Epigenetic Landscape and Therapeutic Implication of Gene Isoforms of Doublecortin-Like Kinase 1 for Cancer Stem Cells
Abstract
:1. Introduction
2. Epigenetics and Cancer Progression
2.1. DNA Methylation in Cancer
2.2. Interplay between Histone Modifications and DNA Methylation
2.3. Regulation of Alternative Promoters through Epigenetics
2.4. Epigenetics and Alternative Splicing
3. Gene Isoforms in Cancer: A Double-Edged Sword
4. Epigenetic Regulation of Alternative Isoforms in Cancer
4.1. RASSF1
4.2. TP63/TP73
4.3. DCLK1
5. The Interplay of Gene Isoforms, Epigenetics, and Cancer Stem Cells
6. The Future of Cancer Treatment: Isoform-Specific Therapies and Epigenetic Modulation
7. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed]
- Sabit, H.; Cevik, E.; Tombuloglu, H. Colorectal cancer: The epigenetic role of microbiome. World J. Clin. Cases 2019, 7, 3683–3697. [Google Scholar] [CrossRef] [PubMed]
- Sun, D.; Chen, Y.; Fang, J.Y. Influence of the microbiota on epigenetics in colorectal cancer. Natl. Sci. Rev. 2019, 6, 1138–1148. [Google Scholar] [CrossRef] [PubMed]
- Wu, Q.; Ni, X. ROS-mediated DNA methylation pattern alterations in carcinogenesis. Curr. Drug Targets 2015, 16, 13–19. [Google Scholar] [CrossRef]
- Das, D.; Karthik, N.; Taneja, R. Crosstalk Between Inflammatory Signaling and Methylation in Cancer. Front. Cell Dev. Biol. 2021, 9, 756458. [Google Scholar] [CrossRef]
- O’Hagan, H.M.; Mohammad, H.P.; Baylin, S.B. Double strand breaks can initiate gene silencing and SIRT1-dependent onset of DNA methylation in an exogenous promoter CpG island. PLoS Genet. 2008, 4, e1000155. [Google Scholar] [CrossRef]
- Baylin, S.B.; Jones, P.A. A decade of exploring the cancer epigenome—Biological and translational implications. Nat. Rev. Cancer 2011, 11, 726–734. [Google Scholar] [CrossRef]
- Easwaran, H.; Tsai, H.C.; Baylin, S.B. Cancer epigenetics: Tumor heterogeneity, plasticity of stem-like states, and drug resistance. Mol. Cell 2014, 54, 716–727. [Google Scholar] [CrossRef]
- Kim, J.H.; Park, S.Y.; Jeon, S.E.; Choi, J.H.; Lee, C.J.; Jang, T.Y.; Yun, H.J.; Lee, Y.; Kim, P.; Cho, S.H.; et al. DCLK1 promotes colorectal cancer stemness and aggressiveness via the XRCC5/COX2 axis. Theranostics 2022, 12, 5258–5271. [Google Scholar] [CrossRef]
- Kalantari, E.; Ghods, R.; Saeednejad Zanjani, L.; Rahimi, M.; Eini, L.; Razmi, M.; Asadi-Lari, M.; Madjd, Z. Cytoplasmic expression of DCLK1-S, a novel DCLK1 isoform, is associated with tumor aggressiveness and worse disease-specific survival in colorectal cancer. Cancer Biomark. 2022, 33, 277–289. [Google Scholar] [CrossRef]
- Wang, L.; Zhao, L.; Lin, Z.; Yu, D.; Jin, M.; Zhou, P.; Ren, J.; Cheng, J.; Yang, K.; Wu, G.; et al. Targeting DCLK1 overcomes 5-fluorouracil resistance in colorectal cancer through inhibiting CCAR1/beta-catenin pathway-mediated cancer stemness. Clin. Transl. Med. 2022, 12, e743. [Google Scholar] [CrossRef] [PubMed]
- Chandrakesan, P.; Weygant, N.; May, R.; Qu, D.; Chinthalapally, H.R.; Sureban, S.M.; Ali, N.; Lightfoot, S.A.; Umar, S.; Houchen, C.W. DCLK1 facilitates intestinal tumor growth via enhancing pluripotency and epithelial mesenchymal transition. Oncotarget 2014, 5, 9269–9280. [Google Scholar] [CrossRef] [PubMed]
- Vedeld, H.M.; Skotheim, R.I.; Lothe, R.A.; Lind, G.E. The recently suggested intestinal cancer stem cell marker DCLK1 is an epigenetic biomarker for colorectal cancer. Epigenetics 2014, 9, 346–350. [Google Scholar] [CrossRef]
- Burgess, H.A.; Reiner, O. Alternative splice variants of doublecortin-like kinase are differentially expressed and have different kinase activities. J. Biol. Chem. 2002, 277, 17696–17705. [Google Scholar] [CrossRef] [PubMed]
- O’Connell, M.R.; Sarkar, S.; Luthra, G.K.; Okugawa, Y.; Toiyama, Y.; Gajjar, A.H.; Qiu, S.; Goel, A.; Singh, P. Epigenetic changes and alternate promoter usage by human colon cancers for expressing DCLK1-isoforms: Clinical Implications. Sci. Rep. 2015, 5, 14983. [Google Scholar] [CrossRef] [PubMed]
- Moore, L.D.; Le, T.; Fan, G. DNA Methylation and Its Basic Function. Neuropsychopharmacology 2013, 38, 23–38. [Google Scholar] [CrossRef]
- Feinberg, A.P.; Vogelstein, B. Hypomethylation distinguishes genes of some human cancers from their normal counterparts. Nature 1983, 301, 89–92. [Google Scholar] [CrossRef]
- Herman, J.G.; Baylin, S.B. Gene silencing in cancer in association with promoter hypermethylation. N. Engl. J. Med. 2003, 349, 2042–2054. [Google Scholar] [CrossRef]
- Ehrlich, M. DNA hypomethylation in cancer cells. Epigenomics 2009, 1, 239–259. [Google Scholar] [CrossRef]
- Jones, P.A. Functions of DNA methylation: Islands, start sites, gene bodies and beyond. Nat. Rev. Genet. 2012, 13, 484–492. [Google Scholar] [CrossRef]
- Bestor, T.H.; Edwards, J.R.; Boulard, M. Notes on the role of dynamic DNA methylation in mammalian development. Proc. Natl. Acad. Sci. USA 2015, 112, 6796–6799. [Google Scholar] [CrossRef] [PubMed]
- Saravanaraman, P.; Selvam, M.; Ashok, C.; Srijyothi, L.; Baluchamy, S. De novo methyltransferases: Potential players in diseases and new directions for targeted therapy. Biochimie 2020, 176, 85–102. [Google Scholar] [CrossRef]
- Du, Q.; Luu, P.L.; Stirzaker, C.; Clark, S.J. Methyl-CpG-binding domain proteins: Readers of the epigenome. Epigenomics 2015, 7, 1051–1073. [Google Scholar] [CrossRef] [PubMed]
- Kohli, R.M.; Zhang, Y. TET enzymes, TDG and the dynamics of DNA demethylation. Nature 2013, 502, 472–479. [Google Scholar] [CrossRef]
- Bannister, A.J.; Kouzarides, T. Regulation of chromatin by histone modifications. Cell Res. 2011, 21, 381–395. [Google Scholar] [CrossRef]
- Lee, K.K.; Workman, J.L. Histone acetyltransferase complexes: One size doesn’t fit all. Nat. Rev. Mol. Cell Biol. 2007, 8, 284–295. [Google Scholar] [CrossRef]
- Yu, Y.; Li, X.; Jiao, R.; Lu, Y.; Jiang, X.; Li, X. H3K27me3-H3K4me1 transition at bivalent promoters instructs lineage specification in development. Cell Biosci. 2023, 13, 66. [Google Scholar] [CrossRef]
- Kong, S.; Lu, Y.; Tan, S.; Li, R.; Gao, Y.; Li, K.; Zhang, Y. Nucleosome-Omics: A Perspective on the Epigenetic Code and 3D Genome Landscape. Genes 2022, 13, 1114. [Google Scholar] [CrossRef]
- Cedar, H.; Bergman, Y. Linking DNA methylation and histone modification: Patterns and paradigms. Nat. Rev. Genet. 2009, 10, 295–304. [Google Scholar] [CrossRef]
- Viré, E.; Brenner, C.; Deplus, R.; Blanchon, L.; Fraga, M.; Didelot, C.; Morey, L.; Van Eynde, A.; Bernard, D.; Vanderwinden, J.M.; et al. The Polycomb group protein EZH2 directly controls DNA methylation. Nature 2006, 439, 871–874. [Google Scholar] [CrossRef]
- Agarwal, S.; Bonefas, K.M.; Garay, P.M.; Brookes, E.; Murata-Nakamura, Y.; Porter, R.S.; Macfarlan, T.S.; Ren, B.; Iwase, S. KDM1A maintains genome-wide homeostasis of transcriptional enhancers. Genome Res. 2021, 31, 186–197. [Google Scholar] [CrossRef]
- Wang, J.; Hevi, S.; Kurash, J.K.; Lei, H.; Gay, F.; Bajko, J.; Su, H.; Sun, W.; Chang, H.; Xu, G.; et al. The lysine demethylase LSD1 (KDM1) is required for maintenance of global DNA methylation. Nat. Genet. 2009, 41, 125–129. [Google Scholar] [CrossRef] [PubMed]
- Shen, H.; Xu, W.; Guo, R.; Rong, B.; Gu, L.; Wang, Z.; He, C.; Zheng, L.; Hu, X.; Hu, Z.; et al. Suppression of Enhancer Overactivation by a RACK7-Histone Demethylase Complex. Cell 2016, 165, 331–342. [Google Scholar] [CrossRef] [PubMed]
- Davuluri, R.V.; Suzuki, Y.; Sugano, S.; Plass, C.; Huang, T.H. The functional consequences of alternative promoter use in mammalian genomes. Trends Genet. 2008, 24, 167–177. [Google Scholar] [CrossRef]
- Juven-Gershon, T.; Kadonaga, J.T. Regulation of gene expression via the core promoter and the basal transcriptional machinery. Dev. Biol. 2010, 339, 225–229. [Google Scholar] [CrossRef] [PubMed]
- D’Alessio, J.A.; Wright, K.J.; Tjian, R. Shifting players and paradigms in cell-specific transcription. Mol. Cell 2009, 36, 924–931. [Google Scholar] [CrossRef]
- Jones, P.A.; Issa, J.-P.J.; Baylin, S. Targeting the cancer epigenome for therapy. Nat. Rev. Genet. 2016, 17, 630–641. [Google Scholar] [CrossRef]
- Vo Ngoc, L.; Wang, Y.L.; Kassavetis, G.A.; Kadonaga, J.T. The punctilious RNA polymerase II core promoter. Genes Dev. 2017, 31, 1289–1301. [Google Scholar] [CrossRef]
- Spitz, F.; Furlong, E.E.M. Transcription factors: From enhancer binding to developmental control. Nat. Rev. Genet. 2012, 13, 613–626. [Google Scholar] [CrossRef]
- Maunakea, A.K.; Nagarajan, R.P.; Bilenky, M.; Ballinger, T.J.; D’Souza, C.; Fouse, S.D.; Johnson, B.E.; Hong, C.; Nielsen, C.; Zhao, Y.; et al. Conserved role of intragenic DNA methylation in regulating alternative promoters. Nature 2010, 466, 253–257. [Google Scholar] [CrossRef]
- Demircioglu, D.; Cukuroglu, E.; Kindermans, M.; Nandi, T.; Calabrese, C.; Fonseca, N.A.; Kahles, A.; Lehmann, K.V.; Stegle, O.; Brazma, A.; et al. A Pan-cancer Transcriptome Analysis Reveals Pervasive Regulation through Alternative Promoters. Cell 2019, 178, 1465–1477.e17. [Google Scholar] [CrossRef] [PubMed]
- Dong, Y.; Liu, X.; Jiang, B.; Wei, S.; Xiang, B.; Liao, R.; Wang, Q.; He, X. A Genome-Wide Investigation of Effects of Aberrant DNA Methylation on the Usage of Alternative Promoters in Hepatocellular Carcinoma. Front. Oncol. 2021, 11, 780266. [Google Scholar] [CrossRef] [PubMed]
- Wu, J.; Zhang, L.; Kuchi, A.; Otohinoyi, D.; Hicks, C. CpG Site-Based Signature Predicts Survival of Colorectal Cancer. Biomedicines 2022, 10, 3163. [Google Scholar] [CrossRef] [PubMed]
- Nepal, C.; Andersen, J.B. Alternative promoters in CpG depleted regions are prevalently associated with epigenetic misregulation of liver cancer transcriptomes. Nat. Commun. 2023, 14, 2712. [Google Scholar] [CrossRef] [PubMed]
- Masalmeh, R.H.A.; Taglini, F.; Rubio-Ramon, C.; Musialik, K.I.; Higham, J.; Davidson-Smith, H.; Kafetzopoulos, I.; Pawlicka, K.P.; Finan, H.M.; Clark, R.; et al. De novo DNA methyltransferase activity in colorectal cancer is directed towards H3K36me3 marked CpG islands. Nat. Commun. 2021, 12, 694. [Google Scholar] [CrossRef]
- Neri, F.; Rapelli, S.; Krepelova, A.; Incarnato, D.; Parlato, C.; Basile, G.; Maldotti, M.; Anselmi, F.; Oliviero, S. Intragenic DNA methylation prevents spurious transcription initiation. Nature 2017, 543, 72–77. [Google Scholar] [CrossRef]
- Ibrahim, A.E.; Arends, M.J.; Silva, A.L.; Wyllie, A.H.; Greger, L.; Ito, Y.; Vowler, S.L.; Huang, T.H.; Tavaré, S.; Murrell, A.; et al. Sequential DNA methylation changes are associated with DNMT3B overexpression in colorectal neoplastic progression. Gut 2011, 60, 499–508. [Google Scholar] [CrossRef]
- Nosho, K.; Shima, K.; Irahara, N.; Kure, S.; Baba, Y.; Kirkner, G.J.; Chen, L.; Gokhale, S.; Hazra, A.; Spiegelman, D.; et al. DNMT3B expression might contribute to CpG island methylator phenotype in colorectal cancer. Clin. Cancer Res. 2009, 15, 3663–3671. [Google Scholar] [CrossRef]
- Xin, D.; Hu, L.; Kong, X. Alternative promoters influence alternative splicing at the genomic level. PLoS ONE 2008, 3, e2377. [Google Scholar] [CrossRef]
- Kolathur, K.K. Role of promoters in regulating alternative splicing. Gene 2021, 782, 145523. [Google Scholar] [CrossRef]
- Maunakea, A.K.; Chepelev, I.; Cui, K.; Zhao, K. Intragenic DNA methylation modulates alternative splicing by recruiting MeCP2 to promote exon recognition. Cell Res. 2013, 23, 1256–1269. [Google Scholar] [CrossRef] [PubMed]
- Luco, R.F.; Pan, Q.; Tominaga, K.; Blencowe, B.J.; Pereira-Smith, O.M.; Misteli, T. Regulation of alternative splicing by histone modifications. Science 2010, 327, 996–1000. [Google Scholar] [CrossRef]
- Bieberstein, N.I.; Carrillo Oesterreich, F.; Straube, K.; Neugebauer, K.M. First exon length controls active chromatin signatures and transcription. Cell Rep. 2012, 2, 62–68. [Google Scholar] [CrossRef] [PubMed]
- Naftelberg, S.; Schor, I.E.; Ast, G.; Kornblihtt, A.R. Regulation of alternative splicing through coupling with transcription and chromatin structure. Annu. Rev. Biochem. 2015, 84, 165–198. [Google Scholar] [CrossRef] [PubMed]
- Knudson, A.G. Two genetic hits (more or less) to cancer. Nat. Rev. Cancer 2001, 1, 157–162. [Google Scholar] [CrossRef] [PubMed]
- Vogelstein, B.; Papadopoulos, N.; Velculescu, V.E.; Zhou, S.; Diaz, L.A., Jr.; Kinzler, K.W. Cancer genome landscapes. Science 2013, 339, 1546–1558. [Google Scholar] [CrossRef] [PubMed]
- Rodriguez Calleja, L.; Lavaud, M.; Tesfaye, R.; Brounais-Le-Royer, B.; Baud’huin, M.; Georges, S.; Lamoureux, F.; Verrecchia, F.; Ory, B. The p53 Family Members p63 and p73 Roles in the Metastatic Dissemination: Interactions with microRNAs and TGFbeta Pathway. Cancers 2022, 14, 5948. [Google Scholar] [CrossRef] [PubMed]
- Xu, Y.; Yang, X.; Xiong, Q.; Han, J.; Zhu, Q. The dual role of p63 in cancer. Front. Oncol. 2023, 13, 1116061. [Google Scholar] [CrossRef]
- Donninger, H.; Vos, M.D.; Clark, G.J. The RASSF1A tumor suppressor. J. Cell Sci. 2007, 120, 3163–3172. [Google Scholar] [CrossRef]
- Chhetri, D.; Vengadassalapathy, S.; Venkadassalapathy, S.; Balachandran, V.; Umapathy, V.R.; Veeraraghavan, V.P.; Jayaraman, S.; Patil, S.; Iyaswamy, A.; Palaniyandi, K.; et al. Pleiotropic effects of DCLK1 in cancer and cancer stem cells. Front. Mol. Biosci. 2022, 9, 965730. [Google Scholar] [CrossRef]
- Nakanishi, Y.; Seno, H.; Fukuoka, A.; Ueo, T.; Yamaga, Y.; Maruno, T.; Nakanishi, N.; Kanda, K.; Komekado, H.; Kawada, M.; et al. Dclk1 distinguishes between tumor and normal stem cells in the intestine. Nat. Genet. 2013, 45, 98–103. [Google Scholar] [CrossRef] [PubMed]
- Bailey, J.M.; Alsina, J.; Rasheed, Z.A.; McAllister, F.M.; Fu, Y.Y.; Plentz, R.; Zhang, H.; Pasricha, P.J.; Bardeesy, N.; Matsui, W.; et al. DCLK1 marks a morphologically distinct subpopulation of cells with stem cell properties in preinvasive pancreatic cancer. Gastroenterology 2014, 146, 245–256. [Google Scholar] [CrossRef] [PubMed]
- Westphalen, C.B.; Asfaha, S.; Hayakawa, Y.; Takemoto, Y.; Lukin, D.J.; Nuber, A.H.; Brandtner, A.; Setlik, W.; Remotti, H.; Muley, A.; et al. Long-lived intestinal tuft cells serve as colon cancer-initiating cells. J. Clin. Investig. 2014, 124, 1283–1295. [Google Scholar] [CrossRef]
- Sureban, S.M.; May, R.; Lightfoot, S.A.; Hoskins, A.B.; Lerner, M.; Brackett, D.J.; Postier, R.G.; Ramanujam, R.; Mohammed, A.; Rao, C.V.; et al. DCAMKL-1 regulates epithelial-mesenchymal transition in human pancreatic cells through a miR-200a-dependent mechanism. Cancer Res. 2011, 71, 2328–2338. [Google Scholar] [CrossRef] [PubMed]
- Standing, D.; Arnold, L.; Dandawate, P.; Ottemann, B.; Snyder, V.; Ponnurangam, S.; Sayed, A.; Subramaniam, D.; Srinivasan, P.; Choudhury, S.; et al. Doublecortin-like kinase 1 is a therapeutic target in squamous cell carcinoma. Mol. Carcinog. 2023, 62, 145–159. [Google Scholar] [CrossRef]
- Vijai, M.; Baba, M.; Ramalingam, S.; Thiyagaraj, A. DCLK1 and its interaction partners: An effective therapeutic target for colorectal cancer. Oncol. Lett. 2021, 22, 850. [Google Scholar] [CrossRef]
- Liu, H.; Yan, R.; Xiao, Z.; Huang, X.; Yao, J.; Liu, J.; An, G.; Ge, Y. Targeting DCLK1 attenuates tumor stemness and evokes antitumor immunity in triple-negative breast cancer by inhibiting IL-6/STAT3 signaling. Breast Cancer Res. 2023, 25, 43. [Google Scholar] [CrossRef] [PubMed]
- Roy, B.C.; Ahmed, I.; Stubbs, J.; Zhang, J.; Attard, T.; Septer, S.; Welch, D.; Anant, S.; Sampath, V.; Umar, S. DCLK1 isoforms and aberrant Notch signaling in the regulation of human and murine colitis. Cell Death Discov. 2021, 7, 169. [Google Scholar] [CrossRef] [PubMed]
- Dammann, R.; Li, C.; Yoon, J.H.; Chin, P.L.; Bates, S.; Pfeifer, G.P. Epigenetic inactivation of a RAS association domain family protein from the lung tumour suppressor locus 3p21.3. Nat. Genet. 2000, 25, 315–319. [Google Scholar] [CrossRef]
- Pfeifer, G.P.; Yoon, J.H.; Liu, L.; Tommasi, S.; Wilczynski, S.P.; Dammann, R. Methylation of the RASSF1A gene in human cancers. Biol. Chem. 2002, 383, 907–914. [Google Scholar] [CrossRef]
- da Costa Prando, E.; Cavalli, L.R.; Rainho, C.A. Evidence of epigenetic regulation of the tumor suppressor gene cluster flanking RASSF1 in breast cancer cell lines. Epigenetics 2011, 6, 1413–1424. [Google Scholar] [CrossRef]
- Zhang, T.; Li, Y.; Zhang, H.; Wang, X.; Liu, X.; Li, L. The Role of RASSF1 Methylation in Lung Carcinoma. Adv. Exp. Med. Biol. 2020, 1255, 99–108. [Google Scholar] [CrossRef]
- Rattanapan, Y.; Korkiatsakul, V.; Kongruang, A.; Chareonsirisuthigul, T.; Rerkamnuaychoke, B.; Wongkularb, A.; Wilailak, S. EGFL7 and RASSF1 promoter hypermethylation in epithelial ovarian cancer. Cancer Genet. 2018, 224–225, 37–40. [Google Scholar] [CrossRef] [PubMed]
- Dubois, F.; Bergot, E.; Levallet, G. Cancer and RASSF1A/RASSF1C, the Two Faces of Janus. Trends Cancer 2019, 5, 662–665. [Google Scholar] [CrossRef] [PubMed]
- Reeves, M.E.; Baldwin, S.W.; Baldwin, M.L.; Chen, S.T.; Moretz, J.M.; Aragon, R.J.; Li, X.; Strong, D.D.; Mohan, S.; Amaar, Y.G. Ras-association domain family 1C protein promotes breast cancer cell migration and attenuates apoptosis. BMC Cancer 2010, 10, 562. [Google Scholar] [CrossRef] [PubMed]
- Ortiz-Vega, S.; Khokhlatchev, A.; Nedwidek, M.; Zhang, X.F.; Dammann, R.; Pfeifer, G.P.; Avruch, J. The putative tumor suppressor RASSF1A homodimerizes and heterodimerizes with the Ras-GTP binding protein Nore1. Oncogene 2002, 21, 1381–1390. [Google Scholar] [CrossRef]
- Osterburg, C.; Dotsch, V. Structural diversity of p63 and p73 isoforms. Cell Death Differ. 2022, 29, 921–937. [Google Scholar] [CrossRef] [PubMed]
- Lai, J.; Nie, W.; Zhang, W.; Wang, Y.; Xie, R.; Wang, Y.; Gu, J.; Xu, J.; Song, W.; Yang, F.; et al. Transcriptional regulation of the p73 gene by Nrf-2 and promoter CpG methylation in human breast cancer. Oncotarget 2014, 5, 6909–6922. [Google Scholar] [CrossRef]
- Pokorna, Z.; Hrabal, V.; Tichy, V.; Vojtesek, B.; Coates, P.J. DNA Demethylation Switches Oncogenic ΔNp63 to Tumor Suppressive TAp63 in Squamous Cell Carcinoma. Front. Oncol. 2022, 12, 924354. [Google Scholar] [CrossRef]
- Ding, L.; Yang, Y.; Ge, Y.; Lu, Q.; Yan, Z.; Chen, X.; Du, J.; Hafizi, S.; Xu, X.; Yao, J.; et al. Inhibition of DCLK1 with DCLK1-IN-1 Suppresses Renal Cell Carcinoma Invasion and Stemness and Promotes Cytotoxic T-Cell-Mediated Anti-Tumor Immunity. Cancers 2021, 13, 5729. [Google Scholar] [CrossRef]
- Patel, O.; Roy, M.J.; Kropp, A.; Hardy, J.M.; Dai, W.; Lucet, I.S. Structural basis for small molecule targeting of Doublecortin Like Kinase 1 with DCLK1-IN-1. Commun. Biol. 2021, 4, 1105. [Google Scholar] [CrossRef] [PubMed]
- Ferguson, F.M.; Liu, Y.; Harshbarger, W.; Huang, L.; Wang, J.; Deng, X.; Capuzzi, S.J.; Muratov, E.N.; Tropsha, A.; Muthuswamy, S.; et al. Synthesis and Structure-Activity Relationships of DCLK1 Kinase Inhibitors Based on a 5,11-Dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one Scaffold. J. Med. Chem. 2020, 63, 7817–7826. [Google Scholar] [CrossRef]
- Sureban, S.M.; Berahovich, R.; Zhou, H.; Xu, S.; Wu, L.; Ding, K.; May, R.; Qu, D.; Bannerman-Menson, E.; Golubovskaya, V.; et al. DCLK1 Monoclonal Antibody-Based CAR-T Cells as a Novel Treatment Strategy against Human Colorectal Cancers. Cancers 2019, 12, 54. [Google Scholar] [CrossRef] [PubMed]
- Ge, Y.; Weygant, N.; Qu, D.; May, R.; Berry, W.L.; Yao, J.; Chandrakesan, P.; Zheng, W.; Zhao, L.; Zhao, K.L.; et al. Alternative splice variants of DCLK1 mark cancer stem cells, promote self-renewal and drug-resistance, and can be targeted to inhibit tumorigenesis in kidney cancer. Int. J. Cancer 2018, 143, 1162–1175. [Google Scholar] [CrossRef]
- Omori, Y.; Suzuki, M.; Ozaki, K.; Harada, Y.; Nakamura, Y.; Takahashi, E.; Fujiwara, T. Expression and chromosomal localization of KIAA0369, a putative kinase structurally related to Doublecortin. J. Hum. Genet. 1998, 43, 169–177. [Google Scholar] [CrossRef]
- Burgess, H.A.; Martinez, S.; Reiner, O. KIAA0369, doublecortin-like kinase, is expressed during brain development. J. Neurosci. Res. 1999, 58, 567–575. [Google Scholar] [CrossRef]
- Matsumoto, N.; Pilz, D.T.; Ledbetter, D.H. Genomic structure, chromosomal mapping, and expression pattern of human DCAMKL1 (KIAA0369), a homologue of DCX (XLIS). Genomics 1999, 56, 179–183. [Google Scholar] [CrossRef] [PubMed]
- Sossey-Alaoui, K.; Srivastava, A.K. DCAMKL1, a brain-specific transmembrane protein on 13q12.3 that is similar to doublecortin (DCX). Genomics 1999, 56, 121–126. [Google Scholar] [CrossRef]
- Moores, C.A.; Perderiset, M.; Francis, F.; Chelly, J.; Houdusse, A.; Milligan, R.A. Mechanism of microtubule stabilization by doublecortin. Mol. Cell 2004, 14, 833–839. [Google Scholar] [CrossRef]
- Lin, P.T.; Gleeson, J.G.; Corbo, J.C.; Flanagan, L.; Walsh, C.A. DCAMKL1 encodes a protein kinase with homology to doublecortin that regulates microtubule polymerization. J. Neurosci. 2000, 20, 9152–9161. [Google Scholar] [CrossRef]
- Silverman, M.A.; Benard, O.; Jaaro, H.; Rattner, A.; Citri, Y.; Seger, R. CPG16, a novel protein serine/threonine kinase downstream of cAMP-dependent protein kinase. J. Biol. Chem. 1999, 274, 2631–2636. [Google Scholar] [CrossRef] [PubMed]
- Burgess, H.A.; Reiner, O. Cleavage of doublecortin-like kinase by calpain releases an active kinase fragment from a microtubule anchorage domain. J. Biol. Chem. 2001, 276, 36397–36403. [Google Scholar] [CrossRef] [PubMed]
- Hardt, R.; Dehghani, A.; Schoor, C.; Gödderz, M.; Cengiz Winter, N.; Ahmadi, S.; Sharma, R.; Schork, K.; Eisenacher, M.; Gieselmann, V.; et al. Proteomic investigation of neural stem cell to oligodendrocyte precursor cell differentiation reveals phosphorylation-dependent Dclk1 processing. Cell. Mol. Life Sci. 2023, 80, 260. [Google Scholar] [CrossRef] [PubMed]
- Bergoglio, E.; Suzuki, I.K.; Togashi, K.; Tsuji, M.; Takeuchi, S.; Koizumi, H.; Emoto, K. Spatial and temporal diversity of DCLK1 isoforms in developing mouse brain. Neurosci. Res. 2021, 170, 154–165. [Google Scholar] [CrossRef]
- Sarkar, S.; Popov, V.L.; O’Connell, M.R.; Stevenson, H.L.; Lee, B.S.; Obeid, R.A.; Luthra, G.K.; Singh, P. A novel antibody against cancer stem cell biomarker, DCLK1-S, is potentially useful for assessing colon cancer risk after screening colonoscopy. Lab. Investig. 2017, 97, 1245–1261. [Google Scholar] [CrossRef] [PubMed]
- Vedeld, H.M.; Andresen, K.; Eilertsen, I.A.; Nesbakken, A.; Seruca, R.; Gladhaug, I.P.; Thiis-Evensen, E.; Rognum, T.O.; Boberg, K.M.; Lind, G.E. The novel colorectal cancer biomarkers CDO1, ZSCAN18 and ZNF331 are frequently methylated across gastrointestinal cancers. Int. J. Cancer 2015, 136, 844–853. [Google Scholar] [CrossRef]
- Powrozek, T.; Krawczyk, P.; Nicos, M.; Kuznar-Kaminska, B.; Batura-Gabryel, H.; Milanowski, J. Methylation of the DCLK1 promoter region in circulating free DNA and its prognostic value in lung cancer patients. Clin. Transl. Oncol. 2016, 18, 398–404. [Google Scholar] [CrossRef]
- Engels, B.M.; Schouten, T.G.; van Dullemen, J.; Gosens, I.; Vreugdenhil, E. Functional differences between two DCLK splice variants. Brain Res. Mol. Brain Res. 2004, 120, 103–114. [Google Scholar] [CrossRef]
- Sarkar, S.; O’Connell, M.R.; Okugawa, Y.; Lee, B.S.; Toiyama, Y.; Kusunoki, M.; Daboval, R.D.; Goel, A.; Singh, P. FOXD3 Regulates CSC Marker, DCLK1-S, and Invasive Potential: Prognostic Implications in Colon Cancer. Mol. Cancer Res. 2017, 15, 1678–1691. [Google Scholar] [CrossRef]
- Park, S.Y.; Kim, J.Y.; Choi, J.H.; Kim, J.H.; Lee, C.J.; Singh, P.; Sarkar, S.; Baek, J.H.; Nam, J.S. Inhibition of LEF1-Mediated DCLK1 by Niclosamide Attenuates Colorectal Cancer Stemness. Clin. Cancer Res. 2019, 25, 1415–1429. [Google Scholar] [CrossRef]
- Kalantari, E.; Razmi, M.; Tajik, F.; Asadi-Lari, M.; Ghods, R.; Madjd, Z. Oncogenic functions and clinical significances of DCLK1 isoforms in colorectal cancer: A systematic review and meta-analysis. Cancer Cell Int. 2022, 22, 217. [Google Scholar] [CrossRef]
- Qu, D.; Weygant, N.; Yao, J.; Chandrakesan, P.; Berry, W.L.; May, R.; Pitts, K.; Husain, S.; Lightfoot, S.; Li, M.; et al. Overexpression of DCLK1-AL Increases Tumor Cell Invasion, Drug Resistance, and KRAS Activation and Can Be Targeted to Inhibit Tumorigenesis in Pancreatic Cancer. J. Oncol. 2019, 2019, 6402925. [Google Scholar] [CrossRef] [PubMed]
- Chandrakesan, P.; Panneerselvam, J.; May, R.; Weygant, N.; Qu, D.; Berry, W.R.; Pitts, K.; Stanger, B.Z.; Rao, C.V.; Bronze, M.S.; et al. DCLK1-Isoform2 Alternative Splice Variant Promotes Pancreatic Tumor Immunosuppressive M2-Macrophage Polarization. Mol. Cancer Ther. 2020, 19, 1539–1549. [Google Scholar] [CrossRef] [PubMed]
- Cheng, L.; Yang, Z.; Guo, W.; Wu, C.; Liang, S.; Tong, A.; Cao, Z.; Thorne, R.F.; Yang, S.Y.; Yu, Y.; et al. DCLK1 autoinhibition and activation in tumorigenesis. Innovation 2022, 3, 100191. [Google Scholar] [CrossRef] [PubMed]
- Cao, Z.; Weygant, N.; Chandrakesan, P.; Houchen, C.W.; Peng, J.; Qu, D. Tuft and Cancer Stem Cell Marker DCLK1: A New Target to Enhance Anti-Tumor Immunity in the Tumor Microenvironment. Cancers 2020, 12, 3801. [Google Scholar] [CrossRef]
- Agulto, R.L.; Rogers, M.M.; Tan, T.C.; Ramkumar, A.; Downing, A.M.; Bodin, H.; Castro, J.; Nowakowski, D.W.; Ori-McKenney, K.M. Autoregulatory control of microtubule binding in doublecortin-like kinase 1. eLife 2021, 10, e60126. [Google Scholar] [CrossRef]
- Takiyama, A.; Tanaka, T.; Kazama, S.; Nagata, H.; Kawai, K.; Hata, K.; Otani, K.; Nishikawa, T.; Sasaki, K.; Kaneko, M.; et al. DCLK1 Expression in Colorectal Polyps Increases with the Severity of Dysplasia. In Vivo 2018, 32, 365–371. [Google Scholar] [CrossRef] [PubMed]
- Ge, Y.; Fan, X.; Huang, X.; Weygant, N.; Xiao, Z.; Yan, R.; Liu, H.; Liu, J.; An, G.; Yao, J. DCLK1-Short Splice Variant Promotes Esophageal Squamous Cell Carcinoma Progression via the MAPK/ERK/MMP2 Pathway. Mol. Cancer Res. 2021, 19, 1980–1991. [Google Scholar] [CrossRef]
- Ge, Y.; Liu, H.; Zhang, Y.; Liu, J.; Yan, R.; Xiao, Z.; Fan, X.; Huang, X.; An, G. Inhibition of DCLK1 kinase reverses epithelial-mesenchymal transition and restores T-cell activity in pancreatic ductal adenocarcinoma. Transl. Oncol. 2022, 17, 101317. [Google Scholar] [CrossRef]
- Batlle, E.; Clevers, H. Cancer stem cells revisited. Nat. Med. 2017, 23, 1124–1134. [Google Scholar] [CrossRef]
- Han, H.; Cortez, C.C.; Yang, X.; Nichols, P.W.; Jones, P.A.; Liang, G. DNA methylation directly silences genes with non-CpG island promoters and establishes a nucleosome occupied promoter. Hum. Mol. Genet. 2011, 20, 4299–4310. [Google Scholar] [CrossRef]
- Brown, R.L.; Reinke, L.M.; Damerow, M.S.; Perez, D.; Chodosh, L.A.; Yang, J.; Cheng, C. CD44 splice isoform switching in human and mouse epithelium is essential for epithelial-mesenchymal transition and breast cancer progression. J. Clin. Investig. 2011, 121, 1064–1074. [Google Scholar] [CrossRef]
- Chen, C.; Zhao, S.; Karnad, A.; Freeman, J.W. The biology and role of CD44 in cancer progression: Therapeutic implications. J. Hematol. Oncol. 2018, 11, 64. [Google Scholar] [CrossRef]
- Andresen, K.; Boberg, K.M.; Vedeld, H.M.; Honne, H.; Hektoen, M.; Wadsworth, C.A.; Clausen, O.P.; Karlsen, T.H.; Foss, A.; Mathisen, O.; et al. Novel target genes and a valid biomarker panel identified for cholangiocarcinoma. Epigenetics 2012, 7, 1249–1257. [Google Scholar] [CrossRef] [PubMed]
- Yoo, J.; Jeon, Y.H.; Cho, H.Y.; Lee, S.W.; Kim, G.W.; Lee, D.H.; Kwon, S.H. Advances in Histone Demethylase KDM3A as a Cancer Therapeutic Target. Cancers 2020, 12, 1098. [Google Scholar] [CrossRef] [PubMed]
- Cai, C.; He, H.H.; Chen, S.; Coleman, I.; Wang, H.; Fang, Z.; Chen, S.; Nelson, P.S.; Liu, X.S.; Brown, M.; et al. Androgen receptor gene expression in prostate cancer is directly suppressed by the androgen receptor through recruitment of lysine-specific demethylase 1. Cancer Cell 2011, 20, 457–471. [Google Scholar] [CrossRef]
- Dandawate, P.; Ghosh, C.; Palaniyandi, K.; Paul, S.; Rawal, S.; Pradhan, R.; Sayed, A.A.A.; Choudhury, S.; Standing, D.; Subramaniam, D.; et al. The Histone Demethylase KDM3A, Increased in Human Pancreatic Tumors, Regulates Expression of DCLK1 and Promotes Tumorigenesis in Mice. Gastroenterology 2019, 157, 1646–1659.e11. [Google Scholar] [CrossRef]
- Das, N.D.; Niwa, H.; Umehara, T. Chemical Inhibitors Targeting the Histone Lysine Demethylase Families with Potential for Drug Discovery. Epigenomes 2023, 7, 7. [Google Scholar] [CrossRef]
- Cheng, Y.; He, C.; Wang, M.; Ma, X.; Mo, F.; Yang, S.; Han, J.; Wei, X. Targeting epigenetic regulators for cancer therapy: Mechanisms and advances in clinical trials. Signal Transduct. Target. Ther. 2019, 4, 62. [Google Scholar] [CrossRef]
- Thorpe, L.M.; Yuzugullu, H.; Zhao, J.J. PI3K in cancer: Divergent roles of isoforms, modes of activation and therapeutic targeting. Nat. Rev. Cancer 2015, 15, 7–24. [Google Scholar] [CrossRef]
- Winkler, D.G.; Faia, K.L.; DiNitto, J.P.; Ali, J.A.; White, K.F.; Brophy, E.E.; Pink, M.M.; Proctor, J.L.; Lussier, J.; Martin, C.M.; et al. PI3K-δ and PI3K-γ inhibition by IPI-145 abrogates immune responses and suppresses activity in autoimmune and inflammatory disease models. Chem. Biol. 2013, 20, 1364–1374. [Google Scholar] [CrossRef] [PubMed]
- Bradley, R.; Braybrooke, J.; Gray, R.; Hills, R.; Liu, Z.; Peto, R.; Davies, L.; Dodwell, D.; McGale, P.; Pan, H.; et al. Trastuzumab for early-stage, HER2-positive breast cancer: A meta-analysis of 13,864 women in seven randomised trials. Lancet Oncol. 2021, 22, 1139–1150. [Google Scholar] [CrossRef] [PubMed]
- Amaar, Y.G.; Reeves, M.E. The Role of RASSF1C in the Tumor Microenvironment. Curr. Issues Mol. Biol. 2023, 45, 1113–1126. [Google Scholar] [CrossRef]
- Yang, W.Q.; Zhao, W.J.; Zhu, L.L.; Xu, S.J.; Zhang, X.L.; Liang, Y.; Ding, X.F.; Kiselyov, A.; Chen, G. XMD-17-51 Inhibits DCLK1 Kinase and Prevents Lung Cancer Progression. Front. Pharmacol. 2021, 12, 603453. [Google Scholar] [CrossRef]
- Jang, D.M.; Lim, H.J.; Hahn, H.; Lee, Y.; Kim, H.K.; Kim, H.S. Structural Basis of Inhibition of DCLK1 by Ruxolitinib. Int. J. Mol. Sci. 2021, 22, 8488. [Google Scholar] [CrossRef]
- Sureban, S.M.; May, R.; Weygant, N.; Qu, D.; Chandrakesan, P.; Bannerman-Menson, E.; Ali, N.; Pantazis, P.; Westphalen, C.B.; Wang, T.C.; et al. XMD8-92 inhibits pancreatic tumor xenograft growth via a DCLK1-dependent mechanism. Cancer Lett. 2014, 351, 151–161. [Google Scholar] [CrossRef]
- Weygant, N.; Qu, D.; Berry, W.L.; May, R.; Chandrakesan, P.; Owen, D.B.; Sureban, S.M.; Ali, N.; Janknecht, R.; Houchen, C.W. Small molecule kinase inhibitor LRRK2-IN-1 demonstrates potent activity against colorectal and pancreatic cancer through inhibition of doublecortin-like kinase 1. Mol. Cancer 2014, 13, 103. [Google Scholar] [CrossRef] [PubMed]
- Majchrzak-Celińska, A.; Warych, A.; Szoszkiewicz, M. Novel Approaches to Epigenetic Therapies: From Drug Combinations to Epigenetic Editing. Genes 2021, 12, 208. [Google Scholar] [CrossRef]
- Sahafnejad, Z.; Ramazi, S.; Allahverdi, A. An Update of Epigenetic Drugs for the Treatment of Cancers and Brain Diseases: A Comprehensive Review. Genes 2023, 14, 873. [Google Scholar] [CrossRef]
- Kaminskas, E.; Farrell, A.; Abraham, S.; Baird, A.; Hsieh, L.-S.; Lee, S.-L.; Leighton, J.K.; Patel, H.; Rahman, A.; Sridhara, R.; et al. Approval Summary: Azacitidine for Treatment of Myelodysplastic Syndrome Subtypes. Clin. Cancer Res. 2005, 11, 3604–3608. [Google Scholar] [CrossRef]
- Erdmann, A.; Halby, L.; Fahy, J.; Arimondo, P.B. Targeting DNA methylation with small molecules: What’s next? J. Med. Chem. 2015, 58, 2569–2583. [Google Scholar] [CrossRef] [PubMed]
- Balch, C.; Yan, P.; Craft, T.; Young, S.; Skalnik, D.G.; Huang, T.H.-M.; Nephew, K.P. Antimitogenic and chemosensitizing effects of the methylation inhibitor zebularine in ovarian cancer. Mol. Cancer Ther. 2005, 4, 1505–1514. [Google Scholar] [CrossRef]
- Ma, P.; Jia, G.; Song, Z. Monobenzone, a Novel and Potent KDM1A Inhibitor, Suppresses Migration of Gastric Cancer Cells. Front. Pharmacol. 2021, 12, 640949. [Google Scholar] [CrossRef]
- Li, F.; Qasim, S.; Li, D.; Dou, Q.P. Updated review on green tea polyphenol epigallocatechin-3-gallate as a cancer epigenetic regulator. Semin. Cancer Biol. 2022, 83, 335–352. [Google Scholar] [CrossRef]
- Sultana, S.; Munir, N.; Mahmood, Z.; Riaz, M.; Akram, M.; Rebezov, M.; Kuderinova, N.; Moldabayeva, Z.; Shariati, M.A.; Rauf, A.; et al. Molecular targets for the management of cancer using Curcuma longa Linn. phytoconstituents: A Review. Biomed. Pharmacother. 2021, 135, 111078. [Google Scholar] [CrossRef] [PubMed]
- Mohamadian, M.; Bahrami, A.; Moradi Binabaj, M.; Asgharzadeh, F.; Ferns, G.A. Molecular Targets of Curcumin and Its Therapeutic Potential for Ovarian Cancer. Nutr. Cancer 2022, 74, 2713–2730. [Google Scholar] [CrossRef]
- Chatterjee, B.; Ghosh, K.; Kanade, S.R. Resveratrol modulates epigenetic regulators of promoter histone methylation and acetylation that restores BRCA1, p53, p21CIP1 in human breast cancer cell lines. BioFactors 2019, 45, 818–829. [Google Scholar] [CrossRef]
- Hassan, F.-u.; Rehman, M.S.-u.; Khan, M.S.; Ali, M.A.; Javed, A.; Nawaz, A.; Yang, C. Curcumin as an Alternative Epigenetic Modulator: Mechanism of Action and Potential Effects. Front. Genet. 2019, 10, 514. [Google Scholar] [CrossRef]
- Ming, T.; Tao, Q.; Tang, S.; Zhao, H.; Yang, H.; Liu, M.; Ren, S.; Xu, H. Curcumin: An epigenetic regulator and its application in cancer. Biomed. Pharmacother. 2022, 156, 113956. [Google Scholar] [CrossRef]
- Kantara, C.; O’Connell, M.; Sarkar, S.; Moya, S.; Ullrich, R.; Singh, P. Curcumin promotes autophagic survival of a subset of colon cancer stem cells, which are ablated by DCLK1-siRNA. Cancer Res. 2014, 74, 2487–2498. [Google Scholar] [CrossRef]
- Farhan, M.; Ullah, M.F.; Faisal, M.; Farooqi, A.A.; Sabitaliyevich, U.Y.; Biersack, B.; Ahmad, A. Differential Methylation and Acetylation as the Epigenetic Basis of Resveratrol’s Anticancer Activity. Medicines 2019, 6, 24. [Google Scholar] [CrossRef]
- Fernandes, G.F.S.; Silva, G.D.B.; Pavan, A.R.; Chiba, D.E.; Chin, C.M.; Dos Santos, J.L. Epigenetic Regulatory Mechanisms Induced by Resveratrol. Nutrients 2017, 9, 1201. [Google Scholar] [CrossRef]
- Kumar, A.; Levenson, A.S. Chapter 8—Epigenetic Mechanisms of Resveratrol and Its Analogs in Cancer Prevention and Treatment. In Epigenetics of Cancer Prevention; Bishayee, A., Bhatia, D., Eds.; Academic Press: Cambridge, MA, USA, 2019; Volume 8, pp. 169–186. [Google Scholar]
- Kaufman-Szymczyk, A.; Majewski, G.; Lubecka-Pietruszewska, K.; Fabianowska-Majewska, K. The Role of Sulforaphane in Epigenetic Mechanisms, Including Interdependence between Histone Modification and DNA Methylation. Int. J. Mol. Sci. 2015, 16, 29732–29743. [Google Scholar] [CrossRef] [PubMed]
- Sundaram, M.K.; Ansari, M.Z.; Al Mutery, A.; Ashraf, M.; Nasab, R.; Rai, S.; Rais, N.; Hussain, A. Genistein Induces Alterations of Epigenetic Modulatory Signatures in Human Cervical Cancer Cells. Anticancer Agents Med. Chem. 2018, 18, 412–421. [Google Scholar] [CrossRef]
- Lee, M.; Son, M.; Ryu, E.; Shin, Y.S.; Kim, J.G.; Kang, B.W.; Cho, H.; Kang, H. Quercetin-induced apoptosis prevents EBV infection. Oncotarget 2015, 6, 12603–12624. [Google Scholar] [CrossRef]
- Li, Y.; Wang, Z.; Ajani, J.A.; Song, S. Drug resistance and Cancer stem cells. Cell Commun. Signal. 2021, 19, 19. [Google Scholar] [CrossRef] [PubMed]
- Zhou, H.-M.; Zhang, J.-G.; Zhang, X.; Li, Q. Targeting cancer stem cells for reversing therapy resistance: Mechanism, signaling, and prospective agents. Signal Transduct. Target. Ther. 2021, 6, 62. [Google Scholar] [CrossRef]
- Panneerselvam, J.; Mohandoss, P.; Patel, R.; Gillan, H.; Li, M.; Kumar, K.; Nguyen, D.; Weygant, N.; Qu, D.; Pitts, K.; et al. DCLK1 Regulates Tumor Stemness and Cisplatin Resistance in Non-small Cell Lung Cancer via ABCD-Member-4. Mol. Ther. Oncolytics 2020, 18, 24–36. [Google Scholar] [CrossRef] [PubMed]
- Weygant, N.; Qu, D.; May, R.; Tierney, R.M.; Berry, W.L.; Zhao, L.; Agarwal, S.; Chandrakesan, P.; Chinthalapally, H.R.; Murphy, N.T.; et al. DCLK1 is a broadly dysregulated target against epithelial-mesenchymal transition, focal adhesion, and stemness in clear cell renal carcinoma. Oncotarget 2015, 6, 2193–2205. [Google Scholar] [CrossRef] [PubMed]
- Yan, R.; Fan, X.; Xiao, Z.; Liu, H.; Huang, X.; Liu, J.; Zhang, S.; Yao, J.; An, G.; Ge, Y. Inhibition of DCLK1 sensitizes resistant lung adenocarcinomas to EGFR-TKI through suppression of Wnt/beta-Catenin activity and cancer stemness. Cancer Lett. 2022, 531, 83–97. [Google Scholar] [CrossRef]
- Dawson, M.A.; Kouzarides, T. Cancer epigenetics: From mechanism to therapy. Cell 2012, 150, 12–27. [Google Scholar] [CrossRef] [PubMed]
Drug | Target | Type | Reference |
---|---|---|---|
XMD-17-51 | Kinase Domain | Small molecule | Yang et al., 2021 [124] |
DCLK1-IN-1 | Kinase Domain | Small molecule | Ferguson et al., 2020 [82] |
Ruxolitinib | Kinase Domain | Small molecule | Jang et al., 2021 [125] |
XMD8-92 | Kinase Domain | Small molecule | Sureban et al., 2014 [126] |
LRRK2-IN-1 | Kinase Domain | Small molecule | Weygant et al., 2014 [127] |
CBT-15/CBT-511 | Unique C-terminus of DCLK1 isoforms 2/4 | Monoclonal antibody/CAR-T | Sureban et al., 2019 [83] |
Drug | Target | FDA Approved | Reference |
---|---|---|---|
Azacitidine | DNMT1 | Yes | Kaminskas et al. [130] |
Decitabine | DNMT1 | Yes | Erdmann et al. [131] |
Zebularine | DNMT1 | No | Balch et al. [132] |
Monobenzone | KDM1 | No | Ma et al. [133] |
Epigallocatechin gallate | Broad | No | Li et al. [134] |
Curcumin | Broad | No | Sultana et al. [135], Mohamadian et al. [136] |
Resveratrol | Broad | No | Chatterjee et al. [137] |
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. |
© 2023 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
Moore, L.L.; Houchen, C.W. Epigenetic Landscape and Therapeutic Implication of Gene Isoforms of Doublecortin-Like Kinase 1 for Cancer Stem Cells. Int. J. Mol. Sci. 2023, 24, 16407. https://doi.org/10.3390/ijms242216407
Moore LL, Houchen CW. Epigenetic Landscape and Therapeutic Implication of Gene Isoforms of Doublecortin-Like Kinase 1 for Cancer Stem Cells. International Journal of Molecular Sciences. 2023; 24(22):16407. https://doi.org/10.3390/ijms242216407
Chicago/Turabian StyleMoore, Landon L., and Courtney W. Houchen. 2023. "Epigenetic Landscape and Therapeutic Implication of Gene Isoforms of Doublecortin-Like Kinase 1 for Cancer Stem Cells" International Journal of Molecular Sciences 24, no. 22: 16407. https://doi.org/10.3390/ijms242216407
APA StyleMoore, L. L., & Houchen, C. W. (2023). Epigenetic Landscape and Therapeutic Implication of Gene Isoforms of Doublecortin-Like Kinase 1 for Cancer Stem Cells. International Journal of Molecular Sciences, 24(22), 16407. https://doi.org/10.3390/ijms242216407