Skip to main content
Genes & Diseases logoLink to Genes & Diseases
. 2019 Jun 19;6(3):247–257. doi: 10.1016/j.gendis.2019.06.002

PRMT5 in gene regulation and hematologic malignancies

Fen Zhu 1, Lixin Rui 1,
PMCID: PMC6997592  PMID: 32042864

Abstract

Arginine methylation is a common posttranslational modification that governs important cellular processes and impacts development, cell growth, proliferation, and differentiation. Arginine methylation is catalyzed by protein arginine methyltransferases (PRMTs), which are classified as type I and type II enzymes responsible for the formation of asymmetric and symmetric dimethylarginine, respectively. PRMT5 is the main type II enzyme that catalyzes symmetric dimethylarginine of histone proteins to induce gene silencing by generating repressive histone marks, including H2AR3me2s, H3R8me2s, and H4R3me2s. PRMT5 can also methylate nonhistone proteins such as the transcription factors p53, E2F1 and p65. Modifications of these proteins by PRMT5 are involved in diverse cellular processes, including transcription, translation, DNA repair, RNA processing, and metabolism. A growing literature demonstrates that PRMT5 expression is upregulated in hematologic malignancies, including leukemia and lymphoma, where PRMT5 regulates gene expression to promote cancer cell proliferation. Targeting PRMT5 by specific inhibitors has emerged as a potential therapeutic strategy to treat these diseases.

Keywords: Gene regulation, Hematologic malignancies, Metabolism, Pathogenesis, PRMT5

Introduction

Protein arginine methylation is a type of post-translational modification that occurs as common as phosphorylation and ubiquitination.1 Arginine side chain has 2 terminal guanidino (NH2) groups that can be subjected to methylation. Depending on where the methyl group is added, there are 3 types of arginine methylation in mammalian cells: ω-NG-monomethylation, ω-NG,N′G-symmetric dimethylation and ω-NG,NG-asymmetric dimethylation (Fig. 1). Arginine methylation does not change the charge of proteins but increases hydrophobicity and reduces hydrogen bond donor. Therefore, arginine methylation can significantly alter protein-protein, protein-DNA, and protein-RNA interactions and has profound biological significance. Arginine methylation of histones (H2AR3, H3R8, H3R2 and H4R3) coordinates the formation of a transcriptional complex that regulates gene transcription. Arginine methylation of transcription factors and other signaling molecules alters their transcriptional activities. Arginine methylation of spliceosomal proteins is required for the assembly of spliceosomal complex and proper gene splicing. As such, arginine methylation governs important cellular processes and impacts development, cell growth, proliferation, and differentiation. To date, 9 protein arginine methyltransferases (PRMTs) have been identified, and they are classified as type I and type II enzymes responsible for the formation of asymmetric and symmetric dimethylarginine, respectively. In this review, we will focus on PRMT5, a main type II enzyme, and discuss its role in gene regulation and hematologic malignancies.

Figure 1.

Figure 1

Arginine methylation catalyzed by PRMTs. *Whether PRMT7 catalyzes symmetric dimethylation (Type II) or monomethylation of arginines (Type III) is controversial.34, 85, 86, 87, 88

Discovery and characterization of PRMT5

PRMT5 is a highly conserved gene from unicellular eukaryotes to higher organisms. In search of second-site mutations that were synthetic lethal in combination with the deletion of the Histone H3 amino terminus in budding yeast Saccharomyces cerevisiae, a new gene product histone synthetic lethal 7 (Hsl7) was identified.2 Hsl7 was not essential for growth, but it regulated morphology of budding yeast. Deletion of Hsl7 led to elongated buds and undivided nuclei.2 This may be explained by the activation of a morphogenesis checkpoint that delays nuclear division.2, 3, 4 Skb1 is a homolog of Saccharomyces cerevisiae Hsl7 in the fission yeast Schizosaccharomyces pombe. Skb1 was identified as an interactor and modulator of shk1 kinase, which was an essential component of the Ras1/Cdc42 signaling cascade in controlling the viability and normal morphology of the fission yeast.5 Similar to Hsl7, Skb1 was not essential for growth and it participated in controlling cell morphology. Deletion of Skb1 led to premature exit from G2 and less elongated cells, which was opposite to the role of Hsl7 in controlling cell morphology.5, 6 Xenopus Hsl7 homologue promoted mitotic entry, which is controlled by the DNA replication checkpoint.7

The human homologue of skb1 (Skb1Hs) was characterized as a Janus Kinase 2 (JAK2) interacting protein in the yeast two-hybrid system, termed Jak binding protein 1(JBP1).8 JBP1 was shown to possess methyltransferase activity towards histone H4 and H2A, but whether the methylation is arginine specific was not clear.8 Another group independently cloned Skb1Hs in an attempt to identify proteins interacting with nonstructural protein-3 (NS3) of the hepatitis C virus (HCV).9 They discovered that Skb1Hs is an arginine methyltransferase termed PRMT5 that contains conserved domains of AdoMet-dependent protein arginine methyltransferases, and exerts arginine methyltransferase activity towards myelin basic protein (MBP). Although MBP was the first protein found with arginines symmetrically dimethylated,10, 11, 12 whether MBP is a direct target of PRMT5 was not determined in this work.9 Later in the same year, a study using amino acid analysis of the in vitro methylated MBP revealed that PRMT5 catalyzes the formation of symmetric dimethylarginines.13 Thus, PRMT5 was identified as the first type II arginine methyltransferase. However, the yeast Hsl7 was shown to catalyze only monomethylation of calf histone H2A in vitro, and no Hsl7-dependent methylation of endogenous yeast histones was observed in vivo.14

Biochemical, structural and enzymological studies further demonstrated the methylation mechanism of PRMT5. PRMT5 functions in a methylosome complex, and several cofactors are important for PRMT5 activity, stability and substrate specification. Methylosome protein 50 (MEP50) is a key cofactor for PRMT5 activity, and depletion of MEP50 significantly reduces methylosome activity.15, 16 pICln associates with and assists the methylation of Sm proteins by PRMT5.17, 18 While Riok1 competes with pICln for binding to PRMT5 and directs the activity of PRMT5 towards the RNA binding protein nucleolin.19 PRMT5 contains a two domain structure containing an N terminal domain and C terminal catalytic domain.20 The N terminal domain has a TIM barrel structure which interacts with the C terminal catalytic domain of adjacent dimers and other cofactors that are important for PRMT5 activity.20 MEP50 forms a hetero-octameric complex with PRMT5. In this complex 4 PRMT5 molecules form a tetramer in a head-to-tail fashion with 4 MEP50 molecules binding to the N terminal domain of PRMT5 molecules.20 The C terminal catalytic domain of PRMT5 contains a Rossmann fold structure for SAM binding followed by a β barrel for substrate binding.20

Several key residues regulating the activity of PRMT5 are revealed by this structure analysis. Glu435 and Glu444 are highly conserved active site residues that form salt bridges with the guanidine side-chain of the substrate arginine and poises ω-NG nitrogen for methyl transfer.20 Glu444 mutation to Gln creates the enzymatically dead version of PRMT5.21, 22 Oncogenic JAK2V617F mutant is shown to phosphorylate PRMT5 at multiple tyrosine residues (280, 283, 286, 297, 304, and 307).23 Structural analysis revealed that Tyr304 and 307 are involved in substrate binding,20 suggesting a possible mechanism by which phosphorylation by JAK2 inhibits methyltransferase activity of PRMT5. A study in Caenorhabditis elegans found that Phe379 is the key to determine the type II activity of CePRMT5, as F379M mutation not only increases the activity of CePRMT5 but also makes CePRMT5 capable of catalyzing both symmetric and asymmetric dimethylation of arginines.24 The corresponding F327M mutation in human PRMT5 also results in the gaining of asymmetric arginine dimethylase activity.24

PRMT5 methylates substrates in a nonprocessive manner, which allows for the release of the monomethylated substrate from PRMT5 but facilitates dimethylation when the concentration of monomethylated substrates exceeds that of unmethylated substrates.16, 20, 21 The domain structure of PRMT5 is illustrated in Fig. 2. CePRMT5 shares high sequence homology with human PRMT5.25 However, CePRMT5 does not associate with a MEP50 homologue and functions as a homodimer.16, 24, 25

Figure 2.

Figure 2

Functional domains of PRMT5.

Gene regulation and cellular functions mediated by PRMT5

Since the discovery of PRMT5, researchers have identified its diverse substrates, from histone proteins to non-histone nuclear and cytoplasmic proteins. Methylation of these substrates by PRMT5 is involved in many cellular processes, including transcription, DNA repair, RNA processing, proliferation and metabolism.

Regulation of gene transcription

In 2002, the first direct experimental evidence demonstrated that PRMT5, as a type II arginine methylase, is involved in controlling gene transcription and cell proliferation.26 PRMT5 is a component of the cyclin E1 repressive complex (CERC) based on mass spectrometry analysis in NIH3T3 cells.26 Chromatin immunoprecipitation (ChIP) revealed that PRMT5 and dimethylated H4R3 are present at the transcription start site of cyclin E1, leading to the suppression of cyclin E1 gene transcription and cell proliferation.26 The first large scale interrogation of gene expression regulated by PRMT5 was done by microarray analysis in PRMT5 knockdown NIH3T3 cells, which revealed 227 up-regulated genes, including tumor suppressors and cell cycle inducers, and 43 down-regulated genes.27 PRMT5 was shown to directly methylate H3R8 and H4R3 in vitro. The overall effect on the proliferation of NIH3T3 cells was to stimulate cell proliferation and promote anchorage dependent cell growth,27 which contradicts the previous study.26 Nevertheless, these two and other studies all suggested that PRMT5 catalyzes histone methylation that suppresses gene transcription.26, 27, 28, 29, 30, 31 However, genome wide studies have yielded controversial results. ChIP sequencing (ChIP-seq) analysis in CD4+ T cells using antibody recognizing both H2A and H4 (symmetric dimethyl R3) demonstrated no preference of those marks for either active or silent promoters.32 In mouse embryonic stem cells and mouse embryonic fibroblasts, H4R3me2s is a hall mark of GC rich regions and is generally independent of transcriptional levels or DNA methylation.33 PRMT5 catalyzed H3R2me2s supports euchromatin maintenance and marks active promoters in B cells.34, 35 Additionally, gene expression profiling in human embryonic stem cells (hESCs) revealed changes in the expression of only 78 genes after PRMT5 knockdown, 62 of which were downregulated,36 suggesting a major role for PRMT5 in promoting gene expression.

In addition to histones and chromatin remodeling complexes, PRMT5 controls gene transcription through methylating transcription factors and other signaling molecules. PRMT5 mediated methylation of Arg 1175 of epidermal growth factor receptor (EGFR) positively modulates EGF-induced EGFR trans-autophosphorylation at Tyr1173, which has suppressive effects on extracellular signal–regulated kinase (ERK) activation and cell proliferation.37 Methylation of p65 at Arg30 by PRMT5 in response to IL-1β stimulation promotes the ability of NF-κB to bind to kB elements for gene transcription.38 Methylation of the transcriptional corepressor KRAB-associated protein 1 (KAP1) contributes to gene repression.39 Epstein–Barr virus (EBV) nuclear antigen 2 (EBNA2) has an arginine glycine repeat domain that can be methylated by PRMT5. Methylation of EBNA2 by PRMT5 has been shown to stimulate EBNA2-dependent transcription and may be important for EBV induced transformation of B cells.40 PRMT5 can also methylate EBNA1, but this methylation appears to regulate the cellular localization of EBNA1 rather than to change the transcriptional activity of EBNA1.41 Methylation of Krüppel-like factor 4 (KLF4) (Arg374) by PRMT5 leads to stabilization and accumulation of KLF4 protein, which antagonizes γ–radiation induced apoptosis and promotes tumorigenesis.42 Methylation of B cell lymphoma 6 (BCL6) at Arg305 by PRMT5 contributes to the suppressive activity of BCL6, which is required for germinal center formation.43

In addition to its suppressive or active role in gene transcription, PRMT5 can also regulate transcriptional specificity of target genes in the context of cellular conditions. During the DNA damaging response, methylation of p53 at Arg333, Arg335, and Arg337 by PRMT5 has an important functional consequence on p53 activity.44 p53 arginine methylation promotes the expression of genes such as p21 that induce cell cycle arrest, while PRMT5 depletion increases apoptosis.44 Arginine methylation affects the promotor specificity based on p53 ChIP analysis of its target genes p21, GADD45 and PUMA. Methylation of RAF proteins by PRMT5 increases their degradation and limits the amplitude and duration of ERK signaling in response to growth factors, affecting the evoked biological response.45 Inhibition of PRMT5-mediated RAF methylation enhances ERK signaling, redirecting the cellular response from proliferation to differentiation.45 Methylation of E2F1 at Arg111 and Arg113 by PRMT1 promotes cell apoptosis, while methylation by PRMT5 locks E2F1 into its cell cycle progression mode.46, 47 Methylation of androgen receptor (AR) at Arg761 by PRMT5 attenuates AR recruitment to its target genes and inhibits transcription of genes expressed in the differentiated prostate epithelium.48 Methylation of the methyl DNA binding domain protein 2 (MBD2) by PRMT5 reduces the affinity of MBD2 to histone deacetylase (HDAC) silencing complex, thus impairing the transcriptional repression function of MBD2 in cells.49

DNA replication and repair

Flap endonuclease 1 (FEN1) is a multifunctional enzyme involved in DNA replication, DNA repair and apoptotic DNA fragmentation. Methylation of FEN1 by PRMT5 promotes its interaction with PCNA and recruitment to DNA replication loci.50 Rad9 is critical for DNA repair and cell cycle checkpoint control. Methylation of Rad9 by PRMT5 is required for cellular resistance to DNA damage stresses, and for activation of S/M and G2/M checkpoints.51 TIP60 is a histone acetyltransferase involved in chromatin remodeling and homologous recombination mediated DNA repair. PRMT5 methylates RUVBL1, a cofactor that regulates TIP60 activity.52 PRMT5 can also directly regulate TIP60 activity through alternative splicing.22 PRMT5 depletion leads to defects in DNA replication and repair, resulting in DNA damage accumulation.22, 50, 52

Transcription elongation and termination

Transcription elongation factors SPT5 and FCP1 are substrates of PRMT5. Methylation of SPT5 by PRMT5 at Arg698 negatively regulates association of SPT5 with RNA polymerase II and gene promoters.28 Methylation of FCP1 at Arg913 and Arg916 by PRMT5 does not change the affinity between FCP1 and RNA polymerase II, but FCP1-associated PRMT5 can methylate histones.53 RNA polymerase II subunit POLR2A is subjected to methylation by PRMT5. Symmetric dimethylation of Arg1810 in the POLR2A C terminal domain promotes recruitment of SMN and RNA–DNA helicase senataxin, which are required for resolving RNA-DNA hybrids created by RNA polymerase II and transcription termination.54

RNA processing

Methylation of SmD1, SmD3 and SmB by pICln-associated PRMT5 directs the Sm proteins to the survival of motor neuron (SMN) proteins, where they are associated with newly exported snRNA to form spliceosomal snRNPs.17, 18, 19, 55, 56, 57 In the Eμ-myc transgenic mouse model, PRMT5 is a key enzymatic component of Myc promoted transcription of core snRNP assembly genes.58 PRMT5 regulates the splicing of genes involved in DNA repair pathway.22, 59, 60 Knockout of PRMT5 reduces the productive transcript levels of DNA repair genes, leading to DNA damage accumulation.22, 60 Cells with wild type p53 is more sensitive to PRMT5 inhibition.59

Metabolism

PRMT5 is involved in metabolism of bile acid, glucose and lipid. In response to the bile acid signal, the small heterodimer partner (SHP) is stabilized and activated to suppress the synthesis and production of bile acid, fatty acid and glucose. In this process, PRMT5 has been shown to increase SHP activity by methylating SHP at Arg57.61 In response to the fasting signal, activation of cAMP response element binding (CREB) protein promotes the transcription of gluconeogenic genes and glucose production(62). PRMT5 is recruited to CREB target genes by CREB regulated transcriptional coactivator 2 (CRTC2) and catalyzes H3R2 symmetric dimethylation, thus promoting chromatin accessibility and protein kinase A (PKA) catalyzed phosphorylation and activation of CREB protein.62 Upon glucose induction in hepatocellular carcinoma (HCC), PRMT5 competes with cyclin dependent kinase inhibitor 2A (CDKN2A) for biding to cyclin dependent kinase 4 (CDK4) and promotes cell cycle transition from G1 to S phase.63 Arg 24 of CDK4 is critical for its binding to both CDKN2A and PRMT5, but is not symmetrically dimethylated, suggesting methyltransferase activity independent function of PRMT5.63

Sterol regulatory element-binding protein (SREBP) is a transcription factor controlling de novo lipogenesis in tumors.64 PRMT5 methylates SREBP1a at Arg321, which is required for SREBP1a transcriptional activity. This methylation can also prevent GSK3β mediated phosphorylation of SREBP1a on S430, which promotes proteasomal degradation of SREBP1.65 Thus, PRMT5 promotes lipogenesis and tumor growth through both activating and stabilizing SREBP1a. In addition, symmetric dimethylation of SREBP1a at Arg321 is increased in human hepatocellular carcinoma tissue and correlates with poor prognosis.65 In response to a high fat diet, PRMT5 expression is upregulated by unknown mechanisms. Increased PRMT5 expression promotes activation of the PI3K-AKT pathway, which suppresses the expression of master transcription factors of mitochondrial biogenesis and promotes high fat induced steatosis.66 Our whole transcriptome profiling in human diffuse large B cell lymphoma (DLBCL) cell lines has also revealed that PRMT5 regulates PI3K-AKT activity, glycolysis and cholesterol homeostasis.79 However, the mechanism by which PRMT5 regulates metabolism in DLBCL is largely unknown.

Current dogma is that PRMTs catalyze methylation of arginine-glycine rich motifs, but proteome-wide analysis of arginine methylation has revealed that methylation is independent of such a sequence context.1 The methylation sites of known substrates of PRMT5 indicate that arginine methylation can occur without having glycine next to it (Table 1).

Table 1.

Substrate methylation by PRMT5.

Substrate Methylation site Biological function reference
MBP R107: KGRGL Myelin integrity 10, 11, 12, 89, 90, 91, 92
SmD1, SmD3, SmB C terminal arginine- and glycine-rich domains Gene splicing 17, 18, 19, 55, 56, 57
H3R8 R8: TARKS Transcriptional repression 27, 74, 78
H3R2 R2: ARTKQ Transcriptional activation 34, 35
H4/H2AR3 R3: SGRGK Transcriptional repression or independent of transcription 26, 27, 31, 32, 33, 74, 78, 93
p53 R333/335/337: QIRGRERFE Apoptosis, cell cycle 44
FEN1 R192: LMRHL DNA replication, DNA repair 50
EGFR R1175: YLRVA Signaling activation 37
CRAF R563: VGRGY Signaling activation 45
SHP R57: TCREA Bile acid biosynthesis, lipid synthesis, glucose production 61
E2F1 R111/113: RGRGRHP Apoptosis, cell cycle 46, 47
p65 R30: KQRGM NFκB activation 38
EBNA1, EBNA2 arginine- and glycine repeat domains Gene transcription, cellular localization 40, 41
KLF4 R374/376/377: PKRGRRSW Protein stability 42
POLR2A R1810: SPRYT Transcription termination 54
AR R761: NSRML Transcriptional activity 48
RUVBL1 R205: QGRCD DNA repair 52
BCL6 R305: EERPS Transcriptional activity 43
SPT5 R698: RGRGR Suppress transcriptional elongation 28
FCP1 R913/916: GGRGPRGH unknown 53
MBD2 N terminal arginine- and glycine-rich domains Suppress transcription repression function of MBD2 49
KAP1 R308: NKRGR
R523: IERGA
Transcriptional repression 39
MYCN R242: GGRQT Protein stability 94
CF I m68 R202/204/206: GGRGRGRFP Not determined 95
FGF-223 N terminal arginine- and glycine-rich domains Nuclear shuttling 96
RPS10 R158/160: FGRGRGQ Ribosome assembly 97
G3BP1 C terminal arginine- and glycine-rich domains Repress stress granule assembly 98
ASK1 R89: GSRRT Inhibits apoptosis 99
PDCD4 R110:KGRGL Cell proliferation 100
srGAPs R927: AGRSK Cell spreading, migration 101
PDGFRα R554:KPRYE Protein stability 102
HOXA9 R140: SARRG Induction of endothelial cell leukocyte adhesion molecules 103
GM130 R6:ETRQS
R18:KLREY
R23: QQRNS
Golgi apparatus ribbon formation 104
Piwi proteins N terminal RG/RA motifs Compartmentalization of small RNA pathway 105
Rad9 R172/174/175: IGRGRRVI DNA damage 51
TDH R180: SPRNP Somatic cell reprogramming 106
SREBP1 R321: QSRGE Lipogenesis 65
GLI1 R515:GTRGL
R990:PPRCG
R1018:VGRLG
Protein stability 107

PRMT5 in hematologic malignancies

PRMT5 in normal hematopoiesis

Several mouse models have been established to investigate the importance of PRMT5 in hematopoiesis. Deletion of PRMT5 is embryonic lethal due to the abrogation of pluripotent cells in blastocysts.67 Conditional knockout of PRMT5 in mouse bone marrow hematopoietic stem and progenitor cells (HSPCs) disrupts quiescent cell status and leads to exhaustion of HSPCs.21, 60 Consequently, erythroid differentiation, T cell and B cell development are impaired, resulting in severe pancytopenia.21 Deletion of p53 significantly delays PRMT5 knockout induced HSC exhaustion.60 Deletion of PRMT5 just prior to the start of fetal liver hematopoiesis causes severe block in erythroid development and is also embryonic lethal.22 Ablation of PRMT5 in pro-B cells blocks B cell development at the pro-B stage, leading to absence of mature B cells in spleen. Concurrent deletion of p53 fully recovers the number of pro-B cells, but not pre-B cells, suggesting a p53 independent role of PRMT5 in pre-B cell development.68 PRMT5 protects activated B cells from apoptosis and promotes proliferation of activated B cells, which are independent of p53 activation.68 During germinal center (GC) formation, the methylation of BCL6 by PRMT5 is necessary for the full transcriptional repressive effects of BCL6.43 Deletion of PRMT5 in GC B cells impairs GC formation and immunoglobulin affinity maturation.43, 68 PRMT5 also prevents premature plasma cell differentiation.68 The role of PRMT5 in mouse B cell development is briefly summarized in Fig. 3. However, it should be kept in mind that mouse models may not recapitulate human biology. PRMT5 has been shown to regulate human embryonic stem cell proliferation but not pluripotency.36 Knockdown of PRMT5 expression in human CD34 + cord blood cells promotes progenitor cell expansion and accelerates erythroid differentiation.23 Knockdown of PRMT5 in a B cell progenitor acute lymphoblastic leukemia cell line Nalm6 induces cell differentiation from the pre-B to immature B stage. While overexpression of PRMT5 in human mature B cells promotes dedifferentiation back to the pre-B/immature B stages.69

Figure 3.

Figure 3

Role of PRMT5 in murine B cell development.21, 43, 60, 68 GC: germinal center; LZ: light zone.

PRMT5 in the pathogenesis of hematologic malignancies

Leukemia

After the identification of PRMT5 as a JAK2 binding protein, the functional consequence of this interaction was not reported until 12 years later. Liu F. et al. found that JAK2 harboring activating mutations (V717F, K539L) interacts with PRMT5 more strongly than wild type JAK2, and is able to phosphorylate PRMT5. Phosphorylation of PRMT5 by constitutively active JAK2 disrupts the interaction of PRMT5 with its co-activator MEP50-, thus impairing methyltransferase activity of PRMT5. Downregulation of PRMT5 activity contributes to mutant JAK2 induced myeloproliferative disease.23 PRMT5 is expressed in acute myeloid leukemia (AML) cell lines at various levels, and inhibition of PRMT5 leads to DNA damage accumulation and reduces AML cell viability.22 In AML, PRMT5 has been shown to epigenetically silence the expression of miR-29b. As a result, transcription factor Sp1, one of the targets of miR-29, is upregulated by PRMT5 indirectly. Then, PRMT5 and Sp1 participate in a transcriptional complex that activates the transcription of receptor tyrosine kinase FLT3.70 PRMT5 is upregulated in human T-cell leukemia virus type-1 (HTLV-1) mediated T cell transformation as well as in T-cell leukemia/lymphoma cell lines and patient PBMCs.71 Inhibition of PRMT5 decreases cancer cell proliferation.71 In pediatric acute lymphoblastic leukemia (ALL), PRMT5 was found to be expressed at initial diagnosis while likely to be absent at complete remission,69 suggesting an oncogenic role of PRMT5 in ALL development. PRMT5 is highly expressed in CD34 + cells originated from bone marrow of chronic myelogenous leukemia (CML), compared to that from normal bone marrow.72 BCR-ABL upregulates PRMT5 expression in CML CD34 + cells, while PRMT5 in turn upregulates BCR-ABL expression in CML cells. Genetic or pharmacologic inhibition of PRMT5 severely impairs the proliferation of CML CD34 + cells both in vitro and in vivo.72

Lymphoma

PRMT5 expression is upregulated in lymphoma.58, 73, 74, 75, 76 PRMT5 knockdown in DLBCL- and MCL- cell lines reactivates the retinoblastoma tumor suppressor pathway and silences polycomb repressor complex 2 (PRC2), leading to lymphoma cell death.74 The same group also found that PRMT5 directly silences the expression of axin-related protein (AXIN2) and WNT inhibitory factor 1 (WIF1), which are negative regulators of the pro-survival WNT/β-catenin signaling pathway and AKT/GSK3β pathway.76 In a transgenic mouse model, PRMT5 cooperates with Cyclin D1T286A to induce aggressive T cell Lymphoma/Leukemia.77 PRMT5 expression is required for tumor development driven by multiple oncogenes (NOTCH1, c-MYC, and MLL–AF9).77 This study also demonstrates that arginine methylation of p53 by PRMT5 reduces expression of proapoptotic p53 transcriptional targets.77 PRMT5 is required for tumor maintenance in the Eμ-myc transgenic mouse model since homozygous deletion of PRMT5 results in full disease free survival.58 PRMT5 is also important for virus induced lymphomagenesis.78 Inhibition of PRMT5 blocks EBV induced B cell transformation. During EBV infection of B cells, PRMT5 is upregulated and contributes to initiation and maintenance of EBV induced B cell transformation.78 We also found that PRMT5 is highly expressed in DLBCL, MCL, Hodgkin lymphoma and Burkitt lymphoma cell lines when compared to naive B cells.79 The level of PRMT5 expression is also elevated in DLBCL and MCL patient samples. PRMT5 knockout inhibits the proliferation of DLBCL cell lines both in vitro and in xenograft mouse models.79 Moreover, inhibition of PRMT5 using a small molecule inhibitor significantly reduces tumor volume and improves animal survival in a DLBCL patient derived xenograft mouse model.79

It is now well accepted that PRMT5 is upregulated in different types of lymphoma, however the mechanism by which it is upregulated remains largely unknown. In MCL, the decreased miR-92b and miR-96 expression accelerates PRMT5 translation.75 We discovered that BTK–NF–kB signaling induces PRMT5 transcription in activated B cell-like (ABC) DLBCL cells while BCR downstream PI3K-AKT-MYC signaling upregulates PRMT5 expression in both ABC and germinal center B cell-like (GCB) DLBCL cells.79

Multiple myeloma

In multiple myeloma (MM), high PRMT5 expression is associated with decreased progression-free survival and overall survival.80 Inhibition of PRMT5 by a specific inhibitor inhibits proliferation of both MM cell lines and patient MM cells, and decreases tumor growth in a xenograft mouse model.80 PRMT5 interacts with Tripartite motif-containing protein 21 (TRIM21), an E3 ubiquitin ligase for IKKβ, to block TRIM21-mediated degradation of IKKβ, allowing activation of NF-κB signaling.80

Targeting PRMT5 in hematologic malignancies

Given the important role PRMT5 plays in cancer development, targeting PRMT5 by specific inhibitors holds promise in clinical application. Methylthioadenosine (MTA) is an analog of the methyl donor S-adenosyl methionine (SAM), and specifically inhibits PRMT5 enzymatic activity.81, 82 5-Methylthioadenosine phosphorylase (MTAP) is one of the enzymes involved in the methionine salvage pathway that metabolizes MTA to adenine and methionine. Due to its proximity to the tumor suppressor gene CDKN2A, MTAP was reported to be frequently co-deleted with CDKN2A in many human tumors, which leads to the accumulation of MTA in cells and sensitizes cells to further PRMT5 inhibition.81, 82

To date, several small molecule inhibitors targeting PRMT5 have been developed by different groups with various selectivity and potency.70, 71, 73, 78, 83, 84 EPZ015666 (GSK3235025) is an SAM cooperative inhibitor of PRMT5 that binds at the peptide binding site of PRMT5.73 In preclinical studies, EPZ015666 efficiently inhibits MCL growth both in vitro and in cell line derived xenograft mouse models with IC50 at a low nanomolar range.73, 83 EPZ015666 also significantly inhibits growth of MM cells.80 The similar compound EPZ015866 (GSK3203591) has been investigated in in vitro studies.43, 59, 83 A more potent PRMT5 inhibitor, EPZ015938 (GSK3326595), is being used for two phase I trials (NCT02783300, NCT03614728) in solid tumors, non-Hodgkin lymphoma and leukemia.84

Given the importance of PRMT5 for normal physiology, targeting PRMT5 might elicit severe side effects. Combination therapy by lowering the dosage of PRMT5 inhibitors is a way to reduce drug toxicity and avoid drug resistance. In human AML cell lines, deletion of PRMT5 leads to defects in homologous recombination due to aberrant splicing of key factors involved in chromatin modification and DNA repair.22 As a result, cells rely more on poly(ADP-ribose) polymerase (PARP) to repair their DNA. Inhibition of both PRMT5 and PARP synergistically inhibits the growth of AML cells while proliferation of normal human CD34 positive cord blood cells is less affected at similar concentrations.22 In DLBCL patient derived primary cancer cells, targeting PRMT5 enhances the effectiveness of BCL6 inhibitors in inhibiting cell proliferation.43 Our work reveals PI3K-AKT dependent expression of PRMT5 in DLBCL.79 Genomic and biochemical analyses demonstrate that PRMT5 promotes cell cycle progression and activates PI3K-AKT signaling, suggesting a feedback regulatory mechanism to enhance cell survival and proliferation.79 Co-targeting PRMT5 and AKT by their specific inhibitors synergistically inhibits the proliferation of both DLBCL cell lines and DLBCL primary cancer cells.79

Conclusions and future directions

PRMT5 plays an important role in both the normal physiology and the development of human cancer including hematologic malignancies. Rational design of combination targeted therapies with PRMT5 inhibitors has emerged as an effective cancer treatment strategy. Our current knowledge about the mechanism by which PRMT5 activity or expression is deregulated in cancer cells is still limited. Future work should include identifying PRMT5 substrates and characterizing the pathologic role of PRMT5 in different types of human cancer. Gaining more insights into PRMT5 tumorigenesis will allow for the development of effective targeted therapeutic strategies for cancer treatment.

Conflicts of interest

The authors have no conflicts of interest to disclose.

Acknowledgement

This work was supported by the National Institutes of Health/National Cancer Institute (NIH/NCI) grant R01 CA187299.

Footnotes

Peer review under responsibility of Chongqing Medical University.

Abbreviations

AR

androgen receptor

ASK1

apoptosis signal–regulating kinase 1

ALL

acute lymphoblastic leukemia

AML

acute myeloid leukemia

AXIN2

axin related protein

BCL6

B cell lymphoma 6

CDK4

cyclin dependent kinase 4

CDKN2A

cyclin dependent kinase inhibitor 2A

CERC

cyclin E1 repressive complex

CF I m68

mammalian cleavage factor I 68kDa

ChIP

chromatin immunoprecipitation

CML

chronic myelogenous leukemia

CREB

cAMP response element binding

CRTC2

CREB regulated transcriptional coactivator 2

DLBCL

diffuse large B cell lymphoma

ABC DLBCL

activated B cell like DLBCL

GCB DLBCL

germinal center B cell like DLBCL

EBNA

Epstein–Barr virus (EBV) nuclear antigen

EGFR

epidermal growth factor receptor

ERK

extracellular signal–regulated kinases

FEN1

flap endonuclease 1

FGF-223

fibroblast growth factor 2 23kDa

G3BP1

Ras GTPase-activating protein-binding protein 1

GADD45

growth arrest and DNA damage 45

GC

germinal center

GLI1

Glioma-Associated Oncogene 1

GM130

130 kDa cis-Golgi matrix protein

GSK3β

glycogen synthase kinase 3 beta

H2AR3

histone H2A arginine 3

H3R8

histone H3 arginine 8

H3R8

histone H3 arginine 2

H4R3

histone H4 arginine 3

HCC

hepatocellular carcinoma

HCV

hepatitis C virus

HDAC

histone deacetylase

hESC

human embryonic stem cells

HOXA9

homeobox protein Hox-A3

HSPCs

hematopoietic stem and progenitor cells

HTLV-1

human T-cell leukemia virus type-1

JAK2

janus kinase 2

JBP1

jak binding protein 1

KAP1

KRAB-associated protein 1

KLF4

Krüppel-like factor 4

KRAB

Krüppel associated box

MBD2

methyl DNA binding domain protein 2

MBP

myelin basic protein

MCL

mantle cell lymphoma

MEP50

methylosome protein 50

MM

multiple myeloma

MTA

methylthioadenosine

MTAP

5-methylthioadenosine phosphorylase

PARP

poly(ADP-ribose) polymerase

PDCD4

programmed cell death 4

PDGFRα

platelet-derived growth factor receptor alpha

PI3K

phosphoinositide 3-kinases

PKA

protein kinase A

PRC2

polycomb repressor complex 2

PRMT

protein arginine methyltransfease

PUMA

p53 upregulated modulator of apoptosis

RAF

rapidly accelerated fibrosarcoma

RPS10

ribosomal protein s10

RUVBL1

RuvB-like 1

SAM

S-adenosylmethionine

SHP

small heterodimer partner

SMN

survival of motor neurons

SREBP

sterol regulatory element-binding protein

srGAPs

slit-Robo GTPase-activating proteins

TDH

L-threonine dehydrogenase

TRIM21

tripartite motif-containing protein 21

WIF1

WNT inhibitory factor 1

References

  • 1.Larsen S.C., Sylvestersen K.B., Mund A. Proteome-wide analysis of arginine monomethylation reveals widespread occurrence in human cells. Sci Signal. 2016;9(443):rs9. doi: 10.1126/scisignal.aaf7329. [DOI] [PubMed] [Google Scholar]
  • 2.Ma X.J., Lu Q., Grunstein M. A search for proteins that interact genetically with histone H3 and H4 amino termini uncovers novel regulators of the Swe1 kinase in Saccharomyces cerevisiae. Genes Dev. 1996;10(11):1327–1340. doi: 10.1101/gad.10.11.1327. [DOI] [PubMed] [Google Scholar]
  • 3.Lew D.J., Reed S.I. A cell cycle checkpoint monitors cell morphogenesis in budding yeast. J Cell Biol. 1995;129(3):739–749. doi: 10.1083/jcb.129.3.739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.McMillan J.N., Longtine M.S., Sia R.A. The morphogenesis checkpoint in Saccharomyces cerevisiae: cell cycle control of Swe1p degradation by Hsl1p and Hsl7p. Mol Cell Biol. 1999;19(10):6929–6939. doi: 10.1128/mcb.19.10.6929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Gilbreth M., Yang P., Wang D. The highly conserved skb1 gene encodes a protein that interacts with Shk1, a fission yeast Ste20/PAK homolog. Proc Natl Acad Sci U S A. 1996;93(24):13802–13807. doi: 10.1073/pnas.93.24.13802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Gilbreth M., Yang P., Bartholomeusz G. Negative regulation of mitosis in fission yeast by the shk1 interacting protein skb1 and its human homolog, Skb1Hs. Proc Natl Acad Sci U S A. 1998;95(25):14781–14786. doi: 10.1073/pnas.95.25.14781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Yamada A., Duffy B., Perry J.A., Kornbluth S. DNA replication checkpoint control of Wee1 stability by vertebrate Hsl7. J Cell Biol. 2004;167(5):841–849. doi: 10.1083/jcb.200406048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Pollack B.P., Kotenko S.V., He W., Izotova L.S., Barnoski B.L., Pestka S. The human homologue of the yeast proteins Skb1 and Hsl7p interacts with Jak kinases and contains protein methyltransferase activity. J Biol Chem. 1999;274(44):31531–31542. doi: 10.1074/jbc.274.44.31531. [DOI] [PubMed] [Google Scholar]
  • 9.Rho J., Choi S., Seong Y.R., Cho W.K., Kim S.H., Im D.S. Prmt5, which forms distinct homo-oligomers, is a member of the protein-arginine methyltransferase family. J Biol Chem. 2001;276(14):11393–11401. doi: 10.1074/jbc.M008660200. [DOI] [PubMed] [Google Scholar]
  • 10.Baldwin G.S., Carnegie P.R. Isolation and partial characterization of methylated arginines from the encephalitogenic basic protein of myelin. Biochem J. 1971;123(1):69–74. doi: 10.1042/bj1230069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Baldwin G.S., Carnegie P.R. Specific enzymic methylation of an arginine in the experimental allergic encephalomyelitis protein from human myelin. Science. 1971;171(3971):579–581. doi: 10.1126/science.171.3971.579. [DOI] [PubMed] [Google Scholar]
  • 12.Eylar E.H., Brostoff S., Hashim G., Caccam J., Burnett P. Basic A1 protein of the myelin membrane. The complete amino acid sequence. J Biol Chem. 1971;246(18):5770–5784. [PubMed] [Google Scholar]
  • 13.Branscombe T.L., Frankel A., Lee J.H. PRMT5 (Janus kinase-binding protein 1) catalyzes the formation of symmetric dimethylarginine residues in proteins. J Biol Chem. 2001;276(35):32971–32976. doi: 10.1074/jbc.M105412200. [DOI] [PubMed] [Google Scholar]
  • 14.Miranda T.B., Sayegh J., Frankel A., Katz J.E., Miranda M., Clarke S. Yeast Hsl7 (histone synthetic lethal 7) catalyses the in vitro formation of omega-N(G)-monomethylarginine in calf thymus histone H2A. Biochem J. 2006;395(3):563–570. doi: 10.1042/BJ20051771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Friesen W.J., Wyce A., Paushkin S. A novel WD repeat protein component of the methylosome binds Sm proteins. J Biol Chem. 2002;277(10):8243–8247. doi: 10.1074/jbc.M109984200. [DOI] [PubMed] [Google Scholar]
  • 16.Burgos E.S., Wilczek C., Onikubo T. Histone H2A and H4 N-terminal tails are positioned by the MEP50 WD repeat protein for efficient methylation by the PRMT5 arginine methyltransferase. J Biol Chem. 2015;290(15):9674–9689. doi: 10.1074/jbc.M115.636894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Friesen W.J., Paushkin S., Wyce A. The methylosome, a 20S complex containing JBP1 and pICln, produces dimethylarginine-modified Sm proteins. Mol Cell Biol. 2001;21(24):8289–8300. doi: 10.1128/MCB.21.24.8289-8300.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Prusty A.B., Meduri R., Prusty B.K., Vanselow J., Schlosser A., Fischer U. Impaired spliceosomal UsnRNP assembly leads to Sm mRNA down-regulation and Sm protein degradation. J Cell Biol. 2017;216(8):2391–2407. doi: 10.1083/jcb.201611108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Guderian G., Peter C., Wiesner J. RioK1, a new interactor of protein arginine methyltransferase 5 (PRMT5), competes with pICln for binding and modulates PRMT5 complex composition and substrate specificity. J Biol Chem. 2011;286(3):1976–1986. doi: 10.1074/jbc.M110.148486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Antonysamy S., Bonday Z., Campbell R.M. Crystal structure of the human PRMT5:MEP50 complex. Proc Natl Acad Sci Unit States Am. 2012;109(44):17960–17965. doi: 10.1073/pnas.1209814109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Liu F., Cheng G., Hamard P.-J. Arginine methyltransferase PRMT5 is essential for sustaining normal adult hematopoiesis. J Clin Investig. 2015;125(9):3532–3544. doi: 10.1172/JCI81749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Hamard P.-J., Santiago G.E., Liu F. PRMT5 regulates DNA repair by controlling the alternative splicing of histone-modifying enzymes. Cell Rep. 2018;24(10):2643–2657. doi: 10.1016/j.celrep.2018.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Liu F., Zhao X., Perna F. JAK2V617F-mediated phosphorylation of PRMT5 downregulates its methyltransferase activity and promotes myeloproliferation. Cancer Cell. 2011;19(2):283–294. doi: 10.1016/j.ccr.2010.12.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Sun L., Wang M., Lv Z. Structural insights into protein arginine symmetric dimethylation by PRMT5. Proc Natl Acad Sci U S A. 2011;108(51):20538–20543. doi: 10.1073/pnas.1106946108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Wang M., Fuhrmann J., Thompson P.R. Protein arginine methyltransferase 5 catalyzes substrate dimethylation in a distributive fashion. Biochemistry (Mosc) 2014;53(50):7884–7892. doi: 10.1021/bi501279g. [DOI] [PubMed] [Google Scholar]
  • 26.Fabbrizio E., Messaoudi S.E., Polanowska J. Negative regulation of transcription by the type II arginine methyltransferase PRMT5. EMBO Rep. 2002;3(7):641–645. doi: 10.1093/embo-reports/kvf136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Pal S., Vishwanath S.N., Erdjument-Bromage H., Tempst P., Sif S. Human SWI/SNF-associated PRMT5 methylates histone H3 arginine 8 and negatively regulates expression of ST7 and NM23 tumor suppressor genes. Mol Cell Biol. 2004;24(21):9630–9645. doi: 10.1128/MCB.24.21.9630-9645.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Kwak Y.T., Guo J., Prajapati S. Methylation of SPT5 regulates its interaction with RNA polymerase II and transcriptional elongation properties. Mol Cell. 2003;11(4):1055–1066. doi: 10.1016/s1097-2765(03)00101-1. [DOI] [PubMed] [Google Scholar]
  • 29.Pal S., Yun R., Datta A. mSin3A/Histone deacetylase 2- and PRMT5-containing Brg1 complex is involved in transcriptional repression of the myc target gene cad. Mol Cell Biol. 2003;23(21):7475–7487. doi: 10.1128/MCB.23.21.7475-7487.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Tae S., Karkhanis V., Velasco K. Bromodomain protein 7 interacts with PRMT5 and PRC2, and is involved in transcriptional repression of their target genes. Nucleic Acids Res. 2011;39(13):5424–5438. doi: 10.1093/nar/gkr170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Zhao Q., Rank G., Tan Y.T. PRMT5-mediated methylation of histone H4R3 recruits DNMT3A, coupling histone and DNA methylation in gene silencing. Nat Struct Mol Biol. 2009;16(3):304–311. doi: 10.1038/nsmb.1568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Barski A., Cuddapah S., Cui K. High-resolution profiling of histone methylations in the human genome. Cell. 2007;129(4):823–837. doi: 10.1016/j.cell.2007.05.009. [DOI] [PubMed] [Google Scholar]
  • 33.Girardot M., Hirasawa R., Kacem S. PRMT5-mediated histone H4 arginine-3 symmetrical dimethylation marks chromatin at G + C-rich regions of the mouse genome. Nucleic Acids Res. 2014;42(1):235–248. doi: 10.1093/nar/gkt884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Migliori V., Müller J., Phalke S. Symmetric dimethylation of H3R2 is a newly identified histone mark that supports euchromatin maintenance. Nat Struct Mol Biol. 2012;19(2):136–144. doi: 10.1038/nsmb.2209. [DOI] [PubMed] [Google Scholar]
  • 35.Yuan C.-C., Matthews A.G.W., Jin Y. Histone H3R2 symmetric dimethylation and histone H3K4 trimethylation are tightly correlated in eukaryotic genomes. Cell Rep. 2012;1(2):83–90. doi: 10.1016/j.celrep.2011.12.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Gkountela S., Li Z., Chin C.J., Lee S.A., Clark A.T. PRMT5 is required for human embryonic stem cell proliferation but not pluripotency. Stem Cell Rev Rep. 2014;10(2):230–239. doi: 10.1007/s12015-013-9490-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Hsu J.-M., Chen C.-T., Chou C.-K. Crosstalk between Arg 1175 methylation and Tyr 1173 phosphorylation negatively modulates EGFR-mediated ERK activation. Nat Cell Biol. 2011;13(2):174–181. doi: 10.1038/ncb2158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Wei H., Wang B., Miyagi M. PRMT5 dimethylates R30 of the p65 subunit to activate NF-κB. Proc Natl Acad Sci U S A. 2013;110(33):13516–13521. doi: 10.1073/pnas.1311784110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.di Caprio R., Ciano M., Montano G., Costanzo P., Cesaro E. KAP1 is a novel substrate for the arginine methyltransferase PRMT5. Biology. 2015;4(1):41–49. doi: 10.3390/biology4010041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Liu C.-D., Cheng C.-P., Fang J.-S. Modulation of Epstein–Barr virus nuclear antigen 2-dependent transcription by protein arginine methyltransferase 5. Biochem Biophys Res Commun. 2013;430(3):1097–1102. doi: 10.1016/j.bbrc.2012.12.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Shire K., Kapoor P., Jiang K. Regulation of the EBNA1 Epstein-Barr virus protein by serine phosphorylation and arginine methylation. J Virol. 2006;80(11):5261–5272. doi: 10.1128/JVI.02682-05. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Hu D., Gur M., Zhou Z. Interplay between arginine methylation and ubiquitylation regulates KLF4-mediated genome stability and carcinogenesis. Nat Commun. 2015;6:8419. doi: 10.1038/ncomms9419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Lu X., Fernando T.M., Lossos C. PRMT5 interacts with the BCL6 oncoprotein and is required for germinal center formation and lymphoma cell survival. Blood. 2018;132(19):2026–2039. doi: 10.1182/blood-2018-02-831438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Jansson M., Durant S.T., Cho E.-C. Arginine methylation regulates the p53 response. Nat Cell Biol. 2008;10(12):1431–1439. doi: 10.1038/ncb1802. [DOI] [PubMed] [Google Scholar]
  • 45.Andreu-Pérez P., Esteve-Puig R., de Torre-Minguela C. Protein arginine methyltransferase 5 regulates ERK1/2 signal transduction amplitude and cell fate through CRAF. Sci Signal. 2011;4(190):ra58. doi: 10.1126/scisignal.2001936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Cho E.-C., Zheng S., Munro S. Arginine methylation controls growth regulation by E2F-1. EMBO J. 2012;31(7):1785–1797. doi: 10.1038/emboj.2012.17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Zheng S., Moehlenbrink J., Lu Y.-C. Arginine methylation-dependent reader-writer interplay governs growth control by E2F-1. Mol Cell. 2013;52(1):37–51. doi: 10.1016/j.molcel.2013.08.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Mounir Z., Korn J.M., Westerling T. ERG signaling in prostate cancer is driven through PRMT5-dependent methylation of the Androgen Receptor. eLife. 2016:5. doi: 10.7554/eLife.13964. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Tan C.P., Nakielny S. Control of the DNA methylation system component MBD2 by protein arginine methylation. Mol Cell Biol. 2006;26(19):7224–7235. doi: 10.1128/MCB.00473-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Guo Z., Zheng L., Xu H. Methylation of FEN1 suppresses nearby phosphorylation and facilitates PCNA binding. Nat Chem Biol. 2010;6(10):766–773. doi: 10.1038/nchembio.422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.He W., Ma X., Yang X., Zhao Y., Qiu J., Hang H. A role for the arginine methylation of Rad9 in checkpoint control and cellular sensitivity to DNA damage. Nucleic Acids Res. 2011;39(11):4719–4727. doi: 10.1093/nar/gkq1264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Clarke T.L., Sanchez-Bailon M.P., Chiang K. PRMT5-Dependent methylation of the TIP60 coactivator RUVBL1 is a key regulator of homologous recombination. Mol Cell. 2017;65(5):900–916. doi: 10.1016/j.molcel.2017.01.019. e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Amente S., Napolitano G., Licciardo P. Identification of proteins interacting with the RNAPII FCP1 phosphatase: FCP1 forms a complex with arginine methyltransferase PRMT5 and it is a substrate for PRMT5-mediated methylation. FEBS Lett. 2005;579(3):683–689. doi: 10.1016/j.febslet.2004.12.045. [DOI] [PubMed] [Google Scholar]
  • 54.Zhao D.Y., Gish G., Braunschweig U. SMN and symmetric arginine dimethylation of RNA polymerase II C-terminal domain control termination. Nature. 2016;529(7584):48–53. doi: 10.1038/nature16469. [DOI] [PubMed] [Google Scholar]
  • 55.Massenet S., Pellizzoni L., Paushkin S., Mattaj I.W., Dreyfuss G. The SMN complex is associated with snRNPs throughout their cytoplasmic assembly pathway. Mol Cell Biol. 2002;22(18):6533–6541. doi: 10.1128/MCB.22.18.6533-6541.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Neuenkirchen N., Chari A., Fischer U. Deciphering the assembly pathway of Sm-class U snRNPs. FEBS Lett. 2008;582(14):1997–2003. doi: 10.1016/j.febslet.2008.03.009. [DOI] [PubMed] [Google Scholar]
  • 57.Pesiridis G.S., Diamond E., Van Duyne G.D. Role of pICLn in methylation of Sm proteins by PRMT5. J Biol Chem. 2009;284(32):21347–21359. doi: 10.1074/jbc.M109.015578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Koh C.M., Bezzi M., Low D.H.P. MYC regulates the core pre-mRNA splicing machinery as an essential step in lymphomagenesis. Nature. 2015;523(7558):96–100. doi: 10.1038/nature14351. [DOI] [PubMed] [Google Scholar]
  • 59.Gerhart S.V., Kellner W.A., Thompson C. Activation of the p53-MDM4 regulatory axis defines the anti-tumour response to PRMT5 inhibition through its role in regulating cellular splicing. Sci Rep. 2018;8(1):9711. doi: 10.1038/s41598-018-28002-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Tan D.Q., Li Y., Yang C. PRMT5 modulates splicing for genome integrity and preserves proteostasis of hematopoietic stem cells. Cell Rep. 2019;26(9):2316–2328. doi: 10.1016/j.celrep.2019.02.001. e6. [DOI] [PubMed] [Google Scholar]
  • 61.Kanamaluru D., Xiao Z., Fang S. Arginine methylation by PRMT5 at a naturally occurring mutation site is critical for liver metabolic regulation by small heterodimer partner. Mol Cell Biol. 2011;31(7):1540–1550. doi: 10.1128/MCB.01212-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Tsai W.-W., Niessen S., Goebel N., Yates J.R., Guccione E., Montminy M. PRMT5 modulates the metabolic response to fasting signals. Proc Natl Acad Sci. 2013;110(22):8870–8875. doi: 10.1073/pnas.1304602110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Yang H., Zhao X., Zhao L. PRMT5 competitively binds to CDK4 to promote G1-S transition upon glucose induction in hepatocellular carcinoma. OncoTarget. 2016;7(44):72131–72147. doi: 10.18632/oncotarget.12351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Horton J.D., Goldstein J.L., Brown M.S. SREBPs: activators of the complete program of cholesterol and fatty acid synthesis in the liver. J Clin Investig. 2002;109(9):1125–1131. doi: 10.1172/JCI15593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Liu L., Zhao X., Zhao L. Arginine methylation of SREBP1a via PRMT5 promotes de novo lipogenesis and tumor growth. Cancer Res. 2016;76(5):1260–1272. doi: 10.1158/0008-5472.CAN-15-1766. [DOI] [PubMed] [Google Scholar]
  • 66.Huang L., Liu J., Zhang X.-O. Inhibition of protein arginine methyltransferase 5 enhances hepatic mitochondrial biogenesis. J Biol Chem. 2018;293(28):10884–10894. doi: 10.1074/jbc.RA118.002377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Tee W.-W., Pardo M., Theunissen T.W. Prmt5 is essential for early mouse development and acts in the cytoplasm to maintain ES cell pluripotency. Genes Dev. 2010;24(24):2772–2777. doi: 10.1101/gad.606110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Litzler L.C., Zahn A., Meli A.P. PRMT5 is essential for B cell development and germinal center dynamics. Nat Commun. 2019;10(1):22. doi: 10.1038/s41467-018-07884-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Mei M., Zhang R., Zhou Z.-W. PRMT5-mediated H4R3sme2 confers cell differentiation in pediatric B-cell precursor acute lymphoblastic leukemia. Clin Cancer Res Off J Am Assoc Cancer Res. 2019;25(8):2633–2643. doi: 10.1158/1078-0432.CCR-18-2342. [DOI] [PubMed] [Google Scholar]
  • 70.Tarighat S.S., Santhanam R., Frankhouser D. The dual epigenetic role of PRMT5 in acute myeloid leukemia: gene activation and repression via histone arginine methylation. Leukemia. 2016;30(4):789. doi: 10.1038/leu.2015.308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Panfil A.R., Al-Saleem J., Howard C.M. PRMT5 is upregulated in HTLV-1-mediated T-cell transformation and selective inhibition alters viral gene expression and infected cell survival. Viruses. 2015;8(1) doi: 10.3390/v8010007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Jin Y., Zhou J., Xu F. Targeting methyltransferase PRMT5 eliminates leukemia stem cells in chronic myelogenous leukemia. J Clin Investig. 2016;126(10):3961–3980. doi: 10.1172/JCI85239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Chan-Penebre E., Kuplast K.G., Majer C.R. A selective inhibitor of PRMT5 with in vivo and in vitro potency in MCL models. Nat Chem Biol. 2015;11(6):432–437. doi: 10.1038/nchembio.1810. [DOI] [PubMed] [Google Scholar]
  • 74.Chung J., Karkhanis V., Tae S. Protein arginine methyltransferase 5 (PRMT5) inhibition induces lymphoma cell death through reactivation of the retinoblastoma tumor suppressor pathway and polycomb repressor complex 2 (PRC2) silencing. J Biol Chem. 2013;288(49):35534–35547. doi: 10.1074/jbc.M113.510669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Pal S., Baiocchi R.A., Byrd J.C., Grever M.R., Jacob S.T., Sif S. Low levels of miR-92b/96 induce PRMT5 translation and H3R8/H4R3 methylation in mantle cell lymphoma. EMBO J. 2007;26(15):3558–3569. doi: 10.1038/sj.emboj.7601794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Chung J., Karkhanis V., Baiocchi R.A., Sif S. Protein arginine methyltransferase 5 (PRMT5) promotes survival of lymphoma cells via activation of WNT/β-CATENIN and AKT/GSK3β proliferative signaling. J Biol Chem. 2019;294(19):7692–7710. doi: 10.1074/jbc.RA119.007640. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Li Y., Chitnis N., Nakagawa H. PRMT5 is required for lymphomagenesis triggered by multiple oncogenic drivers. Cancer Discov. 2015;5(3):288–303. doi: 10.1158/2159-8290.CD-14-0625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Alinari L., Mahasenan K.V., Yan F. Selective inhibition of protein arginine methyltransferase 5 blocks initiation and maintenance of B-cell transformation. Blood. 2015;125(16):2530–2543. doi: 10.1182/blood-2014-12-619783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Zhu F., Guo H., Bates P.D. 2019. PRMT5 is Upregulated by B-Cell Receptor Signaling and Forms a Positive-Feedback Loop with PI3K/AKT in Lymphoma Cells. Leukemia. [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Gullà A., Hideshima T., Bianchi G. Protein arginine methyltransferase 5 has prognostic relevance and is a druggable target in multiple myeloma. Leukemia. 2018;32(4):996–1002. doi: 10.1038/leu.2017.334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Kryukov G.V., Wilson F.H., Ruth J.R. MTAP deletion confers enhanced dependency on the PRMT5 arginine methyltransferase in cancer cells. Science. 2016;351(6278):1214–1218. doi: 10.1126/science.aad5214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Mavrakis K.J., McDonald E.R., Schlabach M.R. Disordered methionine metabolism in MTAP/CDKN2A-deleted cancers leads to dependence on PRMT5. Science. 2016;351(6278):1208–1213. doi: 10.1126/science.aad5944. [DOI] [PubMed] [Google Scholar]
  • 83.Duncan K.W., Rioux N., Boriack-Sjodin P.A. Structure and property guided design in the identification of PRMT5 tool compound EPZ015666. ACS Med Chem Lett. 2016;7(2):162–166. doi: 10.1021/acsmedchemlett.5b00380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Inc E. GlobeNewswire News Room; 2016. Epizyme Earns $6 Million Milestone Payment from GlaxoSmithKline for Initiation of Clinical Development with First-In-Class PRMT5 Inhibitor.http://globenewswire.com/news-release/2016/09/15/872059/0/en/Epizyme-Earns-6-Million-Milestone-Payment-from-GlaxoSmithKline-for-Initiation-of-Clinical-Development-with-First-in-Class-PRMT5-Inhibitor.html [Internet] [cited 2019 Feb 1]. Available from: [Google Scholar]
  • 85.Miranda T.B., Miranda M., Frankel A., Clarke S. PRMT7 is a member of the protein arginine methyltransferase family with a distinct substrate specificity. J Biol Chem. 2004;279(22):22902–22907. doi: 10.1074/jbc.M312904200. [DOI] [PubMed] [Google Scholar]
  • 86.Lee J.-H., Cook J.R., Yang Z.-H. PRMT7, a new protein arginine methyltransferase that synthesizes symmetric dimethylarginine. J Biol Chem. 2005;280(5):3656–3664. doi: 10.1074/jbc.M405295200. [DOI] [PubMed] [Google Scholar]
  • 87.Zurita-Lopez C.I., Sandberg T., Kelly R., Clarke S.G. Human protein arginine methyltransferase 7 (PRMT7) is a type III enzyme forming ω-NG-monomethylated arginine residues. J Biol Chem. 2012;287(11):7859–7870. doi: 10.1074/jbc.M111.336271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Feng Y., Hadjikyriacou A., Clarke S.G. Substrate specificity of human protein arginine methyltransferase 7 (PRMT7): the importance of acidic residues in the double E loop. J Biol Chem. 2014;289(47):32604–32616. doi: 10.1074/jbc.M114.609271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Chanderkar L.P., Paik W.K., Kim S. Studies on myelin-basic-protein methylation during mouse brain development. Biochem J. 1986;240(2):471–479. doi: 10.1042/bj2400471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Crang A.J., Jacobson W. The relationship of myelin basic protein (arginine) methyltransferase to myelination in mouse spinal cord. J Neurochem. 1982;39(1):244–247. doi: 10.1111/j.1471-4159.1982.tb04726.x. [DOI] [PubMed] [Google Scholar]
  • 91.DesJardins K.C., Morell P. Phosphate groups modifying myelin basic proteins are metabolically labile; methyl groups are stable. J Cell Biol. 1983;97(2):438–446. doi: 10.1083/jcb.97.2.438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Ghosh S.K., Rawal N., Syed S.K., Paik W.K., Kim S.D. Enzymic methylation of myelin basic protein in myelin. Biochem J. 1991;275(Pt 2):381–387. doi: 10.1042/bj2750381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Ancelin K., Lange U.C., Hajkova P. Blimp1 associates with Prmt5 and directs histone arginine methylation in mouse germ cells. Nat Cell Biol. 2006;8(6):623–630. doi: 10.1038/ncb1413. [DOI] [PubMed] [Google Scholar]
  • 94.Park J.H., Szemes M., Vieira G.C. Protein arginine methyltransferase 5 is a key regulator of the MYCN oncoprotein in neuroblastoma cells. Mol Oncol. 2015;9(3):617–627. doi: 10.1016/j.molonc.2014.10.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Martin G., Ostareck-Lederer A., Chari A. Arginine methylation in subunits of mammalian pre-mRNA cleavage factor I. RNA. 2010;16(8):1646–1659. doi: 10.1261/rna.2164210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Bruns A.-F., Grothe C., Claus P. Fibroblast growth factor 2 (FGF-2) is a novel substrate for arginine methylation by PRMT5. Biol Chem. 2009;390(1):59–65. doi: 10.1515/BC.2009.001. [DOI] [PubMed] [Google Scholar]
  • 97.Ren J., Wang Y., Liang Y., Zhang Y., Bao S., Xu Z. Methylation of ribosomal protein S10 by protein-arginine methyltransferase 5 regulates ribosome biogenesis. J Biol Chem. 2010;285(17):12695–12705. doi: 10.1074/jbc.M110.103911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Tsai W.-C., Gayatri S., Reineke L.C., Sbardella G., Bedford M.T., Lloyd R.E. Arginine demethylation of G3BP1 promotes stress granule assembly. J Biol Chem. 2016;291(43):22671–22685. doi: 10.1074/jbc.M116.739573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Chen M., Qu X., Zhang Z. Cross-talk between Arg methylation and Ser phosphorylation modulates apoptosis signal–regulating kinase 1 activation in endothelial cells. Mol Biol Cell. 2016;27(8):1358–1366. doi: 10.1091/mbc.E15-10-0738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Powers M.A., Fay M.M., Factor R.E., Welm A.L., Ullman K.S. Protein arginine methyltransferase 5 accelerates tumor growth by arginine methylation of the tumor suppressor programmed cell death 4. Cancer Res. 2011;71(16):5579–5587. doi: 10.1158/0008-5472.CAN-11-0458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Guo S., Bao S. srGAP2 arginine methylation regulates cell migration and cell spreading through promoting dimerization. J Biol Chem. 2010;285(45):35133–35141. doi: 10.1074/jbc.M110.153429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Calabretta S., Vogel G., Yu Z. Loss of PRMT5 promotes PDGFRα degradation during oligodendrocyte differentiation and myelination. Dev Cell. 2018;46(4):426–440. doi: 10.1016/j.devcel.2018.06.025. e5. [DOI] [PubMed] [Google Scholar]
  • 103.Bandyopadhyay S., Harris D.P., Adams G.N. HOXA9 methylation by PRMT5 is essential for endothelial cell expression of leukocyte adhesion molecules. Mol Cell Biol. 2012;32(7):1202–1213. doi: 10.1128/MCB.05977-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Zhou Z., Sun X., Zou Z. PRMT5 regulates Golgi apparatus structure through methylation of the golgin GM130. Cell Res. 2010;20(9):1023–1033. doi: 10.1038/cr.2010.56. [DOI] [PubMed] [Google Scholar]
  • 105.Vagin V.V., Wohlschlegel J., Qu J. Proteomic analysis of murine Piwi proteins reveals a role for arginine methylation in specifying interaction with Tudor family members. Genes Dev. 2009;23(15):1749–1762. doi: 10.1101/gad.1814809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Han C., Gu H., Wang J., Lu W., Mei Y., Wu M. Regulation of L-threonine dehydrogenase in somatic cell reprogramming. Stem Cells Dayt Ohio. 2013;31(5):953–965. doi: 10.1002/stem.1335. [DOI] [PubMed] [Google Scholar]
  • 107.Abe Y., Suzuki Y., Kawamura K., Tanaka N. MEP50/PRMT5-mediated methylation activates GLI1 in Hedgehog signalling through inhibition of ubiquitination by the ITCH/NUMB complex. Commun Biol. 2019;2(1):23. doi: 10.1038/s42003-018-0275-4. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Genes & Diseases are provided here courtesy of Chongqing Medical University

RESOURCES