Export of microRNAs and microRNA-protective protein by mammalian cells

Kai Wang, Shile Zhang, Jessica Weber, David Baxter, David J Galas, Kai Wang, Shile Zhang, Jessica Weber, David Baxter, David J Galas

Abstract

The discovery of microRNAs (miRNAs) as a new class of regulators of gene expression has triggered an explosion of research activities, but has left many unanswered questions about how this regulation functions and how it is integrated with other regulatory mechanisms. A number of miRNAs have been found to be present in plasma and other body fluids of humans and mice in surprisingly high concentrations. This observation was unexpected in two respects: first, the fact that these molecules are present at all outside the cell at significant concentrations and second, that these molecules appear to be stable outside of the cell. In light of this it has been suggested that the biological function of miRNAs may also extend outside of the cell and mediate cell-cell communication. We report here that after serum deprivation several human cell lines tested promptly export a substantial amount of miRNAs into the culture medium and the export process is largely energy dependent. The exported miRNAs are found both within and outside of the 16.5 and 120 K centrifugation pellets which contain most of the known cell-derived vesicles, the microvesicles and exosomes. We have identified some candidate proteins involved in this system, and one of these proteins may also play a role in protecting extracellular miRNAs from degradation. Our results point to a hitherto unrecognized and uncharacterized miRNA trafficking system in mammalian cells that is consistent with the cell-cell communication hypothesis.

Figures

Figure 1.
Figure 1.
Differential miRNA spectra between cultured cells and the medium. (a) Intra- and extracellular miRNA spectra for six different cell lines in culture as measured by miRNA arrays. The cell lines are as indicated (T98 is derived from human glioblastoma multiforma, BEAS2B is derived from lung bronchial epithelium and HPF are primary pulmonary fibroblast cells isolated from human lung tissue). Heat maps represent the levels of miRNA for the cell lines as indicated (red highest, green lowest). The brackets indicating ‘in’ or ‘out’ mark groups of miRNAs that show striking differences between the intra (‘in’) and extra (‘out’) cellular concentrations specifically for HepG2 cells. (b) qPCR measurements of selected miRNAs shown in 1a that illustrate different profiles for HepG2 (red) and A549 (blue). The histogram shows the external versus internal levels of selected miRNAs. The extracellular levels are indicated above, the intracellular below.
Figure 2.
Figure 2.
Time course of levels of miRNA exported into the medium. Samples were taken at various indicated time points after serum deprivation. The serum free medium contains no proteins or miRNAs. Most of the miRNA measured shown very similar behavior. The levels of 16 miRNAs measured by quantitative PCR from A549 (left frame) and HepG2 (right frame). The points are the means of three biological replicates. All vertical axes, the levels of miRNAs, are in ΔCt.
Figure 3.
Figure 3.
Decay of miRNAs in the medium. The cells in culture were serum deprived for 2 h, then the medium was removed and incubated under the same conditions for varying periods of time. Specific miRNA levels were measured (three biological replicates) with quantitative PCR on each sample. (a) The time course measurements of four selected miRNAs with standard derivations showing diverse profiles. (b) Model kinetics for the export response of the majority of the miRNAs considered here, showing the rate of export as a function of time (in red) and the accumulated level of extracellular miRNA as a function of time (in blue). (c) the intracellular response in HepG2 cells of three selected miRNAs.
Figure 4.
Figure 4.
miRNA levels in fractionated serum-free culture medium for A549 and HepG2. The medium, 2 h after serum deprivation, was fractionated by differential centrifugation into four fractions, microvesicles (size range: 100–1000 nm), exosomes (size range: 30–100 nm), one higher g spin pellet (220K for 1 h), and the remaining supernatant. The final supernant were concentrated to a final volume of 0.5 ml and 200 µl of the concentrated media were used for miRNA analysis. Pellets were also resuspended in 0.5 ml of PBS and 200 µl of the solution were used for miRNA isolation. (a) Experimental design for fractionation of medium. The levels of miRNA distribution from A549 (b) and HepG2 (c) were grouped into distinct distribution profiles.
Figure 4.
Figure 4.
miRNA levels in fractionated serum-free culture medium for A549 and HepG2. The medium, 2 h after serum deprivation, was fractionated by differential centrifugation into four fractions, microvesicles (size range: 100–1000 nm), exosomes (size range: 30–100 nm), one higher g spin pellet (220K for 1 h), and the remaining supernatant. The final supernant were concentrated to a final volume of 0.5 ml and 200 µl of the concentrated media were used for miRNA analysis. Pellets were also resuspended in 0.5 ml of PBS and 200 µl of the solution were used for miRNA isolation. (a) Experimental design for fractionation of medium. The levels of miRNA distribution from A549 (b) and HepG2 (c) were grouped into distinct distribution profiles.
Figure 4.
Figure 4.
miRNA levels in fractionated serum-free culture medium for A549 and HepG2. The medium, 2 h after serum deprivation, was fractionated by differential centrifugation into four fractions, microvesicles (size range: 100–1000 nm), exosomes (size range: 30–100 nm), one higher g spin pellet (220K for 1 h), and the remaining supernatant. The final supernant were concentrated to a final volume of 0.5 ml and 200 µl of the concentrated media were used for miRNA analysis. Pellets were also resuspended in 0.5 ml of PBS and 200 µl of the solution were used for miRNA isolation. (a) Experimental design for fractionation of medium. The levels of miRNA distribution from A549 (b) and HepG2 (c) were grouped into distinct distribution profiles.
Figure 5.
Figure 5.
Protein exported upon serum deprivation. (a) NPM1 protein is observed in western analysis of the concentrated medium from HepG2 cells. This protein is exported into the medium, but is not found in any fraction except the supernatant (as defined in legend to Figure 4) as shown by westerns on the fractions. (b) The relative concentrations of all miRNAs measured in this work were averaged for each fraction to obtain a relative miRNA levels in each fraction. These averages for HepG2 cells are shown in the histogram using a scale of log to base 2. (c) NPM1 protein protects miRNA from RNase degradation. Synthetic mir-122 RNA (100 pmol) was mixed with different proteins, NPM1 (black bars, 3 pmol), TGF-β (gray bars, 4 pmol) or BSA (open bars, 1.5 nmol) for 30 min followed by adding RNase A (7 nmol) for another 30 min of incubation at 37°C. The miRNA levels were determined by qPCR. Control experiments: omitting RNase A, protein, or miRNA were also included, as indicated on top of the graph. The scale is Ct of relative mir-122 levels.

References

    1. Lim LP, Glasner ME, Yekta S, Burge CB, Bartel DP. Vertebrate microRNA genes. Science. 2003;299:1540.
    1. Lau NC, Lim LP, Weinstein EG, Bartel DP. An abundant class of tiny RNAs with probable regulatory roles in Caenorhabditis elegans. Science. 2001;294:858–862.
    1. Kiss T. Small nucleolar RNAs: an abundant group of noncoding RNAs with diverse cellular functions. Cell. 2002;109:145–148.
    1. Omer AD, Janas MM, Novina CD. The chicken or the egg: microRNA-mediated regulation of mRNA translation or mRNA stability. Mol. Cell. 2009;35:739–740.
    1. Jayaswal V, Lutherborrow M, Ma DD, Yang HY. Identification of microRNAs with regulatory potential using a matched microRNA-mRNA time-course data. Nucleic Acids Res. 2009;37:e60.
    1. Bushati N, Cohen SM. microRNA functions. Ann. Rev. Cell Dev. Biol. 2007;23:175–205.
    1. Wang Y, Russell I, Chen C. MicroRNA and stem cell regulation. Curr. Opin Mol. Ther. 2009;11:292–298.
    1. Bhaskaran M, Wang Y, Zhang H, Weng T, Baviskar P, Guo Y, Gou D, Liu L. MicroRNA-127 modulates fetal lung development. Physiol. Genom. 2009;37:268–278.
    1. Christensen M, Schratt GM. microRNA involvement in developmental and functional aspects of the nervous system and in neurological diseases. Neurosci. Lett. 2009;466:55–62.
    1. Peng X, Li Y, Walters KA, Rosenzweig ER, Lederer SL, Aicher LD, Proll S, Katze MG. Computational identification of hepatitis C virus associated microRNA-mRNA regulatory modules in human livers. BMC Genomics. 2009;10:373.
    1. Godshalk SE, Bhaduri-McIntosh S, Slack FJ. Epstein-Barr virus-mediated dysregulation of human microRNA expression. Cell Cycle. 2008;7:3595–3600.
    1. Croce CM. Causes and consequences of microRNA dysregulation in cancer. Nat. Rev. Genet. 2009;10:704–714.
    1. Drakaki A, Iliopoulos D. MicroRNA gene networks in oncogenesis. Curr. Genomics. 2009;10:35–41.
    1. Israel A, Sharan R, Ruppin E, Galun E. Increased microRNA activity in human cancers. PloS One. 2009;4:e6045.
    1. Hanson EK, Lubenow H, Ballantyne J. Identification of forensically relevant body fluids using a panel of differentially expressed microRNAs. Anal. Biochem. 2009;387:303–314.
    1. Ai J, Zhang R, Li Y, Pu J, Lu Y, Jiao J, Li K, Yu B, Li Z, Wang R, et al. Circulating microRNA-1 as a potential novel biomarker for acute myocardial infarction. Biochem. Biophys. Res. Commun. 2009;391:73–77.
    1. Valadi H, Ekstrom K, Bossios A, Sjostrand M, Lee JJ, Lotvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 2007;9:654–659.
    1. Michael A, Bajracharya SD, Yuen PS, Zhou H, Star RA, Illei GG, Alevizos I. Exosomes from human saliva as a source of microRNA biomarkers. Oral Dis. 2009;16:34–38.
    1. Simpson RJ, Lim JW, Moritz RL, Mathivanan S. Exosomes: proteomic insights and diagnostic potential. Expert Rev. Proteomics. 2009;6:267–283.
    1. Hanke M, Hoefig K, Merz H, Feller AC, Kausch I, Jocham D, Warnecke JM, Sczakiel G. A robust methodology to study urine microRNA as tumor marker: microRNA-126 and microRNA-182 are related to urinary bladder cancer. Urol. Oncol. 2009 (in press)
    1. Wang K, Zhang S, Marzolf B, Troisch P, Brightman A, Hu Z, Hood LE, Galas DJ. Circulating microRNAs, potential biomarkers for drug-induced liver injury. Procd. Natl Acad. Sci. USA. 2009;106:4402–4407.
    1. Higuchi A, Shimmura S, Takeuchi T, Suematsu M, Tsubota K. Elucidation of apoptosis induced by serum deprivation in cultured conjunctival epithelial cells. Br. J. Ophthalmol. 2006;90:760–764.
    1. Yang MH, Yoo KH, Yook YJ, Park EY, Jeon JO, Choi SH, Park SY, Woo YM, Lee MJ, Park JH. The gene expression profiling in murine cortical cells undergoing programmed cell death (PCD) induced by serum deprivation. J. Biochem. Mol. Biol. 2007;40:277–285.
    1. Zhuge J, Cederbaum AI. Serum deprivation-induced HepG2 cell death is potentiated by CYP2E1. Free Rad. Biol. Med. 2006;40:63–74.
    1. Knowles BB, Howe CC, Aden DP. Human hepatocellular carcinoma cell lines secrete the major plasma proteins and hepatitis B surface antigen. Science. 1980;209:497–499.
    1. Giard DJ, Aaronson SA, Todaro GJ, Arnstein P, Kersey JH, Dosik H, Parks WP. In vitro cultivation of human tumors: establishment of cell lines derived from a series of solid tumors. J. Natl Cancer Inst. 1973;51:1417–1423.
    1. Kesimer M, Scull M, Brighton B, DeMaria G, Burns K, O'Neal W, Pickles RJ, Sheehan JK. Characterization of exosome-like vesicles released from human tracheobronchial ciliated epithelium: a possible role in innate defense. FASEB J. 2009;23:1858–1868.
    1. Zhou H, Cheruvanky A, Hu X, Matsumoto T, Hiramatsu N, Cho ME, Berger A, Leelahavanichkul A, Doi K, Chawla LS, et al. Urinary exosomal transcription factors, a new class of biomarkers for renal disease. Kidney Int. 2008;74:613–621.
    1. Tyler DD, Gonze J, Lamy F, Dumont JE. Influence of mitochondrial inhibitors on the respiration and energy-dependent uptake of iodide by thyroid slices. Biochem. J. 1968;106:123–133.
    1. Misumi Y, Misumi Y, Miki K, Takatsuki A, Tamura G, Ikehara Y. Novel blockade by brefeldin A of intracellular transport of secretory proteins in cultured rat hepatocytes. J. Biol. Chem. 1986;261:11398–11403.
    1. Gupta S, Knowlton AA. HSP60 trafficking in adult cardiac myocytes: role of the exosomal pathway. Am. J. Physiol. Heart Circ. Physiol. 2007;292:H3052–3056.
    1. Borer RA, Lehner CF, Eppenberger HM, Nigg EA. Major nucleolar proteins shuttle between nucleus and cytoplasm. Cell. 1989;56:379–390.
    1. Maggi LB, Jr, Kuchenruether M, Dadey DY, Schwope RM, Grisendi S, Townsend RR, Pandolfi PP, Weber JD. Nucleophosmin serves as a rate-limiting nuclear export chaperone for the Mammalian ribosome. Mol. Cell. Biol. 2008;28:7050–7065.
    1. Biggiogera M, Burki K, Kaufmann SH, Shaper JH, Gas N, Amalric F, Fakan S. Nucleolar distribution of proteins B23 and nucleolin in mouse preimplantation embryos as visualized by immunoelectron microscopy. Development. 1990;110:1263–1270.
    1. Leask A. Hijacking ZIP codes: posttanscriptional regulation of CCN2 by nucleophosmin. J. Cell Commun. Signal. 2009;3:85–86.
    1. Nawa Y, Kawahara K, Tancharoen S, Meng X, Sameshima H, Ito T, Masuda Y, Imaizumi H, Hashiguchi T, Maruyama I. Nucleophosmin may act as an alarmin: implications for severe sepsis. J. Leukocyte Biol. 2009;86:645–653.

Source: PubMed

3
Subskrybuj