A. C. Gingeras, T. R. Haerty, and W. , Author response, vol.3, pp.1002248-1002262, 2014.

K. Beilstein, A. Wittmann, M. Grez, and B. Suess, Conditional Control of Mammalian Gene Expression by Tetracycline-Dependent Hammerhead Ribozymes, ACS Synth. Biol, vol.4, pp.526-534, 2014.

L. Bertolini, M. Bertolini, G. Anderson, E. Maga, K. Madden et al., , 2007.

, Transient depletion of Ku70 and Xrcc4 by RNAi as a means to manipulate the non-homologous end-joining pathway, Journal of Biotechnology, vol.128, pp.246-257

S. Berghmans, R. D. Murphey, E. Wienholds, D. Neuberg, J. L. Kutok et al., mutant zebrafish develop malignant peripehral nerve sheath tumors, Proc Natl. Acad. Sci. USA, vol.11, pp.407-412, 2005.

A. Bhan, M. Soleimani, and S. S. , Long Noncoding RNA and Cancer:A New Paradigm, Cancer Research AACR, 2017.

P. Bhattaram, A. P. Ndez, .. E. Sock, E. Colmenares, C. Kaneko et al., Organogenesis relies on SoxC transcription factors for the survival of neural and mesenchymal progenitors, Nature Communications, vol.1, pp.1-12, 2010.

A. Bitetti, A. C. Mallory, E. Golini, C. Carrieri, H. C. Gutiérrez et al., MicroRNA degradation by a conserved target RNA regulates animal behavior, Nature Structural &Amp, 2018.

D. Branzei and M. Foiani, Regulation of DNA repair throughout the cell cycle, Nat Rev Mol Cell Biol, vol.9, pp.297-308, 2008.

N. Brockdorff, A. Ashworth, G. F. Kay, P. Cooper, S. Smith et al., Conservation of position and exclusive expression of mouse Xist from the inactive X chromosome, p.351, 1991.

C. J. Ceol, Y. Houvras, J. Jane-valbuena, S. Bilodeau, D. A. Orlando et al., The histone methyltransferase SETDB1 is recurrently amplified in melanoma and accelerates its onset, Nature, vol.471, pp.513-517, 2011.

M. Cesana, D. Cacchiarelli, I. Legnini, T. Santini, O. Sthandier et al., A Long Noncoding RNA Controls Muscle Differentiation by Functioning as a Competing Endogenous RNA, Cell, vol.147, pp.358-369, 2011.

A. Chapman, L. F. Del-ama, J. Ferguson, J. Kamarashev, C. Wellbrock et al., , 2014.

, Heterogeneous Tumor Subpopulations Cooperate to Drive Invasion, CellReports, vol.8, pp.688-695

L. Chen, H. Yang, Y. Xiao, X. Tang, Y. Li et al., , 2016.

, Lentiviral-mediated overexpression of long non-coding RNA GAS5 reduces invasion by mediating MMP2 expression and activity in human melanoma cells, International Journal of Oncology, vol.48, pp.1509-1518

W. Chen, J. Wu, L. Li, Z. Zhang, J. Ren et al., Ppm1b negatively regulates necroptosis through dephosphorylating Rip3, vol.17, pp.434-444, 2015.

X. Chen, G. Gao, S. Liu, L. Yu, D. Yan et al., Research Article, Biomed Res Int, pp.1-9, 2017.

Z. Chen, L. Huang, Y. Wu, W. Zhai, P. Zhu et al., LncSox4 promotes the selfrenewal of liver tumour-initiating cells through Stat3-mediated Sox4 expression, Nature Communications, vol.7, pp.12598-12611, 2016.

S. W. Cho, J. Xu, R. Sun, M. R. Mumbach, A. C. Carter et al., Promoter of lncRNA Gene PVT1 Is a Tumor-Suppressor DNA Boundary Element, pp.1-38, 2018.

R. A. Chodroff, L. Goodstadt, T. M. Sirey, P. L. Oliver, K. E. Davies et al., Long noncoding RNA genes: conservation of sequence and brain expression among diverse amniotes, Genome Biol, vol.11, pp.72-88, 2010.

V. T. Chu, T. Weber, B. Wefers, W. Wurst, S. Sander et al., Increasing the efficiency of homology-directed repair for CRISPR-Cas9-induced precise gene editing in mammalian cells, Nat Biotechnol, vol.33, pp.543-548, 2015.

Y. Chudnovsky, A. E. Adams, P. B. Robbins, Q. Lin, and P. A. Khavari, Use of human tissue to assess the oncogenic activity of melanoma-associated mutations, Nat Genet, vol.37, pp.745-749, 2005.

W. Cizelsky, A. Hempel, M. Metzig, S. Tao, T. Hollemann et al., sox4 And sox11 Function during Xenopus laevis Eye Development, PLoS ONE, vol.8, pp.69372-69384, 2013.

T. Colombo, L. Farina, G. Macino, and P. Paci, PVT1: A Rising Star among Oncogenic Long Noncoding RNAs, Biomed Res Int, issue.10, 2015.

L. Cong, F. A. Ran, D. Cox, S. Lin, R. Barretto et al., Multiplex Genome Engineering Using CRISPR/Cas Systems, Science, vol.339, pp.819-823, 2013.

D. Corallo, S. Candiani, M. Ori, S. Aveic, and G. P. Tonini, The zebrafish as a model for studying neuroblastoma, Cancer Cell International, pp.1-9, 2016.

M. S. Cunnington, M. Santibanez-koref, B. M. Mayosi, J. Burn, and B. Keavney, Chromosome 9p21, 2010.

, SNPs Associated with Multiple Disease Phenotypes Correlate with ANRIL Expression, PLoS Genet, vol.6, pp.1000899-1000916

C. V. Dang, MYC on the Path to, Cancer. Cell, vol.149, pp.22-35, 2012.

M. P. Davis, C. Carrieri, H. K. Saini, S. Van-dongen, T. Leonardi et al., Transposon-driven transcription is a conserved feature of vertebrate spermatogenesis and transcript evolution, EMBO Reports, vol.18, pp.1231-1247, 2017.

M. J. Delás and G. J. Hannon, lncRNAs in development and disease: from functions to mechanisms, Open Biol, vol.7, pp.170121-170131, 2017.

T. Derrien, R. Johnson, G. Bussotti, A. Tanzer, S. Djebali et al., The GENCODE v7 catalog of human long noncoding RNAs: Analysis of their gene structure, evolution, and expression, Genome Research, vol.22, pp.1775-1789, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01205054

X. Ding, X. Wang, M. Lin, Y. Xing, S. Ge et al., PAUPARlncRNA suppresses tumourigenesis by H3K4 demethylation in uveal melanoma, FEBS Letters, vol.590, pp.1729-1738, 2016.

D. Dominguez, P. Freese, M. S. Alexis, A. Su, M. Hochman et al., Sequence, Structure, and Context Preferences of Human RNA Binding Proteins, Molecular Cell, vol.70, pp.854-867, 2018.

M. Dovey, R. M. White, and L. I. Zon, Oncogenic NRAS Cooperates with p53 Loss to Generate Melanoma in Zebrafish, Zebrafish, issue.4, pp.397-404, 2009.

Z. Du, T. Fei, R. G. Verhaak, Z. Su, Y. Zhang et al., Integrative genomic analyses reveal clinically relevant long noncoding RNAs in human cancer, Nat Struct Mol Biol, vol.20, pp.908-913, 2013.

M. Eißmann, T. Gutschner, M. Hämmerle, S. Günther, M. Caudron-herger et al., Loss of the abundant nuclear non-coding RNA MALAT1is compatible with life and development, RNA Biology, vol.9, pp.1076-1087, 2012.

J. M. Engreitz, N. Ollikainen, and M. Guttman, Long non-coding RNAs: spatial amplifiers that control nuclear structure and gene expression, Nature Publishing Group, vol.17, pp.756-770, 2016.

J. Etchin, J. P. Kanki, and A. T. Look, Zebrafish as a Model for the Study of Human Cancer, 2011.

M. A. Faghihi, F. Modarresi, A. M. Khalil, D. E. Wood, B. G. Sahagan et al., Expression of a noncoding RNA is elevated in Alzheimer's disease and drives rapid feed-forward regulation of ?-secretase, Nat Med, vol.14, pp.723-730, 2008.

M. J. Fedor and J. R. Williamson, The catalytic diversity of RNAs, Nat Rev Mol Cell Biol, vol.6, pp.399-412, 2005.

I. V. Fedorenko, G. T. Gibney, and K. S. Smalley, NRAS mutant melanoma: biological behavior and future strategies for therapeutic management, Oncogene, vol.32, pp.3009-3018, 2012.

A. Fedoruk-wyszomirska, M. Szyma?-ski, E. Wyszko, M. Z. Barciszewska, and J. Barciszewski, Highly active low magnesium hammerhead ribozyme, J. Biochem, vol.145, pp.451-459, 2009.

T. R. Fernando, J. R. Contreras, M. Zampini, N. I. Rodriguez-malave, M. O. Alberti et al., The lncRNA CASC15 regulates SOX4 expression in RUNX1-rearranged acute leukemia, pp.1-15, 2017.

R. Fior, V. Póvoa, R. V. Mendes, T. Carvalho, A. Gomes et al., Singlecell functional and chemosensitive profiling of combinatorial colorectal therapy in zebrafish xenografts, Proc Natl Acad Sci, vol.114, pp.8234-8243, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02438810

G. V. Fitzpatrick, P. D. Soloway, and M. J. Higgins, Regional loss of imprinting and growth deficiency in mice with a targeted deletion of KvDMR1, Nat Genet, vol.32, pp.426-431, 2002.

R. J. Flockhart, D. E. Webster, K. Qu, N. Mascarenhas, J. Kovalski et al., , 2012.

, BRAF V600Eremodels the melanocyte transcriptome and induces BANCRto regulate melanoma cell migration

, Genome Research, vol.22, pp.1006-1014

G. Fornabaio, R. L. Barnhill, C. Lugassy, L. A. Bentolila, N. Cassoux et al., Angiotropism and extravascular migratory metastasis in cutaneous and uveal melanoma progression in a zebrafish model, Sci Rep, vol.8, issue.1, p.10448, 2018.

S. Ganesh and P. Svoboda, Retrotransposon-associated long non-coding RNAs in mice and men, Pflügers Archiv -European Journal of Physiology, pp.1-12, 2016.

J. E. Garneau, M. Dupuis, M. Villion, D. A. Romero, R. Barrangou et al., The CRISPR/Cas bacterial immune system cleaves bacteriophage and plasmid DNA, Nature, vol.468, pp.67-71, 2010.

S. F. Gilbert, Early Development in fish, 2000.

G. Glusman, S. Qin, M. R. El-gewely, A. F. Siegel, J. C. Roach et al., RMEL3, A novel BRAFV600E-associated long noncoding RNA is required for MAPK and PI3K signaling in melanoma, PLoS Comput Biol Goedert, vol.7, p.24, 2006.

M. Gouzardi, K. Berg, L. M. Pieper, and A. F. Schier, Long non-coding RNAs are largely dispensable for zebrafish embryogenesis, viability and fertility, 2018.

A. Graindorge, I. Pinheiro, P. Tsvetkov, C. Carolis, F. Buchholz et al., revision) Systematic Identification and Quantitative Measurement of RNA-Protein Interaction by incPRINT

C. Grill, L. , and L. , NRAS, NRAS, Which Mutation Is Fairest of Them All, Journal of Investigative Dermatology, vol.136, pp.1936-1938, 2016.

A. F. Groff, D. B. Sanchez-gomez, M. M. Soruco, C. Gerhardinger, A. R. Barutcu et al., In Vivo Characterization of Linc-p21 Reveals Functional cis -Regulatory DNA Elements, Cell Reports, vol.16, pp.2178-2186, 2016.

P. Grote and B. G. Herrmann, The long non-coding RNA Fendrrlinks epigenetic control mechanisms to gene regulatory networks in mammalian embryogenesis, RNA Biology, vol.10, pp.1579-1585, 2014.

P. Grote, L. Wittler, D. Hendrix, F. Koch, S. Währisch et al., The Tissue-Specific lncRNA Fendrr Is an Essential Regulator of Heart and Body Wall Development in the Mouse, Developmental Cell, vol.24, pp.206-214, 2013.

S. Guil and M. Esteller, Cis-acting noncoding RNAs: friends and foes, Nature Structural &Amp, 2012.

, Molecular Biology, vol.19, pp.1068-1075

T. Gutschner, M. Baas, and S. Diederichs, Noncoding RNA gene silencing through genomic integration of RNA destabilizing elements using zinc finger nucleases, Genome Research, vol.21, pp.1944-1954, 2011.

M. Haemmerle and T. Gutschner, Long Non-Coding RNAs in Cancer and Development: Where Do We Go from Here? Ijms, vol.16, pp.1395-1405, 2015.

W. Haerty and C. P. Ponting, Mutations within lncRNAs are effectively selected against in fruitfly but not in human, Genome Biol, vol.14, p.49, 2013.

C. R. Hale, P. Zhao, S. Olson, M. O. Duff, B. R. Graveley et al., , 2009.

, RNA-Guided RNA Cleavage by a CRISPR RNA-Cas Protein Complex, Cell, vol.139, pp.945-956

C. Hammann and W. E. , Searching genomes for ribozymes and riboswitches, Genome Biol, vol.8, issue.4, p.210, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00167549

J. S. Hartig, Ribozymes, Methods and Protocols. Meth in Mol Biol, vol.848, 2012.

M. He, F. Zhang, H. Wang, H. Wang, Z. Zhu et al., Efficient ligase 3-dependent microhomology-mediated end joining repair of DNA double-strand breaks in zebrafish embryos, Mutation Research -Fundamental and Molecular Mechanisms of Mutagenesis, vol.780, pp.86-96, 2015.

H. Hezroni, D. Koppstein, M. G. Schwartz, A. Avrutin, D. P. Bartel et al., Principles of Long Noncoding RNA Evolution Derived from Direct Comparison of Transcriptomes in 17 Species, Cell Reports, vol.11, pp.1110-1122, 2015.

K. Hiom, Coping with DNA double strand breaks, DNA Repair, vol.9, pp.1256-1263, 2010.

M. Hirata, K. Nakamura, and S. Kondo, Pigment cell distributions in different tissues of the zebrafish, with special reference to the striped pigment pattern, Dev. Dyn, vol.234, pp.293-300, 2005.

M. Hirata, K. Nakamura, T. Kanemaru, Y. Shibata, and S. Kondo, Pigment cell organization in the hypodermis of zebrafish, Dev. Dyn, vol.227, pp.497-503, 2003.

Y. Hisano, T. Sakuma, S. Nakade, R. Ohga, S. Ota et al., Precise in-frame integration of exogenous DNA mediated by CRISPR/Cas9 system in zebrafish, Sci. Rep, vol.5, pp.8841-8847, 2015.

S. Hombach and M. Kretz, The non-coding skin: Exploring the roles of long non-coding RNAs in epidermal homeostasis and disease, BioEssays, vol.35, pp.1093-1100, 2013.

J. Y. Hoo, Y. Kumari, M. F. Shaikh, S. M. Hue, and B. H. Goh, Zebrafish: A Versatile Animal Model for Fertility Research, Biomed Res Int, pp.1-20, 2016.

K. Hoshijima, M. J. Jurynec, and D. J. Grunwald, Precise genome editing by homologous recombination, The Zebrafish -Genetics, pp.121-147, 2016.

Y. Hosono, Y. S. Niknafs, J. R. Prensner, M. K. Iyer, S. M. Dhanasekaran et al., Oncogenic Role of THOR, a Conserved Cancer/Testis Long Noncoding RNA, Cell, vol.171, pp.1559-1561, 2017.

A. Hua-van, A. Le-rouzic, T. S. Boutin, J. Filée, and P. Capy, The struggle for life of the genome's selfish architects, Biology Direct, vol.6, p.19, 2011.

H. Huang, Y. Cheng, W. Liao, Y. Tien, C. J. Yang et al., , 2012.

, SOX4 Transcriptionally Regulates Multiple SEMA3/Plexin Family Members and Promotes Tumor Growth in Pancreatic Cancer, PLoS ONE, vol.7, pp.48637-48649

J. Huang, M. Arsenault, M. Kann, C. Lopez-mendez, M. Saleh et al., The transcription factor sry-related HMG box-4 (SOX4) is required for normal renal development in vivo, Dev. Dyn, vol.242, pp.790-799, 2013.

C. J. Hutchins, P. D. Rathjen, A. C. Forster, and R. H. Symons, Self-cleavage of plus and minus RNA transcripts of avocado sunblotch viroid, Nucleic Acids Research, vol.14, issue.9, 1986.

W. Y. Hwang, Y. Fu, D. Reyon, M. L. Maeder, P. Kaini et al., Heritable and Precise Zebrafish Genome Editing Using a CRISPR-Cas System, PLoS ONE, vol.8, pp.68708-68709, 2013.

Y. Ichigozaki, S. Fukushima, M. Jinnin, A. Miyashita, S. Nakahara et al., Serum long non-coding RNA, snoRNA host gene 5 level as a new tumor marker of malignant melanoma, Exp Dermatol, vol.25, pp.67-69, 2015.

I. A. Ilik, J. J. Quinn, P. Georgiev, F. Tavares-cadete, D. Maticzka et al., Tandem Stem-Loops in roX RNAs Act Together to Mediate X Chromosome Dosage Compensation in Drosophila, Molecular Cell, vol.51, pp.156-173, 2013.

M. K. Iyer, Y. S. Niknafs, R. Malik, U. Singhal, A. Sahu et al., The landscape of long noncoding RNAs in the human transcriptome, Nat Genet, vol.47, pp.199-208, 2015.

S. Izraely, O. Sagi-assif, A. Klein, T. Meshel, G. Tsarfaty et al., The metastatic microenvironment: Brain-residing melanoma metastasis and dormant micrometastasis, Int. J. Cancer, vol.131, pp.1071-1082, 2011.

F. Jacob and J. Monod, Genetic regulatory mechanisms in the synthesis of proteins, J. Mol. Biol, vol.3, pp.318-356, 1961.

M. Jinek, K. Chylinski, I. Fonfara, M. Hauer, J. A. Doudna et al., A Programmable Dual-RNA-GuidedDNA Endonuclease in AdaptiveBacterial Immunity, Science, vol.337, issue.6096, pp.816-837, 2012.

J. , Zebrafish pigmentation mutations and the processes of neural crest development, pp.1-21, 1997.

D. B. Johnson and I. Puzanov, Treatment of NRAS-Mutant Melanoma, Curr. Treat. Options in Oncol, vol.16, pp.15-29, 2015.

J. Joung, J. M. Engreitz, S. Konermann, O. O. Abudayyeh, V. K. Verdine et al., Genome-scale activation screen identifies a lncRNA locus regulating a gene neighbourhood, Nature, vol.548, pp.343-346, 2017.

B. Ju, Oncogenic KRAS promotes malignant brain tumors in zebrafish, pp.1-11, 2015.

M. Kalkat, J. De-melo, K. Hickman, C. Lourenco, C. Redel et al., MYC Deregulation in Primary Human Cancers, Genes, vol.8, pp.151-181, 2017.

G. J. Kapral, S. Jain, J. Noeske, J. A. Doudna, D. C. Richardson et al., New tools Activating RNAs associate with Mediator to enhance chromatin architecture and transcription, Nature, vol.494, pp.497-501, 2014.

F. Lai, U. A. Orom, M. Cesaroni, M. Beringer, D. J. Taatjes et al., , 2013.

, Activating RNAs associate with Mediator to enhance chromatin architecture and transcription, Nature, vol.494, pp.497-501

K. V. Lai, G. Gong, A. Atanasio, J. Rojas, J. Quispe et al., Diverse Phenotypes and Specific Transcription Patterns in Twenty Mouse Lines with Ablated LincRNAs, PLoS ONE, vol.10, pp.125522-125543, 2015.

D. M. Langenau, D. Traver, A. A. Ferrando, J. L. Kutok, J. C. Aster et al., Myc-Induced T Cell Leukemia in Transgenic Zebrafish, vol.299, pp.887-90, 2003.

G. Lavorgna, R. Vago, M. Sarmini, F. Montorsi, A. Salonia et al., Long non-coding RNAs as novel therapeutic targets in cancer, Pharmacological Research, vol.110, pp.131-138, 2016.

R. T. Lee, A. S. Ng, and P. W. Ingham, Ribozyme Mediated gRNA Generation for In Vitro and In Vivo CRISPR/Cas9 Mutagenesis, PLoS ONE, vol.11, pp.166020-166032, 2016.

B. Lee, A. Sahoo, J. Marchica, E. Holzhauser, X. Chen et al., The long noncoding RNA SPRIGHTLY acts as an intranuclear organizing hub for pre-mRNA molecules, Sci Adv, vol.3, issue.5, p.1602505, 2017.

L. Lessard, M. Liu, D. M. Marzese, H. Wang, K. Chong et al., The CASC15 Long Intergenic Noncoding RNA Locus Is Involved in Melanoma Progression and Phenotype Switching, Journal of Investigative Dermatology, vol.135, pp.2464-2474, 2015.

E. Leucci, R. Vendramin, M. Spinazzi, P. Laurette, M. Fiers et al., Melanoma addiction to the long non-coding RNA SAMMSON, Nature Publishing Group, vol.531, pp.518-522, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02086933

M. Li, L. Zhao, P. S. Page-mccaw, C. , and W. , Zebrafish Genome Engineering Using the CRISPR-Cas9 System, Trends in Genetics, vol.32, pp.815-827, 2016.

R. Li, L. Zhang, L. Jia, Y. Duan, Y. Li et al., Long Non-Coding RNA BANCR Promotes Proliferation in Malignant Melanoma by Regulating MAPK Pathway Activation, PLoS ONE, vol.9, pp.100893-100899, 2014.

Y. Li, W. Zhang, P. Liu, Y. Xu, L. Tang et al., Long non-coding RNA FENDRR inhibits cell proliferation and is associated with good prognosis in breast cancer, vol.11, pp.1403-1412, 2018.

H. Liu, L. Fang, Y. Cheng, and Q. Sun, LncRNA PVT1 regulates prostate cancer cell growth by inducing the methylation of miR-146a, Cancer Med, vol.5, pp.3512-3519, 2016.

S. J. Liu, M. A. Horlbeck, S. W. Cho, H. S. Birk, M. Malatesta et al., CRISPRi-based genome-scale identification of functional long noncoding RNA loci in human cells, Science, vol.355, pp.7111-7119, 2017.

T. Liu, S. Shen, J. Xiong, Y. Xu, H. Zhang et al., Clinical significance of long noncoding RNA SPRY4-IT1 in melanoma patients, FEBS Open Bio, vol.6, pp.147-154, 2016.

W. Luan, L. Li, Y. Shi, X. Bu, Y. Xia et al., Long noncoding RNA MALAT1 acts as a competing endogenous RNA to promote malignant melanoma growth and metastasis by sponging miR-22, Oncotarget, vol.7, issue.39, pp.63901-63912, 2017.

L. H. Maguire, A. R. Thomas, and A. M. Goldstein, Tumors of the neural crest: Common themes in development and cancer, Dev. Dyn, vol.244, pp.311-322, 2014.

Y. Marahens, B. Panning, J. Dausman, W. Strauss, and J. R. , Xist-deficient mice are defective in dosage compensation but not spermatogenesis, Genes Dev, vol.11, issue.2, pp.156-66, 2007.

P. Mali, L. Yang, K. M. Esvelt, J. Aach, M. Guell et al., RNA-guided human genome engineering via Cas9, Science, vol.339, issue.6121, pp.823-826, 2013.

A. Malina, J. R. Mills, R. Cencic, Y. Yan, J. Fraser et al., Repurposing CRISPR/Cas9 for in situ functional assays, Genes Dev, vol.27, pp.2602-2614, 2013.

A. C. Mallory and A. Shkumatava, LncRNAs in vertebrates: Advances and challenges, pp.1-12, 2015.

F. P. Marchese, E. Grossi, O. Marín-béjar, S. K. Bharti, I. Raimondi et al., A Long Noncoding RNA Regulates Sister Chromatid Cohesion, Molecular Cell, vol.63, pp.397-407, 2016.

F. P. Marchese, I. Raimondi, and M. Huarte, The multidimensional mechanisms of long noncoding RNA function, pp.1-13, 2017.

O. Marín-béjar, F. P. Marchese, A. Athie, Y. Sánchez, J. González et al., Pint lincRNA connects the p53 pathway with epigenetic silencing by the Polycomb repressive complex 2, Genome Biol, vol.14, p.104, 2013.

T. Maruyama, S. K. Dougan, M. C. Truttmann, A. M. Bilate, J. R. Ingram et al., Increasing the efficiency of precise genome editing with CRISPR-Cas9 by inhibition of nonhomologous end joining, Nature Biotechnology, pp.1-8, 2015.

M. Matsui and D. R. Corey, Non-coding RNAs as drug targets, Nature Publishing Group, vol.16, pp.167-179, 2016.

J. Mazar, W. Zhao, A. M. Khalil, B. Lee, J. Shelley et al., The Functional Characterization of Long Noncoding RNA SPRY4-IT1 in Human Melanoma Cells, vol.5, p.19, 2014.

A. M. Mcconnell, J. K. Mito, J. Ablain, M. Dang, L. Formichella et al., Neural crest state activation in NRAS driven melanoma, 2018.

S. Meierjohann, S. , and M. , From Mendelian to molecular genetics: the Xiphophorus melanoma model, Trends in Genetics, vol.22, pp.654-661, 2006.

A. Merdrignac, G. Angenard, C. Allain, K. Petitjean, D. Bergeat et al., A novel transforming growth factor beta-induced long noncoding RNA promotes an inflammatory microenvironment in human intrahepatic cholangiocarcinoma, Hepatology Communications, vol.2, pp.254-269, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01744444

Y. Miao, W. Liu, Q. Zhang, and A. Guo, lncRNASNP2: an updated database of functional SNPs and mutations in human and mouse lncRNAs, Nucleic Acids Research, vol.46, pp.276-280, 2017.

C. Michailidou, M. Jones, P. Walker, J. Kamarashev, A. Kelly et al., Dissecting the roles of Raf-and PI3K-signalling pathways in melanoma formation and progression in a zebrafish model, 2009.

K. M. Michalik, X. You, Y. Manavski, A. Doddaballapur, M. Zörnig et al., Long Noncoding RNA MALAT1 Regulates Endothelial Cell Function and Vessel Growth, Circ. Res, vol.114, issue.9, pp.1389-97, 2014.

M. Mimeault and S. K. Batra, Emergence of zebrafish models in oncology for validating novel anticancer drug targets and nanomaterials, Drug Discovery Today, vol.18, pp.128-140, 2013.

F. Mohammad, T. Mondal, N. Guseva, G. K. Pandey, and C. Kanduri, Kcnq1ot1 noncoding RNA mediates transcriptional gene silencing by interacting with Dnmt1, Development, vol.137, pp.2493-2499, 2010.

S. Mohammadin, P. P. Edger, J. C. Pires, and M. E. Schranz, Positionally-conserved but sequencediverged: identification of long non-coding RNAs in the Brassicaceae and Cleomaceae, BMC Plant Biology, pp.1-12, 2015.

T. Mondal, P. K. Juvvuna, A. Kirkeby, S. Mitra, S. T. Kosalai et al., Sense-Antisense lncRNA Pair Encoded by Locus 6p22.3 Determines Neuroblastoma Susceptibility via the USP36-CHD7-SOX9 Regulatory Axis, Cancer Cell, vol.33, pp.417-434, 2018.

S. Nakade, T. Tsubota, Y. Sakane, S. Kume, N. Sakamoto et al., Microhomology-mediated end-joining-dependent integration of donor DNA in cells and animals using TALENs and CRISPR/Cas9, Nature Communications, vol.5, pp.1-8, 2014.

S. Nakagawa, J. Y. Ip, G. Shioi, V. Tripathi, X. Zong et al., Malat1 is not an essential component of nuclear speckles in mice, Rna, vol.18, pp.1487-1499, 2012.

S. Nakagawa, M. Shimada, K. Yanaka, M. Mito, T. Arai et al., The lncRNA Neat1 is required for corpus luteum formation and the establishment of pregnancy in a subpopulation of mice, Development, vol.141, pp.4618-4627, 2014.

S. Nakagawa, Lessons from reverse-genetic studies of lncRNAs. BBA -Gene Regulatory Mechanisms 1859, pp.177-183, 2016.

A. Necsulea, M. Soumillon, M. Warnefors, A. Liechti, T. Daish et al., The evolution of lncRNA repertoires and expression patterns in tetrapods, Nature, vol.505, pp.635-640, 2015.

J. V. Neiswender, R. L. Kortum, C. Bourque, M. Kasheta, L. I. Zon et al., , 2017.

, KIT Suppresses BRAF V600E-Mutant Melanoma by Attenuating Oncogenic RAS/MAPK Signaling, Cancer Res, vol.77, pp.5820-5830

B. L. Nelms and P. A. Labosky, A predicted hairpin cluster correlate with barriers to PCR, sequencing and possibly BAC recombineering, 2011.

S. Nicoli, D. Ribatti, F. Cotelli, and M. Presta, Mammalian Tumor Xenografts Induce Neovascularization in Zebrafish Embryos, Cancer Res, vol.67, pp.2927-2931, 2007.

J. A. Nilsson, C. , and J. L. , Myc pathways provoking cell suicide and cancer, Oncogene, vol.22, pp.9007-9021, 2003.

F. Nishikawa, M. Shirai, and S. Nishikawa, Site-specific modification of functional groups in genomic hepatitis delta virus (HDV) ribozyme, European Journal of Biochemistry, vol.269, pp.5792-5803, 2002.

L. S. Nissen-meyer, R. Jemtland, V. T. Gautvik, M. E. Pedersen, R. Paro et al., Osteopenia, decreased bone formation and impaired osteoblast development in Sox4 heterozygous mice, Journal of Cell Science, vol.120, pp.2785-2795, 2007.

Y. Nomura, L. Zhou, A. Miu, Y. , and Y. , Controlling Mammalian Gene Expression by Allosteric Hepatitis Delta Virus Ribozymes, ACS Synth. Biol, vol.2, pp.684-689, 2013.

V. R. Paralkar, C. C. Taborda, P. Huang, Y. Yao, A. V. Kossenkov et al., Unlinking an lncRNA from Its Associated cis Element, Molecular Cell, vol.62, pp.104-110, 2016.

E. E. Patton, H. R. Widlund, J. L. Kutok, K. R. Kopani, J. F. Amatruda et al., BRAF Mutations Are Sufficient to Promote Nevi Formation and Cooperate with p53 in the Genesis of Melanoma, Current Biology, vol.15, pp.249-254, 2005.

A. Pauli, J. L. Rinn, and A. F. Schier, Non-coding RNAs as regulators of embryogenesis, Nat Rev Genet, vol.12, pp.136-149, 2011.

D. Pei and P. R. Strauss, Mutation Research/Fundamental and MolecularMechanisms of Mutagenesis. Mutation Research -Fundamental and Molecular Mechanisms of Mutagenesis 743-744, pp.151-159, 2013.

R. B. Perry, .. Ulitsky, and I. , The functions of long noncoding RNAs in development and stem cells, Development, vol.143, pp.3882-3894, 2016.

V. A. Pollack, E. Alvarez, K. F. Tse, M. Y. Torgov, S. Xie et al., Treatment parameters modulating regression of human melanoma xenografts by an antibody-drug conjugate (CR011-vcMMAE) targeting GPNMB, Cancer Chemother Pharmacol, vol.60, pp.423-435, 2007.

C. Posch, M. Sanlorenzo, I. Vujic, J. A. Oses-prieto, B. D. Cholewa et al., Phosphoproteomic Analyses of NRAS(G12) and NRAS(Q61) Mutant Melanocytes Reveal Increased CK2&alpha, 2016.

, Kinase Levels in NRAS(Q61) Mutant Cells, pp.1-8

S. Prajapati, U. Verma, Y. Yamamoto, Y. T. Kwak, G. et al., Protein Phosphatase 2C? Association with the I?B Kinase Complex Is Involved in Regulating NF-?B Activity, J. Biol. Chem, vol.279, pp.1739-1746, 2004.

J. R. Prensner and A. M. Chinnaiyan, The emergence of lncRNAs in cancer biology, Cancer Discovery, vol.1, pp.391-407, 2011.

J. J. Quinn, Q. C. Zhang, P. Georgiev, I. A. Ilik, A. Akhtar et al., Rapid evolutionary turnover underlies conserved lncRNA-genome interactions, Genes Dev, vol.30, pp.191-207, 2016.

A. D. Ramos, R. E. Andersen, S. J. Liu, T. J. Nowakowski, S. J. Hong et al., The Long Noncoding RNA Pnky Regulates Neuronal Differentiation of Embryonic and Postnatal Neural Stem Cells, Cell Stem Cell, vol.16, pp.439-447, 2015.

K. D. Rasmussen and D. Carroll, The miR-144/451eGFP allele, a novel tool for resolving the erythroid potential of hematopoietic precursors, Blood, vol.118, pp.2988-2992, 2011.

P. Reautschnig, P. Vogel, and T. Stafforst, The notorious R.N.A. in the spotlight-drug or target for the treatment, RNA Biol, vol.14, pp.651-668, 2017.

G. Richtig, B. Ehall, E. Richtig, A. Aigelsreiter, T. Gutschner et al., Function and Clinical Implications of Long Non-Coding RNAs in Melanoma, Ijms, vol.18, pp.715-719, 2017.

G. Riddihough, In the Forests of RNA Dark Matter, Science, vol.309, pp.1507-1507, 2005.

D. Rudel and R. J. Sommer, The evolution of developmental mechanisms, Developmental Biology, vol.264, pp.15-37, 2003.

M. R. Russell, A. Penikis, D. A. Oldridge, J. R. Alvarez-dominguez, L. Mcdaniel et al., CASC15-S Is a Tumor Suppressor lncRNA at the 6p22 Neuroblastoma Susceptibility Locus, Cancer Res, 2015.

T. Sado, Z. Wang, H. Sasaki, and L. E. , Regulation of imprinted X-chromosome inactivation in mice by Tsix, Development, vol.128, pp.1275-1286, 2001.

A. Sandru, S. Voinea, E. Panaitescu, and A. Blidaru, Survival rates of patients with metastatic malignant melanoma, Journ. of Med. and Life, vol.7, issue.4, pp.572-576, 2014.

C. Santoriello, E. Gennaro, V. Anelli, M. Distel, A. Kelly et al.,

, Kita Driven Expression of Oncogenic HRAS Leads to Early Onset and Highly Penetrant Melanoma in Zebrafish, PLoS ONE, vol.5, pp.15170-15181

M. Sauvageau, L. A. Goff, S. Lodato, B. Bonev, A. F. Groff et al.,

P. Arlotta and J. L. Rinn, Multiple knockout mouse models reveals lincRNAs are required for life and brain development, 2013.

M. Sasaki, M. Ohnishi, F. Tashiro, H. Niwa, A. Suzuki et al., Disruption of the mouse protein Ser/Thr phosphatase 2C? gene leads to early pre-implantation lethality, Mechanisms of Development, vol.124, pp.489-499, 2007.

C. M. Scahill, Z. Digby, I. M. Sealy, S. Wojciechowska, R. J. White et al., Loss of the chromatin modifier Kdm2aa causes BrafV600E-independent spontaneous melanoma in zebrafish, PLoS Genet, vol.13, pp.1006959-1006983, 2017.

B. Schmid and C. Haass, Genomic editing opens new avenues for zebrafish as a model for neurodegeneration, J. Neurochem, vol.127, pp.461-470, 2013.

K. Schmidt, C. E. Joyce, F. Buquicchio, A. Brown, J. Ritz et al., The lncRNA SLNCR1 Mediates Melanoma Invasion through a Conserved SRA1-like Region, CellReports, vol.15, pp.2025-2037, 2016.

S. Schulte-merker and D. Y. Stainier, Out with the old, in with the new: reassessing morpholino knockdowns in light of genome editing technology, Development, vol.141, pp.3103-3104, 2014.

A. H. Shain and B. C. Bastian, From melanocytes to melanomas, Nature Publishing Group, vol.16, pp.345-358, 2016.

L. Shen, F. Chen, Y. Zhang, L. Cao, Y. Kuang et al., MYCN Transgenic Zebrafish Model with the Characterization of Acute Myeloid Leukemia and Altered Hematopoiesis, PLoS ONE, vol.8, pp.59070-59082, 2013.

V. Singh, R. Govindarajan, S. Naik, and A. Kumar, The effect of hairpin structure on PCR amplification efficiency, Molecular Biology Today, vol.1, issue.3, pp.67-69, 2000.

F. Sleutels, . Zwart, and D. P. Barlow, The non-coding Air RNA is required for silencing autosomal imprinted genes, Nature, vol.14, issue.6873, pp.810-813, 2002.

L. Standaert, C. Adriaens, E. Radaelli, A. Van-keymeulen, C. Blanpain et al., The long noncoding RNA Neat1 is required for mammary gland development and lactation, Rna, vol.20, pp.1844-1849, 2014.

C. H. Stuelten, C. A. Parent, and D. J. Montell, Cell motility in cancer invasion and metastasis: insights from simple model organisms, p.17, 2018.

R. J. Sullivan and K. Flaherty, MAP kinase signaling and inhibition in melanoma, vol.32, pp.2373-2379, 2012.

L. Sun, P. Sun, Q. Y. Zhou, X. Gao, and H. Q. , Long noncoding RNA MALAT1 promotes uveal melanoma cell growth and invasion by silencing of miR-140, Am J Transl Res, vol.8, issue.9, pp.3939-3946, 2016.

L. Tang, W. Zhang, B. Su, Y. , and B. , Long Noncoding RNA HOTAIR Is Associated with Motility, Invasion, and Metastatic Potential of Metastatic Melanoma, Biomed Res Int, vol.2013, pp.1-7, 2013.

A. M. Taylor and L. I. Zon, Zebrafish Tumor Assays: The State of Transplantation, Zebrafish, issue.4, pp.339-385, 2009.

K. J. Teittinen, T. Grönroos, M. Parikka, M. Rämet, and O. Lohi, The zebrafish as a tool in leukemia research, Leukemia Research, vol.36, pp.1082-1088, 2012.

Y. Teng, X. Xie, S. Walker, D. T. White, J. S. Mumm et al., Evaluating human cancer cell metastasis in zebrafish, BMC Cancer, vol.13, pp.1-1, 2013.

B. Thisse and C. Thisse, Situ Hybridization on Whole-Mount Zebrafish Embryos and Young Larvae In Situ Hybridization Protocols, pp.53-67, 2014.

Y. Tian, X. Zhang, Y. Hao, Z. Fang, and Y. He, Potential roles of abnormally expressed long noncoding RNA UCA1 and Malat-1 in metastasis of melanoma, Melanoma Research, vol.24, pp.335-341, 2014.

N. Tiwari, V. K. Tiwari, L. Waldmeier, P. J. Balwierz, P. Arnold et al., Sox4 Is a Master Regulator of Epithelial-Mesenchymal Transition by Controlling Ezh2 Expression and Epigenetic Reprogramming, Cancer Cell, vol.23, pp.768-783, 2013.

V. Tripathi, J. D. Ellis, Z. Shen, D. Y. Song, Q. Pan et al., The Nuclear-Retained Noncoding RNA MALAT1 Regulates Alternative Splicing by Modulating SR Splicing Factor Phosphorylation, Molecular Cell, vol.39, pp.925-938, 2010.

V. Tripathi, J. D. Ellis, Z. Shen, D. Y. Song, Q. Pan et al., The Nuclear-Retained Noncoding RNA MALAT1 Regulates Alternative Splicing by Modulating SR Splicing Factor Phosphorylation, Molecular Cell, vol.39, pp.925-938, 2010.

Y. Tseng and A. Bagchi, The PVT1-MYC duet in cancer, Molecular & Cellular Oncology, vol.2, pp.974467-974470, 2015.

Y. Tseng, B. S. Moriarity, W. Gong, R. Akiyama, A. Tiwari et al., , 2014.

Y. Tseng, B. S. Moriarity, W. Gong, R. Akiyama, A. Tiwari et al., PVT1 dependence in cancer with MYC copy-number increase, Nature, vol.512, pp.82-86, 2015.

I. Ulitsky, Evolution to the rescue: using comparative genomics to understand long non-coding RNAs, Nature Publishing Group, vol.17, pp.601-614, 2016.

I. Ulitsky and D. P. Bartel, lincRNAs: Genomics, Evolution, and Mechanisms, Cell, vol.154, pp.26-46, 2013.

I. Ulitsky, A. Shkumatava, C. H. Jan, H. Sive, and D. P. Bartel, Conserved function of lincRNAs in vertebrate embryonic development despite rapid sequence evolution, Cell, vol.147, pp.1537-1550, 2011.

V. Villegas and P. Zaphiropoulos, Neighboring Gene Regulation by Antisense Long Non-Coding RNAs, Ijms, vol.16, pp.3251-3266, 2015.

S. A. Vinores, Pegatinib in the treatment of wet, age-related macular degeneration, IJN, vol.1, issue.3, pp.263-268, 2006.

I. A. Viringipurampeer, X. Shan, K. Gregory-evans, J. P. Zhang, Z. Mohammadi et al., Rip3 knockdown rescues photoreceptor cell death in blind pde6c zebrafish, vol.21, pp.665-675, 2014.

F. Wang, X. Li, X. Xie, L. Zhao, C. et al., UCA1, a non-protein-coding RNA up-regulated in bladder carcinoma and embryo, influencing cell growth and promoting invasion, FEBS Letters, vol.582, pp.1919-1927, 2008.

M. S. Werner, M. A. Sullivan, R. N. Shah, R. D. Nadadur, A. T. Grzybowski et al., Chromatin-enriched lncRNAs can act as cell-type specific activators of proximal gene transcription, Nat Struct Mol Biol, vol.24, pp.596-603, 2017.

C. Wellbrock, A. Gomez, and M. Scharti, Melanoma development and pigment cell transformation in xiphophorus, Microsc Res Tech, vol.58, issue.6, pp.456-63, 2002.

R. M. White, A. Sessa, C. Burke, T. Bowman, J. Leblanc et al., , 2008.

, Transparent adult zebrafish as a tool for in vivo transplantation analysis, Cell Stem Cell, vol.7, issue.2, pp.183-189

R. M. White, J. Cech, S. Ratanasirintrawoot, C. Y. Lin, P. B. Rahl et al., DHODH modulates transcriptional elongation in the neural crest and melanoma, Nature, vol.471, pp.518-522, 2011.

M. Won and I. B. Dawid, PCR artifact in testing for homologous recombination in genomic editing in zebrafish, PLOS ONE, 2017.

C. Wu, G. Tan, C. Ma, L. , and L. , The Non-Coding RNA Llme23 Drives the Malignant Property of Human Melanoma Cells, pp.1-10, 2018.

Q. Wu, S. Xiang, J. Ma, P. Hui, T. Wang et al., Long non-coding RNA CASC15 regulates gastric cancer cell proliferation, migration and epithelial mesenchymal transition by targeting CDKN1A and ZEB1, Mol Oncol, pp.1-44, 2018.

Y. Xing, X. Wen, X. Ding, J. Fan, P. Chai et al., CANT1 lncRNA Triggers Efficient Therapeutic Efficacy by Correcting Aberrant lncing Cascade in Malignant Uveal Melanoma, Molecular Therapy, vol.25, pp.1209-1221, 2017.

T. Xu, M. Huang, R. Xia, X. Liu, M. Sun et al., Decreased expression of the long non-coding RNA FENDRR is associated with poor prognosis in gastric cancer and FENDRR regulates gastric cancer cell metastasis by affecting fibronectin1 expression, vol.7, pp.1-15, 2014.

X. Yan, Z. Hu, Y. Feng, X. Hu, J. Yuan et al., , 2015.

, Comprehensive Genomic Characterization of Long Non-coding RNAs across Human Cancers, Cancer Cell, vol.28, pp.529-540

R. J. Yanez and A. C. Porter, Gene targeting is enhanced in human cells overexpressing hRAD51, Gene Ther, vol.6, pp.1282-1290, 1999.

K. L. Yap, S. Li, A. M. Muñoz-cabello, S. Raguz, L. Zeng et al.,

, Molecular Interplay of the Noncoding RNA ANRIL and Methylated Histone H3 Lysine 27 by Polycomb CBX7 in Transcriptional Silencing of INK4a, vol.38, pp.662-674

J. Yen, R. M. White, and D. L. Stemple, ScienceDirectZebrafish models of cancer: progress and future challenges, Current Opinion in Genetics & Development, vol.24, pp.38-45, 2014.

J. Yoon, K. Abdelmohsen, and M. Gorospe, Posttranscriptional Gene Regulation by Long Noncoding RNA, J. Mol. Biol, vol.425, pp.3723-3730, 2013.

Y. Yu, Z. Hu, M. Li, B. Li, Z. Wang et al., Cardiac hypertrophy is positively regulated by long non-coding RNA PVT1, Int J Clin Exp Pathol, vol.8, pp.2582-2589, 2015.

B. Zhang, G. Arun, Y. S. Mao, Z. Lazar, G. Hung et al., The lncRNA Malat1 Is Dispensable for Mouse Development but Its Transcription Plays a cis-Regulatory Role in the Adult, Cell Reports, vol.2, pp.111-123, 2012.

H. Zhang, M. Alberich-jorda, G. Amabile, H. Yang, P. B. Staber et al., Sox4 Is a Key Oncogenic Target in C/EBP? Mutant Acute Myeloid Leukemia, Cancer Cell, vol.24, pp.575-588, 2013.

Y. Zhang, H. Huang, B. Zhang, L. , and S. , TALEN-and CRISPR-enhanced DNA homologous recombination for gene editing in zebrafish, 2016.

L. Zhao, M. Arsenault, E. T. Ng, E. Longmuss, T. Chau et al., SOX4 regulates gonad morphogenesis and promotes male germ cell differentiation in mice, Developmental Biology, vol.423, pp.46-56, 2017.

W. Zhao, J. Mazar, B. Lee, J. Sawada, J. Li et al., The Long Noncoding RNA SPRIGHTLY Regulates Cell Proliferation in Primary Human Melanocytes, Journal of Investigative Dermatology, vol.136, pp.819-828, 2016.

X. Zheng, H. Hu, and S. Li, High expression of lncRNA PVT1 promotes invasion by inducing epithelial-to-mesenchymal transition in esophageal cancer, Oncol Lett, pp.1-6, 2016.

L. Zhigui, H. Shuang, Y. Hongqiang, G. Jing, and Y. Zhuo, Autophagy ameliorates cognitive impairment through activation of PVT1 and apoptosis in diabetes mice, Behavioural Brain Research, vol.305, pp.265-277, 2016.

C. Ziegler and M. Kretz, The More the Merrier-Complexity in Long Non-Coding RNA Loci, products through PCR and nested PCR, subcloned into the PCR BLUNT II TOPO vector (Invitrogen) and transformed in the NEB TOP-10, vol.8, pp.101-106, 2017.

A. R. Amandio, A. Necsulea, E. Joye, B. Mascrez, and D. Duboule, , 2016.

, Hotair Is Dispensible for Mouse Development, PLoS Genet, vol.12, p.1006232

K. M. Anderson, D. M. Anderson, J. R. Mcanally, J. M. Shelton, R. Bassel-duby et al., Transcription of the non-coding RNA upperhand controls Hand2 expression and heart development, Nature, vol.539, pp.433-436, 2016.

M. Ballarino, A. Cipriano, R. Tita, T. Santini, F. Desideri et al., Deficiency in the nuclear long noncoding RNA Charme causes myogenic defects and heart remodeling in mice, EMBO J, vol.37, 2018.

A. Bitetti, A. C. Mallory, E. Golini, C. Carrieri, H. Carreno-gutierrez et al., MicroRNA degradation by a conserved target RNA regulates animal behavior, Nat Struct Mol Biol, vol.25, pp.244-251, 2018.

A. M. Bond, M. J. Vangompel, E. A. Sametsky, M. F. Clark, J. C. Savage et al., Balanced gene regulation by an embryonic brain ncRNA is critical for adult hippocampal GABA circuitry, Nat Neurosci, vol.12, pp.1020-1027, 2009.

M. Eissmann, T. Gutschner, M. Hammerle, S. Gunther, M. Caudron-herger et al., Loss of the abundant nuclear non-coding RNA MALAT1 is compatible with life and development, RNA Biol, vol.9, pp.1076-1087, 2012.

G. V. Fitzpatrick, P. D. Soloway, and M. J. Higgins, Regional loss of imprinting and growth deficiency in mice with a targeted deletion of KvDMR1, Nat Genet, vol.32, pp.426-431, 2002.

L. A. Goff and J. L. Rinn, Linking RNA biology to lncRNAs, Genome Res, vol.25, pp.1456-1465, 2015.

P. Grote, L. Wittler, D. Hendrix, F. Koch, S. Wahrisch et al., The tissue-specific lncRNA Fendrr is an essential regulator of heart and body wall development in the mouse, Dev Cell, vol.24, pp.206-214, 2013.

V. Haberle, N. Li, Y. Hadzhiev, C. Plessy, C. Previti et al., , 2014.

P. Han, W. Li, C. H. Lin, J. Yang, C. Shang et al., A long noncoding RNA protects the heart from pathological hypertrophy, Nature, vol.514, pp.102-106, 2014.

X. Han, S. Luo, G. Peng, J. Y. Lu, G. Cui et al., Mouse knockout models reveal largely dispensable but context-dependent functions of lncRNAs during development, J Mol Cell Biol, vol.10, pp.175-178, 2018.

H. Hezroni, D. Koppstein, M. G. Schwartz, A. Avrutin, D. P. Bartel et al., Principles of long noncoding RNA evolution derived from direct comparison of transcriptomes in 17 species, Cell Rep, vol.11, pp.1110-1122, 2015.

Y. Hosono, Y. S. Niknafs, J. R. Prensner, M. K. Iyer, S. M. Dhanasekaran et al., , 2017.

, Oncogenic Role of THOR, a Conserved Cancer/Testis Long Non-coding RNA, Cell, vol.171, p.1520

W. Y. Hwang, Y. Fu, D. Reyon, M. L. Maeder, P. Kaini et al., Heritable and precise zebrafish genome editing using a CRISPR-Cas system, PLoS One, vol.8, p.68708, 2013.

J. Y. Ip, M. Sone, C. Nashiki, Q. Pan, K. Kitaichi et al., Gomafu lncRNA knockout mice exhibit mild hyperactivity with enhanced responsiveness to the psychostimulant methamphetamine, 2016.

T. Isoda, A. J. Moore, Z. He, V. Chandra, M. Aida et al., Non-coding Transcription Instructs Chromatin Folding and Compartmentalization to Dictate Enhancer-Promoter Communication and T Cell Fate, Cell, vol.171, pp.103-119, 2017.

C. B. Kimmel, W. W. Ballard, S. R. Kimmel, B. Ullmann, and T. F. Schilling, Stages of embryonic development of the zebrafish, Dev Dyn, vol.203, pp.253-310, 1995.

B. Kleaveland, C. Y. Shi, J. Stefano, and D. P. Bartel, A Network of Noncoding Regulatory RNAs Acts in the Mammalian Brain, Cell, vol.174, pp.350-362, 2018.

F. O. Kok, M. Shin, C. W. Ni, A. Gupta, A. S. Grosse et al., Reverse genetic screening reveals poor correlation between morpholinoinduced and mutant phenotypes in zebrafish, Dev Cell, vol.32, pp.97-108, 2015.

J. J. Kotzin, S. P. Spencer, S. J. Mccright, D. B. Kumar, M. A. Collet et al., The long non-coding RNA Morrbid regulates Bim and short-lived myeloid cell lifespan, Nature, vol.537, pp.239-243, 2016.

K. M. Lai, G. Gong, A. Atanasio, J. Rojas, J. Quispe et al., Diverse Phenotypes and Specific Transcription Patterns in Twenty Mouse Lines with Ablated LincRNAs, PLoS One, vol.10, p.125522, 2015.

P. A. Leighton, R. S. Ingram, J. Eggenschwiler, A. Efstratiadis, and S. M. Tilghman, Disruption of imprinting caused by deletion of the H19 gene region in mice, Nature, vol.375, pp.34-39, 1995.

L. Li, B. Liu, O. L. Wapinski, M. C. Tsai, K. Qu et al., Targeted disruption of Hotair leads to homeotic transformation and gene derepression, Cell Rep, vol.5, pp.3-12, 2013.

Y. Marahrens, B. Panning, J. Dausman, W. Strauss, J. et al.,

, Xist-deficient mice are defective in dosage compensation but not spermatogenesis, Genes Dev, vol.11, pp.156-166

S. Nakagawa, J. Y. Ip, G. Shioi, V. Tripathi, X. Zong et al., Malat1 is not an essential component of nuclear speckles in mice, RNA, vol.18, pp.1487-1499, 2012.

S. Nakagawa, M. Shimada, K. Yanaka, M. Mito, T. Arai et al., The lncRNA Neat1 is required for corpus luteum formation and the establishment of pregnancy in a subpopulation of mice, Development, vol.141, pp.4618-4627, 2014.

A. Necsulea, M. Soumillon, M. Warnefors, A. Liechti, T. Daish et al., The evolution of lncRNA repertoires and expression patterns in tetrapods, Nature, vol.505, pp.635-640, 2014.

C. Nepal, Y. Hadzhiev, C. Previti, V. Haberle, N. Li et al., , 2013.

, Genome Res, vol.23, pp.1938-1950

Y. A. Perez-rico, V. Boeva, A. C. Mallory, A. Bitetti, S. Majello et al., Comparative analyses of super-enhancers reveal conserved elements in vertebrate genomes, Genome Res, vol.27, pp.259-268, 2017.

M. A. Ripoche, C. Kress, F. Poirier, and L. Dandolo, Deletion of the H19 transcription unit reveals the existence of a putative imprinting control element, Genes Dev, vol.11, pp.1596-1604, 1997.

T. Sado, Z. Wang, H. Sasaki, and E. Li, Regulation of imprinted Xchromosome inactivation in mice by Tsix, Development, vol.128, pp.1275-1286, 2001.

M. A. Sarangdhar, D. Chaubey, N. Srikakulam, and B. Pillai, Parentally inherited long non-coding RNA Cyrano is involved in zebrafish neurodevelopment, Nucleic Acids Res, 2018.

M. Sauvageau, L. A. Goff, S. Lodato, B. Bonev, A. F. Groff et al., , 2013.

, Multiple knockout mouse models reveal lincRNAs are required for life and brain development, vol.2, p.1749

F. Sleutels, R. Zwart, and D. P. Barlow, The non-coding Air RNA is required for silencing autosomal imprinted genes, Nature, vol.415, pp.810-813, 2002.

I. Ulitsky, A. Shkumatava, C. H. Jan, H. Sive, and D. P. Bartel, Conserved function of lincRNAs in vertebrate embryonic development despite rapid sequence evolution, Cell, vol.147, pp.1537-1550, 2011.

M. Wu, S. Zhang, X. Chen, H. Xu, and X. Li, Expression and function of lncRNA MALAT-1 in the embryonic development of zebrafish, Gene, 2018.

B. Zhang, G. Arun, Y. S. Mao, Z. Lazar, G. Hung et al., , 2012.