Transcriptome characterization by RNA sequencing identifies a major molecular and clinical subdivision in chronic lymphocytic leukemia

Pedro G Ferreira, Pedro Jares, Daniel Rico, Gonzalo Gómez-López, Alejandra Martínez-Trillos, Neus Villamor, Simone Ecker, Abel González-Pérez, David G Knowles, Jean Monlong, Rory Johnson, Victor Quesada, Sarah Djebali, Panagiotis Papasaikas, Mónica López-Guerra, Dolors Colomer, Cristina Royo, Maite Cazorla, Magda Pinyol, Guillem Clot, Marta Aymerich, Maria Rozman, Marta Kulis, David Tamborero, Anaïs Gouin, Julie Blanc, Marta Gut, Ivo Gut, Xose S Puente, David G Pisano, José Ignacio Martin-Subero, Nuria López-Bigas, Armando López-Guillermo, Alfonso Valencia, Carlos López-Otín, Elías Campo, Roderic Guigó, Pedro G Ferreira, Pedro Jares, Daniel Rico, Gonzalo Gómez-López, Alejandra Martínez-Trillos, Neus Villamor, Simone Ecker, Abel González-Pérez, David G Knowles, Jean Monlong, Rory Johnson, Victor Quesada, Sarah Djebali, Panagiotis Papasaikas, Mónica López-Guerra, Dolors Colomer, Cristina Royo, Maite Cazorla, Magda Pinyol, Guillem Clot, Marta Aymerich, Maria Rozman, Marta Kulis, David Tamborero, Anaïs Gouin, Julie Blanc, Marta Gut, Ivo Gut, Xose S Puente, David G Pisano, José Ignacio Martin-Subero, Nuria López-Bigas, Armando López-Guillermo, Alfonso Valencia, Carlos López-Otín, Elías Campo, Roderic Guigó

Abstract

Chronic lymphocytic leukemia (CLL) has heterogeneous clinical and biological behavior. Whole-genome and -exome sequencing has contributed to the characterization of the mutational spectrum of the disease, but the underlying transcriptional profile is still poorly understood. We have performed deep RNA sequencing in different subpopulations of normal B-lymphocytes and CLL cells from a cohort of 98 patients, and characterized the CLL transcriptional landscape with unprecedented resolution. We detected thousands of transcriptional elements differentially expressed between the CLL and normal B cells, including protein-coding genes, noncoding RNAs, and pseudogenes. Transposable elements are globally derepressed in CLL cells. In addition, two thousand genes-most of which are not differentially expressed-exhibit CLL-specific splicing patterns. Genes involved in metabolic pathways showed higher expression in CLL, while genes related to spliceosome, proteasome, and ribosome were among the most down-regulated in CLL. Clustering of the CLL samples according to RNA-seq derived gene expression levels unveiled two robust molecular subgroups, C1 and C2. C1/C2 subgroups and the mutational status of the immunoglobulin heavy variable (IGHV) region were the only independent variables in predicting time to treatment in a multivariate analysis with main clinico-biological features. This subdivision was validated in an independent cohort of patients monitored through DNA microarrays. Further analysis shows that B-cell receptor (BCR) activation in the microenvironment of the lymph node may be at the origin of the C1/C2 differences.

Figures

Figure 1.
Figure 1.
CLL transcriptional landscape. (A) Distribution of differentially expressed genes between tumor and normal samples according to their coding potential. (B) Normalized expression of transposable elements (TEs). Blue indicates low expression and red high expression, with some of the TEs differentially expressed highlighted. (C) Genes with condition-specific splicing ratios. Two of the genes with the most significant differences in the relative abundance of alternatively spliced isoforms between tumor and normal samples are shown. The boxplots correspond to the distribution of the relative abundances for each transcript (represented with a specific color) in the normal (left, N) and tumor (right, T) populations. The exonic structure of each transcript is represented using the same color scheme. (D) Allele-specific expression of somatic mutations. The relative expression of the reference allele, as derived from RNA-seq reads, was binned, and the number of cases in each bin plotted. The color gradient reflects the relative expression of the two alleles (the value that labels the bin in the x-axis).
Figure 2.
Figure 2.
Splicing changes in the BCR pathway between normal (N) and tumor (T) samples. (A) Partial view of the BCR signaling pathway with representation of genes with significant changes in alternative splicing. (B) Distribution of alternative splicing ratios between tumor and normal samples for three example genes in the BCR pathway highlighted in A. (C) ATM splicing in SF3B1-mutated samples. (Top left) Location of CLL-specific novel splice junction in the ATM gene. The novel 3′ splice site extends 20 bp upstream into the intron. Red dashed line indicates the truncation of the C-terminal end of ATM caused by the frame shift introduced by the novel splice site. (Bottom right) Expression levels of the putative and annotated junctions analyzed by qPCR. The log2 scale of the relative gene expression is represented for cases with SF3B1 mutation (dark gray), cases with ATM mutation and/or 11q deletion (dark gray), and cases without these genetic alterations. Light gray indicates unknown status.
Figure 3.
Figure 3.
Chimeric junctions between FCRL2–FCRL3 and GAB1–SMARCA5. (A) Schematic representation of the chimeric genes, associated ORFs, and junction sequences. (Top) Black boxes represent exons skipped by the chimeric junction. ORFs in the three possible frames are indicated in yellow. (Bottom) Sanger sequencing for the junction part of each chimera. The number in the square corresponds to the CLL sequenced sample. (B) Number of split-mapped (single reads split and mapped independently) and paired-end (two single reads from both ends of the same fragment) reads supporting the two chimeras and six previously described cases. TTTY15–USP9Y is reported twice because of the presence of two distinct fusion points. We should note that, although split-mapped and paired-end reads are shown separately, each chimera is supported by a combination of both.
Figure 4.
Figure 4.
Major transcriptional CLL subgroups. (A) Clustering of CLL and normal samples. Dendrogram obtained by hierarchical clustering of CLL and normal samples. (B) Consensus cluster. The matrix shows a clear and robust separation between CLL samples in C1 and C2. Dark blue regions indicate cluster partitions for samples that always cluster together (high consensus) and white indicate partitions with low consensus. (C) Multidimensional scaling of CLL and normal samples according to gene expression. (D) Enrichment score plot by GSEA comparing the RNA-seq based clustering with the clustering of an independent set of 124 samples, profiled with expression arrays. The plot compares the ranking correlation of the list of genes from the two clustering solutions. The vertical dark lines indicate where the genes in one list appear in the other ranked list of genes. An accumulation at the extremes indicates an agreement between the two lists. (E) Enrichment score plot by GSEA comparing the clustering in the RNA-seq and the previously published data sets. The three previously published data sets (Fabris et al. 2008; Friedman et al. 2009; Herold et al. 2011) contained 60, 40, and 106 samples, respectively.
Figure 5.
Figure 5.
Clinical behavior of the C1 (green) and C2 (red) subgroups. (A) Distribution of clinico-biological features in the RNA-seq profiled patients. (B) Time to treatment in the RNA-seq profiled patients at Binet stages A and B. (C) Distribution of clinico-biological features in the microarray profiled patients from the independent validation series. (D) Time to treatment in the microarray-profiled patients from the independent validation series at Binet stages A and B.
Figure 6.
Figure 6.
Interaction of CLL cells and the lymph node microenvironment. Stimulation of the BCR complex and other receptor and cell surface genes (CD79B, CD22, CD83, FCRG2B) leads to downstream changes in regulation. Affected genes such as those of the DUSP family, involved in the regulation of the MAPK pathway, may explain transcriptional differences observed for this pathway. Up-regulation of transcriptional regulators, like FOS and JUN, may trigger proliferation and inflammation processes that could be at the origin of C2 cells. Other genes involved in cell–cell signaling are also up-regulated in C2.

References

    1. Anczukow O, Rosenberg AZ, Akerman M, Das S, Zhan L, Karni R, Muthuswamy SK, Krainer AR 2012. The splicing factor SRSF1 regulates apoptosis and proliferation to promote mammary epithelial cell transformation. Nat Struct Mol Biol 19: 220–228
    1. Antonov AV 2011. : Analytical web portal for high-throughput cell biology. Nucleic Acids Res 39: W323–W327
    1. Berger MF, Levin JZ, Vijayendran K, Sivachenko A, Adiconis X, Maguire J, Johnson LA, Robinson J, Verhaak RG, Sougnez C, et al. 2010. Integrative analysis of the melanoma transcriptome. Genome Res 20: 413–427
    1. Buchner M, Fuchs S, Prinz G, Pfeifer D, Bartholome K, Burger M, Chevalier N, Vallat L, Timmer J, Gribben JG, et al. 2009. Spleen tyrosine kinase is overexpressed and represents a potential therapeutic target in chronic lymphocytic leukemia. Cancer Res 69: 5424–5432
    1. Burger JA, Gandhi V 2009. The lymphatic tissue microenvironments in chronic lymphocytic leukemia: In vitro models and the significance of CD40-CD154 interactions. Blood 114: 2560–2561, author reply 2561–2562
    1. Chuang H, Rassenti L, Salcedo M, Licon K, Kohlmann A, Haferlach T, Foa R, Ideker T, Kipps T 2012. Subnetwork-based analysis of chronic lymphocytic leukemia identifies pathways that associate with disease progression. Blood 120: 2639–2649
    1. Clauset A, Newman ME, Moore C 2004. Finding community structure in very large networks. Phys Rev E 70: 066111
    1. Corrionero A, Minana B, Valcarcel J 2011. Reduced fidelity of branch point recognition and alternative splicing induced by the anti-tumor drug spliceostatin A. Genes Dev 25: 445–459
    1. Coufal NG, Garcia-Perez JL, Peng GE, Yeo GW, Mu Y, Lovci MT, Morell M, O'Shea KS, Moran JV, Gage FH 2009. L1 retrotransposition in human neural progenitor cells. Nature 460: 1127–1131
    1. Cox D 1972. Regression models and life tables. J R Stat Soc Series B Stat Methodol 34: 187–220
    1. Djebali S, Davis C, Merkel A, Dobin A, Lassmann T, Mortazavi A, Tanzer A, Lagarde J, Lin W, Schlesinger F, et al. 2012. Landscape of transcription in human cells. Nature 489: 101–108
    1. Dühren-von Minden M, Übelhart R, Schneider D, Wossning T, Bach M, Buchner M, Hofmann D, Surova E, Follo M, Kohler F, et al. 2012. Chronic lymphocytic leukaemia is driven by antigen-independent cell-autonomous signalling. Nature 489: 309–312
    1. Fabbri G, Rasi S, Rossi D, Trifonov V, Khiabanian H, Ma J, Grunn A, Fangazio M, Capello D, Monti S, et al. 2011. Analysis of the chronic lymphocytic leukemia coding genome: Role of NOTCH1 mutational activation. J Exp Med 208: 1389–1401
    1. Fabris S, Mosca L, Todoerti K, Cutrona G, Lionetti M, Intini D, Matis S, Colombo M, Agnelli L, Gentile M, et al. 2008. Molecular and transcriptional characterization of 17p loss in B-cell chronic lymphocytic leukemia. Genes Chromosomes Cancer 47: 781–793
    1. Friedman J, Hastie T, Tibshirani R 2008. Sparse inverse covariance estimation with the graphical lasso. Biostatistics 9: 432–441
    1. Friedman DR, Weinberg JB, Barry WT, Goodman BK, Volkheimer AD, Bond KM, Chen Y, Jiang N, Moore JO, Gockerman JP, et al. 2009. A genomic approach to improve prognosis and predict therapeutic response in chronic lymphocytic leukemia. Clin Cancer Res 15: 6947–6955
    1. Gonzalez-Porta M, Calvo M, Sammeth M, Guigo R 2011. Estimation of alternative splicing variability in human populations. Genome Res 22: 528–538
    1. Guarini A, Chiaretti S, Tavolaro S, Maggio R, Peragine N, Citarella F, Ricciardi MR, Santangelo S, Marinelli M, De Propris MS, et al. 2008. BCR ligation induced by IgM stimulation results in gene expression and functional changes only in IgV H unmutated chronic lymphocytic leukemia (CLL) cells. Blood 112: 782–792
    1. Gundem G, Lopez-Bigas N 2012. Sample-level enrichment analysis unravels shared stress phenotypes among multiple cancer types. Genome Med 4: 28.
    1. Harrow J, Denoeud F, Frankish A, Reymond A, Chen CK, Chrast J, Lagarde J, Gilbert JG, Storey R, Swarbreck D et al. 2006. GENCODE: Producing a reference annotation for ENCODE. Genome Biol 7: S4.1–S4.9
    1. Herishanu Y, Perez-Galan P, Liu D, Biancotto A, Pittaluga S, Vire B, Gibellini F, Njuguna N, Lee E, Stennett L, et al. 2011. The lymph node microenvironment promotes B-cell receptor signaling, NF-κB activation, and tumor proliferation in chronic lymphocytic leukemia. Blood 117: 563–574
    1. Herold T, Jurinovic V, Mulaw M, Seiler T, Dufour A, Schneider S, Kakadia PM, Feuring-Buske M, Braess J, Spiekermann K, et al. 2011. Expression analysis of genes located in the minimally deleted regions of 13q14 and 11q22-23 in chronic lymphocytic leukemia-unexpected expression pattern of the RHO GTPase activator ARHGAP20. Genes Chromosomes Cancer 50: 546–558
    1. Huang da W, Sherman BT, Lempicki RA 2009. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc 4: 44–57
    1. Jurka J, Kapitonov VV, Pavlicek A, Klonowski P, Kohany O, Walichiewicz J 2005. Repbase Update, a database of eukaryotic repetitive elements. Cytogenet Genome Res 110: 462–467
    1. Kapp AV, Tibshirani R 2007. Are clusters found in one dataset present in another dataset? Biostatistics 8: 9–31
    1. Klein U, Tu Y, Stolovitzky GA, Mattioli M, Cattoretti G, Husson H, Freedman A, Inghirami G, Cro L, Baldini L, et al. 2001. Gene expression profiling of B cell chronic lymphocytic leukemia reveals a homogeneous phenotype related to memory B cells. J Exp Med 194: 1625–1638
    1. Knowles DG, Roder M, Merkel A, Guigo R 2013. Grape RNA-Seq analysis pipeline environment. Bioinformatics 29: 614–621
    1. Kulis M, Heath S, Bibikova M, Queiros AC, Navarro A, Clot G, Martinez-Trillos A, Castellano G, Brun-Heath I, Pinyol M, et al. 2012. Epigenomic analysis detects widespread gene-body DNA hypomethylation in chronic lymphocytic leukemia. Nat Genet 44: 1236–1242
    1. Lefebvre C, Rajbhandari P, Alvarez MJ, Bandaru P, Lim WK, Sato M, Wang K, Sumazin P, Kustagi M, Bisikirska BC, et al. 2010. A human B-cell interactome identifies MYB and FOXM1 as master regulators of proliferation in germinal centers. Mol Syst Biol 6: 377.
    1. Li FJ, Ding S, Pan J, Shakhmatov MA, Kashentseva E, Wu J, Li Y, Soong SJ, Chiorazzi N, Davis RS 2008. FCRL2 expression predicts IGHV mutation status and clinical progression in chronic lymphocytic leukemia. Blood 112: 179–187
    1. Maher C, Palanisamy N, Brenner J, Cao X, Kalyana-Sundaram S, Luo S, Khrebtukova I, Barrette T, Grasso C, Yu J, et al. 2009. Chimeric transcript discovery by paired-end transcriptome sequencing. Proc Natl Acad Sci 106: 12353–12358
    1. Marco-Sola S, Sammeth M, Guigó R, Ribeca P 2012. The GEM mapper: Fast, accurate and versatile alignment by filtration. Nat Methods 9: 1185–1188
    1. McCall MN, Irizarry RA 2011. Thawing frozen Robust Multi-array Analysis (fRMA). BMC Bioinformatics 12: 369.
    1. Mihailovich M, Militti C, Gabaldon T, Gebauer F 2010. Eukaryotic cold shock domain proteins: Highly versatile regulators of gene expression. Bioessays 32: 109–118
    1. Mitelman F, Johansson B, Mertens F 2007. The impact of translocations and gene fusions on cancer causation. Nat Rev Cancer 7: 233–245
    1. Montgomery SB, Sammeth M, Gutierrez-Arcelus M, Lach RP, Ingle C, Nisbett J, Guigo R, Dermitzakis ET 2010. Transcriptome genetics using second generation sequencing in a Caucasian population. Nature 464: 773–777
    1. Monti S, Tamayo P, Mesirov J, Golub T 2003. Consensus clustering: A resampling-based method for class discovery and visualization of gene expression microarray data. Mach Learn 52: 91–118
    1. Mortazavi A, Williams BA, McCue K, Schaeffer L, Wold B 2008. Mapping and quantifying mammalian transcriptomes by RNA-Seq. Nat Methods 5: 621–628
    1. Oscier D, Rose-Zerilli M, Winkelmann N, Gonzalez de Castro D, Gomez B, Forster J, Parker H, Parker A, Gardiner A, Collins A, et al. 2012. The clinical significance of NOTCH1 and SF3B1 mutations in the UK LRF CLL4 trial. Blood 121: 468–475
    1. Pajares M, Ezponda T, Catena R, Calvo A, Pio R, Montuenga L 2007. Alternative splicing: An emerging topic in molecular and clinical oncology. Lancet Oncol 8: 349–357
    1. Papaemmanuil E, Cazzola M, Boultwood J, Malcovati L, Vyas P, Bowen D, Pellagatti A, Wainscoat JS, Hellstrom-Lindberg E, Gambacorti-Passerini C, et al. 2011. Somatic SF3B1 mutation in myelodysplasia with ring sideroblasts. N Engl J Med 365: 1384–1395
    1. Perez-Llamas C, Lopez-Bigas N 2011. Gitools: Analysis and visualisation of genomic data using interactive heat-maps. PLoS ONE 6: e19541.
    1. Peto R, Pike MC 1973. Conservatism of the approximation sigma (O-E)2-E in the logrank test for survival data or tumor incidence data. Biometrics 29: 579–584
    1. Poliseno L, Salmena L, Zhang J, Carver B, Haveman WJ, Pandolfi PP 2010. A coding-independent function of gene and pseudogene mRNAs regulates tumour biology. Nature 465: 1033–1038
    1. Prinos P, Garneau D, Lucier JF, Gendron D, Couture S, Boivin M, Brosseau JP, Lapointe E, Thibault P, Durand M, et al. 2011. Alternative splicing of SYK regulates mitosis and cell survival. Nat Struct Mol Biol 18: 673–679
    1. Puente XS, Pinyol M, Quesada V, Conde L, Ordonez GR, Villamor N, Escaramis G, Jares P, Bea S, Gonzalez-Diaz M, et al. 2011. Whole-genome sequencing identifies recurrent mutations in chronic lymphocytic leukaemia. Nature 475: 101–105
    1. Quesada V, Conde L, Villamor N, Ordonez GR, Jares P, Bassaganyas L, Ramsay AJ, Bea S, Pinyol M, Martinez-Trillos A, et al. 2011. Exome sequencing identifies recurrent mutations of the splicing factor SF3B1 gene in chronic lymphocytic leukemia. Nat Genet 44: 47–52
    1. Quiroga MP, Balakrishnan K, Kurtova AV, Sivina M, Keating MJ, Wierda WG, Gandhi V, Burger JA 2009. B-cell antigen receptor signaling enhances chronic lymphocytic leukemia cell migration and survival: Specific targeting with a novel spleen tyrosine kinase inhibitor, R406. Blood 114: 1029–1037
    1. Raa DT, Derks IAM, Luijks DM, van Laar J, Monsuur H, Oldreive C, Jethwa A, Hüllein J, Stankovic T, Zenz T, et al. 2012. SF3B1 mutations in CLL are equivalent to p53/ATM dysfunction and cause defective puma upregulation in response to chemotherapy. In 54th ASH Annual Meeting and Exposition, Abstract 711. American Society of Hematology, Atlanta, USA
    1. Ramsay AJ, Quesada V, Foronda M, Conde L, Martinez-Trillos A, Villamor N, Rodriguez D, Kwarciak A, Garabaya C, Gallardo M, et al. 2013. POT1 mutations cause telomere dysfunction in chronic lymphocytic leukemia. Nat Genet 45: 526–530
    1. Refaeli Y, Young RM, Turner BC, Duda J, Field KA, Bishop JM 2008. The B cell antigen receptor and overexpression of MYC can cooperate in the genesis of B cell lymphomas. PLoS Biol 6: e152.
    1. Reich M, Liefeld T, Gould J, Lerner J, Tamayo P, Mesirov JP 2006. GenePattern 2.0. Nat Genet 38: 500–501
    1. Ren S, Peng Z, Mao JH, Yu Y, Yin C, Gao X, Cui Z, Zhang J, Yi K, Xu W, et al. 2012. RNA-seq analysis of prostate cancer in the Chinese population identifies recurrent gene fusions, cancer-associated long noncoding RNAs and aberrant alternative splicings. Cell Res 22: 806–821
    1. Rosenwald A, Alizadeh AA, Widhopf G, Simon R, Davis RE, Yu X, Yang L, Pickeral OK, Rassenti LZ, Powell J, et al. 2001. Relation of gene expression phenotype to immunoglobulin mutation genotype in B cell chronic lymphocytic leukemia. J Exp Med 194: 1639–1647
    1. Shapiro IM, Cheng AW, Flytzanis NC, Balsamo M, Condeelis JS, Oktay MH, Burge CB, Gertler FB 2011. An EMT-driven alternative splicing program occurs in human breast cancer and modulates cellular phenotype. PLoS Genet 7: e1002218.
    1. Simon R, Lam A, Li MC, Ngan M, Menenzes S, Zhao Y 2007. Analysis of gene expression data using BRB-ArrayTools. Cancer Inform 3: 11–17
    1. Smeenk G, Wiegant WW, Marteijn JA, Luijsterburg MS, Sroczynski N, Costelloe T, Romeijn RJ, Pastink A, Mailand N, Vermeulen W, et al. 2013. Poly(ADP-ribosyl)ation links the chromatin remodeler SMARCA5/SNF2H to RNF168-dependent DNA damage signaling. J Cell Sci 126: 889–903
    1. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, Paulovich A, Pomeroy SL, Golub TR, Lander ES, et al. 2005. Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci 102: 15545–15550
    1. Ting DT, Lipson D, Paul S, Brannigan BW, Akhavanfard S, Coffman EJ, Contino G, Deshpande V, Iafrate AJ, Letovsky S, et al. 2011. Aberrant overexpression of satellite repeats in pancreatic and other epithelial cancers. Science 331: 593–596
    1. Triqueneaux G, Velten M, Franzon P, Dautry F, Jacquemin-Sablon H 1999. RNA binding specificity of Unr, a protein with five cold shock domains. Nucleic Acids Res 27: 1926–1934
    1. Velusamy T, Palanisamy N, Kalyana-Sundaram S, Sahasrabuddhe AA, Maher CA, Robinson DR, Bahler DW, Cornell TT, Wilson TE, Lim MS, et al. 2013. Recurrent reciprocal RNA chimera involving YPEL5 and PPP1CB in chronic lymphocytic leukemia. Proc Natl Acad Sci 110: 3035–3040
    1. Wang ET, Sandberg R, Luo S, Khrebtukova I, Zhang L, Mayr C, Kingsmore SF, Schroth GP, Burge CB 2008. Alternative isoform regulation in human tissue transcriptomes. Nature 456: 470–476
    1. Wang L, Lawrence MS, Wan Y, Stojanov P, Sougnez C, Stevenson K, Werner L, Sivachenko A, DeLuca DS, Zhang L, et al. 2011. SF3B1 and other novel cancer genes in chronic lymphocytic leukemia. N Engl J Med 365: 2497–2506
    1. Wilkerson MD, Hayes DN 2010. ConsensusClusterPlus: A class discovery tool with confidence assessments and item tracking. Bioinformatics 26: 1572–1573
    1. Yoshida K, Sanada M, Shiraishi Y, Nowak D, Nagata Y, Yamamoto R, Sato Y, Sato-Otsubo A, Kon A, Nagasaki M, et al. 2011. Frequent pathway mutations of splicing machinery in myelodysplasia. Nature 478: 64–69
    1. Zenz T, Mertens D, Kuppers R, Dohner H, Stilgenbauer S 2009. From pathogenesis to treatment of chronic lymphocytic leukaemia. Nat Rev Cancer 10: 37–50
    1. Zhuo L, Gong J, Yang R, Sheng Y, Zhou L, Kong X, Cao K 2009. Inhibition of proliferation and differentiation and promotion of apoptosis by cyclin L2 in mouse embryonic carcinoma P19 cells. Biochem Biophys Res Commun 390: 451–457

Source: PubMed

3
Tilaa