue, L.K.; Gillespie, S.M.; Flavahan, W.A.; Miller, T.E.; Venteicher, A.S.; Hebert, C.H.; Carey, C.D.; Rodig, S.J.; et al. Adaptive Chromatin Remodeling Drives Glioblastoma Stem Cell Plasticity and Drug Tolerance. Cell Stem Cell 2017, 20, 233–246.e7. [Google Scholar] [CrossRef]
- Maynard, A.; McCoach, C.E.; Rotow, J.K.; Harris, L.; Haderk, F.; Kerr, D.L.; Yu, E.A.; Schenk, E.L.; Tan, W.; Zee, A.; et al. Therapy-Induced Evolution of Human Lung Cancer Revealed by Single-Cell RNA Sequencing. Cell 2020, 182, 1232–1251.e22. [Google Scholar] [CrossRef] [PubMed]
- Aissa, A.F.; Islam, A.B.M.M.K.; Ariss, M.M.; Go, C.C.; Rader, A.E.; Conrardy, R.D.; Gajda, A.M.; Rubio-Perez, C.; Valyi-Nagy, K.; Pasquinelli, M.; et al. Single-cell transcriptional changes associated with drug tolerance and response to combination therapies in cancer. Nat. Commun. 2021, 12, 1628. [Google Scholar] [CrossRef] [PubMed]
- Mikubo, M.; Inoue, Y.; Liu, G.; Tsao, M.-S. Mechanism of Drug Tolerant Persister Cancer Cells: The Landscape and Clinical Implication for Therapy. J. Thorac. Oncol. 2021, 16, 1798–1809. [Google Scholar] [CrossRef] [PubMed]
- Mattick, J.S.; Amaral, P.P.; Carninci, P.; Carpenter, S.; Chang, H.Y.; Chen, L.-L.; Chen, R.; Dean, C.; Dinger, M.E.; Fitzgerald, K.A.; et al. Long non-coding RNAs: Definitions, functions, challenges and recommendations. Nat. Rev. Mol. Cell Biol. 2023, 24, 430–447. [Google Scholar] [CrossRef] [PubMed]
- Statello, L.; Guo, C.J.; Chen, L.L.; Huarte, M. Gene regulation by long non-coding RNAs and its biological functions. Nat. Rev. Mol. Cell Biol. 2021, 22, 96–118. [Google Scholar] [CrossRef]
- Morris, K.V.; Mattick, J.S. The rise of regulatory RNA. Nat. Rev. Genet. 2014, 15, 423–437. [Google Scholar] [CrossRef] [PubMed]
- van Bakel, H.; Nislow, C.; Blencowe, B.J.; Hughes, T.R. Most “Dark Matter” Transcripts Are Associated with Known Genes. PLoS Biol. 2010, 8, e1000371. [Google Scholar] [CrossRef]
- Anastasiadou, E.; Jacob, L.S.; Slack, F.J. Non-coding RNA networks in cancer. Nat. Rev. Cancer 2018, 18, 5–18. [Google Scholar] [CrossRef] [PubMed]
- Gencel-Augusto, J.; Wu, W.; Bivona, T.G. Long Non-Coding RNAs as Emerging Targets in Lung Cancer. Cancers 2023, 15, 3135. [Google Scholar] [CrossRef]
- Fatica, A.; Bozzoni, I. Long non-coding RNAs: New players in cell differentiation and development. Nat. Rev. Genet. 2014, 15, 7–21. [Google Scholar] [CrossRef]
- Kitagawa, M.; Kitagawa, K.; Kotake, Y.; Niida, H.; Ohhata, T. Cell cycle regulation by long non-coding RNAs. Cell. Mol. Life Sci. 2013, 70, 4785–4794. [Google Scholar] [CrossRef] [PubMed]
- Ghafouri-Fard, S.; Shoorei, H.; Anamag, F.T.; Taheri, M. The Role of Non-Coding RNAs in Controlling Cell Cycle Related Proteins in Cancer Cells. Front. Oncol. 2020, 10, 608975. [Google Scholar] [CrossRef] [PubMed]
- Mercer, T.R.; Mattick, J.S. Structure and function of long noncoding RNAs in epigenetic regulation. Nat. Struct. Mol. Biol. 2013, 20, 300–307. [Google Scholar] [CrossRef] [PubMed]
- Chowdhury, D.; Choi, Y.E.; Brault, M.E. Charity begins at home: Non-coding RNA functions in DNA repair. Nat. Rev. Mol. Cell Biol. 2013, 14, 181–189. [Google Scholar] [CrossRef] [PubMed]
- Taiana, E.; Ronchetti, D.; Todoerti, K.; Nobili, L.; Tassone, P.; Amodio, N.; Neri, A. LncRNA NEAT1 in Paraspeckles: A Structural Scaffold for Cellular DNA Damage Response Systems? Noncoding RNA 2020, 6, 26. [Google Scholar] [CrossRef] [PubMed]
- Guler, G.D.; Tindell, C.A.; Pitti, R.; Wilson, C.; Nichols, K.; Cheung, T.K.; Kim, H.-J.; Wongchenko, M.; Yan, Y.; Haley, B.; et al. Repression of Stress-Induced LINE-1 Expression Protects Cancer Cell Subpopulations from Lethal Drug Exposure. Cancer Cell 2017, 32, 221–237.e13. [Google Scholar] [CrossRef] [PubMed]
- Vinogradova, M.; Gehling, V.S.; Gustafson, A.; Arora, S.; A Tindell, C.; Wilson, C.; E Williamson, K.; Guler, G.D.; Gangurde, P.; Manieri, W.; et al. An inhibitor of KDM5 demethylases reduces survival of drug-tolerant cancer cells. Nat. Chem. Biol. 2016, 12, 531–538. [Google Scholar] [CrossRef] [PubMed]
- Gao, Y.; Huang, P.; Zhang, J. Hypermethylation of MEG3 promoter correlates with inactivation of MEG3 and poor prognosis in patients with retinoblastoma. J. Transl. Med. 2017, 15, 268. [Google Scholar] [CrossRef] [PubMed]
- Yang, Z.; Wang, Z.; Duan, Y. LncRNA MEG3 inhibits non-small cell lung cancer via interaction with DKC1 protein. Oncol. Lett. 2020, 20, 2183–2190. [Google Scholar] [CrossRef]
- Schmitt, A.M.; Chang, H.Y. Long Noncoding RNAs in Cancer Pathways. Cancer Cell 2016, 29, 452–463. [Google Scholar] [CrossRef]
- Zhou, Y.; Zhong, Y.; Wang, Y.; Zhang, X.; Batista, D.L.; Gejman, R.; Ansell, P.J.; Zhao, J.; Weng, C.; Klibanski, A. Activation of p53 by MEG3 non-coding RNA. J. Biol. Chem. 2007, 282, 24731–24742. [Google Scholar] [CrossRef] [PubMed]
- Uroda, T.; Anastasakou, E.; Rossi, A.; Teulon, J.-M.; Pellequer, J.-L.; Annibale, P.; Pessey, O.; Inga, A.; Chillón, I.; Marcia, M. Conserved Pseudoknots in lncRNA MEG3 Are Essential for Stimulation of the p53 Pathway. Mol. Cell 2019, 75, 982–995.e9. [Google Scholar] [CrossRef] [PubMed]
- Li, X.; Fu, X.-D. Chromatin-associated RNAs as facilitators of functional genomic interactions. Nat. Rev. Genet. 2019, 20, 503–519. [Google Scholar] [CrossRef] [PubMed]
- Limouse, C.; Smith, O.K.; Jukam, D.; Fryer, K.A.; Greenleaf, W.J.; Straight, A.F. Global mapping of RNA-chromatin contacts reveals a proximity-dominated connectivity model for ncRNA-gene interactions. Nat. Commun. 2023, 14, 6073. [Google Scholar] [CrossRef]
- Dueva, R.; Akopyan, K.; Pederiva, C.; Trevisan, D.; Dhanjal, S.; Lindqvist, A.; Farnebo, M. Neutralization of the Positive Charges on Histone Tails by RNA Promotes an Open Chromatin Structure. Cell Chem. Biol. 2019, 26, 1436–1449.e5. [Google Scholar] [CrossRef]
- Chu, C.; Qu, K.; Zhong, F.L.; Artandi, S.E.; Chang, H.Y. Genomic maps of long noncoding RNA occupancy reveal principles of RNA-chromatin interactions. Mol. Cell 2011, 44, 667–678. [Google Scholar] [CrossRef]
- Oh, H.J.; Aguilar, R.; Kesner, B.; Lee, H.-G.; Kriz, A.J.; Chu, H.-P.; Lee, J.T. Jpx RNA regulates CTCF anchor site selection and formation of chromosome loops. Cell 2021, 184, 6157–6173.e24. [Google Scholar] [CrossRef] [PubMed]
- Huang, W.; Li, H.; Yu, Q.; Xiao, W.; Wang, D.O. LncRNA-mediated DNA methylation: An emerging mechanism in cancer and beyond. J. Exp. Clin. Cancer Res. 2022, 41, 100. [Google Scholar] [CrossRef] [PubMed]
- Merry, C.R.; Forrest, M.E.; Sabers, J.N.; Beard, L.; Gao, X.-H.; Hatzoglou, M.; Jackson, M.W.; Wang, Z.; Markowitz, S.D.; Khalil, A.M. DNMT1-associated long non-coding RNAs regulate global gene expression and DNA methylation in colon cancer. Hum. Mol. Genet. 2015, 24, 6240–6253. [Google Scholar] [CrossRef]
- Long, Y.; Wang, X.; Youmans, D.T.; Cech, T.R. How do lncRNAs regulate transcription? Sci. Adv. 2017, 3, eaao2110. [Google Scholar] [CrossRef]
- Bose, D.A.; Donahue, G.; Reinberg, D.; Shiekhattar, R.; Bonasio, R.; Berger, S.L. RNA Binding to CBP Stimulates Histone Acetylation and Transcription. Cell 2017, 168, 135–149.e22. [Google Scholar] [CrossRef]
- Piunti, A.; Shilatifard, A. The roles of Polycomb repressive complexes in mammalian development and cancer. Nat. Rev. Mol. Cell Biol. 2021, 22, 326–345. [Google Scholar] [CrossRef] [PubMed]
- Khalil, A.M.; Guttman, M.; Huarte, M.; Garber, M.; Raj, A.; Morales, D.R.; Thomas, K.; Presser, A.; Bernstein, B.E.; van Oudenaarden, A.; et al. Many human large intergenic noncoding RNAs associate with chromatin-modifying complexes and affect gene expression. Proc. Natl. Acad. Sci. USA 2009, 106, 11667–11672. [Google Scholar] [CrossRef]
- Davidovich, C.; Zheng, L.; Goodrich, K.J.; Cech, T.R. Promiscuous RNA binding by Polycomb repressive complex 2. Nat. Struct. Mol. Biol. 2013, 20, 1250–1257. [Google Scholar] [CrossRef]
- Long, Y.; Hwang, T.; Gooding, A.R.; Goodrich, K.J.; Rinn, J.L.; Cech, T.R. RNA is essential for PRC2 chromatin occupancy and function in human pluripotent stem cells. Nat. Genet. 2020, 52, 931–938. [Google Scholar] [CrossRef]
- Rinn, J.L.; Kertesz, M.; Wang, J.K.; Squazzo, S.L.; Xu, X.; Brugmann, S.A.; Goodnough, L.H.; Helms, J.A.; Farnham, P.J.; Segal, E.; et al. Functional demarcation of active and silent chromatin domains in human HOX loci by noncoding RNAs. Cell 2007, 129, 1311–1323. [Google Scholar] [CrossRef] [PubMed]
- De Kumar, B.; Krumlauf, R. HOXs and lincRNAs: Two sides of the same coin. Sci. Adv. 2016, 2, e1501402. [Google Scholar] [CrossRef] [PubMed]
- Shah, N.; Sukumar, S. The HOX genes and their roles in oncogenesis. Nat. Rev. Cancer 2010, 10, 361–371. [Google Scholar] [CrossRef]
- Li, L.; Zhang, X.; Liu, Q.; Yin, H.; Diao, Y.; Zhang, Z.; Wang, Y.; Gao, Y.; Ren, X.; Li, J.; et al. Emerging role of HOX genes and their related long noncoding RNAs in lung cancer. Crit. Rev. Oncol. Hematol. 2019, 139, 1–6. [Google Scholar] [CrossRef]
- Steens, J.; Klein, D. HOX genes in stem cells: Maintaining cellular identity and regulation of differentiation. Front. Cell Dev. Biol. 2022, 10, 1002909. [Google Scholar] [CrossRef]
- Battistelli, C.; Garbo, S.; Riccioni, V.; Montaldo, C.; Santangelo, L.; Vandelli, A.; Strippoli, R.; Tartaglia, G.G.; Tripodi, M.; Cicchini, C. Design and Functional Validation of a Mutant Variant of the LncRNA HOTAIR to Counteract Snail Function in Epithelial-to-Mesenchymal Transition. Cancer Res. 2021, 81, 103–113. [Google Scholar] [CrossRef] [PubMed]
- Gupta, R.A.; Shah, N.; Wang, K.C.; Kim, J.; Horlings, H.M.; Wong, D.J.; Tsai, M.-C.; Hung, T.; Argani, P.; Rinn, J.L.; et al. Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature 2010, 464, 1071–1076. [Google Scholar] [CrossRef]
- Hajjari, M.; Salavaty, A. HOTAIR: An oncogenic long non-coding RNA in different cancers. Cancer Biol. Med. 2015, 12, 1–9. [Google Scholar] [CrossRef] [PubMed]
- Tsai, M.-C.; Manor, O.; Wan, Y.; Mosammaparast, N.; Wang, J.K.; Lan, F.; Shi, Y.; Segal, E.; Chang, H.Y. Long noncoding RNA as modular scaffold of histone modification complexes. Science 2010, 329, 689–693. [Google Scholar] [CrossRef] [PubMed]
- Jarroux, J.; Foretek, D.; Bertrand, C.; Gabriel, M.; Szachnowski, U.; Saci, Z.; Guo, S.; Londoño-Vallejo, A.; Pinskaya, M.; Morillon, A. HOTAIR lncRNA promotes epithelial-mesenchymal transition by redistributing LSD1 at regulatory chromatin regions. EMBO Rep. 2021, 22, e50193. [Google Scholar] [CrossRef]
- Liu, X.H.; Liu, Z.L.; Sun, M.; Liu, J.; Wang, Z.X.; De, W. The long non-coding RNA HOTAIR indicates a poor prognosis and promotes metastasis in non-small cell lung cancer. BMC Cancer 2013, 13, 464. [Google Scholar] [CrossRef] [PubMed]
- Liu, M.Y.; Li, X.Q.; Gao, T.H.; Cui, Y.; Ma, N.; Zhou, Y.; Zhang, G.J. Elevated HOTAIR expression associated with cisplatin resistance in non-small cell lung cancer patients. J. Thorac. Dis. 2016, 8, 3314–3322. [Google Scholar] [CrossRef] [PubMed]
- Wang, K.C.; Yang, Y.W.; Liu, B.; Sanyal, A.; Corces-Zimmerman, R.; Chen, Y.; Lajoie, B.R.; Protacio, A.; Flynn, R.A.; Gupta, R.A.; et al. A long noncoding RNA maintains active chromatin to coordinate homeotic gene expression. Nature 2011, 472, 120–124. [Google Scholar] [CrossRef] [PubMed]
- Yu, H.; Xu, Q.; Liu, F.; Ye, X.; Wang, J.; Meng, X. Identification and validation of long noncoding RNA biomarkers in human non-small-cell lung carcinomas. J. Thorac. Oncol. 2015, 10, 645–654. [Google Scholar] [CrossRef]
- Navarro, A.; Moises, J.; Santasusagna, S.; Marrades, R.M.; Viñolas, N.; Castellano, J.J.; Canals, J.; Muñoz, C.; Ramírez, J.; Molins, L.; et al. Clinical significance of long non-coding RNA HOTTIP in early-stage non-small-cell lung cancer. BMC Pulm. Med. 2019, 19, 55. [Google Scholar] [CrossRef]
- Hata, A.N.; Niederst, M.J.; Archibald, H.L.; Gomez-Caraballo, M.; Siddiqui, F.M.; Mulvey, H.E.; Maruvka, Y.E.; Ji, F.; Bhang, H.-E.C.; Radhakrishna, V.K.; et al. Tumor cells can follow distinct evolutionary paths to become resistant to epidermal growth factor receptor inhibition. Nat. Med. 2016, 22, 262–269. [Google Scholar] [CrossRef] [PubMed]
- Balaban, N.Q.; Merrin, J.; Chait, R.; Kowalik, L.; Leibler, S. Bacterial persistence as a phenotypic switch. Science 2004, 305, 1622–1625. [Google Scholar] [CrossRef]
- Harms, A.; Maisonneuve, E.; Gerdes, K. Mechanisms of bacterial persistence during stress and antibiotic exposure. Science 2016, 354, aaf4268. [Google Scholar] [CrossRef]
- Tripathi, V.; Shen, Z.; Chakraborty, A.; Giri, S.; Freier, S.M.; Wu, X.; Zhang, Y.; Gorospe, M.; Prasanth, S.G.; Lal, A.; et al. Long noncoding RNA MALAT1 controls cell cycle progression by regulating the expression of oncogenic transcription factor B-MYB. PLoS Genet. 2013, 9, e1003368. [Google Scholar] [CrossRef]
- Marín-Béjar, O.; Marchese, F.P.; Athie, A.; Sánchez, Y.; González, J.; Segura, V.; Huang, L.; Moreno, I.; Navarro, A.; Monzó, M.; et al. Pint lincRNA connects the p53 pathway with epigenetic silencing by the Polycomb repressive complex 2. Genome Biol. 2013, 14, R104. [Google Scholar] [CrossRef]
- Xu, Y.; Wang, H.; Li, F.; Heindl, L.M.; He, X.; Yu, J.; Yang, J.; Ge, S.; Ruan, J.; Jia, R.; et al. Long Non-coding RNA LINC-PINT Suppresses Cell Proliferation and Migration of Melanoma via Recruiting EZH2. Front. Cell Dev. Biol. 2019, 7, 350. [Google Scholar] [CrossRef] [PubMed]
- Marín-Béjar, O.; Mas, A.M.; González, J.; Martinez, D.; Athie, A.; Morales, X.; Galduroz, M.; Raimondi, I.; Grossi, E.; Guo, S.; et al. The human lncRNA LINC-PINT inhibits tumor cell invasion through a highly conserved sequence element. Genome Biol. 2017, 18, 202. [Google Scholar] [CrossRef] [PubMed]
- Brown, N.R.; Korolchuk, S.; Martin, M.P.; Stanley, W.A.; Moukhametzianov, R.; Noble, M.E.M.; Endicott, J.A. CDK1 structures reveal conserved and unique features of the essential cell cycle CDK. Nat. Commun. 2015, 6, 6769. [Google Scholar] [CrossRef]
- Zhang, E.-B.; Yin, D.-D.; Sun, M.; Kong, R.; Liu, X.-H.; You, L.-H.; Han, L.; Xia, R.; Wang, K.-M.; Yang, J.-S.; et al. P53-regulated long non-coding RNA TUG1 affects cell proliferation in human non-small cell lung cancer, partly through epigenetically regulating HOXB7 expression. Cell Death Dis. 2014, 5, e1243. [Google Scholar] [CrossRef]
- Zhang, E.; He, X.; Yin, D.; Han, L.; Qiu, M.; Xu, T.; Xia, R.; Xu, L.; Yin, R.; De, W. Increased expression of long noncoding RNA TUG1 predicts a poor prognosis of gastric cancer and regulates cell proliferation by epigenetically silencing of p57. Cell Death Dis. 2016, 7, e2109. [Google Scholar] [CrossRef]
- Niu, Y.; Ma, F.; Huang, W.; Fang, S.; Li, M.; Wei, T.; Guo, L. Long non-coding RNA TUG1 is involved in cell growth and chemoresistance of small cell lung cancer by regulating LIMK2b via EZH2. Mol. Cancer 2017, 16, 5. [Google Scholar] [CrossRef] [PubMed]
- Liao, W.-T.; Jiang, D.; Yuan, J.; Cui, Y.-M.; Shi, X.-W.; Chen, C.-M.; Bian, X.-W.; Deng, Y.-J.; Ding, Y.-Q. HOXB7 as a Prognostic Factor and Mediator of Colorectal Cancer Progression. Clin. Cancer Res. 2011, 17, 3569–3578. [Google Scholar] [CrossRef] [PubMed]
- Kunimasa, K.; Nagano, T.; Shimono, Y.; Dokuni, R.; Kiriu, T.; Tokunaga, S.; Tamura, D.; Yamamoto, M.; Tachihara, M.; Kobayashi, K.; et al. Glucose metabolism-targeted therapy and withaferin A are effective for epidermal growth factor receptor tyrosine kinase inhibitor-induced drug-tolerant persisters. Cancer Sci. 2017, 108, 1368–1377. [Google Scholar] [CrossRef] [PubMed]
- Kurppa, K.J.; Liu, Y.; To, C.; Zhang, T.; Fan, M.; Vajdi, A.; Knelson, E.H.; Xie, Y.; Lim, K.; Cejas, P.; et al. Treatment-Induced Tumor Dormancy through YAP-Mediated Transcriptional Reprogramming of the Apoptotic Pathway. Cancer Cell 2020, 37, 104–122.e12. [Google Scholar] [CrossRef] [PubMed]
- Leonce, C.; Saintigny, P.; Ortiz-Cuaran, S. Cell-Intrinsic Mechanisms of Drug Tolerance to Systemic Therapies in Cancer. Mol. Cancer Res. 2022, 20, 11–29. [Google Scholar] [CrossRef] [PubMed]
- Montes, M.; Nielsen, M.M.; Maglieri, G.; Jacobsen, A.; Højfeldt, J.; Agrawal-Singh, S.; Hansen, K.; Helin, K.; van de Werken, H.J.G.; Pedersen, J.S.; et al. The lncRNA MIR31HG regulates p16INK4A expression to modulate senescence. Nat. Commun. 2015, 6, 6967. [Google Scholar] [CrossRef] [PubMed]
- Montes, M.; Lubas, M.; Arendrup, F.S.; Mentz, B.; Rohatgi, N.; Tumas, S.; Harder, L.M.; Skanderup, A.J.; Andersen, J.S.; Lund, A.H. The long non-coding RNA MIR31HG regulates the senescence associated secretory phenotype. Nat. Commun. 2021, 12, 2459. [Google Scholar] [CrossRef] [PubMed]
- Cipponi, A.; Goode, D.L.; Bedo, J.; McCabe, M.J.; Pajic, M.; Croucher, D.R.; Rajal, A.G.; Junankar, S.R.; Saunders, D.N.; Lobachevsky, P.; et al. MTOR signaling orchestrates stress-induced mutagenesis, facilitating adaptive evolution in cancer. Science 2020, 368, 1127–1131. [Google Scholar] [CrossRef] [PubMed]
- Faget, D.V.; Ren, Q.; Stewart, S.A. Unmasking senescence: Context-dependent effects of SASP in cancer. Nat. Rev. Cancer 2019, 19, 439–453. [Google Scholar] [CrossRef] [PubMed]
- Yap, K.L.; Li, S.; Muñoz-Cabello, A.M.; Raguz, S.; Zeng, L.; Mujtaba, S.; Gil, J.; Walsh, M.J.; Zhou, M.-M. Molecular interplay of the noncoding RNA ANRIL and methylated histone H3 lysine 27 by polycomb CBX7 in transcriptional silencing of INK4a. Mol. Cell 2010, 38, 662–674. [Google Scholar] [CrossRef]
- Muniz, L.; Lazorthes, S.; Delmas, M.; Ouvrard, J.; Aguirrebengoa, M.; Trouche, D.; Nicolas, E. Circular ANRIL isoforms switch from repressors to activators of p15/CDKN2B expression during RAF1 oncogene-induced senescence. RNA Biol. 2021, 18, 404–420. [Google Scholar] [CrossRef] [PubMed]
- Yu, W.; Gius, D.; Onyango, P.; Muldoon-Jacobs, K.; Karp, J.; Feinberg, A.P.; Cui, H. Epigenetic silencing of tumour suppressor gene p15 by its antisense RNA. Nature 2008, 451, 202–206. [Google Scholar] [CrossRef] [PubMed]
- Pasmant, E.; Laurendeau, I.; Héron, D.; Vidaud, M.; Vidaud, D.; Bièche, I. Characterization of a germ-line deletion, including the entire INK4/ARF locus, in a melanoma-neural system tumor family: Identification of ANRIL, an antisense noncoding RNA whose expression coclusters with ARF. Cancer Res. 2007, 67, 3963–3969. [Google Scholar] [CrossRef] [PubMed]
- Kotake, Y.; Nakagawa, T.; Kitagawa, K.; Suzuki, S.; Liu, N.; Kitagawa, M.; Xiong, Y. Long non-coding RNA ANRIL is required for the PRC2 recruitment to and silencing of p15(INK4B) tumor suppressor gene. Oncogene 2011, 30, 1956–1962. [Google Scholar] [CrossRef] [PubMed]
- Kong, Y.; Hsieh, C.H.; Alonso, L.C. ANRIL: A lncRNA at the CDKN2A/B Locus with Roles in Cancer and Metabolic Disease. Front. Endocrinol. 2018, 9, 405. [Google Scholar] [CrossRef]
- Wan, G.; Mathur, R.; Hu, X.; Liu, Y.; Zhang, X.; Peng, G.; Lu, X. Long non-coding RNA ANRIL (CDKN2B-AS) is induced by the ATM-E2F1 signaling pathway. Cell Signal. 2013, 25, 1086–1095. [Google Scholar] [CrossRef] [PubMed]
- Menon, D.R.; Das, S.; Krepler, C.; Vultur, A.; Rinner, B.; Schauer, S.; Kashofer, K.; Wagner, K.; Zhang, G.; Rad, E.B.; et al. A stress-induced early innate response causes multidrug tolerance in melanoma. Oncogene 2015, 34, 4448–4459. [Google Scholar] [CrossRef]
- Takahashi, K.; Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006, 126, 663–676. [Google Scholar] [CrossRef]
- Shaffer, S.M.; Dunagin, M.C.; Torborg, S.R.; Torre, E.A.; Emert, B.; Krepler, C.; Beqiri, M.; Sproesser, K.; Brafford, P.A.; Xiao, M.; et al. Rare cell variability and drug-induced reprogramming as a mode of cancer drug resistance. Nature 2017, 546, 431–435. [Google Scholar] [CrossRef]
- Pauli, A.; Rinn, J.L.; Schier, A.F. Non-coding RNAs as regulators of embryogenesis. Nat. Rev. Genet. 2011, 12, 136–149. [Google Scholar] [CrossRef]
- Mehravar, M.; Ghaemimanesh, F.; Poursani, E.M. An Overview on the Complexity of OCT4: At the Level of DNA, RNA and Protein. Stem Cell Rev. Rep. 2021, 17, 1121–1136. [Google Scholar] [CrossRef] [PubMed]
- Shahryari, A.; Jazi, M.S.; Samaei, N.M.; Mowla, S.J. Long non-coding RNA SOX2OT: Expression signature, splicing patterns, and emerging roles in pluripotency and tumorigenesis. Front. Genet. 2015, 6, 196. [Google Scholar] [CrossRef] [PubMed]
- Fantes, J.; Ragge, N.K.; Lynch, S.-A.; McGill, N.I.; Collin, J.R.O.; Howard-Peebles, P.N.; Hayward, C.; Vivian, A.J.; Williamson, K.; van Heyningen, V.; et al. Mutations in SOX2 cause anophthalmia. Nat. Genet. 2003, 33, 462–463. [Google Scholar] [CrossRef] [PubMed]
- Shahryari, A.; Rafiee, M.R.; Fouani, Y.; Oliae, N.A.; Samaei, N.M.; Shafiee, M.; Semnani, S.; Vasei, M.; Mowla, S.J. Two Novel Splice Variants of SOX2OT, SOX2OT-S1, and SOX2OT-S2 are Coupregulated with SOX2 and OCT4 in Esophageal Squamous Cell Carcinoma. Stem Cells 2014, 32, 126–134. [Google Scholar] [CrossRef] [PubMed]
- Hou, Z.; Zhao, W.; Zhou, J.; Shen, L.; Zhan, P.; Xu, C.; Chang, C.; Bi, H.; Zou, J.; Yao, X.; et al. A long noncoding RNA Sox2ot regulates lung cancer cell proliferation and is a prognostic indicator of poor survival. Int. J. Biochem. Cell Biol. 2014, 53, 380–388. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; He, L.; Du, Y.; Zhu, P.; Huang, G.; Luo, J.; Yan, X.; Ye, B.; Li, C.; Xia, P.; et al. The Long Noncoding RNA lncTCF7 Promotes Self-Renewal of Human Liver Cancer Stem Cells through Activation of Wnt Signaling. Cell Stem Cell 2015, 16, 413–425. [Google Scholar] [CrossRef] [PubMed]
- Jiang, Y.; Jiang, Y.-Y.; Xie, J.-J.; Mayakonda, A.; Hazawa, M.; Chen, L.; Xiao, J.-F.; Li, C.-Q.; Huang, M.-L.; Ding, L.-W.; et al. Co-activation of super-enhancer-driven CCAT1 by TP63 and SOX2 promotes squamous cancer progression. Nat. Commun. 2018, 9, 3619. [Google Scholar] [CrossRef] [PubMed]
- Flores, E.R.; Tsai, K.Y.; Crowley, D.; Sengupta, S.; Yang, A.; McKeon, F.; Jacks, T. p63 and p73 are required for p53-dependent apoptosis in response to DNA damage. Nature 2002, 416, 560–564. [Google Scholar] [CrossRef] [PubMed]
- Jen, J.; Tang, Y.A.; Lu, Y.H.; Lin, C.C.; Lai, W.W.; Wang, Y.C. Oct4 transcriptionally regulates the expression of long non-coding RNAs NEAT1 and MALAT1 to promote lung cancer progression. Mol. Cancer 2017, 16, 104. [Google Scholar] [CrossRef] [PubMed]
- Tang, D.; Yang, Z.; Long, F.; Luo, L.; Yang, B.; Zhu, R.; Sang, X.; Cao, G.; Wang, K. Long noncoding RNA MALAT1 mediates stem cell-like properties in human colorectal cancer cells by regulating miR-20b-5p/Oct4 axis. J. Cell Physiol. 2019, 234, 20816–20828. [Google Scholar] [CrossRef]
- Zhu, Y.; Hu, H.; Yuan, Z.; Zhang, Q.; Xiong, H.; Hu, Z.; Wu, H.; Huang, R.; Wang, G.; Tang, Q. LncRNA NEAT1 remodels chromatin to promote the 5-Fu resistance by maintaining colorectal cancer stemness. Cell Death Dis. 2020, 11, 962. [Google Scholar] [CrossRef] [PubMed]
- Jiang, P.; Chen, A.; Wu, X.; Zhou, M.; Ul Haq, I.; Mariyam, Z.; Feng, Q. NEAT1 acts as an inducer of cancer stem cell-like phenotypes in NSCLC by inhibiting EGCG-upregulated CTR1. J. Cell Physiol. 2018, 233, 4852–4863. [Google Scholar] [CrossRef] [PubMed]
- Takakuwa, H.; Yamazaki, T.; Souquere, S.; Adachi, S.; Yoshino, H.; Fujiwara, N.; Yamamoto, T.; Natsume, T.; Nakagawa, S.; Pierron, G.; et al. Shell protein composition specified by the lncRNA NEAT1 domains dictates the formation of paraspeckles as distinct membraneless organelles. Nat. Cell Biol. 2023, 25, 1664–1675. [Google Scholar] [CrossRef] [PubMed]
- Miao, H.; Wu, F.; Li, Y.; Qin, C.; Zhao, Y.; Xie, M.; Dai, H.; Yao, H.; Cai, H.; Wang, Q.; et al. MALAT1 modulates alternative splicing by cooperating with the splicing factors PTBP1 and PSF. Sci. Adv. 2022, 8, eabq7289. [Google Scholar] [CrossRef]
- Guru, S.C.; Agarwal, S.K.; Manickam, P.; Olufemi, S.-E.; Crabtree, J.S.; Weisemann, J.M.; Kester, M.B.; Kim, Y.S.; Wang, Y.; Emmert-Buck, M.R.; et al. A transcript map for the 2.8-Mb region containing the multiple endocrine neoplasia type 1 locus. Genome Res. 1997, 7, 725–735. [Google Scholar] [CrossRef] [PubMed]
- Brown, J.A.; Valenstein, M.L.; Yario, T.A.; Tycowski, K.T.; Steitz, J.A. Formation of triple-helical structures by the 3’-end sequences of MALAT1 and MENβ noncoding RNAs. Proc. Natl. Acad. Sci. USA 2012, 109, 19202–19207. [Google Scholar] [CrossRef] [PubMed]
- Hutchinson, J.N.; Ensminger, A.W.; Clemson, C.M.; Lynch, C.R.; Lawrence, J.B.; Chess, A. A screen for nuclear transcripts identifies two linked noncoding RNAs associated with SC35 splicing domains. BMC Genom. 2007, 8, 39. [Google Scholar] [CrossRef] [PubMed]
- Tano, K.; Mizuno, R.; Okada, T.; Rakwal, R.; Shibato, J.; Masuo, Y.; Ijiri, K.; Akimitsu, N. MALAT-1 enhances cell motility of lung adenocarcinoma cells by influencing the expression of motility-related genes. FEBS Lett. 2010, 584, 4575–4580. [Google Scholar] [CrossRef]
- Gutschner, T.; Hämmerle, M.; Eißmann, M.; Hsu, J.; Kim, Y.; Hung, G.; Revenko, A.; Arun, G.; Stentrup, M.; Groß, M.; et al. The Noncoding RNA MALAT1 Is a Critical Regulator of the Metastasis Phenotype of Lung Cancer Cells. Cancer Res. 2013, 73, 1180–1189. [Google Scholar] [CrossRef] [PubMed]
- Sasaki, Y.T.; Ideue, T.; Sano, M.; Mituyama, T.; Hirose, T. MENε/β noncoding RNAs are essential for structural integrity of nuclear paraspeckles. Proc. Natl. Acad. Sci. USA 2009, 106, 2525–2530. [Google Scholar] [CrossRef]
- Yamazaki, T.; Nakagawa, S.; Hirose, T. Architectural RNAs for Membraneless Nuclear Body Formation. Cold Spring Harb. Symp. Quant. Biol. 2019, 84, 227–237. [Google Scholar] [CrossRef]
- Clemson, C.M.; Hutchinson, J.N.; Sara, S.A.; Ensminger, A.W.; Fox, A.H.; Chess, A.; Lawrence, J.B. An architectural role for a nuclear noncoding RNA: NEAT1 RNA is essential for the structure of paraspeckles. Mol. Cell 2009, 33, 717–726. [Google Scholar] [CrossRef]
- Fan, X.-J.; Wang, Y.-L.; Zhao, W.-W.; Bai, S.-M.; Ma, Y.; Yin, X.-K.; Feng, L.-L.; Feng, W.-X.; Wang, Y.-N.; Liu, Q.; et al. NONO phase separation enhances DNA damage repair by accelerating nuclear EGFR-induced DNA-PK activation. Am. J. Cancer Res. 2021, 11, 2838–2852. [Google Scholar]
- Levone, B.R.; Lenzken, S.C.; Antonaci, M.; Maiser, A.; Rapp, A.; Conte, F.; Reber, S.; Mechtersheimer, J.; Ronchi, A.E.; Mühlemann, O.; et al. FUS-dependent liquid-liquid phase separation is important for DNA repair initiation. J. Cell Biol. 2021, 220, e202008030. [Google Scholar] [CrossRef]
- Desai, T.J.; Brownfield, D.G.; Krasnow, M.A. Alveolar progenitor and stem cells in lung development, renewal and cancer. Nature 2014, 507, 190–194. [Google Scholar] [CrossRef]
- Zepp, J.A.; Morrisey, E.E. Cellular crosstalk in the development and regeneration of the respiratory system. Nat. Rev. Mol. Cell Biol. 2019, 20, 551–566. [Google Scholar] [CrossRef]
- Zhao, W.; Luo, J.; Jiao, S. Comprehensive characterization of cancer subtype associated long non-coding RNAs and their clinical implications. Sci. Rep. 2014, 4, 6591. [Google Scholar] [CrossRef]
- Teng, F.; Zhang, J.; Chen, Y.; Shen, X.; Su, C.; Guo, Y.; Wang, P.; Shi, C.; Lei, M.; Cao, Y.; et al. LncRNA NKX2-1-AS1 promotes tumor progression and angiogenesis via upregulation of SERPINE1 expression and activation of the VEGFR-2 signaling pathway in gastric cancer. Mol. Oncol. 2021, 15, 1234–1255. [Google Scholar] [CrossRef]
- Singh, N.; Ramnarine, V.R.; Song, J.H.; Pandey, R.; Padi, S.K.R.; Nouri, M.; Olive, V.; Kobelev, M.; Okumura, K.; McCarthy, D.; et al. The long noncoding RNA H19 regulates tumor plasticity in neuroendocrine prostate cancer. Nat. Commun. 2021, 12, 7349. [Google Scholar] [CrossRef]
- Faubert, B.; Solmonson, A.; DeBerardinis, R.J. Metabolic reprogramming and cancer progression. Science 2020, 368, eaaw5473. [Google Scholar] [CrossRef]
- Hangauer, M.J.; Viswanathan, V.S.; Ryan, M.J.; Bole, D.; Eaton, J.K.; Matov, A.; Galeas, J.; Dhruv, H.D.; Berens, M.E.; Schreiber, S.L.; et al. Drug-tolerant persister cancer cells are vulnerable to GPX4 inhibition. Nature 2017, 551, 247–250. [Google Scholar] [CrossRef]
- Cargill, K.R.; Hasken, W.L.; Gay, C.M.; Byers, L.A. Alternative Energy: Breaking Down the Diverse Metabolic Features of Lung Cancers. Front. Oncol. 2021, 11, 757323. [Google Scholar] [CrossRef]
- Vander Heiden, M.G.; Cantley, L.C.; Thompson, C.B. Understanding the Warburg Effect: The Metabolic Requirements of Cell Proliferation. Science 2009, 324, 1029–1033. [Google Scholar] [CrossRef]
- Li, Y.; Chen, H.; Xie, X.; Yang, B.; Wang, X.; Zhang, J.; Qiao, T.; Guan, J.; Qiu, Y.; Huang, Y.-X.; et al. PINK1-Mediated Mitophagy Promotes Oxidative Phosphorylation and Redox Homeostasis to Induce Drug-Tolerant Persister Cancer Cells. Cancer Res. 2023, 83, 398–413. [Google Scholar] [CrossRef]
- Roesch, A.; Vultur, A.; Bogeski, I.; Wang, H.; Zimmermann, K.M.; Speicher, D.; Körbel, C.; Laschke, M.W.; Gimotty, P.A.; Philipp, S.E.; et al. Overcoming intrinsic multidrug resistance in melanoma by blocking the mitochondrial respiratory chain of slow-cycling JARID1B(high) cells. Cancer Cell 2013, 23, 811–825. [Google Scholar] [CrossRef]
- Connerty, P.; Lock, R.B.; de Bock, C.E. Long Non-coding RNAs: Major Regulators of Cell Stress in Cancer. Front. Oncol. 2020, 10, 285. [Google Scholar] [CrossRef]
- Fan, N.; Fu, H.; Feng, X.; Chen, Y.; Wang, J.; Wu, Y.; Bian, Y.; Li, Y. Long non-coding RNAs play an important regulatory role in tumorigenesis and tumor progression through aerobic glycolysis. Front. Mol. Biosci. 2022, 9, 941653. [Google Scholar] [CrossRef]
- Yang, F.; Zhang, H.; Mei, Y.; Wu, M. Reciprocal regulation of HIF-1α and lincRNA-p21 modulates the Warburg effect. Mol. Cell 2014, 53, 88–100. [Google Scholar] [CrossRef]
- Park, M.K.; Zhang, L.; Min, K.-W.; Cho, J.-H.; Yeh, C.-C.; Moon, H.; Hormaechea-Agulla, D.; Mun, H.; Ko, S.; Lee, J.W.; et al. NEAT1 is essential for metabolic changes that promote breast cancer growth and metastasis. Cell Metab. 2021, 33, 2380–2397.e9. [Google Scholar] [CrossRef]
- Zhu, Y.; Jin, L.; Shi, R.; Li, J.; Wang, Y.; Liang, C.-Z.; Narayana, V.K.; De Souza, D.P.; Thorne, R.F.; Zhang, L.R.; et al. The long noncoding RNA glycoLINC assembles a lower glycolytic metabolon to promote glycolysis. Mol. Cell 2022, 82, 542–554.e6. [Google Scholar] [CrossRef]
- Xiang, S.; Gu, H.; Jin, L.; Thorne, R.F.; Zhang, X.D.; Wu, M. LncRNA IDH1-AS1 links the functions of c-Myc and HIF1α via IDH1 to regulate the Warburg effect. Proc. Natl. Acad. Sci. USA 2018, 115, E1465–E1474. [Google Scholar] [CrossRef]
- Mao, C.; Liu, X.; Zhang, Y.; Lei, G.; Yan, Y.; Lee, H.; Koppula, P.; Wu, S.; Zhuang, L.; Fang, B.; et al. DHODH-mediated ferroptosis defence is a targetable vulnerability in cancer. Nature 2021, 593, 586–590. [Google Scholar] [CrossRef]
- Leon, L.M.; Gautier, M.; Allan, R.; Ilié, M.; Nottet, N.; Pons, N.; Paquet, A.; Lebrigand, K.; Truchi, M.; Fassy, J.; et al. The nuclear hypoxia-regulated NLUCAT1 long non-coding RNA contributes to an aggressive phenotype in lung adenocarcinoma through regulation of oxidative stress. Oncogene 2019, 38, 7146–7165. [Google Scholar] [CrossRef]
- Russo, M.; Sogari, A.; Bardelli, A. Adaptive Evolution: How Bacteria and Cancer Cells Survive Stressful Conditions and Drug Treatment. Cancer Discov. 2021, 11, 1886–1895. [Google Scholar] [CrossRef]
- Russo, M.; Pompei, S.; Sogari, A.; Corigliano, M.; Crisafulli, G.; Puliafito, A.; Lamba, S.; Erriquez, J.; Bertotti, A.; Gherardi, M.; et al. A modified fluctuation-test framework characterizes the population dynamics and mutation rate of colorectal cancer persister cells. Nat. Genet. 2022, 54, 976–984. [Google Scholar] [CrossRef]
- Juvekar, A.; Burga, L.N.; Hu, H.; Lunsford, E.P.; Ibrahim, Y.H.; Balmaña, J.; Rajendran, A.; Papa, A.; Spencer, K.; Lyssiotis, C.A.; et al. Combining a PI3K inhibitor with a PARP inhibitor provides an effective therapy for BRCA1-related breast cancer. Cancer Discov. 2012, 2, 1048–1063. [Google Scholar] [CrossRef] [PubMed]
- Temprine, K.; Campbell, N.R.; Huang, R.; Langdon, E.M.; Simon-Vermot, T.; Mehta, K.; Clapp, A.; Chipman, M.; White, R.M. Regulation of the error-prone DNA polymerase Polκ by oncogenic signaling and its contribution to drug resistance. Sci. Signal. 2020, 13, eaau1453. [Google Scholar] [CrossRef]
- Gay, C.M.; Balaji, K.; Byers, L.A. Giving AXL the axe: Targeting AXL in human malignancy. Br. J. Cancer 2017, 116, 415–423. [Google Scholar] [CrossRef]
- Ghafouri-Fard, S.; Khoshbakht, T.; Taheri, M.; Mokhtari, M. A review on the role of GAS6 and GAS6-AS1 in the carcinogenesis. Pathol. Res. Pract. 2021, 226, 153596. [Google Scholar] [CrossRef]
- Zhang, P.; Dong, Q.; Zhu, H.; Li, S.; Shi, L.; Chen, X. Long non-coding antisense RNA GAS6-AS1 supports gastric cancer progression via increasing GAS6 expression. Gene 2019, 696, 1–9. [Google Scholar] [CrossRef]
- Luo, J.; Wang, H.; Wang, L.; Wang, G.; Yao, Y.; Xie, K.; Li, X.; Xu, L.; Shen, Y.; Ren, B. lncRNA GAS6-AS1 inhibits progression and glucose metabolism reprogramming in LUAD via repressing E2F1-mediated transcription of GLUT1. Mol. Ther. Nucleic Acids 2021, 25, 11–24. [Google Scholar] [CrossRef] [PubMed]
- Wu, Z.R.; Yan, L.; Liu, Y.T.; Cao, L.; Guo, Y.H.; Zhang, Y.; Yao, H.; Cai, L.; Shang, H.B.; Rui, W.W.; et al. Inhibition of mTORC1 by lncRNA H19 via disrupting 4E-BP1/Raptor interaction in pituitary tumours. Nat. Commun. 2018, 9, 4624. [Google Scholar] [CrossRef]
- Li, Z.; Li, X.; Wu, S.; Xue, M.; Chen, W. Long non-coding RNA UCA1 promotes glycolysis by upregulating hexokinase 2 through the mTOR-STAT3/microRNA143 pathway. Cancer Sci. 2014, 105, 951–955. [Google Scholar] [CrossRef] [PubMed]
- Malakar, P.; Shilo, A.; Mogilevsky, A.; Stein, I.; Pikarsky, E.; Nevo, Y.; Benyamini, H.; Elgavish, S.; Zong, X.; Prasanth, K.V.; et al. Long Noncoding RNA MALAT1 Promotes Hepatocellular Carcinoma Development by SRSF1 Upregulation and mTOR Activation. Cancer Res. 2017, 77, 1155–1167. [Google Scholar] [CrossRef] [PubMed]
- Crooke, S.T.; Baker, B.F.; Crooke, R.M.; Liang, X.-H. Antisense technology: An overview and prospectus. Nat. Rev. Drug Discov. 2021, 20, 427–453. [Google Scholar] [CrossRef] [PubMed]
- Winkle, M.; El-Daly, S.M.; Fabbri, M.; Calin, G.A. Noncoding RNA therapeutics—Challenges and potential solutions. Nat. Rev. Drug Discov. 2021, 20, 629–651. [Google Scholar] [CrossRef] [PubMed]
- Barry, G.; Briggs, J.A.; Hwang, D.W.; Nayler, S.P.; Fortuna, P.R.J.; Jonkhout, N.; Dachet, F.; Maag, J.L.V.; Mestdagh, P.; Singh, E.M.; et al. The long non-coding RNA NEAT1 is responsive to neuronal activity and is associated with hyperexcitability states. Sci. Rep. 2017, 7, 40127. [Google Scholar] [CrossRef]
- Perez-Pinera, P.; Kocak, D.D.; Vockley, C.M.; Adler, A.F.; Kabadi, A.M.; Polstein, L.R.; Thakore, P.I.; Glass, K.A.; Ousterout, D.G.; Leong, K.W.; et al. RNA-guided gene activation by CRISPR-Cas9-based transcription factors. Nat. Methods 2013, 10, 973–976. [Google Scholar] [CrossRef] [PubMed]
- Qi, L.S.; Larson, M.H.; Gilbert, L.A.; Doudna, J.A.; Weissman, J.S.; Arkin, A.P.; Lim, W.A. Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell 2013, 152, 1173–1183. [Google Scholar] [CrossRef]
- Abudayyeh, O.O.; Gootenberg, J.S.; Essletzbichler, P.; Han, S.; Joung, J.; Belanto, J.J.; Verdine, V.; Cox, D.B.T.; Kellner, M.J.; Regev, A.; et al. RNA targeting with CRISPR-Cas13. Nature 2017, 550, 280–284. [Google Scholar] [CrossRef]
- Aparicio-Prat, E.; Arnan, C.; Sala, I.; Bosch, N.; Guigó, R.; Johnson, R. DECKO: Single-oligo, dual-CRISPR deletion of genomic elements including long non-coding RNAs. BMC Genom. 2015, 16, 846. [Google Scholar] [CrossRef] [PubMed]
- Paunovska, K.; Loughrey, D.; Dahlman, J.E. Drug delivery systems for RNA therapeutics. Nat. Rev. Genet. 2022, 23, 265–280. [Google Scholar] [CrossRef] [PubMed]
- Ross, S.J.; Revenko, A.S.; Hanson, L.L.; Ellston, R.; Staniszewska, A.; Whalley, N.; Pandey, S.K.; Revill, M.; Rooney, C.; Buckett, L.K.; et al. Targeting KRAS-dependent tumors with AZD4785, a high-affinity therapeutic antisense oligonucleotide inhibitor of KRAS. Sci. Transl. Med. 2017, 9, eaal5253. [Google Scholar] [CrossRef] [PubMed]
- Lee, M.S.; Pant, S. Personalizing Medicine with Germline and Somatic Sequencing in Advanced Pancreatic Cancer: Current Treatments and Novel Opportunities. Am. Soc. Clin. Oncol. Educ. Book 2021, 41, e153–e165. [Google Scholar] [CrossRef] [PubMed]
- Wiesenfarth, M.; Dorst, J.; Brenner, D.; Elmas, Z.; Parlak, Ö.; Uzelac, Z.; Kandler, K.; Mayer, K.; Weiland, U.; Herrmann, C.; et al. Effects of tofersen treatment in patients with SOD1-ALS in a “real-world” setting—A 12-month multicenter cohort study from the German early access program. eClinicalMedicine 2024, 69, 102495. [Google Scholar] [CrossRef] [PubMed]
- Durcan, N.; Murphy, C.; Cryan, S.-A. Inhalable siRNA: Potential as a Therapeutic Agent in the Lungs. Mol. Pharm. 2008, 5, 559–566. [Google Scholar] [CrossRef]
- Shaffer, C. Mist begins to clear for lung delivery of RNA. Nat. Biotechnol. 2020, 38, 1110–1112. [Google Scholar] [CrossRef]
- Qiu, Y.; Lam, J.K.W.; Leung, S.W.S.; Liang, W. Delivery of RNAi Therapeutics to the Airways—From Bench to Bedside. Molecules 2016, 21, 1249. [Google Scholar] [CrossRef] [PubMed]
- Wilbie, D.; Walther, J.; Mastrobattista, E. Delivery Aspects of CRISPR/Cas for in Vivo Genome Editing. Acc. Chem. Res. 2019, 52, 1555–1564. [Google Scholar] [CrossRef]
- Abulwerdi, F.A.; Xu, W.; Ageeli, A.A.; Yonkunas, M.J.; Arun, G.; Nam, H.; Schneekloth, J.J.S.; Dayie, T.K.; Spector, D.; Baird, N.; et al. Selective Small-Molecule Targeting of a Triple Helix Encoded by the Long Noncoding RNA, MALAT1. ACS Chem. Biol. 2019, 14, 223–235. [Google Scholar] [CrossRef]
- Childs-Disney, J.L.; Yang, X.; Gibaut, Q.M.R.; Tong, Y.; Batey, R.T.; Disney, M.D. Targeting RNA structures with small molecules. Nat. Rev. Drug Discov. 2022, 21, 736–762. [Google Scholar] [CrossRef] [PubMed]
- Jones, A.N.; Sattler, M. Challenges and perspectives for structural biology of lncRNAs-the example of the Xist lncRNA A-repeats. J. Mol. Cell Biol. 2019, 11, 845–859. [Google Scholar] [CrossRef] [PubMed]
- Zhang, J.; Fei, Y.; Sun, L.; Zhang, Q.C. Advances and opportunities in RNA structure experimental determination and computational modeling. Nat. Methods 2022, 19, 1193–1207. [Google Scholar] [CrossRef] [PubMed]
- Flynn, R.A.; Zhang, Q.C.; Spitale, R.C.; Lee, B.; Mumbach, M.R.; Chang, H.Y. Transcriptome-wide interrogation of RNA secondary structure in living cells with icSHAPE. Nat. Protoc. 2016, 11, 273–290. [Google Scholar] [CrossRef] [PubMed]
- Townshend, R.J.L.; Eismann, S.; Watkins, A.M.; Rangan, R.; Karelina, M.; Das, R.; Dror, R.O. Geometric deep learning of RNA structure. Science 2021, 373, 1047–1051. [Google Scholar] [CrossRef] [PubMed]
- Feyder, M.; Goff, L.A. Investigating long noncoding RNAs using animal models. J. Clin. Investig. 2016, 126, 2783–2791. [Google Scholar] [CrossRef]
- Zhang, B.; Mao, Y.S.; Diermeier, S.D.; Novikova, I.V.; Nawrocki, E.P.; Jones, T.A.; Lazar, Z.; Tung, C.-S.; Luo, W.; Eddy, S.R.; et al. Identification and Characterization of a Class of MALAT1-like Genomic Loci. Cell Rep. 2017, 19, 1723–1738. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 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
Davis, W.J.H.; Drummond, C.J.; Diermeier, S.; Reid, G. The Potential Links between lncRNAs and Drug Tolerance in Lung Adenocarcinoma. Genes 2024, 15, 906. https://doi.org/10.3390/genes15070906
Davis WJH, Drummond CJ, Diermeier S, Reid G. The Potential Links between lncRNAs and Drug Tolerance in Lung Adenocarcinoma. Genes. 2024; 15(7):906. https://doi.org/10.3390/genes15070906
Chicago/Turabian StyleDavis, William J. H., Catherine J. Drummond, Sarah Diermeier, and Glen Reid. 2024. "The Potential Links between lncRNAs and Drug Tolerance in Lung Adenocarcinoma" Genes 15, no. 7: 906. https://doi.org/10.3390/genes15070906
APA StyleDavis, W. J. H., Drummond, C. J., Diermeier, S., & Reid, G. (2024). The Potential Links between lncRNAs and Drug Tolerance in Lung Adenocarcinoma. Genes, 15(7), 906. https://doi.org/10.3390/genes15070906