Skip to main content Accessibility help
×
Hostname: page-component-7479d7b7d-m9pkr Total loading time: 0 Render date: 2024-07-11T11:29:06.888Z Has data issue: false hasContentIssue false

41 - Small silencing non-coding RNAs: cancer connections and significance

from Part 2.6 - Molecular pathways underlying carcinogenesis: other pathways

Published online by Cambridge University Press:  05 February 2015

Milena S. Nicoloso
Affiliation:
RNA Interference and Non-coding RNA Center and the Department of Experimental herapeutics, University of Texas, M.D. Anderson Cancer Center, Houston, TX, USA
George A. Calin
Affiliation:
RNA Interference and Non-coding RNA Center and the Department of Experimental herapeutics, University of Texas, M.D. Anderson Cancer Center, Houston, TX, USA
Edward P. Gelmann
Affiliation:
Columbia University, New York
Charles L. Sawyers
Affiliation:
Memorial Sloan-Kettering Cancer Center, New York
Frank J. Rauscher, III
Affiliation:
The Wistar Institute Cancer Centre, Philadelphia
Get access

Summary

Focus of the chapter

Despite a few intuitive theories in the sixties that proposed a regulatory role for RNA in controlling gene expression through base-pair complementarity (1,2), the subsequent discovery of transcription factors quenched any further research in this field. As a consequence, for many years RNA has been regarded exclusively as the intermediary molecule between DNA and protein, with the primary role of carrying the genetic information necessary for protein synthesis. Intriguingly, only about 2% of human DNA accounts for protein-coding genes and the total number of proteins does not vary significantly among different species. On the other hand, the extent of non-protein-coding DNA, regarded for a long time as junk DNA, increases proportionally with developmental complexity (3) and over 90% (4) of the genome is actually transcribed in a developmentally regulated manner to produce non-coding RNA (ncRNA) that can be inter-genic, intronic, or overlapping with protein-coding transcripts (5–7). In addition, ncRNAs display precise tissue-expression patterns (8) and are differentially expressed in pathologic conditions such as cancer, and immune or heart diseases (9; for review see 10).

ncRNAs can be conventionally divided into long and small RNAs. Long ncRNAs include those greater than 200 nucleotides (nt) in length that can reach up to 100kb (11,12). Their function and characterization is still underway; however, it is already clear that this heterogeneous class displays important regulatory functions, as shown in developmental processes where they can regulate expression of homeotic genes, oncogenes, and metabolic genes (10). Despite their smaller size, small RNAs are equally important in development, cell biology, and disease, and their discovery triggered the general interest of the scientific community for ncRNAs. Currently, a total of 7053 small RNAs are annotated by Gencode, 85% of which correspond to four major classes: small nuclear (sn)RNAs, small nucleolar (sno)RNAs, miRNAs, and transfer (t)RNAs (13; for small RNA classification and main characteristics, see Table 41.1). In this chapter we will focus mainly on small silencing RNAs that include: microRNAs, which are the most studied class of ncRNAs; piwi-interacting RNAs (piRNAs), which are germline-specific ncRNAs, and finally a brief reference to the most recently identified class of small silencing RNAs, which consists of endogenous small interfering RNAs (endo-siRNAs).

Type
Chapter
Information
Molecular Oncology
Causes of Cancer and Targets for Treatment
, pp. 481 - 496
Publisher: Cambridge University Press
Print publication year: 2013

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Jacob, F, Monod, J.Genetic regulatory mechanisms in the synthesis of proteins. Journal of Molecular Biology 1961;3:318–56.CrossRefGoogle ScholarPubMed
Britten, RJ, Davidson, EH. Gene regulation for higher cells: a theory. Science 1969;165:349–57.CrossRef
Taft, RJ, Pheasant, M, Mattick, JS. The relationship between non-protein-coding DNA and eukaryotic complexity. Bioessays 2007;29:288–99.CrossRef
Birney, E, Stamatoyannopoulos JA, Dutta A, et al. Identification and analysis of functional elements in 1% of the human genome by the ENCODE pilot project. Nature 2007;447:799–816.CrossRef
Kapranov, P, Willingham, AT, Gingeras, TR. Genome-wide transcription and the implications for genomic organization. Nature Reviews Genetics 2007;8:413–23.CrossRef
Carninci, P, Kasukawa, T, Katayama, S, et al. The transcriptional landscape of the mammalian genome. Science 2005;309:1559–63.CrossRef
Harrow, J, Frankish, A, Gonzalez, JM, et al. GENCODE: the reference human genome annotation for The ENCODE Project. Genome Research 2012;22:1760–74.CrossRef
Mercer, TR, Dinger, ME, Sunkin, SM, Mehler, MF, Mattick, JS. Specific expression of long noncoding RNAs in the mouse brain. Proceedings of the National Academy of Sciences USA 2008;105:716–21.CrossRef
Calin, GA, Liu, CG, Ferracin, M, et al. Ultraconserved regions encoding ncRNAs are altered in human leukemias and carcinomas. Cancer Cell 2007;12:215–29.CrossRef
Mattick, JS. The genetic signatures of noncoding RNAs. PLoS Genetics 2009;5:e1000459.
Ravasi, T, Suzuki, H, Pang, KC, et al. Experimental validation of the regulated expression of large numbers of non-coding RNAs from the mouse genome. Genome Research 2006;16:11–19.CrossRef
Furuno, M, Pang, KC, Ninomiya, N, et al. Clusters of internally primed transcripts reveal novel long noncoding RNAs. PLoS Genetics 2006;2:e37.
Djebali, S, Davis, CA, Merkel, A, et al. Landscape of transcription in human cells. Nature 2012;489:101–8.CrossRef
Zhao, T, Li, G, Mi, S, et al. A complex system of small RNAs in the unicellular green alga Chlamydomonas reinhardtii. Genes and Development 2007;21:1190–203.CrossRef
Molnár, A, Schwach, F, Studholme, DJ, Thuenemann, EC, Baulcombe, DC. miRNAs control gene expression in the single-cell alga Chlamydomonas reinhardtii. Nature 2007;447:1126–9.CrossRef
Valencia-Sanchez, MA, Liu, J, Hannon, GJ, et al. Control of translation and mRNA degradation by miRNAs and siRNAs. Genes and Development 2006;20:515–24.CrossRef
Wightman, B, Ha, I, Ruvkun, G. Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C. elegans. Cell 1993;75:855–62.
Lee, RC, Feinbaum, RL, Ambros, V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 1993;75:843–54.CrossRef
Sempere, LF, Freemantle, S, Pitha-Rowe, I, et al. Expression profiling of mammalian microRNAs uncovers a subset of brain-expressed microRNAs with possible roles in murine and human neuronal differentiation. Genome Biology 2004;5:R13.
Lewis, BP, Shih, IH, Jones-Rhoades, MW, Bartel, DP, Burge, CB. Prediction of mammalian microRNA targets. Cell 2003;115:787–98.CrossRef
Coller, HA, Forman, JJ, Legesse-Miller, A. “Myc'ed messages”: myc induces transcription of E2F1 while inhibiting its translation via a microRNA polycistron. PLoS Genetics, 2007;3:e146.
Bartel, DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 2004;116:281–97.CrossRef
Lee, Y, Jeon, K, Lee, JT, Kim, S, Kim, VN. MicroRNA maturation: stepwise processing and subcellular localization. EMBO Journal 2002;21:4663–70.CrossRef
Cullen, BR. Transcription and processing of human microRNA precursors. Molecular Cell 2004;16:861–5.CrossRef
Han, J, Lee, Y, Yeom, K-H, et al. The Drosha-DGCR8 complex in primary microRNA processing. Genes and Development 2004;18:3016–27.CrossRef
Lee, Y, Ahn, C, Han, J, et al. The nuclear RNase III Drosha initiates microRNA processing. Nature 2003;425:415–19.CrossRef
Lee, Y, Han, J, Yeom, KH, Jin, H, Kim, VN. Drosha in primary microRNA processing. Cold Spring Harbor Symposia on Quantitative Biology 2006;7151–7.
Ladewig, E, Okamura, K, Flynt, AS, et al. Discovery of hundreds of mirtrons in mouse and human small RNA data. Genome Research 2012;22:1634–45.CrossRef
Berezikov, E, Chung, WJ, Willis, J, Cuppen, E, Lai, EC. Mammalian mirtron genes. Molecular Cell 2007;28:328–36.CrossRef
Okamura, K, Hagen, JW, Duan, H, Tyler, DM, Lai, EC. The mirtron pathway generates microRNA-class regulatory RNAs in Drosophila. Cell 2007;130:89–100.CrossRef
Ruby, JG, Jan, CH, Bartel, DP. Intronic microRNA precursors that bypass Drosha processing. Nature 2007;448:83–6.CrossRef
Yi, R, Qin, Y, Macara, IG, Cullen, BR. Exportin-5 mediates the nuclear export of pre-microRNAs and short hairpin RNAs. Genes and Development 2003;17:3011–16.CrossRef
Hutvagner, G, McLachlan, J, Pasquinelli, AE, et al. A cellular function for the RNA-interference enzyme Dicer in the maturation of the let-7 small temporal RNA. Science 2001;293:834–8.CrossRef
Bernstein, E, Caudy, AA, Hammond, SM, Hannon, GJ. Role for a bidentate ribonuclease in the initiation step of RNA interference. Nature 2001;409:363–6.CrossRef
Chendrimada, TP, Gregory, RI, Kumaraswamy, E, et al. TRBP recruits the Dicer complex to Ago2 for microRNA processing and gene silencing. Nature 2005;436:740–4.CrossRef
Gregory, RI, Chendrimada, TP, Cooch, N, Sheikhattar, R. Human RISC couples microRNA biogenesis and posttranscriptional gene silencing. Cell 2005;123: 631–40.CrossRef
Tay, Y, Zhang, J, Thomson, AM, Lim, B, Rigoutsos, I. MicroRNAs to Nanog, Oct4 and Sox2 coding regions modulate embryonic stem cell differentiation. Nature 2008;455: 1124–8.CrossRef
Carrington, JC, Ambros, V. Role of microRNAs in plant and animal development. Science 2003;301:336–8.CrossRef
Pillai, RS, Bhattacharyya, SN, Filipowicz, W. Repression of protein synthesis by miRNAs: how many mechanisms? Trends in Cell Biology 2007;17:118–26.
Selbach, M, Schwanhäusser, B, Thierfelder, N, et al. Widespread changes in protein synthesis induced by microRNAs. Nature 2008;455:58–63.CrossRef
Baek, D, Villén, J, Shin, C, et al. The impact of microRNAs on protein output. Nature 2008;455:64–71.CrossRef
Bernstein, E, Kim, SY, Carmell, MA, et al. Dicer is essential for mouse development. Nature Genetics 2003;35:215–17.CrossRef
Harfe, BD, McManus, MT, Mansfield, JH, Hornstein, E, Tabin, CJ. The RNaseIII enzyme Dicer is required for morphogenesis but not patterning of the vertebrate limb. Proceedings of the National Academy of Sciences USA 2005;102:10898–903.CrossRef
Harris, KS, Zhang, Z, McManus, MT, Harfe, BD, Sun, X. Dicer function is essential for lung epithelium morphogenesis. Proceedings of the National Academy of Sciences USA 2006;103:2208–13.CrossRef
Lynn, FC, Skewes-Cox, P, Kosaka, Y, et al. MicroRNA expression is required for pancreatic islet cell genesis in the mouse. Diabetes 2007;56:2938–45.CrossRef
Zehir, A, Hua, ll, Mask, EL, Morikawa, Y, Cserjesi, P. Dicer is required for survival of differentiating neural crest cells. Developmental Biology;340:459–67.
Kalsotra, A, Wang, K, Li, PF, Cooper, TA. MicroRNAs coordinate an alternative splicing network during mouse postnatal heart development. Genes and Development 2010;24:653–8.CrossRef
da Costa Martins, PA, Bourajjaj, M, Gladka, M, et al. Conditional dicer gene deletion in the postnatal myocardium provokes spontaneous cardiac remodeling. Circulation 2008;118:1567–76.CrossRef
Lei, L, Jin, S, Gonzalez, G, Behringer, RR, Woodruff, TK. The regulatory role of Dicer in folliculogenesis in mice. Molecular Cell Endocrinology 2010;315:63–73.CrossRef
Yi, R, O’Carroll, D, Pasolli, HA, et al. Morphogenesis in skin is governed by discrete sets of differentially expressed microRNAs. Nature Genetics 2006;38:356–62.CrossRef
Zhou, X, Jeker, LT, Fife, BT, et al. Selective miRNA disruption in T reg cells leads to uncontrolled autoimmunity. Journal of Experimental Medicine 2008;205:1983–91.CrossRefGoogle Scholar
Rao, PK, Kumar, RM, Farkhondeh, M, Baskerville, S, Lodish, HF. Myogenic factors that regulate expression of muscle-specific microRNAs. Proceedings of the National Academy of Sciences USA 2006;103:8721–6.CrossRef
Chen, JF, Mandel, EM, Thomson, JM, et al. The role of microRNA-1 and microRNA-133 in skeletal muscle proliferation and differentiation. Nature Genetics 2006;38:228–33.CrossRef
Zhao, Y, Samal, E, Srivastava, D. Serum response factor regulates a muscle-specific microRNA that targets Hand2 during cardiogenesis. Nature 2005;436:214–20.CrossRef
Zhao, Y, Ransom, JF, Li, A, et al. Dysregulation of cardiogenesis, cardiac conduction, and cell cycle in mice lacking miRNA-1–2. Cell 2007;129:303–17.CrossRef
Calin, GA, Dumitru, CD, Shimizu, M, et al. Frequent deletions and down-regulation of micro- RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proceedings of the National Academy of Sciences USA 2002;99:15 524–9.
Croce, CM. Causes and consequences of microRNA dysregulation in cancer. Nature Reviews Genetics 2009;10:704–14.CrossRef
Meng, F, Hensn, R, Wehbe-Janek, H, et al. MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer. Gastroenterology 2007;133:647–58.CrossRef
Cimmino, A, Calin, GA, Fabbri, M, et al. miR-15 and miR-16 induce apoptosis by targeting BCL2. Proceedings of the National Academy of Sciences USA 2005;102:13 944–9.
le Sage, C, Nagel, R, Egan, DA, et al. Regulation of the p27(Kip1) tumor suppressor by miR-221 and miR-222 promotes cancer cell proliferation. EMBO Journal 2007;26:3699–708.CrossRef
Fornari, F, Gramantieri, L, Ferracin, M, et al. MiR-221 controls CDKN1C/p57 and CDKN1B/p27 expression in human hepatocellular carcinoma. Oncogene 2008;27:5651–61.CrossRef
Garofalo, M, Di leva, G, Romano, G, et al. miR-221&222 regulate TRAIL resistance and enhance tumorigenicity through PTEN and TIMP3 downregulation. Cancer Cell 2009;16:498–509.CrossRef
Felli, N, Fontana, L, Pelosi, E, et al. MicroRNAs 221 and 222 inhibit normal erythropoiesis and erythroleukemic cell growth via kit receptor down-modulation. Proceedings of the National Academy of Sciences USA 2005;102:18 081–6.
Calin, GA, Croce, CM. MicroRNAs and chromosomal abnormalities in cancer cells. Oncogene 2006;25:6202–10.CrossRef
Lujambio, A, Calin, GA, Villanueava, A, et al. A microRNA DNA methylation signature for human cancer metastasis. Proceedings of the National Academy of Sciences USA 2008;105:13 556–61.
Bandres, E, Agirre, X, Bitarte, N, et al. Epigenetic regulation of microRNA expression in colorectal cancer. International Journal of Cancer 2009;125:2737–43.CrossRefGoogle ScholarPubMed
Roman-Gomez, J, Agirre, X, Jiménez-Velasco, A, et al. Epigenetic regulation of microRNAs in acute lymphoblastic leukemia. Journal of Clinical Oncology 2009;27:1316–22.CrossRefGoogle ScholarPubMed
He, L, He, X, Lim, LP, et al. A microRNA component of the p53 tumour suppressor network. Nature 2007;447:1130–4.CrossRef
Chang, TC, Yu, D, Lee, YS, et al. Widespread microRNA repression by Myc contributes to tumorigenesis. Nature Genetics 2008;40:43–50.CrossRef
Schulte, JH, Horn, S, Otto, T, et al. MYCN regulates oncogenic MicroRNAs in neuroblastoma. International Journal of Cancer 2007;122:699–704.CrossRefGoogle Scholar
Melo, SA, Ropero, S, Moutinho, C, et al. A TARBP2 mutation in human cancer impairs microRNA processing and DICER1 function. Nature Genetics 2009;41:365–70.CrossRef
He, L, Thomson, JM, Hemann, MT, et al. A microRNA polycistron as a potential human oncogene. Nature 2005;435:828–33.CrossRef
Ventura, A, Young, AG, Winslow, MM, et al. Targeted deletion reveals essential and overlapping functions of the miR-17 through 92 family of miRNA clusters. Cell 2008;132:875–86.CrossRef
Volinia, S, Calin, GA, Liu, CG, et al. A microRNA expression signature of human solid tumors defines cancer gene targets. Proceedings of the National Academy of Sciences USA 2006;103:2257–61.CrossRef
Costinean, S, Zanesi, N, Pekarsky, Y, et al. Pre-B cell proliferation and lymphoblastic leukemia/high-grade lymphoma in E(mu)-miR155 transgenic mice. Proceedings of the National Academy of Sciences USA 2006;103:7024–9.CrossRef
Costinean, S, Sandhu, SK, Pedersen, IM, et al. Src homology 2 domain-containing inositol-5-phosphatase and CCAAT enhancer-binding protein beta are targeted by miR-155 in B cells of Emicro-MiR-155 transgenic mice. Blood 2009;114:1374–82.CrossRef
Viswanathan, SR, Daley, GQ, Gregory, RI. Selective blockade of microRNA processing by Lin28. Science 2008;320:97–100.CrossRef
Heo, I, Joo, C, Cho, J, et al. Lin28 mediates the terminal uridylation of let-7 precursor microRNA. Molecular Cell 2008;32:276–84.CrossRef
Kumar, MS, Erkeland, SJ, Paster, RE, et al. Suppression of non-small cell lung tumor development by the let-7 microRNA family. Proceedings of the National Academy of Sciences USA 2008;105:3903–8.CrossRef
Esquela-Kerscher, A, Trang, P, Wiggins, JF, et al. The let-7 microRNA reduces tumor growth in mouse models of lung cancer. Cell Cycle 2008;7:759–64.CrossRef
Klein, U, Lia, M, Crespo, M, et al. The DLEU2/miR-15a/16–1 cluster controls B cell proliferation and its deletion leads to chronic lymphocytic leukemia. Cancer Cell 2010;17:28–40.CrossRef
Negrini, M, Nicoloso, MS, Calin, GA. MicroRNAs and cancer: new paradigms in molecular oncology. Current Opinion in Cell Biology 2009;21:470–9.CrossRef
Nicoloso, MS, Spizzo, R, Shimizu, M, Rossi, S, Calin, GA. MicroRNAs: the micro steering wheel of tumour metastases. Nature Reviews Cancer 2009;9:293–302.CrossRef
Cole, KA, Attiyeh, EF, Mosse, YP, et al. A functional screen identifies miR-34a as a candidate neuroblastoma tumor suppressor gene. Molecular Cancer Research 2008;6:735–42.CrossRef
Welch, C, Chen, Y, Stallings, RL. MicroRNA-34a functions as a potential tumor suppressor by inducing apoptosis in neuroblastoma cells. Oncogene 2007:26:5017–22.
Yamakuchi, M, Ferlito, M, Lowenstein, CJ. miR-34a repression of SIRT1 regulates apoptosis. Proceedings of the National Academy of Sciences USA 2008;105:13 421–6.
Garofalo, M, Quintavalle, C, Di Leva, G, et al. MicroRNA signatures of TRAIL resistance in human non-small cell lung cancer. Oncogene 2008;27:3845–55.CrossRef
Hua, Z, Lv, Q, Ye, W, et al. MiRNA-directed regulation of VEGF and other angiogenic factors under hypoxia. PLoS One 2006;1: e116.
Fasanaro, P, D’Alessandra, Y, Di Stefano, V, et al. MicroRNA-210 modulates endothelial cell response to hypoxia and inhibits the receptor tyrosine kinase ligand Ephrin-A3. Journal of Biological Chemistry 2008;283:15 878–83.CrossRefGoogle ScholarPubMed
Furney, SJ, Madden, SF, Kisiel, TA, Higgins, DG, Lopez-Bigaz, N. Distinct patterns in the regulation and evolution of human cancer genes. In Silico Biology 2008;8:33–46.
Martello, G, Rosato, A, Perrari, F, et al. A microRNA targeting dicer for metastasis control. Cell 2010;141:1195–207.CrossRef
Valastyan, S, Chang, A, Benaich, N, Reinhardt, F, Weinberg, RA. Concurrent suppression of integrin alpha5, radixin, and RhoA phenocopies the effects of miR-31 on metastasis. Cancer Research 2010;70:5147–54.CrossRef
Tavazoie, SF, Alarcón, C, Oskarsson, T, et al. Endogenous human microRNAs that suppress breast cancer metastasis. Nature 2008;451:147–52.CrossRef
Calin, GA, Ferracin, M, Cimmino, A, et al. A microRNA signature associated with prognosis and progression in chronic lymphocytic leukemia. New England Journal of Medicine 2005;353:1793–801.CrossRefGoogle ScholarPubMed
Jazdzewski, K, Murray, EL, Franssila, K, et al. Common SNP in pre-miR-146a decreases mature miR expression and predisposes to papillary thyroid carcinoma. Proceedings of the National Academy of Sciences USA 2008;105:7269–74.CrossRef
Krichevsky, AM, Gabriely, G.miR-21: a small multi-faceted RNA. Journal of Cellular and Molecular Medicine 2009;13:39–53.CrossRefGoogle ScholarPubMed
Yanaihara, N, Caplen, N, Bowman, E, et al. Unique microRNA molecular profiles in lung cancer diagnosis and prognosis. Cancer Cell 2006;9:189–98.CrossRef
Lu, J, Getz, G, Miska, EA, et al. MicroRNA expression profiles classify human cancers. Nature 2005;435:834–8.CrossRef
Yan, LX, Huang, XF, Shao, Q, et al. MicroRNA miR-21 overexpression in human breast cancer is associated with advanced clinical stage, lymph node metastasis and patient poor prognosis. RNA 2008;14:2348–60.CrossRef
Schetter, AJ, Leung, SY, Sohn, JJ, et al. MicroRNA expression profiles associated with prognosis and therapeutic outcome in colon adenocarcinoma. Journal of the American Medical Association 2008;299:425–36.Google ScholarPubMed
Bianchi, F, Nicasso, F, Marzi, M, et al. A serum circulating miRNA diagnostic test to identify asymptomatic high-risk individuals with early stage lung cancer. EMBO Molecular Medicine 2011;3:495–503.CrossRef
Boeri, M, Verri, C, Conte, D, et al. MicroRNA signatures in tissues and plasma predict development and prognosis of computed tomography detected lung cancer. Proceedings of the National Academy of Sciences of the USA 2011;108:3713–18.CrossRef
Weiss, GJ, Bemis, LT, Nakajima, E, et al. EGFR regulation by microRNA in lung cancer: correlation with clinical response and survival to gefitinib and EGFR expression in cell lines. Annals of Oncology 2008;19:1053–9.CrossRef
Jung, EJ, Santarpia, L, Kim, J, et al. Plasma microRNA 210 levels correlate with sensitivity to trastuzumab and tumor presence in breast cancer patients. Cancer 2012;118:2603–14.CrossRef
Ebert, MS, Neilson, JR, Sharp, PA. MicroRNA sponges: competitive inhibitors of small RNAs in mammalian cells. Nature Methods 2007;4:721–6.CrossRef
Weiler, J, Hunziker, J, Hall, J. Anti-miRNA oligonucleotides (AMOs): ammunition to target miRNAs implicated in human disease? Gene Therapy 2006;13:496–502.
Valastyan, S, Reinhardt, F, Benaich, N, et al. A pleiotropically acting microRNA, miR-31, inhibits breast cancer metastasis. Cell 2009;137:1032–46.CrossRef
Tanaka, T, Mangala, LS, Vivas-Mejia, PE, et al. Sustained small interfering RNA delivery by mesoporous silicon particles. Cancer Research 2010;70:3687–96.CrossRef
McLaughlin, J, Cheng, D, Singer, O, et al. Sustained suppression of Bcr-Abl-driven lymphoid leukemia by microRNA mimics. Proceedings of the National Academy of Sciences USA 2007;104:20 501–6.
Aravin, AA, Naumova, NMTulin, AV, et al. Double-stranded RNA-mediated silencing of genomic tandem repeats and transposable elements in the D. melanogaster germline. Current Biology 2001;11:1017–27.
Kazazian, HH Mobile elements: drivers of genome evolution. Science 2004;303:1626–32.CrossRef
McClintock, B. Chromosome organization and genic expression. Cold Spring Harbor Symposia on Quantitative Biology 1951;16:13–47.CrossRef
Malone, CD, Hannon, GJ. Small RNAs as guardians of the genome. Cell 2009;136:656–68.CrossRef
Vagin, VV, Sigova, A, Li, C, et al. A distinct small RNA pathway silences selfish genetic elements in the germline. Science 2006;313:320–4.CrossRef
Aravin, A, Gaodatzis, D, Pfeffer, S, et al. A novel class of small RNAs bind to MILI protein in mouse testes. Nature 2006;442:203–7.CrossRef
Girard, A, Sachidanandam, R, Hannon, GJ, Carmell, MA. A germline-specific class of small RNAs binds mammalian Piwi proteins. Nature 2006;442:199–202.CrossRef
Grivna, ST, Beyret, E, Wang, Z, Lin, H. A novel class of small RNAs in mouse spermatogenic cells. Genes and Development 2006;20:1709–1714.CrossRef
Hutvagner, G, Simard, MJ. Argonaute proteins: key players in RNA silencing. Nature Reviews Molecular and Cell Biology 2008;9:22–32.CrossRef
Cox, DN, Chao, A, Baker, J, et al. A novel class of evolutionarily conserved genes defined by piwi are essential for stem cell self-renewal. Genes and Development 1998;12:3715–27.CrossRef
Aravin, AA, Sachidanandam, R, Girard, A, Fejes-Toth, K, Hannon, GJ. Developmentally regulated piRNA clusters implicate MILI in transposon control. Science 2007;316:744–7.CrossRef
Aravin, AA, Sachidanandam, R, Bourc'his, D, et al. A piRNA pathway primed by individual transposons is linked to de novo DNA methylation in mice. Molecular Cell 2008;31:785–99.CrossRef
Brennecke, J, Aravin, AA, Stark, A, et al. Discrete small RNA-generating loci as master regulators of transposon activity in Drosophila. Cell 2007;128:1089–103.CrossRef
Robine, N, Lau, NC, Balla, S, et al. A broadly conserved pathway generates 3ʹUTR-directed primary piRNAs. Current Biology 2009;19:2066–76.CrossRef
Gunawardane, LS, Saito, K, Nishida, KM, et al. A slicer-mediated mechanism for repeat-associated siRNA 5ʹ end formation in Drosophila. Science 2007;315:1587–90.CrossRef
Houwing, S, Kamminga, LM, Berezikov, E, et al. A role for Piwi and piRNAs in germ cell maintenance and transposon silencing in Zebrafish. Cell 2007;129:69–82.CrossRef
Visvader, JE, Lindeman, GJ. Cancer stem cells in solid tumours: accumulating evidence and unresolved questions. Nature Reviews Cancer 2008;8:755–68.CrossRef
Chen, L, Shen, R, Ye, Y, et al. Precancerous stem cells have the potential for both benign and malignant differentiation. PLoS One 2007;2:e293.
Qiao, D, Zeeman, AM, Deng, W, Looijenga, LH, Lin, H. Molecular characterization of hiwi, a human member of the piwi gene family whose overexpression is correlated to seminomas. Oncogene 2002;21:3988–99.CrossRef
Lee, JH, Schütte, D, Wulf, G, et al. Stem-cell protein Piwil2 is widely expressed in tumors and inhibits apoptosis through activation of Stat3/Bcl-XL pathway. Human Molecular Genetics 2006;15:201–11.CrossRef
Liu, X, Sun, Y, Guo, J, et al. Expression of hiwi gene in human gastric cancer was associated with proliferation of cancer cells. International Journal of Cancer 2006;118:1922–9.CrossRefGoogle ScholarPubMed
Taubert, H, Greither, T, Kaushal, D, et al. Expression of the stem cell self-renewal gene Hiwi and risk of tumour-related death in patients with soft-tissue sarcoma. Oncogene 2007;26:1098–100.CrossRef
He, W, Wang, Z, Wang, Q, et al. Expression of HIWI in human esophageal squamous cell carcinoma is significantly associated with poorer prognosis. BMC Cancer 2009;9;426.
Li, S, Meng, L, Zhu, C, et al. The universal overexpression of a cancer testis antigen hiwi is associated with cancer angiogenesis. Oncology Reports 2010;23:1063–8.
Wang, QE, Han, C, Milum, K, Wani, AA. Stem cell protein Piwil2 modulates chromatin modifications upon cisplatin treatment. Mutation Research 2011;708:59–68.CrossRef
Fire, A, Xu, S, Montgomery, MK, et al. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature 1998;391:806–11.CrossRef
Hamilton, A, Voinnet, O, Chappell, L, Baulcombe, D. Two classes of short interfering RNA in RNA silencing. EMBO Journal 2002;21:4671–9.CrossRef
Tam, OH, Aravin, AA, Stein, P, et al. Pseudogene-derived small interfering RNAs regulate gene expression in mouse oocytes. Nature 2008;453:534–8.CrossRef
Watanabe, T, Totoki, Y, Toyoda, A, et al. Endogenous siRNAs from naturally formed dsRNAs regulate transcripts in mouse oocytes. Nature 2008;453:539–43.CrossRef
Ghildiyal, M, Seitz, H, Horwich, MD, et al. Endogenous siRNAs derived from transposons and mRNAs in Drosophila somatic cells. Science 2008;320:1077–81.CrossRef
Chung, W-J, Okamura, K, Martin, R, Lai, EC. Endogenous RNA interference provides a somatic defense against Drosophila transposons. Current Biology 2008;18:795–802.CrossRef
Czech, B, Malone, CD, Zhou, R, et al. An endogenous small interfering RNA pathway in Drosophila. Nature 2008;453:798–802.CrossRef
Kawamura, Y, Saito, K, Kin, T, et al. Drosophila endogenous small RNAs bind to Argonaute 2 in somatic cells. Nature 2008;453:793–7.CrossRef
Okamura, K, Chung, WJ, Ruby, JG, et al. The Drosophila hairpin RNA pathway generates endogenous short interfering RNAs. Nature 2008;453:803–6.CrossRef
Okamura, K, Balla, S, Martin, R, Liu, N, Lai, EC. Two distinct mechanisms generate endogenous siRNAs from bidirectional transcription in Drosophila melanogaster. Nature Structural and Molecular Biology 2008;15:581–90.CrossRef

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×