The H19 non-coding RNA is essential for human tumor growth

Imad J Matouk, Nathan DeGroot, Shaul Mezan, Suhail Ayesh, Rasha Abu-lail, Abraham Hochberg, Eithan Galun, Imad J Matouk, Nathan DeGroot, Shaul Mezan, Suhail Ayesh, Rasha Abu-lail, Abraham Hochberg, Eithan Galun

Abstract

Background: Mutations and epigenetic aberrant signaling of growth factors pathways contribute to carcinogenesis. Recent studies reveal that non-coding RNAs are controllers of gene expression. H19 is an imprinted gene that demonstrates maternal monoallelic expression without a protein product; although its expression is shut off in most tissues postnatally, it is re-activated during adult tissue regeneration and tumorigenesis. Moreover, H19 is highly expressed in liver metastasis derived from a range of carcinomas. The objective of this study is to explore the role of H19 in carcinogenesis, and to determine its identification as an anti-tumor target.

Methodology/principle findings: By controlling oxygen pressure during tumor cell growth and H19 expression levels, we investigated the role of H19 expression in vitro and in vivo in hepatocellular (HCC) and bladder carcinoma. Hypoxia upregulates the level of H19 RNA. Ablations of tumorigenicity of HCC and bladder carcinomas in vivo are seen by H19 knockdown which also significantly abrogates anchorage-independent growth after hypoxia recovery, while ectopic H19 expression enhances tumorigenic potential of carcinoma cells in vivo. Knocking-down H19 message in hypoxic stress severely diminishes p57(kip2) induction. We identified a number of potential downstream targets of H19 RNA, including angiogenin and FGF18.

Conclusions: H19 RNA harbors pro-tumorigenic properties, thus the H19 gene behaves as an oncogene and may serve as a potential new target for anti-tumor therapy.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. H19 RNA is largely induced…
Figure 1. H19 RNA is largely induced in response to hypoxic stress and moderately by hypoxia-mimicking condition triggered by CoCl2 in Hep3B.
Hep3B cells were cultured under normal culture conditions for 24 hours before hypoxic or CoCl2 manipulation. Cells were either placed into an aneoropack rectangular jar to create a hypoxic condition within an hour, or left under normal culture conditions. Incubation lasted for 24 hours before RNA extraction. (A) Shown are RT-PCR products of H19 gene (28 PCR cycles) cultured under normal conditions-lane 1, or hypoxic conditions-lane 2 (lane M indicates the marker, and C is a PCR blank without a target). (B) PCR for Histone H3.3 as a positive control for RT-PCR integrity. Shown also are RT-PCR products of both the H19 gene (32 PCR cycles) (C), the GAPDH gene (D) for untreated Hep3B (lane1) and for 50, 100, 200, 300 and 400 µM CoCl2 treated cells (lanes 2, 3, 4, 5 and 6, respectively). Incubation with the indicated concentrations of CoCl2 lasted for an additional 22 hours before RNA extraction. QPCR analysis for H19 RNA levels normalized to β-actin in Hep3B treated CoCl2 is shown in (E) where the numbers above the bars indicate the concentrations of CoCl2 used.
Figure 2. Effect of four different H19…
Figure 2. Effect of four different H19 siRNA duplexes on the expression level of H19 in the Hep3B cell line under normal culture condition (A) and hypoxia-mimicking CoCl2 treatment (B).
Shown is H19 RNA levels (34 PCR cycles) in (A): Hep3B cells transfected with unrelated siRNA duplex that targets luciferase gene (lane 1) and with 4 different H19 siRNA1 to H19 siRNA4 duplexes (lanes 2–5) and an equimolar mixture of the four siRNAs (lane 6); for controls, we performed transfection also with lipofectamine 2000 without siRNA (Mock) (lane 7) and C is PCR blank without a target. (B): Hep3B cells were transfected under normal medium with siRNA duplex that targets the luciferase gene (lanes 1 and 5) and with 3 different H19 siRNA duplexes (lanes 2–4). Twenty four hours post transfection, media was changed, and 100 µM CoCl2 containing media was added except for lane 5 which continued to grow under normal culture media; the incubation lasted for an additional 22 hours. (C): Shown are RT-PCR products of GAPDH gene as a positive control for RT-PCR integrity.
Figure 3. H19 RNA is induced by…
Figure 3. H19 RNA is induced by hypoxic stress in Hep3B cell line and siRNA directed against H19 very efficiently impedes its induction.
Hep3B cells were seeded and transfected either with H19 siRNA or luc siRNA . Twenty four hours post transfection, cells were either placed into an aneoropack rectangular jar , or left under normal oxygen concentration. Incubation lasted for 24 hours before RNA extraction. Shown are RT-PCR analyses for H19 RNA (28 PCR cycles). (A): Hep3B transfected with Luc siRNA (lanes 1, 2) and H19 siRNA (lanes 3, 4) both in normal (lanes 1, 3) and hypoxic (lanes 2, 4) culture conditions, respectively. PCR analysis of a house-keeping gene Histone H3.3 (B), and uPAR (C). In (D) QPCR analysis for H19 RNA levels normalized to β-actin in Hep3B cells is depicted.. A Quantitative SYBR Green RT–PCR was performed on human total RNA for both H19 and β-actin to estimate H19 RNA copy number under different manipulations and efficiency of knockdown under hypoxic conditions.
Figure 4. Transient H19 RNA knockdown in…
Figure 4. Transient H19 RNA knockdown in Hep3B cells inhibited tumorigenicity in vivo.
Hep3B cells were transiently transfected with H19 siRNA3 or Luc siRNA. Forty eight hours post transfection, cells were washed twice with PBS, trypsinized and counted. Equal numbers of cells (1.5×106) were injected subcutaneously into the dorsal part of CD-1 nude mice (n = 7 for both, and 4 for mock transfected). Palpable tumors were observed 15 days post inoculation in mice inoculated with Hep3B, transiently transfected with Luc siRNA. Tumor volumes were followed up and measured using a caliper until day 30 post inoculation, after which mice were sacrificed. Significant (p<0.03) reductions of about 82% of both mean tumor weights (A) (± standard error) and mean tumor volumes (p<0.03) (B) (± standard error) were observed. Values represent end-points just before and after sacrificing animals. Shown are also representative features of tumors in 2 mice of each group (mice 1 and 2 are the H19 siRNA3 treated Hep3B cells, and mice 3 and 4 are the Luc siRNA ) before tumor surgical exposure (C), and after exposure of their internal tumors (D).
Figure 5. The effect of over-expression of…
Figure 5. The effect of over-expression of H19 RNA on the tumorigenic potential of bladder carcinoma cells in vivo.
Equal amounts (2×106) of TA31H19high and TA11H19-ve cells were implanted subcutaneously to CD-1 mice (n = 5, each). Two weeks later, palpable tumors appeared and tumor volumes were measured for an additional two weeks . Shown are end point measurements of the mean tumor volumes of the two groups (upper left pannel), their mean tumor volumes kinetics (upper right), and a representative gross morphology of tumors derived from the TA11H19-ve (lower left) and TA31H19high cells(down and right).
Figure 6. The in vivo effect of…
Figure 6. The in vivo effect of H19 silencing on the tumorigenic potential of human bladder carcinoma cells-UMUC3.
One million UMUC3 cells were injected subcutaneously to athymic mice (n = 3 for GFP siRNA, and 5 for H19 siRNA ), 48 hours after transiently transfected with siRNAs. Palpable tumors were observed 6 weeks later in 2 out of 3 mice of the GFP siRNA group, while in none of the H19 siRNA group . Mice were sacrificed 8 weeks after inoculation. Mean tumor volumes (B, P

Figure 7. H19 RNA knockdown impedes p57Kip2…

Figure 7. H19 RNA knockdown impedes p57Kip2 mRNA induction in response to hypoxic stress, in…

Figure 7. H19 RNA knockdown impedes p57Kip2 mRNA induction in response to hypoxic stress, in Hep3B cell line.
Hep3B cells were seeded and transfected either with H19 siRNA or Luc siRNA as described . Shown are RT-PCR analyses for H19 RNA (28 PCR cycles). (A): Hep3B transfected with luc siRNA (lanes 1, 2) and H19 siRNA (lanes 3, 4) both in normal (lanes 1, 3) and hypoxic (lanes 2, 4) culture conditions, respectively. And similar treatments are assessing the mRNA levels of: (B). p19INK4 (C). p57Kip2 (D). Histone.

Figure 8. Expression levels of selected genes…

Figure 8. Expression levels of selected genes in Hep3B cells transfected either with H19 and…

Figure 8. Expression levels of selected genes in Hep3B cells transfected either with H19 and GFP siRNAs under normoxic and hypoxic conditions.
A few genes that showed variations as a result of H19 knockdown under different manipulations were selected, based on their potential importance in tumorigenesis, for further RT-PCR analysis to confirm the results of the microarray data. In the figure +denotes (+H19) and –(-H19). The genes chosen were (A)-FGF18; (B)-IGFBP4; (C)-ANG; (D)-LBR; and (E)-AKT1 . The knockdown, of H19 and the RT-PCR integrity are shown in Figure 7.
All figures (8)
Figure 7. H19 RNA knockdown impedes p57Kip2…
Figure 7. H19 RNA knockdown impedes p57Kip2 mRNA induction in response to hypoxic stress, in Hep3B cell line.
Hep3B cells were seeded and transfected either with H19 siRNA or Luc siRNA as described . Shown are RT-PCR analyses for H19 RNA (28 PCR cycles). (A): Hep3B transfected with luc siRNA (lanes 1, 2) and H19 siRNA (lanes 3, 4) both in normal (lanes 1, 3) and hypoxic (lanes 2, 4) culture conditions, respectively. And similar treatments are assessing the mRNA levels of: (B). p19INK4 (C). p57Kip2 (D). Histone.
Figure 8. Expression levels of selected genes…
Figure 8. Expression levels of selected genes in Hep3B cells transfected either with H19 and GFP siRNAs under normoxic and hypoxic conditions.
A few genes that showed variations as a result of H19 knockdown under different manipulations were selected, based on their potential importance in tumorigenesis, for further RT-PCR analysis to confirm the results of the microarray data. In the figure +denotes (+H19) and –(-H19). The genes chosen were (A)-FGF18; (B)-IGFBP4; (C)-ANG; (D)-LBR; and (E)-AKT1 . The knockdown, of H19 and the RT-PCR integrity are shown in Figure 7.

References

    1. Farazi PA, DePinho RA. Hepatocellular carcinoma pathogenesis: from genes to environment. Nat Rev Cancer. 2006;6:674–87.
    1. Thorgeirsson SS, Lee JS, Grisham JW. Functional genomics of hepatocellular carcinoma. Hepatology. 2006;43:S145–50.
    1. Poon TC, Wong N, Lai PB, Rattray M, Johnson PJ, et al. A tumor progression model for hepatocellular carcinoma: bioinformatic analysis of genomic data. Gastroenterology. 2006;131:1262–70.
    1. Laurent-Puig P, Zucman-Rossi J. Genetics of hepatocellular tumors. Oncogene. 2006;25:3778–86.
    1. Puisieux A, Lim S, Groopman J, Ozturk M. Selective targeting of p53 gene mutational hotspots in human cancers by etiologically defined carcinogens. Cancer Res. 1991;51:6185–9.
    1. Boccaccio C, Comoglio PM. Invasive growth: a MET-driven genetic programme for cancer and stem cells. Nat Rev Cancer. 2006;6:637–45.
    1. Kaposi-Novak P, Lee JS, Gomez-Quiroz L, Coulouarn C, Factor VM, et al. Met-regulated expression signature defines a subset of human hepatocellular carcinomas with poor prognosis and aggressive phenotype. J Clin Invest. 2006;116:1582–95.
    1. Katzenellenbogen M, Pappo O, Barash H, Klopstock N, Mizrahi L, et al. Multiple adaptive mechanisms to chronic liver disease revealed at early stages of liver carcinogenesis in the Mdr2-knockout mice. Cancer Res. 2006;66:4001–10.
    1. Pikarsky E, Porat RM, Stein I, Abramovitch R, Amit S, et al. NF-kappaB functions as a tumour promoter in inflammation-associated cancer. Nature. 2004;431:461–6.
    1. Wald O, Izhar U, Amir G, Avniel S, Bar-Shavit Y, et al. CD4+CXCR4highCD69+ T Cells Accumulate in Lung Adenocarcinoma. J Immunol. 2006;177:6983–90.
    1. Maeda S, Kamata H, Luo JL, Leffert H, Karin M. IKKbeta couples hepatocyte death to cytokine-driven compensatory proliferation that promotes chemical hepatocarcinogenesis. Cell. 2005;121:977–90.
    1. Ariel I, Miao HQ, Ji XR, Schneider T, Roll D, et al. Imprinted H19 oncofetal RNA is a candidate tumour marker for hepatocellular carcinoma. Mol Pathol. 1998;51:21–5.
    1. Chaumeil J, Le Baccon P, Wutz A, Heard E. A novel role for Xist RNA in the formation of a repressive nuclear compartment into which genes are recruited when silenced. Genes Dev. 2006;20:2223–37.
    1. Calin GA, Croce CM. MicroRNA signatures in human cancers. Nat Rev Cancer. 2006;6:857–66.
    1. Gabory A, Ripoche MA, Yoshimizu T, Dandolo L. The H19 gene: regulation and function of a non-coding RNA. Cytogenet Genome Res. 2006;113:188–93.
    1. Lottin S, Adriaenssens E, Dupressoir T, Berteaux N, Montpellier C, et al. Overexpression of an ectopic H19 gene enhances the tumorigenic properties of breast cancer cells. Carcinogenesis. 2002;23:1885–95.
    1. Berteaux N, Lottin S, Monte D, Pinte S, Quatannens B, et al. H19 mRNA-like noncoding RNA promotes breast cancer cell proliferation through positive control by E2F1. J Biol Chem. 2005;280:29625–36.
    1. Dugimont T, Montpellier C, Adriaenssens E, Lottin S, Dumont L, et al. The H19 TATA-less promoter is efficiently repressed by wild-type tumor suppressor gene product p53. Oncogene. 1998;16:2395–401.
    1. Scott RE, Gao S, Kim CK, Cox R. De-differentiation-derived mesenchymal stem cells demonstrate selective repression in H19 bioregulatory RNA gene expression. Differentiation. 2005;73:294–302.
    1. Yang J, Mani SA, Donaher JL, Ramaswamy S, Itzykson RA, et al. Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell. 2004;117:927–39.
    1. Kaplan R, Luettich K, Heguy A, Hackett NR, Harvey BG, et al. Monoallelic up-regulation of the imprinted H19 gene in airway epithelium of phenotypically normal cigarette smokers. Cancer Res. 2003;63:1475–82.
    1. Elkin M, Ayesh S, Schneider T, de Groot N, Hochberg A, et al. The dynamics of the imprinted H19 gene expression in the mouse model of bladder carcinoma induced by N-butyl-N-(4-hydroxybutyl)nitrosamine. Carcinogenesis. 1998;19:2095–9.
    1. Ariel I, Ayesh S, Gofrit O, Ayesh B, Abdul-Ghani R, et al. Gene expression in the bladder carcinoma rat model. Mol Carcinog. 2004;41:69–76.
    1. Graveel CR, Jatkoe T, Madore SJ, Holt AL, Farnham PJ. Expression profiling and identification of novel genes in hepatocellular carcinomas. Oncogene. 2001;20:2704–12.
    1. Ariel I, Sughayer M, Fellig Y, Pizov G, Ayesh S, et al. The imprinted H19 gene is a marker of early recurrence in human bladder carcinoma. Mol Pathol. 2000;53:320–3.
    1. Dugimont T, Curgy JJ, Wernert N, Delobelle A, Raes MB, et al. The H19 gene is expressed within both epithelial and stromal components of human invasive adenocarcinomas. Biol Cell. 1995;85:117–24.
    1. Barsyte-Lovejoy D, Lau SK, Boutros PC, Khosravi F, Jurisica I, et al. The c-Myc oncogene directly induces the H19 noncoding RNA by allele-specific binding to potentiate tumorigenesis. Cancer Res. 2006;66:5330–7.
    1. Coulouarn C, Gomez-Quiroz LE, Lee JS, Kaposi-Novak P, Conner EA, et al. Oncogene-specific gene expression signatures at preneoplastic stage in mice define distinct mechanisms of hepatocarcinogenesis. Hepatology. 2006;44:1003–11.
    1. Adriaenssens E, Lottin S, Berteaux N, Hornez L, Fauquette W, et al. Cross-talk between mesenchyme and epithelium increases H19 gene expression during scattering and morphogenesis of epithelial cells. Exp Cell Res. 2002;275:215–29.
    1. Iizuka N, Oka M, Yamada-Okabe H, Mori N, Tamesa T, et al. Comparison of gene expression profiles between hepatitis B virus- and hepatitis C virus-infected hepatocellular carcinoma by oligonucleotide microarray data on the basis of a supervised learning method. Cancer Res. 2002;62:3939–44.
    1. Kim KS, Lee YI. Biallelic expression of the H19 and IGF2 genes in hepatocellular carcinoma. Cancer Lett. 1997;119:143–8.
    1. Manoharan H, Babcock K, Willi J, Pitot HC. Biallelic expression of the H19 gene during spontaneous hepatocarcinogenesis in the albumin SV40 T antigen transgenic rat. Mol Carcinog. 2003;38:40–7.
    1. Ayesh S, Matouk I, Schneider T, Ohana P, Laster M, et al. Possible physiological role of H19 RNA. Mol Carcinog. 2002;35:63–74.
    1. Sells MA, Chen ML, Acs G. Production of hepatitis B virus particles in Hep G2 cells transfected with cloned hepatitis B virus DNA. Proc Natl Acad Sci U S A. 1987;84:1005–9.
    1. Aoki Y, Aizaki H, Shimoike T, Tani H, Ishii K, et al. A human liver cell line exhibits efficient translation of HCV RNAs produced by a recombinant adenovirus expressing T7 RNA polymerase. Virology. 1998;250:140–50.
    1. Fellig Y, Almogy G, Galun E, Ketzinel-Gilad M. A hepatocellular carcinoma cell line producing mature hepatitis B viral particles. Biochem Biophys Res Commun. 2004;321:269–74.
    1. Reynolds A, Leake D, Boese Q, Scaringe S, Marshall WS, et al. Rational siRNA design for RNA interference. Nat Biotechnol. 2004;22:326–30.
    1. Elkin M, Shevelev A, Schulze E, Tykocinsky M, Cooper M, et al. The expression of the imprinted H19 and IGF-2 genes in human bladder carcinoma. FEBS Lett. 1995;374:57–61.
    1. Matsuoka S, Thompson JS, Edwards MC, Bartletta JM, Grundy P, et al. Imprinting of the gene encoding a human cyclin-dependent kinase inhibitor, p57KIP2, on chromosome 11p15. Proc Natl Acad Sci U S A. 1996;93:3026–30.
    1. Ito Y, Takeda T, Sakon M, Tsujimoto M, Monden M, et al. Expression of p57/Kip2 protein in hepatocellular carcinoma. Oncology. 2001;61:221–5.
    1. Olson KA, Fett JW, French TC, Key ME, Vallee BL. Angiogenin antagonists prevent tumor growth in vivo. Proc Natl Acad Sci U S A. 1995;92:442–6.
    1. Liu Z, Lavine KJ, Hung IH, Ornitz DM. FGF18 is required for early chondrocyte proliferation, hypertrophy and vascular invasion of the growth plate. Dev Biol 2006
    1. Zhao Y, Qiu Q, Mahdi F, Shariat-Madar Z, Rojkjaer R, et al. Assembly and activation of HK-PK complex on endothelial cells results in bradykinin liberation and NO formation. Am J Physiol Heart Circ Physiol. 2001;280:H1821–9.
    1. Pandey A, Shao H, Marks RM, Polverini PJ, Dixit VM. Role of B61, the ligand for the Eck receptor tyrosine kinase, in TNF-alpha-induced angiogenesis. Science. 1995;268:567–9.
    1. Tang J, Hu G, Hanai J, Yadlapalli G, Lin Y, et al. A critical role for calponin 2 in vascular development. J Biol Chem. 2006;281:6664–72.
    1. Benezra R, Rafii S, Lyden D. The Id proteins and angiogenesis. Oncogene. 2001;20:8334–41.
    1. Ito Y, Oike Y, Yasunaga K, Hamada K, Miyata K, et al. Inhibition of angiogenesis and vascular leakiness by angiopoietin-related protein 4. Cancer Res. 2003;63:6651–7.
    1. Busca R, Berra E, Gaggioli C, Khaled M, Bille K, et al. Hypoxia-inducible factor 1{alpha} is a new target of microphthalmia-associated transcription factor (MITF) in melanoma cells. J Cell Biol. 2005;170:49–59.
    1. Wu MX, Ao Z, Prasad KV, Wu R, Schlossman SF. IEX-1L, an apoptosis inhibitor involved in NF-kappaB-mediated cell survival. Science. 1998;281:998–1001.
    1. Datta K, Li J, Bhattacharya R, Gasparian L, Wang E, et al. Protein kinase C zeta transactivates hypoxia-inducible factor alpha by promoting its association with p300 in renal cancer. Cancer Res. 2004;64:456–62.
    1. Kashatus D, Cogswell P, Baldwin AS. Expression of the Bcl-3 proto-oncogene suppresses p53 activation. Genes Dev. 2006;20:225–35.
    1. Toker A, Yoeli-Lerner M. Akt signaling and cancer: surviving but not moving on. Cancer Res. 2006;66:3963–6.
    1. Nozaki S, Sledge GW, Jr, Nakshatri H. Repression of GADD153/CHOP by NF-kappaB: a possible cellular defense against endoplasmic reticulum stress-induced cell death. Oncogene. 2001;20:2178–85.
    1. Smith P, Syed N, Crook T. Epigenetic inactivation implies a tumor suppressor function in hematologic malignancies for Polo-like kinase 2 but not Polo-like kinase 3. Cell Cycle. 2006;5:1262–4.
    1. Seibold S, Rudroff C, Weber M, Galle J, Wanner C, et al. Identification of a new tumor suppressor gene located at chromosome 8p21.3-22. Faseb J. 2003;17:1180–2.
    1. Cao X, Qin J, Xie Y, Khan O, Dowd F, et al. Regulator of G-protein signaling 2 (RGS2) inhibits androgen-independent activation of androgen receptor in prostate cancer cells. Oncogene. 2006;25:3719–34.
    1. Massoumi R, Chmielarska K, Hennecke K, Pfeifer A, Fassler R. Cyld inhibits tumor cell proliferation by blocking Bcl-3-dependent NF-kappaB signaling. Cell. 2006;125:665–77.
    1. Kiss-Toth E, Bagstaff SM, Sung HY, Jozsa V, Dempsey C, et al. Human tribbles, a protein family controlling mitogen-activated protein kinase cascades. J Biol Chem. 2004;279:42703–8.
    1. Williams M, Lisanti P. Caveolin-1 in oncogenic transformation, cancer, and metastasis. Am J Physiol Cell Physiol. 2005;288:C494–C506.
    1. Guehenneux F, Duret L, Callanan MB, Bouhas R, Hayette S, et al. Cloning of the mouse BTG3 gene and definition of a new gene family (the BTG family) involved in the negative control of the cell cycle. Leukemia. 1997;11:370–5.
    1. Ogushi T, Takahashi S, Takeuchi T, Urano T, Horie-Inoue K, et al. Estrogen receptor-binding fragment-associated antigen 9 is a tumor-promoting and prognostic factor for renal cell carcinoma. Cancer Res. 2005;65:3700–6.
    1. Pelosi G, Pastorino U, Pasini F, Maissoneuve P, Fraggetta F, et al. Independent prognostic value of fascin immunoreactivity in stage I nonsmall cell lung cancer. Br J Cancer. 2003;88:537–47.
    1. Jiang Z, Woda BA, Wu CL, Yang XJ. Discovery and clinical application of a novel prostate cancer marker: alpha-methylacyl CoA racemase (P504S). Am J Clin Pathol. 2004;122:275–89.
    1. Rubin MA, Varambally S, Beroukhim R, Tomlins SA, Rhodes DR, et al. Overexpression, amplification, and androgen regulation of TPD52 in prostate cancer. Cancer Res. 2004;64:3814–22.
    1. Moll R, Wu XR, Lin JH, Sun TT. Uroplakins, specific membrane proteins of urothelial umbrella cells, as histological markers of metastatic transitional cell carcinomas. Am J Pathol. 1995;147:1383–97.
    1. Kelly MG, Alvero AB, Chen R, Silasi DA, Abrahams VM, et al. TLR-4 signaling promotes tumor growth and paclitaxel chemoresistance in ovarian cancer. Cancer Res. 2006;66:3859–68.
    1. Tsuchiya A, Sakamoto M, Yasuda J, Chuma M, Ohta T, et al. Expression profiling in ovarian clear cell carcinoma: identification of hepatocyte nuclear factor-1 beta as a molecular marker and a possible molecular target for therapy of ovarian clear cell carcinoma. Am J Pathol. 2003;163:2503–12.
    1. Meyer AM, Dwyer-Nield LD, Hurteau GJ, Keith RL, O'Leary E, et al. Decreased lung tumorigenesis in mice genetically deficient in cytosolic phospholipase A2. Carcinogenesis. 2004;25:1517–24.
    1. Triest-van B, Pinedo M, Blaauwgeers L. Prognostic role of thymidylate synthase, thymidine phosphorylase/platelet-derived endothelial cell growth factor, and proliferation markers in colorectal cancer. Clin Cancer Res. 2000;6:1063–72.
    1. Lottin S, Vercoutter-Edouart AS, Adriaenssens E, Czeszak X, Lemoine J, et al. Thioredoxin post-transcriptional regulation by H19 provides a new function to mRNA-like non-coding RNA. Oncogene. 2002;21:1625–31.
    1. Rodesch F, Simon P, Donner C, Jauniaux E. Oxygen measurements in endometrial and trophoblastic tissues during early pregnancy. Obstet Gynecol. 1992;80:283–5.
    1. Hao Y, Crenshaw T, Moulton T, Newcomb E, Tycko B. Tumour-suppressor activity of H19 RNA. Nature. 1993;365:764–7.
    1. Moulton T, Crenshaw T, Hao Y, Moosikasuwan J, Lin N, et al. Epigenetic lesions at the H19 locus in Wilms' tumour patients. Nat Genet. 1994;7:440–7.
    1. Matouk I, Ohana P, Ayesh S. The oncofetal H19 RNA in human cancer, from the bench to the patient. Cancer Therapy. 2005;3:249–66.
    1. Moreno MJ, Ball M, Andrade MF, McDermid A, Stanimirovic DB. Insulin-like growth factor binding protein-4 (IGFBP-4) is a novel anti-angiogenic and anti-tumorigenic mediator secreted by dibutyryl cyclic AMP (dB-cAMP)-differentiated glioblastoma cells. Glia. 2006;53:845–57.
    1. He L, Thomson JM, Hemann MT, Hernando-Monge E, Mu D, et al. A microRNA polycistron as a potential human oncogene. Nature. 2005;435:828–33.
    1. O'Donnell KA, Wentzel EA, Zeller KI, Dang CV, Mendell JT. c-Myc-regulated microRNAs modulate E2F1 expression. Nature. 2005;435:839–43.
    1. Cai X, Cullen BR. The imprinted H19 noncoding RNA is a primary microRNA precursor. RNA. 2007;13:313–6.
    1. Harris AL. Hypoxia–a key regulatory factor in tumour growth. Nat Rev Cancer. 2002;2:38–47.

Source: PubMed

Подписаться