Circular non-coding RNA ANRIL modulates ribosomal RNA maturation and atherosclerosis in humans

Lesca M Holdt, Anika Stahringer, Kristina Sass, Garwin Pichler, Nils A Kulak, Wolfgang Wilfert, Alexander Kohlmaier, Andreas Herbst, Bernd H Northoff, Alexandros Nicolaou, Gabor Gäbel, Frank Beutner, Markus Scholz, Joachim Thiery, Kiran Musunuru, Knut Krohn, Matthias Mann, Daniel Teupser, Lesca M Holdt, Anika Stahringer, Kristina Sass, Garwin Pichler, Nils A Kulak, Wolfgang Wilfert, Alexander Kohlmaier, Andreas Herbst, Bernd H Northoff, Alexandros Nicolaou, Gabor Gäbel, Frank Beutner, Markus Scholz, Joachim Thiery, Kiran Musunuru, Knut Krohn, Matthias Mann, Daniel Teupser

Abstract

Circular RNAs (circRNAs) are broadly expressed in eukaryotic cells, but their molecular mechanism in human disease remains obscure. Here we show that circular antisense non-coding RNA in the INK4 locus (circANRIL), which is transcribed at a locus of atherosclerotic cardiovascular disease on chromosome 9p21, confers atheroprotection by controlling ribosomal RNA (rRNA) maturation and modulating pathways of atherogenesis. CircANRIL binds to pescadillo homologue 1 (PES1), an essential 60S-preribosomal assembly factor, thereby impairing exonuclease-mediated pre-rRNA processing and ribosome biogenesis in vascular smooth muscle cells and macrophages. As a consequence, circANRIL induces nucleolar stress and p53 activation, resulting in the induction of apoptosis and inhibition of proliferation, which are key cell functions in atherosclerosis. Collectively, these findings identify circANRIL as a prototype of a circRNA regulating ribosome biogenesis and conferring atheroprotection, thereby showing that circularization of long non-coding RNAs may alter RNA function and protect from human disease.

Figures

Figure 1. CircANRIL expression in human vascular…
Figure 1. CircANRIL expression in human vascular tissue and association with atheroprotection at 9p21.
(a) Schematic of circANRIL at 9p21 and qPCR assays for isoform specific quantification. CircANRIL contains exons 5, 6 and 7. Exon 7 is non-canonically spliced to exon 5. (b) qPCR analysis of circANRIL expression in human tissues, primary cells and cell lines. Beta actin (ACTB), house-keeping gene; FB, adventitial fibroblasts; HEK, HEK-293 cell line; THP1 and MonoMac, human monocytic cell line. Analyses were done in RNA pools of at least three donors. (c) In situ hybridization of circANRIL in human atherosclerotic plaque and co-localization with smooth muscle actin (SMA)-positive cells and macrophages (CD68). Sense control (CTR)-negative control. Representative staining out of three biological replicates. (d) Association of circANRIL with 9p21 haplotypes in PBMC from CAD patients. Protective (A, n=498), heterozygote (H, n=979) and risk (G, n=456) haplotypes. (e) Association of circANRIL with 9p21 haplotypes A (n=49), H (n=114) and G (n=55) in endarterectomy specimens. (f) Association of circ/linANRIL ratio in PBMC with severity of CAD (No, n=745; <50% stenosis, n=392; ≥50% stenosis, n=747). Data are given as mean±s.e.m., and associations were calculated using robust linear regression models.
Figure 2. Atherosclerosis-related cell functions in c…
Figure 2. Atherosclerosis-related cell functions in circANRIL-overexpressing cells.
(a) Schematic of the vector construct for circANRIL stable and transient overexpression. RNA polymerase II (RNAP II). (b) ANRIL isoform expression in HEK-293 cells that stably express circANRIL, linANRIL or empty vector (three biological replicates each). PCR of reverse-transcribed RNA, glyceraldehyde 3-phosphate dehydrogenase (GAPDH), house-keeping gene; OdT, oligo(d)T primers; RH, random hexamer primers. (c) Apoptosis and (d) proliferation in HEK-293 cell line stably overexpressing circANRIL (pool of 3 biological replicates, and 8 and 12 technical replicates per condition, respectively), compared with overexpression of an unrelated circular RNA (circHPRT1). Apoptosis (e) and proliferation (f) after RNAi against circANRIL or non-circANRIL or scrambled (SCR) siRNA control. Quadruplicate measurements per condition. *P<0.05; **P<0.01; ***P<0.001. Comparison of multiple groups was done using analysis of variance, and Tukey was performed as post hoc test in c,e. Two-tailed unpaired Student's t-test in d,f. Data are given as mean±s.e.m.
Figure 3. Role of endogenous circANRIL on…
Figure 3. Role of endogenous circANRIL on apoptosis and cell proliferation.
(a) Schematic of CRISPR/Cas9-mediated knockout of ANRIL in HEK-293 cells. Genomic regions encompassing exons (Ex) 5–20 were deleted. Arrowheads indicate location and orientation of primers used for genotyping. (b) PCR genotyping. Sequencing of PCR products from Ex5–20 assay validated successful deletion in mutant cell lines (Supplementary Table 2). (c) Apoptosis and (d) proliferation in heterozygous and homozygous ANRIL knockout or control cell lines. As control, Cas9 was overexpressed without guide RNAs (gRNAs). (e) Apoptosis and (f) proliferation in homozygous knockout cells following transient re-expression of circANRIL. *P<0.05; **P<0.01; ***P<0.001. (d) Heterozygous (***P<0.001) or homozygous (#P<0.05) ANRIL knockout versus control cell lines, respectively. Comparison of multiple groups was done using analysis of variance, and Tukey was performed as post hoc test in c. Two-tailed unpaired Student's t-tests were applied in df. Data are given as mean±s.e.m.
Figure 4. CircANRIL does not regulate 9p21…
Figure 4. CircANRIL does not regulate 9p21 protein-coding genes and lacks miRNA sponging activity.
(a) Overexpression of circANRIL in HEK-293 cells does not modulate endogenous linANRIL RNA abundance as measured by qPCR (left panel). Expression of p16INK4a and p14ARF, encoded by CDKN2A, and p15INK4b, encoded by CDKN2B, in circANRIL-overexpressing cells (right panel) (pool of three biological replicates, quadruplicate qPCR measurements). (b) Prediction of miRNA-binding sites in circANRIL using miRanda and TargetSpy algorithms. (c) miRNA expression normalized to miR-27b as measured by qPCR (four biological replicates, quadruplicate measurements). (d) RNA immunoprecipitation (RIP) of AGO2. Analysis of precipitated RNAs: CDR1as, MALAT1, positive controls; U1, negative control (RIP was performed in a pool of three biological replicates, quadruplicate qPCR measurements). ***P<0.001. NS, not significantly different. (analysis of variance, and Tukey as post hoc test). Data are given as mean±s.e.m.
Figure 5. Identification of circANRIL- binding proteins.
Figure 5. Identification of circANRIL-binding proteins.
(a) Schematic of λN-peptide-mediated capture of circANRIL-BoxB from cellular lysates of stably overexpressing HEK-293 cell lines, and label-free mass spectrometric quantification. Experiments were performed in a pool of three biological replicates (quadruplicate measurements). (b) Quantification of RNAs by qPCR after λN-peptide capture (quadruplicate measurements). (c) Summary of circANRIL-BoxB-bound proteins according to annotated (KEGG, GO) and published functions. (d) Volcano plot of label-free quantification (LFQ) of circANRIL-BoxB-bound proteins compared with circANRIL input control. (e) RIP of PES1, NOLC1, NOP14 and mouse IgG (mIgG) control (RIP was performed in a pool of three biological replicates, quadruplicate measurements). (f) Western blot of PES1 after λN-peptide-mediated circANRIL-BoxB capture using nuclear extracts from indicated cell lines.
Figure 6. rRNA maturation defects and nucleolar…
Figure 6. rRNA maturation defects and nucleolar stress in circANRIL-overexpressing cells.
(a) Relative quantification of pre-rRNA in circANRIL- or in circHPRT1-overexpressing HEK-293 cells using isoform-specific qPCRs (RNA from a pool of three biological replicates, quadruplicate measurements). (b) Pre-rRNA levels after RNAi against circANRIL or using scrambled siRNA control (quadruplicate measurements per condition). (c) Immunofluorescent staining of PES1 in circANRIL- or in circHPRT1-overexpressing HEK-293 or empty vector control cells. Quantification of (d) nucleoli and (e) nucleolar size in circANRIL- or circHPRT1-overexpressing or empty vector control cells (*** P<0.001). Data are given as mean±s.e.m. (analysis of variance, and Tukey as post hoc test). (f) Immunofluorescent staining of p53 (red) in circANRIL- or circHPRT1-overexpressing or empty vector control cells. (g) Schematic of transcriptome and proteome analyses in circANRIL-expressing HEK-293 cells and in control cells by genome-wide expression arrays and by pulseSILAC, respectively. For results of pathway analyses and procedure, see Supplementary Tables 4–6 and Methods section, respectively. L/M/H, light/medium/heavy medium.
Figure 7. Molecular mechanism of circANRIL controlling…
Figure 7. Molecular mechanism of circANRIL controlling PES1 function.
(a) Homology of circANRIL and precursor 47S rRNA (blue boxes) determined by BLASTn algorithm. (b) Prediction of RNA–protein interaction of circANRIL with PES1 using the catRAPID algorithm. Homology regions (HR1–HR5)—predicted RNA–protein interaction motifs in circANRIL. (c) Secondary structure prediction and HR1–5 of circANRIL using the Vienna RNA package. (d) Schematic of PES1 with functional protein domains. Wild-type PES1 (PES1-WT) and two mutants lacking the 5′ (PES1Δ1–54) or the 3′ (PES1Δ446–588) lysine-rich NLSs. (e) Immunoprecipitation (IP) of PES1 isoforms from HEK-293 cells and quantification of RNA by qPCR (IP was performed in a pool of three biological replicates, quadruplicate measurements). (f) Pre-rRNA binding to PES1-WT and PES1Δ446–588 in circANRIL-overexpressing and control cells (IP was performed in a pool of three biological replicates, quadruplicate measurements). (g) Pre-rRNA and 7SL control RNA in circANRIL-expressing HEK-293 cells after transient PES1-WT or PES1Δ446–588 overexpression (pool of three biological replicates, quadruplicate measurements). (h) p53 western blot, (i), apoptosis and (j) cell proliferation in circANRIL-overexpressing HEK-293 cells with transient overexpression of PES1-WT or of PES1Δ446–588. Quadruplicate measurements per condition. *P<0.05; **P<0.01; ***P<0.001. NS, not significantly different. Analysis of variance, and Tukey as post hoc test in e,g and i. Two-tailed unpaired Student's t-tests were applied in f,j. Data are given mean±s.e.m.
Figure 8. Translation of circANRIL molecular mechanisms…
Figure 8. Translation of circANRIL molecular mechanisms in human primary cells and human cohorts.
(a) CircANRIL overexpression in primary human SMC using the green fluorescent protein (GFP)-encoding, bicistronic pBI-CMV2 vector (green; Fig. 2a) and immunofluorescent staining of PES1 (red). Quantification of nucleoli (b) and nucleolar size (c) in circANRIL-overexpressing (GFP+) SMC and circANRIL-negative (GFP−) cells. AU, arbitrary units. (d) Quantification of pre-rRNAs following circANRIL overexpression in SMC. (e) Apoptosis and (f) proliferation in SMC transfected with circANRIL. Quadruplicate measurements per condition. Correlation of endogenous circANRIL expression in human SMC with (g) apoptosis and (h) proliferation (n=5; quadruplicate measurements each). (i) Quantification of pre-rRNA following circANRIL overexpression in iPSC-derived macrophages (Supplementary Fig. 16). (j) Apoptosis in iPSC-derived macrophages transfected with circANRIL. LI, luminescence intensity. Quadruplicate measurements per condition. (k) Association of 32S rRNA expression with 9p21 A (n=49), H (n=114) and G (n=55) haplotypes in endarterectomy specimens. ***P<0.001. Two-tailed unpaired Student's t-tests were applied in b,e,f and j, and Mann–Whitney U-testing in k. Data are given mean±s.e.m.

References

    1. Jeck W. R. et al. Circular RNAs are abundant, conserved, and associated with ALU repeats. RNA 19, 141–157 (2013).
    1. Memczak S. et al. Circular RNAs are a large class of animal RNAs with regulatory potency. Nature 495, 333–338 (2013).
    1. Salzman J., Gawad C., Wang P. L., Lacayo N. & Brown P. O. Circular RNAs are the predominant transcript isoform from hundreds of human genes in diverse cell types. PLoS ONE 7, e30733 (2012).
    1. Nigro J. M. et al. Scrambled exons. Cell 64, 607–613 (1991).
    1. Dubin R. A., Kazmi M. A. & Ostrer H. Inverted repeats are necessary for circularization of the mouse testis Sry transcript. Gene 167, 245–248 (1995).
    1. Zhang X. O. et al. Complementary sequence-mediated exon circularization. Cell 159, 134–147 (2014).
    1. Capel B. et al. Circular transcripts of the testis-determining gene Sry in adult mouse testis. Cell 73, 1019–1030 (1993).
    1. Jeske Y. W., Bowles J., Greenfield A. & Koopman P. Expression of a linear Sry transcript in the mouse genital ridge. Nat. Genet. 10, 480–482 (1995).
    1. Hansen T. B. et al. Natural RNA circles function as efficient microRNA sponges. Nature 495, 384–388 (2013).
    1. Guo J. U., Agarwal V., Guo H. & Bartel D. P. Expanded identification and characterization of mammalian circular RNAs. Genome Biol. 15, 409 (2014).
    1. Holdt L. M. & Teupser D. From genotype to phenotype in human atherosclerosis—recent findings. Curr. Opin. Lipidol. 24, 410–418 (2013).
    1. Samani N. J. et al. Genome-wide association analysis of coronary artery disease. N. Engl. J. Med. 357, 443–453 (2007).
    1. Schunkert H. et al. Large-scale association analysis identifies 13 new susceptibility loci for coronary artery disease. Nat. Genet. 43, 333–338 (2011).
    1. Burd C. E. et al. Expression of linear and novel circular forms of an INK4/ARF-associated non-coding RNA correlates with atherosclerosis risk. PLoS Genet. 6, e1001233 (2010).
    1. Holdt L. M. & Teupser D. Recent studies of the human chromosome 9p21 locus, which is associated with atherosclerosis in human populations. Arterioscler. Thromb. Vasc. Biol. 32, 196–206 (2012).
    1. Holdt L. M. et al. ANRIL expression is associated with atherosclerosis risk at chromosome 9p21. Arterioscler. Thromb. Vasc. Biol. 30, 620–627 (2010).
    1. Holdt L. M. et al. Alu elements in ANRIL non-coding RNA at chromosome 9p21 modulate atherogenic cell functions through trans-regulation of gene networks. PLoS Genet. 9, e1003588 (2013).
    1. He S., Gu W., Li Y. & Zhu H. ANRIL/CDKN2B-AS shows two-stage clade-specific evolution and becomes conserved after transposon insertions in simians. BMC Evol. Biol. 13, 247 (2013).
    1. Wang Y. & Wang Z. Efficient backsplicing produces translatable circular mRNAs. RNA 21, 172–179 (2015).
    1. Beutner F. et al. Rationale and design of the Leipzig (LIFE) Heart Study: phenotyping and cardiovascular characteristics of patients with coronary artery disease. PLoS ONE 6, e29070 (2011).
    1. Kotake Y. et al. Long non-coding RNA ANRIL is required for the PRC2 recruitment to and silencing of p15(INK4B) tumor suppressor gene. Oncogene 30, 1956–1962 (2011).
    1. Yap K. L. et al. Molecular interplay of the noncoding RNA ANRIL and methylated histone H3 lysine 27 by polycomb CBX7 in transcriptional silencing of INK4a. Mol. Cell 38, 662–674 (2010).
    1. De Gregorio E., Preiss T. & Hentze M. W. Translation driven by an eIF4G core domain in vivo. EMBO J. 18, 4865–4874 (1999).
    1. Milkereit P. et al. A Noc complex specifically involved in the formation and nuclear export of ribosomal 40 S subunits. J. Biol. Chem. 278, 4072–4081 (2003).
    1. Granneman S., Petfalski E. & Tollervey D. A cluster of ribosome synthesis factors regulate pre-rRNA folding and 5.8S rRNA maturation by the Rat1 exonuclease. EMBO J. 30, 4006–4019 (2011).
    1. Tang L., Sahasranaman A., Jakovljevic J., Schleifman E. & Woolford J. L. Jr Interactions among Ytm1, Erb1, and Nop7 required for assembly of the Nop7-subcomplex in yeast preribosomes. Mol. Biol. Cell 19, 2844–2856 (2008).
    1. Takahashi N., Yanagida M., Fujiyama S., Hayano T. & Isobe T. Proteomic snapshot analyses of preribosomal ribonucleoprotein complexes formed at various stages of ribosome biogenesis in yeast and mammalian cells. Mass Spectrom. Rev. 22, 287–317 (2003).
    1. Thomson E., Ferreira-Cerca S. & Hurt E. Eukaryotic ribosome biogenesis at a glance. J. Cell Sci. 126, 4815–4821 (2013).
    1. Grimm T. et al. Dominant-negative Pes1 mutants inhibit ribosomal RNA processing and cell proliferation via incorporation into the PeBoW-complex. Nucleic Acids Res. 34, 3030–3043 (2006).
    1. Holzel M. et al. The BRCT domain of mammalian Pes1 is crucial for nucleolar localization and rRNA processing. Nucleic Acids Res. 35, 789–800 (2007).
    1. Obrdlik A. et al. Nuclear myosin 1 is in complex with mature rRNA transcripts and associates with the nuclear pore basket. FASEB J. 24, 146–157 (2010).
    1. James A., Wang Y., Raje H., Rosby R. & DiMario P. Nucleolar stress with and without p53. Nucleus 5, 402–426 (2014).
    1. Boulon S., Westman B. J., Hutten S., Boisvert F. M. & Lamond A. I. The nucleolus under stress. Mol. Cell 40, 216–227 (2010).
    1. Burger K. & Eick D. Functional ribosome biogenesis is a prerequisite for p53 destabilization: impact of chemotherapy on nucleolar functions and RNA metabolism. Biol. Chem. 394, 1133–1143 (2013).
    1. Castello A. et al. Insights into RNA biology from an atlas of mammalian mRNA-binding proteins. Cell 149, 1393–1406 (2012).
    1. Holzel M. et al. Mammalian WDR12 is a novel member of the Pes1-Bop1 complex and is required for ribosome biogenesis and cell proliferation. J. Cell Biol. 170, 367–378 (2005).
    1. Kathiresan S. et al. Genome-wide association of early-onset myocardial infarction with single nucleotide polymorphisms and copy number variants. Nat. Genet. 41, 334–341 (2009).
    1. Kinoshita Y. et al. Pescadillo, a novel cell cycle regulatory protein abnormally expressed in malignant cells. J. Biol. Chem. 276, 6656–6665 (2001).
    1. Li J. J., Bickel P. J. & Biggin M. D. System wide analyses have underestimated protein abundances and the importance of transcription in mammals. PeerJ 2, e270 (2014).
    1. Ivanov A. et al. Analysis of intron sequences reveals hallmarks of circular RNA biogenesis in animals. Cell Rep. 10, 170–177 (2015).
    1. Liang D. & Wilusz J. E. Short intronic repeat sequences facilitate circular RNA production. Genes Dev. 28, 2233–2247 (2014).
    1. Ashwal-Fluss R. et al. circRNA biogenesis competes with pre-mRNA splicing. Mol. Cell 56, 55–66 (2014).
    1. Rybak-Wolf A. et al. Circular RNAs in the mammalian brain are highly abundant, conserved, and dynamically expressed. Mol. Cell 58, 870–885 (2015).
    1. Conn S. J. et al. The RNA binding protein quaking regulates formation of circRNAs. Cell 160, 1125–1134 (2015).
    1. Di Gesualdo F., Capaccioli S. & Lulli M. A pathophysiological view of the long non-coding RNA world. Oncotarget 5, 10976–10996 (2014).
    1. Visel A. et al. Targeted deletion of the 9p21 non-coding coronary artery disease risk interval in mice. Nature 464, 409–412 (2010).
    1. van Heesch S. et al. Extensive localization of long noncoding RNAs to the cytosol and mono- and polyribosomal complexes. Genome Biol. 15, R6 (2014).
    1. Vlatkovic N., Boyd M. T. & Rubbi C. P. Nucleolar control of p53: a cellular Achilles' heel and a target for cancer therapy. Cell. Mol. Life Sci. 71, 771–791 (2014).
    1. Moore K. J. & Tabas I. Macrophages in the pathogenesis of atherosclerosis. Cell 145, 341–355 (2011).
    1. Guevara N. V., Kim H. S., Antonova E. I. & Chan L. The absence of p53 accelerates atherosclerosis by increasing cell proliferation in vivo. Nat. Med. 5, 335–339 (1999).
    1. Mercer J., Figg N., Stoneman V., Braganza D. & Bennett M. R. Endogenous p53 protects vascular smooth muscle cells from apoptosis and reduces atherosclerosis in ApoE knockout mice. Circ. Res. 96, 667–674 (2005).
    1. Holdt L. M. et al. Expression of Chr9p21 genes CDKN2B (p15(INK4b)), CDKN2A (p16(INK4a), p14(ARF)) and MTAP in human atherosclerotic plaque. Atherosclerosis 214, 264–270 (2011).
    1. Thasler W. E. et al. Charitable state-controlled foundation human tissue and cell research: ethic and legal aspects in the supply of surgically removed human tissue for research in the academic and commercial sector in Germany. Cell Tissue Bank. 4, 49–56 (2003).
    1. Tripathi V. et al. Long noncoding RNA MALAT1 controls cell cycle progression by regulating the expression of oncogenic transcription factor B-MYB. PLoS Genet. 9, e1003368 (2013).
    1. Yanagimachi M. D. et al. Robust and highly-efficient differentiation of functional monocytic cells from human pluripotent stem cells under serum- and feeder cell-free conditions. PLoS ONE 8, e59243 (2013).
    1. Peters D. T., Cowan C. A. & Musunuru K. Genome editing in human pluripotent stem cells. StemBook, ed. The Stem Cell Research Community (2013).
    1. Anderegg U., Saalbach A. & Haustein U. F. Chemokine release from activated human dermal microvascular endothelial cells—implications for the pathophysiology of scleroderma? Arch. Dermatol. Res. 292, 341–347 (2000).
    1. Hirsch J. D. et al. Easily reversible desthiobiotin binding to streptavidin, avidin, and other biotin-binding proteins: uses for protein labeling, detection, and isolation. Anal. Biochem. 308, 343–357 (2002).
    1. Kulak N. A., Pichler G., Paron I., Nagaraj N. & Mann M. Minimal, encapsulated proteomic-sample processing applied to copy-number estimation in eukaryotic cells. Nat. Methods 11, 319–324 (2014).
    1. Scheltema R. A. & Mann M. SprayQc: a real-time LC-MS/MS quality monitoring system to maximize uptime using off the shelf components. J. Proteome Res. 11, 3458–3466 (2012).
    1. Cox J. & Mann M. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat. Biotechnol. 26, 1367–1372 (2008).
    1. Cox J. et al. Andromeda: a peptide search engine integrated into the MaxQuant environment. J. Proteome Res. 10, 1794–1805 (2011).
    1. Cox J. et al. Accurate proteome-wide label-free quantification by delayed normalization and maximal peptide ratio extraction, termed MaxLFQ. Mol. Cell. Proteomics 13, 2513–2526 (2014).
    1. Enright A. J. et al. MicroRNA targets in Drosophila. Genome Biol. 5, R1 (2003).
    1. Sturm M., Hackenberg M., Langenberger D. & Frishman D. TargetSpy: a supervised machine learning approach for microRNA target prediction. BMC Bioinformatics 11, 292 (2010).
    1. Griffiths-Jones S., Grocock R. J., van Dongen S., Bateman A. & Enright A. J. miRBase: microRNA sequences, targets and gene nomenclature. Nucleic Acids Res. 34, D140–D144 (2006).
    1. Bellucci M., Agostini F., Masin M. & Tartaglia G. G. Predicting protein associations with long noncoding RNAs. Nat. Methods 8, 444–445 (2011).
    1. Muppirala U. K., Honavar V. G. & Dobbs D. Predicting RNA-protein interactions using only sequence information. BMC Bioinformatics 12, 489 (2011).
    1. Tacker M., Fontana W., Stadler P. F. & Schuster P. Statistics of RNA melting kinetics. Eur. Biophys. J. 23, 29–38 (1994).
    1. R Core Team. R: a Language and Environment for Statistical Computing. R Foundation for Statistical Computing, Vienna, Austria (2016).

Source: PubMed

Подписаться