MiR675-5p Acts on HIF-1α to Sustain Hypoxic Responses: A New Therapeutic Strategy for Glioma

Alessia Lo Dico, Viviana Costa, Cristina Martelli, Cecilia Diceglie, Francesca Rajata, Aroldo Rizzo, Carmine Mancone, Marco Tripodi, Luisa Ottobrini, Riccardo Alessandro, Alice Conigliaro, Alessia Lo Dico, Viviana Costa, Cristina Martelli, Cecilia Diceglie, Francesca Rajata, Aroldo Rizzo, Carmine Mancone, Marco Tripodi, Luisa Ottobrini, Riccardo Alessandro, Alice Conigliaro

Abstract

Hypoxia is a common feature in solid tumours. In glioma, it is considered the major driving force for tumour angiogenesis and correlates with enhanced resistance to conventional therapies, increased invasiveness and a poor prognosis for patients. Here we describe, for the first time, that miR675-5p, embedded in hypoxia-induced long non-coding RNA H19, plays a mandatory role in establishing a hypoxic response and in promoting hypoxia-mediated angiogenesis. We demonstrated, in vitro and in vivo, that miR675-5p over expression in normoxia is sufficient to induce a hypoxic moreover, miR675-5p depletion in low oxygen conditions, drastically abolishes hypoxic responses including angiogenesis. In addition, our data indicate an interaction of miR675-5p, HIF-1α mRNA and the RNA Binding Protein HuR in hypoxia-induced responses. We suggest the modulation of miR675-5p as a new therapeutic option to promote or abolish hypoxia induced angiogenesis.

Keywords: Angiogenesis; HuR; VHL.; glioma; hypoxia; miRNA675; optical imaging.

Conflict of interest statement

Competing Interests: The authors declare no competing interest.

Figures

Figure 1
Figure 1
U251 glioma cells and HUVEC endothelial cells as suitable models for hypoxia studies. (A) ELISA assay for HIF-1α performed in U251 and HUVEC nuclear extracts after 6h of hypoxia. Data are expressed as Absorbance (ABS) values at 450nm. (B) Real-time PCR performed on U251 and HUVEC cells 6 hours post-hypoxia. Data were normalized for β-actin, and ΔΔct is expressed as fold of induction (FOI) of analyzed genes in hypoxia vs normoxia. The dashed line indicates control sample in normoxia. (C) ELISA assay for VEGF levels in supernatants from both cell lines after 6 hours of hypoxia. Data are expressed as pg/ml of soluble VEGF. (D) Left panel: Real time-PCR for lncRNA H19, after 6h of hypoxia, normalized for β-actin. Data are expressed as FOI of hypoxia-treated cells compared to normoxia samples. Right panel: Real time-PCR for H19's microRNAs, miR675-3p and miR675-5p, after 6h of hypoxia. Data were normalized for RNU48. Data are expressed as FOI compared to scramble-treated cells. Values are presented as the mean ± SD. Hypoxia vs normoxia * p<0.05; **p<0.01;* **p<0.001.
Figure 2
Figure 2
Gain and loss of function suggests a critical role of miR675-5p on hypoxia modulation. (A) ELISA for HIF-1α nuclear level in U251 and HUVECs transfected with miR675-5p inhibitor and exposed to hypoxia for 6h. Data are expressed as ABS values at 450nm. (B) Real-time PCR from U251 and HUVECs exposed to hypoxia after miR675-5p inhibitor or scramble control transfection. Data were normalized for β-actin and ΔΔct is expressed as fold of induction (FOI) of indicated genes after inhibitor transfection compared to scramble control (dashed line). (C) ELISA assay for VEGF level in supernatant from both cell lines transfected with miRNA inhibitor or negative control and exposed to hypoxia for 6h. Data are expressed as pg/ml of soluble VEGF. Values are presented as the mean ± SD. Inhibitor vs scramble * p<0.05; **p<0.0; ***p<0.001. (D) HIF-1α nuclear level analyzed by ELISA assay in U251 and HUVEC cells after 18h of miR675-5p mimic transfection in normoxia. Data are expressed as ABS values at 450nm. (E) Real-time PCR performed on U251 and HUVEC cells after miR675-5p mimic-transfection. Data were normalized for β-actin, and ΔΔct is expressed as fold of induction (FOI) in cells transfected with mimic compared to control (dashed line). (F) ELISA assay for VEGF level in supernatant from both cell lines 18h after mimic transfection. Data are expressed as pg/ml of soluble VEGF. Values are presented as the mean ± SD. Mimic vs scramble * p<0.05; **p<0.01;***p<0.001.
Figure 3
Figure 3
miR675-5p in vivo is able to enhance hypoxia establishment and neo-angiogenesis. (A) Luciferase in vitro assay performed on U251-HRE lysated cells after 18h of miR675-5p mimic-transfection. Data are represented as RLU (luminescent counts normalized to protein content) of mimic-treated cells compared to scramble-treated cells. Mimic vs scramble ***p<0.001. (B) Left panel: Representative (n=5) 2D bioluminescent (rainbow scale) of U251-HRE-mCherry tumours in scramble and miR675-5p mimic-treated mice (one of 5 mice) at day 12 (pre-treatment), day 15 (after two doses) and day 19 (end of treatment). Images are presented with the same scale bar. Right panel: Graphical representation of Luciferase activity over time during treatment. Data are presented as average radiance (photons/s/cm^2/steradian). (C) Left panel: Representative 2D images of mice co-injected with HypoxiSense 680 (BlueHot scale) and IntegriSense 750 (YellowHot scale) and axial images of IntegriSense750 co-registered with CT scan. All scans were performed at the final time point (day 19). Right panel: Quantification of HypoxiSense680 and IntegriSense750 fluorescence by ROI analysis for scramble and mimic-treated mice. Data are expressed as average radiance efficiency [(photons/s/cm^2/steradian)/(μW/cm^2)]. (D) Left panel: Representative (n=5) 2D mCherry fluorescent signal (rainbow scale) of U251-HRE-mCherry tumours in scramble and miR675-5p mimic-treated mice at day 19. Right panel: Graphical representation of mCherry activity at the end of treatment. Data are presented as average radiance efficiency [(photons/s/cm^2/steradian)/(μW/cm^2). Images are presented with the same scale bar. Mimic-treated mice vs scramble- treated mice * p<0.05; **p<0.01; ***p<0.001.
Figure 4
Figure 4
miR675-5p inhibitor counteracts hypoxia-mediated angiogenesis in vivo. (A) Left Panel: Representative (n=5) 2D images of scramble or miR675-5p inhibitor-treated mice co-injected with HypoxiSense 680 (BlueHot scale) and IntegriSense 750 (YellowHot scale) and axial images of IntegriSense750 co registered with CT scan. All scans were performed at the final time point (day 28). Right panel: Quantification of HypoxiSense 680 and IntegriSense 750 fluorescence by ROI analysis for scramble and miR675-5p inhibitor-treated mice (considering five mice for each treatment). Data are expressed as average radiance efficiency [(photons/s/cm^2/steradian)/(μW/cm^2)]. (B) Left Panel: Representative (n=5) 2D mCherry fluorescent signal (rainbow scale) of U251-HRE-mCherry tumours in scramble and inhibitor-treated mice (one of five mouse for each treatment) at day 28. Images are presented with the same scale bar. Right panel: Graphical representation of mCherry activity at the end of treatment considering five mice for each treatment. Data are presented as the average radiance efficiency [(photons/s/cm^2/steradian)/(μW/cm^2). Images are presented with the same scale bar. Inhibitor-treated mice vs scramble-treated mice * p<0.05; **p<0.01; ***p<0.001. (C) Immunoistochemistry for HIF-1α staining in scramble and miR675-inhibitor-treated group (scale bar is represented).
Figure 5
Figure 5
miR675-5p regulates HIF-1α mRNA stability by VHL/HuR axis. (A) Real-time PCR for VHL was done on U251 cells 18h after transfection with 5pM, 15pM, 30pM and 60pM of miR675-5p mimic. The dashed line indicates the control sample (scramble-treated). Values are presented as the mean ± SD. Mimic vs scramble * p<0.05; **p<0.01. (B) Western Blot for VHL and β-actin from U251 cells transfected with miR675-5p mimic (5pM) or scramble negative control. (C) Expression of VHL-regulated miRNA was validated by Luminescence assay kit, a secondary reporter was used for transfection normalization. Data are expressed as Relative Luciferase Activity in pGL3-pVHL-WT construct transfected with miR675-5p or miR21-5p and their relative scrambles. pGL3-control is represented as dashed line. The mean ± SD of three independent experiment is shown, and each sample was assayed in triplicate. (D) Left panel: Real-time PCR for HIF-1α performed on U251 and HUVEC cells 6 hours post-hypoxia. Data were normalized for β-actin, and ΔΔCt is expressed as fold of induction (FOI) of HIF-1α in hypoxia vs normoxia. Right panel: Real-time PCR performed on U251 and HUVEC cells 6 hours post-hypoxia and after miRNA675-5p inhibitor transfection. Data were normalized for β-actin, and ΔΔCt is expressed as fold of induction (FOI) of HIF-1α in hypoxia in inhibitor vs normoxia. (E) Left panel: Western Blot for HuR and β-actin from U251 cells transfected with miR675-5p inhibitor (5pM) or scramble negative control after 6h of hypoxia. Right panel: RNA-immunoprecipitation for HuR. Real time PCR for VEGF and HIF-1α were performed starting from HuR co-immunoprecipitated RNA in different culture conditions. Ct value was normalized to the Input RNA Ct value. Data are expressed as fold enrichment above the input percentage and presented as the mean ± SD. Inhibitor vs scramble negative control ***p<0.001. (F) Molecular mechanism of miRNA 675-5p in hypoxia condition. The dashed line indicates our experimental results.
Figure 6
Figure 6
Time lines represent the schedule of i.v. administration for n=5 mice. Upper panel: mir675-5p mimic/scramble administration. Lower panel: miR675-5p inhibitor/scramble administration.
Figure 6
Figure 6
Time lines represent the schedule of i.v. administration for n=5 mice. Upper panel: mir675-5p mimic/scramble administration. Lower panel: miR675-5p inhibitor/scramble administration.

References

    1. Vaupel P, Harrison L. Tumor hypoxia: causative factors, compensatory mechanisms, and cellular response. Oncologist. 2004;9(Suppl 5):4–9.
    1. Folkman J. Angiogenesis in cancer, vascular, rheumatoid and other disease. Nat Med. 1995;1:27–31.
    1. Mongiardi MP. Angiogenesis and hypoxia in glioblastoma: a focus on cancer stem cells. CNS Neurol Disord Drug Targets. 2012;11:878–83.
    1. Jensen RL. Hypoxia in the tumorigenesis of gliomas and as a potential target for therapeutic measures. Neurosurg Focus. 2006;20:E24.
    1. Zhong H, De Marzo AM, Laughner E, Lim M, Hilton DA, Zagzag D. et al. Overexpression of hypoxia-inducible factor 1alpha in common human cancers and their metastases. Cancer Res. 1999;59:5830–5.
    1. Mack FA, Rathmell WK, Arsham AM, Gnarra J, Keith B, Simon MC. Loss of pVHL is sufficient to cause HIF dysregulation in primary cells but does not promote tumor growth. Cancer Cell. 2003;3:75–88.
    1. Oliver L, Olivier C, Marhuenda FB, Campone M, Vallette FM. Hypoxia and the malignant glioma microenvironment: regulation and implications for therapy. Curr Mol Pharmacol. 2009;2:263–84.
    1. Brat DJ, Van Meir EG. Vaso-occlusive and prothrombotic mechanisms associated with tumor hypoxia, necrosis, and accelerated growth in glioblastoma. Lab Invest. 2004;84:397–405.
    1. Ke Q, Costa M. Hypoxia-inducible factor-1 (HIF-1) Mol Pharmacol. 2006;70:1469–80.
    1. Semenza GL, Nejfelt MK, Chi SM, Antonarakis SE. Hypoxia-inducible nuclear factors bind to an enhancer element located 3' to the human erythropoietin gene. Proc Natl Acad Sci U S A. 1991;88:5680–4.
    1. Berteaux N, Lottin S, Monte D, Pinte S, Quatannens B, Coll J. 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. Berteaux N, Aptel N, Cathala G, Genton C, Coll J, Daccache A. et al. A novel H19 antisense RNA overexpressed in breast cancer contributes to paternal IGF2 expression. Mol Cell Biol. 2008;28:6731–45.
    1. Byun HM, Wong HL, Birnstein EA, Wolff EM, Liang G, Yang AS. Examination of IGF2 and H19 loss of imprinting in bladder cancer. Cancer Res. 2007;67:10753–8.
    1. Luo M, Li Z, Wang W, Zeng Y, Liu Z, Qiu J. Upregulated H19 contributes to bladder cancer cell proliferation by regulating ID2 expression. FEBS J. 2013;280:1709–16.
    1. Kim SJ, Park SE, Lee C, Lee SY, Jo JH, Kim JM. et al. Alterations in promoter usage and expression levels of insulin-like growth factor-II and H19 genes in cervical carcinoma exhibiting biallelic expression of IGF-II. Biochim Biophys Acta. 2002;1586:307–15.
    1. Matouk IJ, DeGroot N, Mezan S, Ayesh S, Abu-lail R, Hochberg A. et al. The H19 non-coding RNA is essential for human tumor growth. PLoS One. 2007;2:e845.
    1. Conigliaro A, Costa V, Lo Dico A, Saieva L, Buccheri S, Dieli F. et al. CD90+ liver cancer cells modulate endothelial cell phenotype through the release of exosomes containing H19 lncRNA. Mol Cancer. 2015;14:155.
    1. Tsang WP, Ng EK, Ng SS, Jin H, Yu J, Sung JJ. et al. Oncofetal H19-derived miR-675 regulates tumor suppressor RB in human colorectal cancer. Carcinogenesis. 2010;31:350–8.
    1. Shi Y, Wang Y, Luan W, Wang P, Tao T, Zhang J. et al. Long non-coding RNA H19 promotes glioma cell invasion by deriving miR-675. PLoS One. 2014;9:e86295.
    1. Zhuang M, Gao W, Xu J, Wang P, Shu Y. The long non-coding RNA H19-derived miR-675 modulates human gastric cancer cell proliferation by targeting tumor suppressor RUNX1. Biochem Biophys Res Commun. 2014;448:315–22.
    1. Li H, Yu B, Li J, Su L, Yan M, Zhu Z. et al. Overexpression of lncRNA H19 enhances carcinogenesis and metastasis of gastric cancer. Oncotarget. 2014;5:2318–29.
    1. Hernandez JM, Elahi A, Clark CW, Wang J, Humphries LA, Centeno B. et al. miR-675 mediates downregulation of Twist1 and Rb in AFP-secreting hepatocellular carcinoma. Ann Surg Oncol. 2013;20(Suppl 3):S625–35.
    1. Zhu M, Chen Q, Liu X, Sun Q, Zhao X, Deng R. et al. lncRNA H19/miR-675 axis represses prostate cancer metastasis by targeting TGFBI. FEBS J. 2014;281:3766–75.
    1. Schmitz KJ, Helwig J, Bertram S, Sheu SY, Suttorp AC, Seggewiss J. et al. Differential expression of microRNA-675, microRNA-139-3p and microRNA-335 in benign and malignant adrenocortical tumours. J Clin Pathol. 2011;64:529–35.
    1. Matouk IJ, Mezan S, Mizrahi A, Ohana P, Abu-Lail R, Fellig Y. et al. The oncofetal H19 RNA connection: hypoxia, p53 and cancer. Biochim Biophys Acta. 2010;1803:443–51.
    1. Lo Dico A, Valtorta S, Martelli C, Belloli S, Gianelli U, Tosi D. et al. Validation of an engineered cell model for in vitro and in vivo HIF-1alpha evaluation by different imaging modalities. Mol Imaging Biol. 2014;16:210–23.
    1. Griffiths-Jones S, Grocock RJ, van Dongen S, Bateman A, Enright AJ. miRBase: microRNA sequences, targets and gene nomenclature. Nucleic Acids Res. 2006;34:D140–4.
    1. Zhang KL, Han L, Chen LY, Shi ZD, Yang M, Ren Y. et al. Blockage of a miR-21/EGFR regulatory feedback loop augments anti-EGFR therapy in glioblastomas. Cancer Lett. 2014;342:139–49.
    1. Galban S, Kuwano Y, Pullmann R Jr, Martindale JL, Kim HH, Lal A. et al. RNA-binding proteins HuR and PTB promote the translation of hypoxia-inducible factor 1alpha. Mol Cell Biol. 2008;28:93–107.
    1. Levy NS, Chung S, Furneaux H, Levy AP. Hypoxic stabilization of vascular endothelial growth factor mRNA by the RNA-binding protein HuR. J Biol Chem. 1998;273:6417–23.
    1. Hockel M, Vaupel P. Tumor hypoxia: definitions and current clinical, biologic, and molecular aspects. J Natl Cancer Inst. 2001;93:266–76.
    1. Parangi S, O'Reilly M, Christofori G, Holmgren L, Grosfeld J, Folkman J. et al. Antiangiogenic therapy of transgenic mice impairs de novo tumor growth. Proc Natl Acad Sci U S A. 1996;93:2002–7.
    1. Calvani M, Rapisarda A, Uranchimeg B, Shoemaker RH, Melillo G. Hypoxic induction of an HIF-1alpha-dependent bFGF autocrine loop drives angiogenesis in human endothelial cells. Blood. 2006;107:2705–12.
    1. Bruick RK, McKnight SL. Building better vasculature. Genes Dev. 2001;15:2497–502.
    1. Kaur B, Khwaja FW, Severson EA, Matheny SL, Brat DJ, Van Meir EG. Hypoxia and the hypoxia-inducible-factor pathway in glioma growth and angiogenesis. Neuro Oncol. 2005;7:134–53.
    1. Rankin EB, Giaccia AJ. The role of hypoxia-inducible factors in tumorigenesis. Cell Death Differ. 2008;15:678–85.
    1. Ayesh S, Matouk I, Schneider T, Ohana P, Laster M, Al-Sharef W. et al. Possible physiological role of H19 RNA. Mol Carcinog. 2002;35:63–74.
    1. Jiang X, Yan Y, Hu M, Chen X, Wang Y, Dai Y, Increased level of H19 long noncoding RNA promotes invasion, angiogenesis, and stemness of glioblastoma cells. J Neurosurg; 2015. pp. 1–8.
    1. Kulshreshtha R, Davuluri RV, Calin GA, Ivan M. A microRNA component of the hypoxic response. Cell Death Differ. 2008;15:667–71.
    1. Kulshreshtha R, Ferracin M, Wojcik SE, Garzon R, Alder H, Agosto-Perez FJ. et al. A microRNA signature of hypoxia. Mol Cell Biol. 2007;27:1859–67.
    1. He D, Wang J, Zhang C, Shan B, Deng X, Li B. et al. Down-regulation of miR-675-5p contributes to tumor progression and development by targeting pro-tumorigenic GPR55 in non-small cell lung cancer. Mol Cancer. 2015;14:73.
    1. Vennin C, Spruyt N, Dahmani F, Julien S, Bertucci F, Finetti P. et al. H19 non coding RNA-derived miR-675 enhances tumorigenesis and metastasis of breast cancer cells by downregulating c-Cbl and Cbl-b. Oncotarget. 2015;6:29209–23.
    1. Maxwell PH, Wiesener MS, Chang GW, Clifford SC, Vaux EC, Cockman ME. et al. The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature. 1999;399:271–5.
    1. Iliopoulos O, Levy AP, Jiang C, Kaelin WG Jr, Goldberg MA. Negative regulation of hypoxia-inducible genes by the von Hippel-Lindau protein. Proc Natl Acad Sci U S A. 1996;93:10595–9.
    1. Shen G, Li X, Jia YF, Piazza GA, Xi Y. Hypoxia-regulated microRNAs in human cancer. Acta Pharmacol Sin. 2013;34:336–41.
    1. Chang SH, Hla T. Gene regulation by RNA binding proteins and microRNAs in angiogenesis. Trends Mol Med. 2011;17:650–8.
    1. Ciafre SA, Galardi S. microRNAs and RNA-binding proteins: a complex network of interactions and reciprocal regulations in cancer. RNA Biol. 2013;10:935–42.
    1. Wu D, Yotnda P. Induction and testing of hypoxia in cell culture. J Vis Exp; 2011.
    1. Lo Dico A, Martelli C, Valtorta S, Raccagni I, Diceglie C, Belloli S. et al. Identification of imaging biomarkers for the assessment of tumour response to different treatments in a preclinical glioma model. Eur J Nucl Med Mol Imaging. 2015;42:1093–105.

Source: PubMed

3
Subscribe