Breast-cancer-secreted miR-122 reprograms glucose metabolism in premetastatic niche to promote metastasis

Miranda Y Fong, Weiying Zhou, Liang Liu, Aileen Y Alontaga, Manasa Chandra, Jonathan Ashby, Amy Chow, Sean Timothy Francis O'Connor, Shasha Li, Andrew R Chin, George Somlo, Melanie Palomares, Zhuo Li, Jacob R Tremblay, Akihiro Tsuyada, Guoqiang Sun, Michael A Reid, Xiwei Wu, Piotr Swiderski, Xiubao Ren, Yanhong Shi, Mei Kong, Wenwan Zhong, Yuan Chen, Shizhen Emily Wang, Miranda Y Fong, Weiying Zhou, Liang Liu, Aileen Y Alontaga, Manasa Chandra, Jonathan Ashby, Amy Chow, Sean Timothy Francis O'Connor, Shasha Li, Andrew R Chin, George Somlo, Melanie Palomares, Zhuo Li, Jacob R Tremblay, Akihiro Tsuyada, Guoqiang Sun, Michael A Reid, Xiwei Wu, Piotr Swiderski, Xiubao Ren, Yanhong Shi, Mei Kong, Wenwan Zhong, Yuan Chen, Shizhen Emily Wang

Abstract

Reprogrammed glucose metabolism as a result of increased glycolysis and glucose uptake is a hallmark of cancer. Here we show that cancer cells can suppress glucose uptake by non-tumour cells in the premetastatic niche, by secreting vesicles that carry high levels of the miR-122 microRNA. High miR-122 levels in the circulation have been associated with metastasis in breast cancer patients, and we show that cancer-cell-secreted miR-122 facilitates metastasis by increasing nutrient availability in the premetastatic niche. Mechanistically, cancer-cell-derived miR-122 suppresses glucose uptake by niche cells in vitro and in vivo by downregulating the glycolytic enzyme pyruvate kinase. In vivo inhibition of miR-122 restores glucose uptake in distant organs, including brain and lungs, and decreases the incidence of metastasis. These results demonstrate that, by modifying glucose utilization by recipient premetastatic niche cells, cancer-derived extracellular miR-122 is able to reprogram systemic energy metabolism to facilitate disease progression.

Conflict of interest statement

The authors declare no conflict of interest.

Figures

Figure 1
Figure 1
MiR-122 is highly secreted by cancer cells. RNA were extracted from the 110,000 ×g medium pellet (a) and PBS-washed cells (b) and analysed for miR-122 by RT-qPCR. Data was normalized to levels of total proteins (secreted; a) or U6 (cellular; b), and compared to the non-tumour line MCF10A (n = 6 extracts). (c) Representative EM images of vesicles in the 110,000 ×g medium pellet. Bar equals 100 nm. (d) Size distribution of vesicles identified in the 110,000 ×g medium pellets (n = 25 vesicles for MCF10A/vec; n = 38 for MCF10A/miR-122; n = 94 for MDA-MB-231). (e) Fractogram (UV absorption at 280 nm) for the AF4 eluates characterizing the MDA-MB-231 110,000 ×g medium pellet. (f) A representative EM image of MDA-MB-231-derived vesicles in the fraction eluted at 18–25 min. The measured diameter of vesicles was shown as mean ± SD (n = 41). Bar equals 100 nm. (g) RT-qPCR-determined levels of miR-122 and miR-16 in MDA-MB-231-derived protein and vesicle fractions separated by AF4 (n = 6 extracts). Absolute miRNA levels are calculated based on standard curves. ND: not detected. (h) After sucrose gradient centrifugation of MDA-MB-231-derived 110,000 ×g medium pellet, absolute miRNA levels in each gradient fraction was determined by RT-qPCR and calculated based on standard curves (n = 6 extracts). A representative EM image of MDA-MB-231-derived vesicles in sucrose fraction 5 (F5) is shown. Bar equals 100 nm. * p < 0.05 for all panels derived from Kruskal-Wallis test. Data are represented as mean ± SD in all panels except (c–e).
Figure 2
Figure 2
MiR-122 suppresses glucose metabolism by downregulating PKM. (a) BrdU uptake in indicated cells were analysed by flow cytometry (n = 6 biological replicates). (b) Quantification of intracellular metabolites by NMR spectroscopy (n = 3 biological replicates). (c) Glycogen staining (red) in MCF10A/vec and MCF10A/miR-122 cells. Bar equals 100 μm. (d) Change of metabolites in the media after 72 h of culture (n = 6 biological replicates). (e) The predicted miR-122 binding site in the 3′UTR of the human PKM and CS gene. The corresponding sequence in the mutated (mt) version is also shown. (f) The psiCHECK reporters containing 3′UTR of human PKM and CS gene with wild-type (wt) or mutated (mt) miR-122 binding site were used to transfect MCF10A cells stably expressing miR-122 or the empty vector (as control). Luciferase activity was analysed at 48 h post-transfection (n = 6 extracts). (g) Determination of PKM isoforms expressed in MCF10A and MDA-MB-231. RNA was subjected to RT-PCR followed by digestion with NcoI (N), PstI (P), or both enzymes (NP), plus an uncut control (U). Products were separated on an agarose gel with Sybr safe. The presence of a PstI digestion site indicates the splicing isoform M2 whereas the NcoI site indicates isoform M1. Size of markers (in bp) are indicated. (h) RT-qPCR analysis showing the relative expression of indicated genes in MCF10A/miR-122 and MCF10A/vec cells (n = 6 extracts). (i) Western blot analysis in MCF10A/miR-122 and MCF10A/vec cells with restored expression of PKM2 and CS. Size of markers (in kDa) are indicated. (j) RT-qPCR analysis in selected colonies with restored expression of PKM2 and CS (n = 6 extracts). (k) PKM activity (unit) in 5 μg proteins in indicated cells (n = 6 extracts). (l) Change of glucose in the media after 72 h of culture of selected clones normalized to cell number (n = 3 biological replicates). * p < 0.05, ** p < 0.01 for all panels derived from Kruskal-Wallis test. Data are represented as mean ± SD in all panels except (c, e, g, i). Uncropped images of blots and gels are shown in Supplementary Fig. 5.
Figure 3
Figure 3
Cancer-secreted miR-122 downregulates glucose uptake in lung fibroblasts. (a) Uptake of DiI-labelled exosome-containing EVs (prepared from the 110,000 ×g medium pellet) at 48 h. Bar equals 60 μm. (b) Levels of miRNAs in fibroblasts pre-treated with 10 μM DRB for 2 h followed by treatment with EVs for 16 h. miR-122 levels were normalized to U6 (n = 6 extracts). (c) Determination of PKM isoforms in fibroblasts and mouse tissues by RT-PCR as indicated in Fig. 2g. Size of markers (in bp) are indicated. (d–h) Fibroblasts were treated with two doses of EVs from indicated producer cells given 48 h apart and transfected with anti-miR-122 oligos or mismatch control oligos, before analysis at 96 h by (d) RT-qPCR (n = 6 extracts), (e) Western blot analysis (with marker size indicated in kDa), (f) PKM activity assay using 5 μg of proteins (n = 5 biological replicates), (g) 2-NBDG uptake (n = 6 biological replicates), and (h) change of glucose in the CM (n = 9 biological replicates). (i) Change of glucose in the CM of fibroblasts transfected with expression plasmids containing the ORF but not 3′UTR of PKM2 or GLUT1, or the empty vector, and treated with EVs from indicated producer cells (n = 5 biological replicates). (j) Western blot of fibroblasts transfected with expression plasmids for PKM2 or GLUT1. Size of markers (in kDa) are indicated. (k) 2-NBDG uptake in siRNA-transfected fibroblasts (n = 5 biological replicates). (l) CM was collected from siRNA-transfected fibroblasts cultured for 72 h and the glucose concentration measured. The CM was then fed to MDA-MB-231-HM cells before proliferation was assessed by BrdU-incorporation at 72 h (n = 6 biological replicates). (m) Circulating EVs were extracted from pooled healthy donor sera and from BC patients’ sera with low or high vesicular miR-122, used to treat fibroblasts and 2-NBDG uptake was measured (n = 8 biological replicates). * p < 0.05, ** p < 0.01 for all panels derived from Kruskal-Wallis test. Data are represented as mean ± SD in all panels except (a, c, e, j). Uncropped images of blots and gels are shown in Supplementary Fig. 5.
Figure 4
Figure 4
Cancer-secreted miR-122 downregulates glucose uptake in astrocytes. (a–b) EV uptake by astrocytes and neurons. Indicated cells were incubated with DiI-labelled EVs (red) for 48 h before fluorescent and phase contrast images were captured. Bar equals 60 μm. (c) Determination of PKM isoforms in primary mouse cells and mouse tissues by RT-PCR as indicated in Fig. 2g. Size of markers (in bp) are indicated. (d–h) Primary mouse astrocytes were treated with two doses of EVs from indicated producer cells given 48 h apart, before subjected to (d–e) RT-qPCR at 72 h for miR-122 (n = 6 extracts) (d) and PKM2 (n = 6 extracts) (e), (f) Western blot analysis for PKM2 and GLUT1 at 96 h (with marker size indicated in kDa), (g) PKM activity assay using 10 μg of proteins (n = 5 biological replicates), and (h) 2-NBDG uptake assay (n = 5 biological replicates). * p < 0.05, ** p < 0.01 for all panels derived from Kruskal-Wallis test. Data are represented as mean ± SD in (d–e & g–h). Uncropped images of blots and gels are shown in Supplementary Fig. 5.
Figure 5
Figure 5
Vesicular transfer of miR-122 alters glucose uptake in niche tissues. Indicated EVs were intravenously injected into the tail vein of NSG mice biweekly for 3.5 weeks. (a) Co-immunofluorescence of exosome marker CD63 (detected by a human-specific antibody; red) with astrocyte marker GFAP (white) or fibroblast marker FSP-1 (white) in brain and lung tissues of mice injected with 2-NBDG (green). Nuclei were counterstained with DAPI (blue). White bar represents 20 μm. (b) Quantification of 2-NBDG uptake in brain (n = 20 fields from 4 mice). (c) Quantification of 2-NBDG uptake in lung (n = 20 fields from 4 mice). (d–e) RT-qPCR in brain (d) and lungs (e) (n = 9 extracts from 3 mice). (f–g) Luciferase qPCR for the detection of metastases in the brain (f) and lungs (g) of mice pre-treated with EVs followed by an intracardiac injection with luciferase-labelled MDA-MB-231-HM tumour cells (n = 15 extracts from 5 mice). (h) MiR-122 levels determined by RT-qPCR in the serum of non-tumour bearing mice (group 1), mice bearing MCFDCIS or MDA-MB-231-HM tumours (groups 2–3), and non-tumour-bearing mice treated with EVs from MCF10A/vec, MCF10A/miR-122, or MDA-MB-231 cells (groups 4–6) (n = 15 extracts from 5 mice). Data was normalized to the levels of miR-16. Data are represented as mean ± SD for all panels except (a). * p < 0.05, ** p < 0.01, *** p < 0.001 for all panels derived from Kruskal-Wallis test.
Figure 6
Figure 6
In vivo effect of miR-122 on primary tumour growth and metastasis. (a) Tumour growth curve in mice carrying MCFDCIS/vec and MCFDCIS/miR-122 orthotopic xenografts (n = 7 mice). (b) BLI imaging at week 3. (c) Luciferase quantification of (b) (n = 7 mice). (d) IHC for Ki67, PKM1/2, and GLUT1 in tumour, brain, and lung sections. Scale bar is 100 μm. (e) Quantification of Ki67+ tumour cells from 3 fields per tumour (n = 6 mice per group). (f) Intratumoural levels of ATP were assessed in tumour lysates by ENLIGHTEN ATP assay (n = 6 mice per group). (g) miR-122 levels in the brain and lungs determined by RT-qPCR (n = 18 extracts from 6 mice). (h) 2-NBDG uptake quantification in the tumour, brain, and lungs (n = 12 fields from 4 mice per group). (i–j) Luciferase qPCR in the brain (i, n = 24 extracts from 8 mice per group) and lungs (j, n = 18 extracts from 6 mice per group) of MCFDCIS tumour-bearing mice. (k) Tumour growth curve in mice carrying orthotopic xenografts of MDA-MB-231 with stable knockdown of miR-122 (MDA-MB-231/122KD) and also receiving EV treatments as indicated (n = 7 mice per group). No significant difference (p > 0.05) between groups based on Kruskal-Wallis test. (l–m) Luciferase qPCR for the detection of metastases in the brain (l) and lungs (m) of mice bearing MDA-MB-231/122KD tumours and treated with indicated EVs (n = 12 extracts from 4 mice per group). * p < 0.05, ** p < 0.01, *** p < 0.001 for all panels derived from Kruskal-Wallis test. Data are represented as mean ± SD in all panels except (b & d).
Figure 7
Figure 7
MiR-122 intervention alleviates cancer-induced glucose reallocation in vivo and reduces metastasis. Luciferase-labelled MDA-MB-231-HM cells were injected into the No. 4 mammary fat pad of NSG mice. Mice were divided into 3 groups (n = 8 mice per group) for treatment with PBS, anti-miR-122, or mismatch control oligos. (a) Tumour growth curve (n = 8 mice). No significant difference (p > 0.05) between groups based on Kruskal-Wallis test. (b) 2-NBDG uptake in the tumour and tumour-adjacent stroma (n = 20 fields from 4 mice per group). (c) Primary tumour sections were analysed by IHC (for PKM2 and GLUT1) and ISH (for miR-122). For 2-NBDG (green) uptake, sections were counterstained with DAPI (blue) to show nuclei. Dotted line delineates tumour (T) from stroma (S). White bar represents 100 μm. (d) BLI at week 5 indicating extensive brain and lung metastases in PBS and mismatch groups and reduced incidence of metastasis in anti-miR-122 group. (e) Quantification of BLI at week 5 (n = 8 mice per group). (f) Representative images of 2-NBDG uptake fluorescence. White bar represents 60 μm. (g) Quantification of 2-NBDG uptake in the brain and lungs of tumour-free (normal) NSG mice and tumour bearing mice that were untreated when sacrificed at week 3 after tumour cell implantation or treated as indicated and sacrificed at week 6 (n = 20 fields from 4 mice per group). (h–i) RT-qPCR in brain (h) and lungs (i) of tumour-free and tumour bearing mice (n = 12 extracts from 4 mice per group). (j) GLUT1 IHC in brain and lungs. Scale bar is 60 μm. Data are represented as mean ± SD in all panels except (c–d, f, & j). * p < 0.05, ** p < 0.01, *** p < 0.001 for all panels derived from Kruskal-Wallis test.

References

    1. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–674.
    1. Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science. 2009;324:1029–1033.
    1. Zhao FQ, Keating AF. Functional properties and genomics of glucose transporters. Curr Genomics. 2007;8:113–128.
    1. DeBerardinis RJ, Lum JJ, Hatzivassiliou G, Thompson CB. The biology of cancer: metabolic reprogramming fuels cell growth and proliferation. Cell Metab. 2008;7:11–20.
    1. Barthel A, et al. Regulation of GLUT1 gene transcription by the serine/threonine kinase Akt1. J Biol Chem. 1999;274:20281–20286.
    1. O’Rourke JF, Pugh CW, Bartlett SM, Ratcliffe PJ. Identification of hypoxically inducible mRNAs in HeLa cells using differential-display PCR. Role of hypoxia-inducible factor-1. Eur J Biochem. 1996;241:403–410.
    1. Semenza GL, Roth PH, Fang HM, Wang GL. Transcriptional regulation of genes encoding glycolytic enzymes by hypoxia-inducible factor 1. J Biol Chem. 1994;269:23757–23763.
    1. Spoden GA, et al. The SUMO-E3 ligase PIAS3 targets pyruvate kinase M2. J Cell Biochem. 2009;107:293–302.
    1. Yang W, et al. ERK1/2-dependent phosphorylation and nuclear translocation of PKM2 promotes the Warburg effect. Nat Cell Biol. 2012;14:1295–1304.
    1. Gao X, Wang H, Yang JJ, Liu X, Liu ZR. Pyruvate kinase M2 regulates gene transcription by acting as a protein kinase. Mol Cell. 2012;45:598–609.
    1. Yang W, et al. Nuclear PKM2 regulates beta-catenin transactivation upon EGFR activation. Nature. 2011;480:118–122.
    1. Lee J, Kim HK, Han YM, Kim J. Pyruvate kinase isozyme type M2 (PKM2) interacts and cooperates with Oct-4 in regulating transcription. Int J Biochem Cell Biol. 2008;40:1043–1054.
    1. Luo W, et al. Pyruvate kinase M2 is a PHD3-stimulated coactivator for hypoxia-inducible factor 1. Cell. 2011;145:732–744.
    1. Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell. 2009;136:215–233.
    1. Calin GA, Croce CM. MicroRNA signatures in human cancers. Nat Rev Cancer. 2006;6:857–866.
    1. Mitchell PS, et al. Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci U S A. 2008;105:10513–10518.
    1. Taylor DD, Gercel-Taylor C. MicroRNA signatures of tumor-derived exosomes as diagnostic biomarkers of ovarian cancer. Gynecol Oncol. 2008;110:13–21.
    1. Wu X, et al. De novo sequencing of circulating miRNAs identifies novel markers predicting clinical outcome of locally advanced breast cancer. J Transl Med. 2012;10:42.
    1. Zhu W, Qin W, Atasoy U, Sauter ER. Circulating microRNAs in breast cancer and healthy subjects. BMC Res Notes. 2009;2:89.
    1. Zen K, Zhang CY. Circulating microRNAs: a novel class of biomarkers to diagnose and monitor human cancers. Med Res Rev. 2012;32:326–348.
    1. Redis RS, Calin S, Yang Y, You MJ, Calin GA. Cell-to-cell miRNA transfer: from body homeostasis to therapy. Pharmacol Ther. 2012;136:169–174.
    1. Valadi H, et al. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007;9:654–659.
    1. Wang K, Zhang S, Weber J, Baxter D, Galas DJ. Export of microRNAs and microRNA-protective protein by mammalian cells. Nucleic Acids Res. 2010;38:7248–7259.
    1. Vickers KC, Remaley AT. Lipid-based carriers of microRNAs and intercellular communication. Curr Opin Lipidol. 2012;23:91–97.
    1. Skog J, et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol. 2008;10:1470–1476.
    1. Peinado H, et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med. 2012;18:883–891.
    1. Zhou W, et al. Cancer-Secreted miR-105 Destroys Vascular Endothelial Barriers to Promote Metastasis. Cancer Cell. 2014;25:501–515.
    1. Rottiers V, Naar AM. MicroRNAs in metabolism and metabolic disorders. Nat Rev Mol Cell Biol. 2012;13:239–250.
    1. Moore KJ, Rayner KJ, Suarez Y, Fernandez-Hernando C. The role of microRNAs in cholesterol efflux and hepatic lipid metabolism. Annu Rev Nutr. 2011;31:49–63.
    1. Boutz DR, et al. Two-tiered approach identifies a network of cancer and liver disease-related genes regulated by miR-122. J Biol Chem. 2011;286:18066–18078.
    1. Palma J, et al. MicroRNAs are exported from malignant cells in customized particles. Nucleic Acids Res. 2012;40:9125–9138.
    1. Ashby J, et al. Distribution Profiling of Circulating MicroRNAs in Serum. Anal Chem. 2014;86:9343–9349.
    1. Lloyd PG, Hardin CD, Sturek M. Examining glucose transport in single vascular smooth muscle cells with a fluorescent glucose analog. Physiol Res. 1999;48:401–410.
    1. Loaiza A, Porras OH, Barros LF. Glutamate triggers rapid glucose transport stimulation in astrocytes as evidenced by real-time confocal microscopy. J Neurosci. 2003;23:7337–7342.
    1. Yamada K, et al. Measurement of glucose uptake and intracellular calcium concentration in single, living pancreatic beta-cells. J Biol Chem. 2000;275:22278–22283.
    1. Itoh Y, Abe T, Takaoka R, Tanahashi N. Fluorometric determination of glucose utilization in neurons in vitro and in vivo. J Cereb Blood Flow Metab. 2004;24:993–1003.
    1. Chuquet J, Quilichini P, Nimchinsky EA, Buzsaki G. Predominant enhancement of glucose uptake in astrocytes versus neurons during activation of the somatosensory cortex. J Neurosci. 2010;30:15298–15303.
    1. Bak LK, Schousboe A, Sonnewald U, Waagepetersen HS. Glucose is necessary to maintain neurotransmitter homeostasis during synaptic activity in cultured glutamatergic neurons. J Cereb Blood Flow Metab. 2006;26:1285–1297.
    1. Hu M, et al. Regulation of in situ to invasive breast carcinoma transition. Cancer Cell. 2008;13:394–406.
    1. Vickers KC, Palmisano BT, Shoucri BM, Shamburek RD, Remaley AT. MicroRNAs are transported in plasma and delivered to recipient cells by high-density lipoproteins. Nat Cell Biol. 2011;13:423–433.
    1. Arroyo JD, et al. Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma. Proc Natl Acad Sci U S A. 2011;108:5003–5008.
    1. Pavlides S, et al. The reverse Warburg effect: aerobic glycolysis in cancer associated fibroblasts and the tumor stroma. Cell Cycle. 2009;8:3984–4001.
    1. Martinez-Outschoorn UE, Lisanti MP, Sotgia F. Catabolic cancer-associated fibroblasts transfer energy and biomass to anabolic cancer cells, fueling tumor growth. Semin Cancer Biol. 2014;25:47–60.
    1. Kaplan RN, Psaila B, Lyden D. Bone marrow cells in the ‘pre-metastatic niche’: within bone and beyond. Cancer Metastasis Rev. 2006;25:521–529.
    1. Kaplan RN, et al. VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature. 2005;438:820–827.
    1. Peinado H, Lavotshkin S, Lyden D. The secreted factors responsible for pre-metastatic niche formation: old sayings and new thoughts. Seminars in cancer biology. 2011;21:139–146.
    1. Sethi N, Kang Y. Unravelling the complexity of metastasis - molecular understanding and targeted therapies. Nat Rev Cancer. 2011;11:735–748.
    1. Hood JL, San RS, Wickline SA. Exosomes released by melanoma cells prepare sentinel lymph nodes for tumor metastasis. Cancer Res. 2011;71:3792–3801.
    1. Grange C, et al. Microvesicles released from human renal cancer stem cells stimulate angiogenesis and formation of lung premetastatic niche. Cancer Res. 2011;71:5346–5356.
    1. Erler JT, et al. Hypoxia-induced lysyl oxidase is a critical mediator of bone marrow cell recruitment to form the premetastatic niche. Cancer Cell. 2009;15:35–44.
    1. Hiratsuka S, et al. Primary tumours modulate innate immune signalling to create pre-metastatic vascular hyperpermeability foci. Nature communications. 2013;4:1853.
    1. Hiratsuka S, et al. Endothelial focal adhesion kinase mediates cancer cell homing to discrete regions of the lungs via E-selectin up-regulation. Proc Natl Acad Sci U S A. 2011;108:3725–3730.
    1. Hiratsuka S, Watanabe A, Aburatani H, Maru Y. Tumour-mediated upregulation of chemoattractants and recruitment of myeloid cells predetermines lung metastasis. Nat Cell Biol. 2006;8:1369–1375.
    1. Hiratsuka S, et al. The S100A8-serum amyloid A3-TLR4 paracrine cascade establishes a pre-metastatic phase. Nat Cell Biol. 2008;10:1349–1355.
    1. Bos R, et al. Biologic correlates of (18)fluorodeoxyglucose uptake in human breast cancer measured by positron emission tomography. J Clin Oncol. 2002;20:379–387.
    1. Kang SS, et al. Clinical significance of glucose transporter 1 (GLUT1) expression in human breast carcinoma. Jpn J Cancer Res. 2002;93:1123–1128.
    1. Elmen J, et al. Antagonism of microRNA-122 in mice by systemically administered LNA-antimiR leads to up-regulation of a large set of predicted target mRNAs in the liver. Nucleic Acids Res. 2008;36:1153–1162.
    1. Elmen J, et al. LNA-mediated microRNA silencing in non-human primates. Nature. 2008;452:896–899.
    1. Rai N. News. Santaris Pharma A/S; Hoersholm, Denmark; San Diego, CA: 2011.
    1. Takanaga H, Frommer WB. Facilitative plasma membrane transporters function during ER transit. FASEB J. 2010;24:2849–2858.
    1. Wang Y, et al. Transforming growth factor-beta regulates the sphere-initiating stem cell-like feature in breast cancer through miRNA-181 and ATM. Oncogene. 2011;30:1470–1480.
    1. Thery C, Amigorena S, Raposo G, Clayton A. Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Curr Protoc Cell Biol. 2006;Chapter 3(Unit 3):22.
    1. Tsuyada A, et al. CCL2 mediates cross-talk between cancer cells and stromal fibroblasts that regulates breast cancer stem cells. Cancer Res. 2012;72:2768–2779.
    1. Yu Y, et al. Context-dependent bidirectional regulation of the MutS homolog 2 by transforming growth factor beta contributes to chemoresistance in breast cancer cells. Mol Cancer Res. 2010;8:1633–1642.
    1. Clower CV, et al. The alternative splicing repressors hnRNP A1/A2 and PTB influence pyruvate kinase isoform expression and cell metabolism. Proc Natl Acad Sci U S A. 2010;107:1894–1899.
    1. Edwards RJ, Watts DC. Specific spectrophotometric assay for the M isoenzymes of pyruvate kinase in plasma samples containing mixtures of the muscle (M) and liver (L) isoenzymes. Clin Chem. 1981;27:906–909.
    1. Cano KE, Li YJ, Chen Y. NMR metabolomic profiling reveals new roles of SUMOylation in DNA damage response. J Proteome Res. 2010;9:5382–5388.
    1. Chida J, Yamane K, Takei T, Kido H. An efficient extraction method for quantitation of adenosine triphosphate in mammalian tissues and cells. Analytica chimica acta. 2012;727:8–12.
    1. Fong MY, et al. Withaferin A Synergizes the Therapeutic Effect of Doxorubicin through ROS-Mediated Autophagy in Ovarian Cancer. PloS one. 2012;7:e42265.
    1. Jorgensen S, Baker A, Moller S, Nielsen BS. Robust one-day in situ hybridization protocol for detection of microRNAs in paraffin samples using LNA probes. Methods. 2010;52:375–381.

Source: PubMed

3
Abonneren