DoxoDB: A Database for the Expression Analysis of Doxorubicin-Induced lncRNA Genes
Abstract
:1. Introduction
2. Results
2.1. Differentially Expressed Genes in Human Pulmonary Fibroblasts Treated with DOX
2.2. Differentially Expressed Genes in Human Foreskin Fibroblasts Treated with DOX
2.3. Differentially Expressed Genes in Human Senescent Cells Treated with DOX
2.4. Differentially Expressed Genes in DOX-Resistant Inflammatory Breast Cancer Cells
2.5. Commonly Dysregulated lncRNA and Protein-Coding Genes
2.6. Loss-of-Function Experiments in Human Cardiac Fibroblasts
2.7. The Web Database for Protein-Coding and lncRNA Genes Induced by DOX: DoxoDB
3. Discussion
4. Materials and Methods
4.1. RNA-Seq Data Analysis and Visualization
4.2. Cell Culture and Treatments
4.3. Isolation of Total RNA and RT-PCR
4.4. Transfection of siRNAs
4.5. Determination of Cell Viability and Apoptosis
4.6. Statistics of In Vitro Experiments
4.7. DoxoDB Web Application
Supplementary Materials
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Ritchie, H.; Spooner, F.; Roser, M. Causes of Death. Our World in Data. 2018. Available online: https://ourworldindata.org/causes-of-death (accessed on 3 June 2023).
- Nukala, S.B.; Jousma, J.; Cho, Y.; Lee, W.H.; Ong, S.G. Long non-coding RNAs and microRNAs as crucial regulators in cardio-oncology. Cell Biosci. 2022, 12, 24. [Google Scholar] [CrossRef]
- Wang, Y.; Wang, Y.; Han, X.; Sun, J.; Li, C.; Adhikari, B.K.; Zhang, J.; Miao, X.; Chen, Z. Cardio-Oncology: A Myriad of Relationships between Cardiovascular Disease and Cancer. Front. Cardiovasc. Med. 2022, 9, 727487. [Google Scholar] [CrossRef]
- Ghosh, A.K.; Walker, J.M. Cardio-Oncology—A new subspecialty with collaboration at its heart. Indian Heart J. 2017, 69, 556–562. [Google Scholar] [CrossRef]
- Koutsoukis, A.; Ntalianis, A.; Repasos, E.; Kastritis, E.; Dimopoulos, M.A.; Paraskevaidis, I. Cardio-oncology: A Focus on Cardiotoxicity. Eur. Cardiol. 2018, 13, 64–69. [Google Scholar] [CrossRef]
- Kostakou, P.M.; Kouris, N.T.; Kostopoulos, V.S.; Damaskos, D.S.; Olympios, C.D. Cardio-oncology: A new and developing sector of research and therapy in the field of cardiology. Heart Fail. Rev. 2019, 24, 91–100. [Google Scholar] [CrossRef] [PubMed]
- Ilieva, M.; Panella, R.; Uchida, S. MicroRNAs in Cancer and Cardiovascular Disease. Cells 2022, 11, 3551. [Google Scholar] [CrossRef] [PubMed]
- van Dalen, E.C.; van der Pal, H.J.; Kok, W.E.; Caron, H.N.; Kremer, L.C. Clinical heart failure in a cohort of children treated with anthracyclines: A long-term follow-up study. Eur. J. Cancer 2006, 42, 3191–3198. [Google Scholar] [CrossRef] [PubMed]
- Mancilla, T.R.; Davis, L.R.; Aune, G.J. Doxorubicin-induced p53 interferes with mitophagy in cardiac fibroblasts. PLoS ONE 2020, 15, e0238856. [Google Scholar] [CrossRef]
- Rawat, P.S.; Jaiswal, A.; Khurana, A.; Bhatti, J.S.; Navik, U. Doxorubicin-induced cardiotoxicity: An update on the molecular mechanism and novel therapeutic strategies for effective management. Biomed. Pharmacother. 2021, 139, 111708. [Google Scholar] [CrossRef]
- Zhao, L.; Zhang, B. Doxorubicin induces cardiotoxicity through upregulation of death receptors mediated apoptosis in cardiomyocytes. Sci. Rep. 2017, 7, 44735. [Google Scholar] [CrossRef] [Green Version]
- Mitry, M.A.; Edwards, J.G. Doxorubicin induced heart failure: Phenotype and molecular mechanisms. Int. J. Cardiol. Heart Vasc. 2016, 10, 17–24. [Google Scholar] [CrossRef] [Green Version]
- Aung, L.H.H.; Chen, X.; Cueva Jumbo, J.C.; Li, Z.; Wang, S.Y.; Zhao, C.; Liu, Z.; Wang, Y.; Li, P. Cardiomyocyte mitochondrial dynamic-related lncRNA 1 (CMDL-1) may serve as a potential therapeutic target in doxorubicin cardiotoxicity. Mol. Ther. Nucleic Acids 2021, 25, 638–651. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; Li, L.; Li, X.; Wu, S. Long noncoding RNA LINC00339 aggravates doxorubicin-induced cardiomyocyte apoptosis by targeting MiR-484. Biochem. Biophys. Res. Commun. 2018, 503, 3038–3043. [Google Scholar] [CrossRef] [PubMed]
- Maejima, Y.; Adachi, S.; Ito, H.; Hirao, K.; Isobe, M. Induction of premature senescence in cardiomyocytes by doxorubicin as a novel mechanism of myocardial damage. Aging Cell 2008, 7, 125–136. [Google Scholar] [CrossRef] [PubMed]
- Zhan, J.; Hu, P.; Wang, Y. lncRNA PVT1 aggravates doxorubicin-induced cardiomyocyte apoptosis by targeting the miR-187-3p/AGO1 axis. Mol. Cell. Probes 2020, 49, 101490. [Google Scholar] [CrossRef] [PubMed]
- Zhang, S.; Yuan, Y.; Zhang, Z.; Guo, J.; Li, J.; Zhao, K.; Qin, Y.; Qiu, C. LncRNA FOXC2-AS1 protects cardiomyocytes from doxorubicin-induced cardiotoxicity through activation of WNT1-inducible signaling pathway protein-1. Biosci. Biotechnol. Biochem. 2019, 83, 653–658. [Google Scholar] [CrossRef]
- Fredj, S.; Bescond, J.; Louault, C.; Delwail, A.; Lecron, J.C.; Potreau, D. Role of interleukin-6 in cardiomyocyte/cardiac fibroblast interactions during myocyte hypertrophy and fibroblast proliferation. J. Cell. Physiol. 2005, 204, 428–436. [Google Scholar] [CrossRef]
- Porter, K.E.; Turner, N.A. Cardiac fibroblasts: At the heart of myocardial remodeling. Pharmacol. Ther. 2009, 123, 255–278. [Google Scholar] [CrossRef]
- Rog-Zielinska, E.A.; Norris, R.A.; Kohl, P.; Markwald, R. The Living Scar—Cardiac Fibroblasts and the Injured Heart. Trends Mol. Med. 2016, 22, 99–114. [Google Scholar] [CrossRef] [Green Version]
- Beermann, J.; Piccoli, M.T.; Viereck, J.; Thum, T. Non-coding RNAs in Development and Disease: Background, Mechanisms, and Therapeutic Approaches. Physiol. Rev. 2016, 96, 1297–1325. [Google Scholar] [CrossRef] [Green Version]
- Chatterjee, S.; Gupta, S.K.; Bar, C.; Thum, T. Noncoding RNAs: Potential regulators in cardioncology. Am. J. Physiol. Heart Circ. Physiol. 2019, 316, H160–H168. [Google Scholar] [CrossRef]
- Lander, E.S.; Linton, L.M.; Birren, B.; Nusbaum, C.; Zody, M.C.; Baldwin, J.; Devon, K.; Dewar, K.; Doyle, M.; FitzHugh, W.; et al. Initial sequencing and analysis of the human genome. Nature 2001, 409, 860–921. [Google Scholar]
- Mercer, T.R.; Dinger, M.E.; Mattick, J.S. Long non-coding RNAs: Insights into functions. Nat. Rev. Genet. 2009, 10, 155–159. [Google Scholar] [CrossRef]
- Li, H.; Liu, X.; Zhang, L.; Li, X. LncRNA BANCR facilitates vascular smooth muscle cell proliferation and migration through JNK pathway. Oncotarget 2017, 8, 114568–114575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Prensner, J.R.; Chinnaiyan, A.M. The emergence of lncRNAs in cancer biology. Cancer Discov. 2011, 1, 391–407. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, W.J.; Wang, Y.M.; Hu, Y.; Lin, Q.; Chen, R.; Liu, H.; Cao, W.Z.; Zhu, H.F.; Tong, C.; Li, L.; et al. HDncRNA: A comprehensive database of non-coding RNAs associated with heart diseases. Database 2018, 2018, bay067. [Google Scholar] [CrossRef] [Green Version]
- Jiang, S.; Li, Z.; Dou, R.; Lin, Z.; Zhang, J.; Zhang, W.; Chen, Z.; Shen, X.; Ji, J.; Qu, M.; et al. Construction and validation of a novel cuproptosis-related long noncoding RNA signature for predicting the outcome of prostate cancer. Front. Genet. 2022, 13, 976850. [Google Scholar] [CrossRef] [PubMed]
- Lu, P.; Jiang, Y.; Xia, Z. Long noncoding RNA TUG1 decreases bladder cancer chemo-sensitivity toward doxorubicin through elevating KPNA2 expression and activating the PI3K/AKT pathway via adsorbing miR-582-5p. Anticancer Drugs 2023, 34, 144–154. [Google Scholar] [CrossRef]
- Xiong, M.; Wu, M.; Dan, P.; Huang, W.; Chen, Z.; Ke, H.; Chen, Z.; Song, W.; Zhao, Y.; Xiang, A.P.; et al. LncRNA DANCR represses Doxorubicin-induced apoptosis through stabilizing MALAT1 expression in colorectal cancer cells. Cell Death Dis. 2021, 12, 24. [Google Scholar] [CrossRef] [PubMed]
- Schmitt, A.M.; Garcia, J.T.; Hung, T.; Flynn, R.A.; Shen, Y.; Qu, K.; Payumo, A.Y.; Peres-da-Silva, A.; Broz, D.K.; Baum, R.; et al. An inducible long noncoding RNA amplifies DNA damage signaling. Nat. Genet. 2016, 48, 1370–1376. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fu, X.; Ravindranath, L.; Tran, N.; Petrovics, G.; Srivastava, S. Regulation of apoptosis by a prostate-specific and prostate cancer-associated noncoding gene, PCGEM1. DNA Cell Biol. 2006, 25, 135–141. [Google Scholar] [CrossRef]
- Li, X.L.; Subramanian, M.; Jones, M.F.; Chaudhary, R.; Singh, D.K.; Zong, X.; Gryder, B.; Sindri, S.; Mo, M.; Schetter, A.; et al. Long Noncoding RNA PURPL Suppresses Basal p53 Levels and Promotes Tumorigenicity in Colorectal Cancer. Cell Rep. 2017, 20, 2408–2423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cai, H.; Tian, P.; Ju, J.; Wang, T.; Chen, X.; Wang, K.; Wang, F.; Yu, X.; Wang, S.; Wang, Y.; et al. Long noncoding RNA NONMMUT015745 inhibits doxorubicin-mediated cardiomyocyte apoptosis by regulating Rab2A-p53 axis. Cell Death Discov. 2022, 8, 364. [Google Scholar] [CrossRef] [PubMed]
- Wang, H.; Lin, X.; Li, J.; Zeng, G.; Xu, T. Long Noncoding RNA SOX2-OT Aggravates Doxorubicin-Induced Apoptosis of Cardiomyocyte by Targeting miR-942-5p/DP5. Drug Des. Devel. Ther. 2021, 15, 481–492. [Google Scholar] [CrossRef]
- Xia, W.; Chen, H.; Xie, C.; Hou, M. Long-noncoding RNA MALAT1 sponges microRNA-92a-3p to inhibit doxorubicin-induced cardiac senescence by targeting ATG4a. Aging 2020, 12, 8241–8260. [Google Scholar] [CrossRef] [PubMed]
- Xie, Z.; Xia, W.; Hou, M. Long intergenic non-coding RNA-p21 mediates cardiac senescence via the Wnt/beta-catenin signaling pathway in doxorubicin-induced cardiotoxicity. Mol. Med. Rep. 2018, 17, 2695–2704. [Google Scholar] [PubMed] [Green Version]
- Dimitrova, N.; Zamudio, J.R.; Jong, R.M.; Soukup, D.; Resnick, R.; Sarma, K.; Ward, A.J.; Raj, A.; Lee, J.T.; Sharp, P.A.; et al. LincRNA-p21 activates p21 in cis to promote Polycomb target gene expression and to enforce the G1/S checkpoint. Mol. Cell 2014, 54, 777–790. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pesce, M.; Duda, G.N.; Forte, G.; Girao, H.; Raya, A.; Roca-Cusachs, P.; Sluijter, J.P.G.; Tschope, C.; Van Linthout, S. Cardiac fibroblasts and mechanosensation in heart development, health and disease. Nat. Rev. Cardiol. 2023, 20, 309–324. [Google Scholar] [CrossRef] [PubMed]
- Ruopp, N.F.; Cockrill, B.A. Diagnosis and Treatment of Pulmonary Arterial Hypertension: A Review. JAMA 2022, 327, 1379–1391. [Google Scholar] [CrossRef]
- Rajagopal, K.; Bryant, A.J.; Sahay, S.; Wareing, N.; Zhou, Y.; Pandit, L.M.; Karmouty-Quintana, H. Idiopathic pulmonary fibrosis and pulmonary hypertension: Heracles meets the Hydra. Br. J. Pharmacol. 2021, 178, 172–186. [Google Scholar] [CrossRef] [PubMed]
- Hennigs, J.K.; Cao, A.; Li, C.G.; Shi, M.; Mienert, J.; Miyagawa, K.; Korbelin, J.; Marciano, D.P.; Chen, P.I.; Roughley, M.; et al. PPARgamma-p53-Mediated Vasculoregenerative Program to Reverse Pulmonary Hypertension. Circ. Res. 2021, 128, 401–418. [Google Scholar] [CrossRef] [PubMed]
- Ilieva, M.; Miller, H.E.; Agarwal, A.; Paulus, G.K.; Madsen, J.H.; Bishop, A.J.R.; Kauppinen, S.; Uchida, S. FibroDB: Expression Analysis of Protein-Coding and Long Non-Coding RNA Genes in Fibrosis. Non-Coding RNA 2022, 8, 13. [Google Scholar] [CrossRef] [PubMed]
- Wang, F.; Liang, R.; Soibam, B.; Yang, J.; Liu, Y. Coregulatory long non-coding RNA and protein-coding genes in serum starved cells. Biochim. Biophys. Acta Gene Regul. Mech. 2019, 1862, 84–95. [Google Scholar] [CrossRef] [PubMed]
- Yang, F.; Kemp, C.J.; Henikoff, S. Doxorubicin enhances nucleosome turnover around promoters. Curr. Biol. 2013, 23, 782–787. [Google Scholar] [CrossRef] [Green Version]
- Pang, B.; Qiao, X.; Janssen, L.; Velds, A.; Groothuis, T.; Kerkhoven, R.; Nieuwland, M.; Ovaa, H.; Rottenberg, S.; van Tellingen, O.; et al. Drug-induced histone eviction from open chromatin contributes to the chemotherapeutic effects of doxorubicin. Nat. Commun. 2013, 4, 1908. [Google Scholar] [CrossRef] [Green Version]
- Yang, F.; Teves, S.S.; Kemp, C.J.; Henikoff, S. Doxorubicin, DNA torsion, and chromatin dynamics. Biochim. Biophys. Acta 2014, 1845, 84–89. [Google Scholar] [CrossRef] [Green Version]
- Schade, A.E.; Fischer, M.; DeCaprio, J.A. RB, p130 and p107 differentially repress G1/S and G2/M genes after p53 activation. Nucleic Acids Res. 2019, 47, 11197–11208. [Google Scholar] [CrossRef]
- Wang, T.W.; Johmura, Y.; Suzuki, N.; Omori, S.; Migita, T.; Yamaguchi, K.; Hatakeyama, S.; Yamazaki, S.; Shimizu, E.; Imoto, S.; et al. Blocking PD-L1-PD-1 improves senescence surveillance and ageing phenotypes. Nature 2022, 611, 358–364. [Google Scholar] [CrossRef]
- Anderson, R.; Lagnado, A.; Maggiorani, D.; Walaszczyk, A.; Dookun, E.; Chapman, J.; Birch, J.; Salmonowicz, H.; Ogrodnik, M.; Jurk, D.; et al. Length-independent telomere damage drives post-mitotic cardiomyocyte senescence. EMBO J. 2019, 38, e100492. [Google Scholar] [CrossRef]
- Booth, L.K.; Redgrave, R.E.; Folaranmi, O.; Gill, J.H.; Richardson, G.D. Anthracycline-induced cardiotoxicity and senescence. Front. Aging 2022, 3, 1058435. [Google Scholar] [CrossRef]
- Demaria, M.; O’Leary, M.N.; Chang, J.; Shao, L.; Liu, S.; Alimirah, F.; Koenig, K.; Le, C.; Mitin, N.; Deal, A.M.; et al. Cellular Senescence Promotes Adverse Effects of Chemotherapy and Cancer Relapse. Cancer Discov. 2017, 7, 165–176. [Google Scholar] [CrossRef] [Green Version]
- Walaszczyk, A.; Dookun, E.; Redgrave, R.; Tual-Chalot, S.; Victorelli, S.; Spyridopoulos, I.; Owens, A.; Arthur, H.M.; Passos, J.F.; Richardson, G.D. Pharmacological clearance of senescent cells improves survival and recovery in aged mice following acute myocardial infarction. Aging Cell 2019, 18, e12945. [Google Scholar] [CrossRef]
- Dookun, E.; Passos, J.F.; Arthur, H.M.; Richardson, G.D. Therapeutic Potential of Senolytics in Cardiovascular Disease. Cardiovasc. Drugs Ther. 2022, 36, 187–196. [Google Scholar] [CrossRef]
- Bientinesi, E.; Lulli, M.; Becatti, M.; Ristori, S.; Margheri, F.; Monti, D. Doxorubicin-induced senescence in normal fibroblasts promotes in vitro tumour cell growth and invasiveness: The role of Quercetin in modulating these processes. Mech. Ageing Dev. 2022, 206, 111689. [Google Scholar] [CrossRef]
- Espitia-Corredor, J.A.; Shamoon, L.; Olivares-Silva, F.; Rimassa-Tare, C.; Munoz-Rodriguez, C.; Espinoza-Perez, C.; Sanchez-Ferrer, C.F.; Peiro, C.; Diaz-Araya, G. Resolvin E1 attenuates doxorubicin-induced cardiac fibroblast senescence: A key role for IL-1beta. Biochim. Biophys. Acta Mol. Basis Dis. 2022, 1868, 166525. [Google Scholar] [CrossRef] [PubMed]
- Coppe, J.P.; Desprez, P.Y.; Krtolica, A.; Campisi, J. The senescence-associated secretory phenotype: The dark side of tumor suppression. Annu. Rev. Pathol. 2010, 5, 99–118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Davalos, A.R.; Coppe, J.P.; Campisi, J.; Desprez, P.Y. Senescent cells as a source of inflammatory factors for tumor progression. Cancer Metastasis Rev. 2010, 29, 273–283. [Google Scholar] [CrossRef] [Green Version]
- Krtolica, A.; Campisi, J. Cancer and aging: A model for the cancer promoting effects of the aging stroma. Int. J. Biochem. Cell Biol. 2002, 34, 1401–1414. [Google Scholar] [CrossRef]
- Levi, N.; Papismadov, N.; Solomonov, I.; Sagi, I.; Krizhanovsky, V. The ECM path of senescence in aging: Components and modifiers. FEBS J. 2020, 287, 2636–2646. [Google Scholar] [CrossRef] [Green Version]
- Wang, C.; Jurk, D.; Maddick, M.; Nelson, G.; Martin-Ruiz, C.; von Zglinicki, T. DNA damage response and cellular senescence in tissues of aging mice. Aging Cell 2009, 8, 311–323. [Google Scholar] [CrossRef] [PubMed]
- Misawa, T.; Tanaka, Y.; Okada, R.; Takahashi, A. Biology of extracellular vesicles secreted from senescent cells as senescence-associated secretory phenotype factors. Geriatr. Gerontol. Int. 2020, 20, 539–546. [Google Scholar] [CrossRef] [PubMed]
- Shin, H.J.; Kwon, H.K.; Lee, J.H.; Gui, X.; Achek, A.; Kim, J.H.; Choi, S. Doxorubicin-induced necrosis is mediated by poly-(ADP-ribose) polymerase 1 (PARP1) but is independent of p53. Sci. Rep. 2015, 5, 15798. [Google Scholar] [CrossRef] [Green Version]
- Arata, Y.; Watanabe, A.; Motosugi, R.; Murakami, R.; Goto, T.; Hori, S.; Hirayama, S.; Hamazaki, J.; Murata, S. Defective induction of the proteasome associated with T-cell receptor signaling underlies T-cell senescence. Genes Cells 2019, 24, 801–813. [Google Scholar] [CrossRef] [PubMed]
- Liu, J.; Zheng, H.; Tang, M.; Ryu, Y.C.; Wang, X. A therapeutic dose of doxorubicin activates ubiquitin-proteasome system-mediated proteolysis by acting on both the ubiquitination apparatus and proteasome. Am. J. Physiol. Heart Circ. Physiol. 2008, 295, H2541–H2550. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ranek, M.J.; Wang, X. Activation of the ubiquitin-proteasome system in doxorubicin cardiomyopathy. Curr. Hypertens. Rep. 2009, 11, 389–395. [Google Scholar] [CrossRef] [Green Version]
- Bhagat, A.; Shrestha, P.; Kleinerman, E.S. The Innate Immune System in Cardiovascular Diseases and Its Role in Doxorubicin-Induced Cardiotoxicity. Int. J. Mol. Sci. 2022, 23, 14649. [Google Scholar] [CrossRef] [PubMed]
- Luu, A.Z.; Chowdhury, B.; Al-Omran, M.; Teoh, H.; Hess, D.A.; Verma, S. Role of Endothelium in Doxorubicin-Induced Cardiomyopathy. JACC Basic Transl. Sci. 2018, 3, 861–870. [Google Scholar] [CrossRef]
- Marchant, D.J.; Boyd, J.H.; Lin, D.C.; Granville, D.J.; Garmaroudi, F.S.; McManus, B.M. Inflammation in myocardial diseases. Circ. Res. 2012, 110, 126–144. [Google Scholar] [CrossRef]
- Todorova, V.K.; Hsu, P.C.; Wei, J.Y.; Lopez-Candales, A.; Chen, J.Z.; Su, L.J.; Makhoul, I. Biomarkers of inflammation, hypercoagulability and endothelial injury predict early asymptomatic doxorubicin-induced cardiotoxicity in breast cancer patients. Am. J. Cancer Res. 2020, 10, 2933–2945. [Google Scholar]
- Stevens, L.E.; Peluffo, G.; Qiu, X.; Temko, D.; Fassl, A.; Li, Z.; Trinh, A.; Seehawer, M.; Jovanovic, B.; Aleckovic, M.; et al. JAK-STAT Signaling in Inflammatory Breast Cancer Enables Chemotherapy-Resistant Cell States. Cancer Res. 2023, 83, 264–284. [Google Scholar] [CrossRef]
- Askarian-Amiri, M.E.; Seyfoddin, V.; Smart, C.E.; Wang, J.; Kim, J.E.; Hansji, H.; Baguley, B.C.; Finlay, G.J.; Leung, E.Y. Emerging role of long non-coding RNA SOX2OT in SOX2 regulation in breast cancer. PLoS ONE 2014, 9, e102140. [Google Scholar] [CrossRef]
- Iranpour, M.; Soudyab, M.; Geranpayeh, L.; Mirfakhraie, R.; Azargashb, E.; Movafagh, A.; Ghafouri-Fard, S. Expression analysis of four long noncoding RNAs in breast cancer. Tumor Biol. 2016, 37, 2933–2940. [Google Scholar] [CrossRef] [PubMed]
- Martin, F.J.; Amode, M.R.; Aneja, A.; Austine-Orimoloye, O.; Azov, A.G.; Barnes, I.; Becker, A.; Bennett, R.; Berry, A.; Bhai, J.; et al. Ensembl 2023. Nucleic Acids Res. 2023, 51, D933–D941. [Google Scholar] [CrossRef] [PubMed]
- Han, W.; Yu, G.; Meng, X.; Hong, H.; Zheng, L.; Wu, X.; Zhang, D.; Yan, B.; Ma, Y.; Li, X.; et al. Potential of C1QTNF1-AS1 regulation in human hepatocellular carcinoma. Mol. Cell. Biochem. 2019, 460, 37–51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, H.; Zhang, B.; Ding, M.; Lu, S.; Zhou, H.; Sun, D.; Wu, G.; Gan, X. C1QTNF1-AS1 regulates the occurrence and development of hepatocellular carcinoma by regulating miR-221-3p/SOCS3. Hepatol. Int. 2019, 13, 277–292. [Google Scholar] [CrossRef] [PubMed]
- Zhong, Q.; Lu, M.; Yuan, W.; Cui, Y.; Ouyang, H.; Fan, Y.; Wang, Z.; Wu, C.; Qiao, J.; Hang, J. Eight-lncRNA signature of cervical cancer were identified by integrating DNA methylation, copy number variation and transcriptome data. J. Transl. Med. 2021, 19, 58. [Google Scholar] [CrossRef]
- Ciocan-Cartita, C.A.; Jurj, A.; Zanoaga, O.; Cojocneanu, R.; Pop, L.A.; Moldovan, A.; Moldovan, C.; Zimta, A.A.; Raduly, L.; Pop-Bica, C.; et al. New insights in gene expression alteration as effect of doxorubicin drug resistance in triple negative breast cancer cells. J. Exp. Clin. Cancer Res. 2020, 39, 241. [Google Scholar] [CrossRef]
- Chen, T.; Guo, Y.; Wang, J.; Ai, L.; Ma, L.; He, W.; Li, Z.; Yu, X.; Li, J.; Fan, X.; et al. LncRNA CTD-2528L19.6 prevents the progression of IPF by alleviating fibroblast activation. Cell Death Dis. 2021, 12, 600. [Google Scholar] [CrossRef]
- Qian, C.; Li, H.; Chang, D.; Wei, B.; Wang, Y. Identification of functional lncRNAs in atrial fibrillation by integrative analysis of the lncRNA-mRNA network based on competing endogenous RNAs hypothesis. J. Cell. Physiol. 2019, 234, 11620–11630. [Google Scholar] [CrossRef]
- Zhang, G.; Sun, H.; Zhang, Y.; Zhao, H.; Fan, W.; Li, J.; Lv, Y.; Song, Q.; Li, J.; Zhang, M.; et al. Characterization of dysregulated lncRNA-mRNA network based on ceRNA hypothesis to reveal the occurrence and recurrence of myocardial infarction. Cell Death Discov. 2018, 4, 35. [Google Scholar] [CrossRef] [Green Version]
- Jacobsen, A.; Bosch, L.J.W.; Martens-de Kemp, S.R.; Carvalho, B.; Sillars-Hardebol, A.H.; Dobson, R.J.; de Rinaldis, E.; Meijer, G.A.; Abeln, S.; Heringa, J.; et al. Aurora kinase A (AURKA) interaction with Wnt and Ras-MAPK signalling pathways in colorectal cancer. Sci. Rep. 2018, 8, 7522. [Google Scholar] [CrossRef] [Green Version]
- Guo, K.; Chen, L.; Wang, Y.; Qian, K.; Zheng, X.; Sun, W.; Sun, T.; Wu, Y.; Wang, Z. Long noncoding RNA RP11-547D24.1 regulates proliferation and migration in papillary thyroid carcinoma: Identification and validation of a novel long noncoding RNA through integrated analysis of TCGA database. Cancer Med. 2019, 8, 3105–3119. [Google Scholar] [CrossRef] [Green Version]
- Wang, P.; Cui, J.; Wen, J.; Guo, Y.; Zhang, L.; Chen, X. Cisplatin induces HepG2 cell cycle arrest through targeting specific long noncoding RNAs and the p53 signaling pathway. Oncol. Lett. 2016, 12, 4605–4612. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, C.; Wang, X.; Liu, T.; Tang, X.; Liu, Y.; Liu, T.; Zhu, J. Overexpression of long non-coding RNA RP11-363E7.4 inhibits proliferation and invasion in gastric cancer. Cell Biochem. Funct. 2020, 38, 921–931. [Google Scholar] [CrossRef] [Green Version]
- Wang, P.; Li, J.; Zhao, W.; Shang, C.; Jiang, X.; Wang, Y.; Zhou, B.; Bao, F.; Qiao, H. A Novel LncRNA-miRNA-mRNA Triple Network Identifies LncRNA RP11-363E7.4 as An Important Regulator of miRNA and Gene Expression in Gastric Cancer. Cell. Physiol. Biochem. 2018, 47, 1025–1041. [Google Scholar] [CrossRef]
- Gu, J.; Li, Y.; Fan, L.; Zhao, Q.; Tan, B.; Hua, K.; Wu, G. Identification of aberrantly expressed long non-coding RNAs in stomach adenocarcinoma. Oncotarget 2017, 8, 49201–49216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mathias, C.; Muzzi, J.C.D.; Antunes, B.B.; Gradia, D.F.; Castro, M.A.A.; Carvalho de Oliveira, J. Unraveling Immune-Related lncRNAs in Breast Cancer Molecular Subtypes. Front. Oncol. 2021, 11, 692170. [Google Scholar] [CrossRef] [PubMed]
- Bhat, A.A.; Syed, N.; Therachiyil, L.; Nisar, S.; Hashem, S.; Macha, M.A.; Yadav, S.K.; Krishnankutty, R.; Muralitharan, S.; Al-Naemi, H.; et al. Claudin-1, A Double-Edged Sword in Cancer. Int. J. Mol. Sci. 2020, 21, 569. [Google Scholar] [CrossRef] [Green Version]
- Akizuki, R.; Maruhashi, R.; Eguchi, H.; Kitabatake, K.; Tsukimoto, M.; Furuta, T.; Matsunaga, T.; Endo, S.; Ikari, A. Decrease in paracellular permeability and chemosensitivity to doxorubicin by claudin-1 in spheroid culture models of human lung adenocarcinoma A549 cells. Biochim. Biophys. Acta Mol. Cell Res. 2018, 1865, 769–780. [Google Scholar] [CrossRef]
- Nasako, H.; Takashina, Y.; Eguchi, H.; Ito, A.; Ishikawa, Y.; Matsunaga, T.; Endo, S.; Ikari, A. Increase in Toxicity of Anticancer Drugs by PMTPV, a Claudin-1-Binding Peptide, Mediated via Down-Regulation of Claudin-1 in Human Lung Adenocarcinoma A549 Cells. Int. J. Mol. Sci. 2020, 21, 5909. [Google Scholar] [CrossRef]
- Kim, H.; Kim, S.H.; Hwang, D.; An, J.; Chung, H.S.; Yang, E.G.; Kim, S.Y. Extracellular pyruvate kinase M2 facilitates cell migration by upregulating claudin-1 expression in colon cancer cells. Biochem. Cell Biol. 2020, 98, 219–226. [Google Scholar] [CrossRef]
- Mahati, S.; Bolati, D.; Yang, Y.; Mao, R.; Zhang, H.; Bao, Y. TMPRSS4 promotes cancer stem cell traits by regulating CLDN1 in hepatocellular carcinoma. Biochem. Biophys. Res. Commun. 2017, 490, 906–912. [Google Scholar] [CrossRef]
- Mattern, J.; Roghi, C.S.; Hurtz, M.; Knauper, V.; Edwards, D.R.; Poghosyan, Z. ADAM15 mediates upregulation of Claudin-1 expression in breast cancer cells. Sci. Rep. 2019, 9, 12540. [Google Scholar] [CrossRef] [Green Version]
- Wang, D.W.; Zhang, W.H.; Danil, G.; Yang, K.; Hu, J.K. The role and mechanism of claudins in cancer. Front. Oncol. 2022, 12, 1051497. [Google Scholar] [CrossRef] [PubMed]
- Sun, Z.; Li, Y.; Tan, X.; Liu, W.; He, X.; Pan, D.; Li, E.; Xu, L.; Long, L. Friend or Foe: Regulation, Downstream Effectors of RRAD in Cancer. Biomolecules 2023, 13, 477. [Google Scholar] [CrossRef] [PubMed]
- Finlin, B.S.; Crump, S.M.; Satin, J.; Andres, D.A. Regulation of voltage-gated calcium channel activity by the Rem and Rad GTPases. Proc. Natl. Acad. Sci. USA 2003, 100, 14469–14474. [Google Scholar] [CrossRef]
- Li, Y.; Chang, Y.; Li, X.; Li, X.; Gao, J.; Zhou, Y.; Wu, F.; Bai, R.; Dong, T.; Ma, S.; et al. RAD-Deficient Human Cardiomyocytes Develop Hypertrophic Cardiomyopathy Phenotypes Due to Calcium Dysregulation. Front. Cell Dev. Biol. 2020, 8, 585879. [Google Scholar] [CrossRef]
- Maguire, J.; Santoro, T.; Jensen, P.; Siebenlist, U.; Yewdell, J.; Kelly, K. Gem: An induced, immediate early protein belonging to the Ras family. Science 1994, 265, 241–244. [Google Scholar] [CrossRef] [Green Version]
- Reynet, C.; Kahn, C.R. Rad: A member of the Ras family overexpressed in muscle of type II diabetic humans. Science 1993, 262, 1441–1444. [Google Scholar] [CrossRef] [PubMed]
- Chang, L.; Zhang, J.; Tseng, Y.H.; Xie, C.Q.; Ilany, J.; Bruning, J.C.; Sun, Z.; Zhu, X.; Cui, T.; Youker, K.A.; et al. Rad GTPase deficiency leads to cardiac hypertrophy. Circulation 2007, 116, 2976–2983. [Google Scholar] [CrossRef] [Green Version]
- Zhang, J.; Chang, L.; Chen, C.; Zhang, M.; Luo, Y.; Hamblin, M.; Villacorta, L.; Xiong, J.W.; Chen, Y.E.; Zhang, J.; et al. Rad GTPase inhibits cardiac fibrosis through connective tissue growth factor. Cardiovasc. Res. 2011, 91, 90–98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nanasi, P., Jr.; Imre, L.; Firouzi Niaki, E.; Bosire, R.; Mocsar, G.; Turk-Mazlo, A.; Ausio, J.; Szabo, G. Doxorubicin induces large-scale and differential H2A and H2B redistribution in live cells. PLoS ONE 2020, 15, e0231223. [Google Scholar] [CrossRef] [PubMed]
- Braunstein, M.; Liao, L.; Lyttle, N.; Lobo, N.; Taylor, K.J.; Krzyzanowski, P.M.; Kalatskaya, I.; Yao, C.Q.; Stein, L.D.; Boutros, P.C.; et al. Downregulation of histone H2A and H2B pathways is associated with anthracycline sensitivity in breast cancer. Breast Cancer Res. 2016, 18, 16. [Google Scholar] [CrossRef]
- Kuznetsov, A.V.; Margreiter, R.; Amberger, A.; Saks, V.; Grimm, M. Changes in mitochondrial redox state, membrane potential and calcium precede mitochondrial dysfunction in doxorubicin-induced cell death. Biochim. Biophys. Acta 2011, 1813, 1144–1152. [Google Scholar] [CrossRef]
- Li, Z.; Liu, L.; Feng, C.; Qin, Y.; Xiao, J.; Zhang, Z.; Ma, L. LncBook 2.0: Integrating human long non-coding RNAs with multi-omics annotations. Nucleic Acids Res. 2023, 51, D186–D191. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Yang, L.; Chen, L.L. Life without A tail: New formats of long noncoding RNAs. Int. J. Biochem. Cell Biol. 2014, 54, 338–349. [Google Scholar] [CrossRef]
- Ilieva, M.; Dao, J.; Miller, H.E.; Madsen, J.H.; Bishop, A.J.R.; Kauppinen, S.; Uchida, S. Systematic Analysis of Long Non-Coding RNA Genes in Nonalcoholic Fatty Liver Disease. Non-Coding RNA 2022, 8, 56. [Google Scholar] [CrossRef]
- Leinonen, R.; Sugawara, H.; Shumway, M.; International Nucleotide Sequence Database, C. The sequence read archive. Nucleic Acids Res. 2011, 39, D19–D21. [Google Scholar] [CrossRef] [Green Version]
- Chen, S.; Zhou, Y.; Chen, Y.; Gu, J. fastp: An ultra-fast all-in-one FASTQ preprocessor. Bioinformatics 2018, 34, i884–i890. [Google Scholar] [CrossRef]
- Dobin, A.; Davis, C.A.; Schlesinger, F.; Drenkow, J.; Zaleski, C.; Jha, S.; Batut, P.; Chaisson, M.; Gingeras, T.R. STAR: Ultrafast universal RNA-seq aligner. Bioinformatics 2013, 29, 15–21. [Google Scholar] [CrossRef]
- Robinson, M.D.; McCarthy, D.J.; Smyth, G.K. edgeR: A Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 2010, 26, 139–140. [Google Scholar] [CrossRef] [Green Version]
- Wickham, H. ggplot2, 1st ed.; Springer: New York, NY, USA, 2009; p. 213. [Google Scholar] [CrossRef]
- Chen, H.; Boutros, P.C. VennDiagram: A package for the generation of highly-customizable Venn and Euler diagrams in R. BMC Bioinform. 2011, 12, 35. [Google Scholar] [CrossRef] [Green Version]
- Conway, J.R.; Lex, A.; Gehlenborg, N. UpSetR: An R package for the visualization of intersecting sets and their properties. Bioinformatics 2017, 33, 2938–2940. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huang, D.W.; Sherman, B.T.; Lempicki, R.A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 2009, 4, 44–57. [Google Scholar] [CrossRef] [PubMed]
- Lawrence, M.; Huber, W.; Pages, H.; Aboyoun, P.; Carlson, M.; Gentleman, R.; Morgan, M.T.; Carey, V.J. Software for computing and annotating genomic ranges. PLoS Comput. Biol. 2013, 9, e1003118. [Google Scholar] [CrossRef]
- Untergasser, A.; Cutcutache, I.; Koressaar, T.; Ye, J.; Faircloth, B.C.; Remm, M.; Rozen, S.G. Primer3—New capabilities and interfaces. Nucleic Acids Res. 2012, 40, e115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chang, W.; Cheng, J.; Allaire, J.J.; Sievert, C.; Schloerke, B.; Xie, Y.; Allen, J.; McPherson, J.; Dipert, A.; Borges, B. shiny: Web Application Framework for R. Available online: https://shiny.rstudio.com/ (accessed on 21 March 2023).
- Gu, Z.; Eils, R.; Schlesner, M. Complex heatmaps reveal patterns and correlations in multidimensional genomic data. Bioinformatics 2016, 32, 2847–2849. [Google Scholar] [CrossRef] [Green Version]
- Raudvere, U.; Kolberg, L.; Kuzmin, I.; Arak, T.; Adler, P.; Peterson, H.; Vilo, J. g:Profiler: A web server for functional enrichment analysis and conversions of gene lists (2019 update). Nucleic Acids Res. 2019, 47, W191–W198. [Google Scholar] [CrossRef] [Green Version]
- Wu, T.; Hu, E.; Xu, S.; Chen, M.; Guo, P.; Dai, Z.; Feng, T.; Zhou, L.; Tang, W.; Zhan, L.; et al. clusterProfiler 4.0: A universal enrichment tool for interpreting omics data. Innovation 2021, 2, 100141. [Google Scholar] [CrossRef]
- Yu, G.; Wang, L.G.; Han, Y.; He, Q.Y. clusterProfiler: An R package for comparing biological themes among gene clusters. OMICS 2012, 16, 284–287. [Google Scholar] [CrossRef]
- Yu, G. enrichplot: Visualization of Functional Enrichment Result. Available online: https://yulab-smu.top/biomedical-knowledge-mining-book/ (accessed on 21 March 2023).
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
Distefano, R.; Ilieva, M.; Madsen, J.H.; Rennie, S.; Uchida, S. DoxoDB: A Database for the Expression Analysis of Doxorubicin-Induced lncRNA Genes. Non-Coding RNA 2023, 9, 39. https://doi.org/10.3390/ncrna9040039
Distefano R, Ilieva M, Madsen JH, Rennie S, Uchida S. DoxoDB: A Database for the Expression Analysis of Doxorubicin-Induced lncRNA Genes. Non-Coding RNA. 2023; 9(4):39. https://doi.org/10.3390/ncrna9040039
Chicago/Turabian StyleDistefano, Rebecca, Mirolyuba Ilieva, Jens Hedelund Madsen, Sarah Rennie, and Shizuka Uchida. 2023. "DoxoDB: A Database for the Expression Analysis of Doxorubicin-Induced lncRNA Genes" Non-Coding RNA 9, no. 4: 39. https://doi.org/10.3390/ncrna9040039
APA StyleDistefano, R., Ilieva, M., Madsen, J. H., Rennie, S., & Uchida, S. (2023). DoxoDB: A Database for the Expression Analysis of Doxorubicin-Induced lncRNA Genes. Non-Coding RNA, 9(4), 39. https://doi.org/10.3390/ncrna9040039