MECOM (Ecotropic Viral Integration Site 1 (EVI1) and Myelodysplastic Syndrome 1 (MDS1-EVI1)

2015-05-01   Rotraud Wieser 

Identity

HGNC
LOCATION
3q26.2
IMAGE
Atlas Image
LEGEND
Genomic locus of the human EVI1 gene, and EVI1 and MDS1/EVI1 mRNA variants. Asterisk, translation initiation codon in EVI1 exon 3; diamond, translation stop codon in EVI1 exon 16. (This figure was reprinted from Gene 396, R. Wieser, The oncogene and developmental regulator EVI1: Expression, biochemical properties, and biological functions, pages 346-357, Copyright Elsevier (2007), with permission from Elsevier. Gene homepage: http://www.sciencedirect.com/science/journal/03781119)
IMAGE
Atlas Image
LEGEND
MECOM (Ecotropic Viral Integration Site 1 (EVI1) and Myelodysplastic Syndrome 1 (MDS1-EVI1) Hybridization with MECOM (EVI1) break apart probe (Kreatech, Leica Biosystems Inc., US) showing the gene on 3q26.2 (red-green or a fused yellow signal) - Courtesy Adriana Zamecnikova
LOCUSID
ALIAS
AML1-EVI-1,EVI1,KMT8E,MDS1,MDS1-EVI1,PRDM3,RUSAT2
FUSION GENES

Abstract

The MECOM locus in chromosome band 3q26.2 gives rise to two major mRNA and protein species, EVI1 and MDS1\/EVI1, of which the former has been characterized far more extensively. EVI1 has been implicated in the maintenance and expansion of normal hematopoietic stem cells. In addition, it plays a role as an oncogene in myeloid leukemia and in certain solid tumors, its overexpression being associated with a dismal prognosis in several of these malignancies. EVI1 exerts its biological effects mainly by regulating gene transcription, and does so by acting as a sequence specific transcription factor, by modulating the activity of other sequence specific transcription factors, and by regulating promoter CpG island methylation.

DNA/RNA

Description

The human EVI1 gene spans approximately 65 kb of genomic DNA. 14 of its 16 exons are coding (Fig. 1A). Transcription can initiate from alternative exons 1a, 1b, 1c, 1d, or 3L (Fig. 1B), and several alternative splice variants of the EVI1 mRNA have been described (Delta324, -Rp9, Delta105; Fig. 1A).
The human MDS1 gene consists of 4 exons spread over a genomic region of more than 500 kb. MDS1 exon 4 is located less than 2 kb upstream of EVI1 exon1a. MDS1 can also be expressed on its own. The MDS1/EVI1 mRNA presumably results from splicing of the second exon of MDS1 to the second exon of EVI1 (Fig. 1B).

Transcription

Telomere to centromere.

Proteins

Atlas Image
EVI1 and MDS1/EVI1 protein domains. Black boxes, zinc finger motifs; RD, repression domain; ac, acidic region; PR, PR domain. This figure was reprinted in modified form from Gene 396, R. Wieser, The oncogene and developmental regulator EVI1: Expression, biochemical properties, and biological functions, pages 346-357, Copyright Elsevier (2007), with permission from Elsevier. Gene homepage: http://www.sciencedirect.com/science/journal/03781119.

Description

Exon 3 of the human EVI1 gene contains two closely spaced ATG codons, either of which may serve as the translation initiation site. Depending on which ATG is used, proteins of 1051 or 1041 amino acids will be formed. EVI1 contains two domains of seven and three zinc finger motifs, respectively, a repression domain between the two sets of zinc fingers, and an acidic domain at its C-terminus. It is a 145 kDa protein that is capable of binding to DNA in a sequence specific manner, and that interacts with transcriptional coactivators and corepressors as well as other sequence specific transcription factors. DNA binding and transcriptional regulation by EVI1 are influenced by posttranslational modifications like phosphorylation, acetylation, and sumoylation (Chakraborty et al, 2001; Shimahara et al, 2010; Bard-Chapeau et al, 2013; Singh et al, 2013; White et al, 2013).
Predicted translation of MDS1-EVI1 adds 188 amino acids to the N-terminus of EVI1. 63 of these additional amino acids are encoded by exon 2 and beginning of exon 3 of EVI1, and the remaining 125 from the MDS1 gene. MDS1-EVI1 contains a PR domain, which is about 40% homologous to the N-terminus of the retinoblastoma-binding protein, RIZ, and the PRDI-BF1 transcription factor. Some biological functions of MDS1/EVI1 were reported to be different from, or even antagonistic to, those of EVI1, while in other cases, EVI1 and MDS1/EVI1 acted in a similar manner. MDS1/EVI1 (PRDM3) has H3K9me1 methyltransferase activity and a role in maintaining heterochromatin integrity (Pinheiro et al, 2012).

Expression

Among human tissues/organs, the EVI1 mRNA is expressed abundantly in kidney, lung, pancreas, stomach, ovaries, uterus, and prostate, to a lesser extent in the small intestine, colon, thymus, spleen, heart, brain, testis, and placenta, and at very low levels in skeletal muscle and bone marrow. The pattern of expression of MDS1-EVI1 is very similar to that of EVI1.
In the adult mouse, the Evi1 mRNA is expressed, at varying levels, in the kidney, lung, stomach, ovary, uterus, intestine, thymus, spleen, heart, brain, and liver. In the mouse embryo, Evi1 mRNA levels are high in the urinary system and Mullerian ducts, the lung, the heart, and the emerging limb buds.
Similar Evi1 expression patterns were also observed in Xenopus, chicken, and zebrafish.
In human and murine hematopoiesis, EVI1 mRNA levels are high in the most immature cell populations and decline in the course of differentiation (Kataoka et al, 2011; Bindels et al, 2012; Steinleitner et al, 2012).
EVI1 expression is regulated by RUNX1 and ELK1, by retinoic acid via RAR/RXR, and by certain MLL fusion proteins (Bingemann et al, 2009; Arai et al, 2011; Maicas et al, 2013).

Localisation

Nuclear; in part in speckles.

Function

Because of the spatially and temporally restricted expression of MECOM, it has been suggested that this gene plays important roles in development and could be involved in organogenesis, cell migration, cell growth, and differentiation.
In the mouse, homozygous disruption of the 6th exon of the Evi1 gene led to embryonic lethality, with widespread hypocellularity, reduced body size, small or absent limb buds, a pale yolk sac and placenta, abnormal development of the nervous system and the heart, and massive haemorrhaging. (Hoyt et al, 1997). Functions of Evi1 and/or Mds1/Evi1 in heart development, spine formation, and, particularly, maintenance and expansion of hematopoietic stem cells (HSCs) have been deduced from, or confirmed through, the phenotypes of additional MECOM knockout models (Goyama et al, 2008; Kataoka et al, 2011; Zhang et al, 2011; Bard-Chapeau et al, 2014; Juneja et al, 2014). A role of Evi1 in HSCs was also corroborated through overexpression and gene marking experiments (Buonamici et al, 2004; Laricchia-Robbio et al, 2008; Dickstein et al, 2010; Kataoka et al, 2011).
Support for a role of Evi1 as a leukemia initiating and promoting oncogene has been obtained through mouse bone marrow transduction/transplantation models, in which overexpression of Evi1 alone caused a myelodysplastic syndrome (MDS) like disease, while its co-expression with other oncogenes led to AML (Buonamici et al, 2004; Jin et al, 2007; Watanabe-Okochi et al, 2008; Watanabe-Okochi et al, 2013). Even more compellingly, in a human gene therapy trial for chronic granulomatous disease, activating integrations of the therapeutic vector into the MECOM locus led to clonal expansion with progression to MDS and, ultimately, AML (Stein et al, 2010). Evi1 was proposed to be essential for AML leukemia stem cell (LSC) function since its experimental down-regulation reduced leukemogenicity in several mouse models of AML (Goyama et al, 2008; Bindels et al, 2012). Evi1 expression was also associated with leukemia initiating capacity in chronic myeloid leukemia (CML) (Sato et al, 2014). A prominent role of EVI1 in therapy resistance was suggested by a number of clinical trials, and illustrated by in vitro data demonstrating that its ectopic expression reduced, and its knockdown enhanced, cellular responsiveness to chemotherapeutic drugs (Bindels et al, 2012; Konantz et al, 2012; Yamakawa et al, 2012; Rommer et al, 2013).
Beyond its roles in normal and malignant hematopoiesis, EVI1 negatively regulated NF-kB dependent inflammation (Xu et al, 2012) and promoted adipocyte differentiation (Ishibashi et al, 2012).
EVI1 exerts its biological functions mainly by acting as a transcription factor. and regulates the expression of both protein coding and miRNA genes. Reported direct EVI1 target genes are MS4A3 (Heller et al, 2015), PLZF (Takahashi and Licht, 2002), Gata2 (Yuasa et al; 2005), Pbx1 (Shimabe et al, 2009), Pten (Yoshimi et al, 2011), Gpr56 (Saito et al, 2013), DeltaNp63 (Nayak et al, 2013), Bcl-xL (Pradhan et al, 2011), Calreticulin (Qiu et al, 2008), Ppargamma2 (Ishibashi et al, 2012), miR-1-2 (Gomez-Benito et al, 2010), miR-9 (Senyuk et al, 2013), miR-124 (Dickstein et al, 2010), and miR-449A (De Weer et al, 2011). ChIP-seq, combined with genome wide gene expression profiling, has been employed for large-scale identification of EVI1 target genes in ovarian cancer and murine myeloid cell lines (Bard-Chapeau et al, 2012; Glass et al, 2013). EVI1 associates with a number of transcriptional cofactors like HDAC1 (Vinatzer et al, 2001), CtBP1 (Palmer et al, 2001), CtBP2 (Turner and Crossley, 1998), CBP, P/CAF (Chakraborty et al, 2001), the histone methyl transferases SUV39H1 and G9a (Spensberger et al, 2008a; Goyama et al, 2010), the ATP dependent helicases BRG1 and BRM (Chi et al, 2003), and the member of histone deacetylase complex, Mbd3b (Spensberger et al, 2008b). EVI1 was also shown to interact with DNA methyl transferases (Lugthart et al, 2011; Senyuk et al, 2011), leading to methylation of CpG islands of some of its target genes, among them, CADM1 (Fisser et al, 2014), miR-9 (Senyuk et al, 2013), and miR-124 (Dickstein et al, 2010). Furthermore, EVI1 interacted with, and modulated the function of, other sequence specific transcription factors, e.g. GATA1 (Laricchia-Robbio et al, 2006), RUNX1/AML1 (Senyuk et al, 2007), PU.1 (Laricchia-Robbio et al, 2009), SMAD3 (Kurokawa et al, 1998; Izutsu et al, 2001; Alliston et al, 2005), FOS (Bard-Chapeau et al, 2012), NFkB (Xu et al, 2012), and RAR/RXR (Bingemann et al, 2009; Steinmetz et al, 2014).
In addition to its activity as a regulator of transcription, EVI1 has been reported to inhibit c-jun N-terminal kinase (Kurokawa et al, 2000), and to stimulate PI3K/AKT signalling (Liu et al, 2006; Yoshimi et al, 2011).

Homology

EVI1 orthologs are present in many species. EVI1 proteins from other mammals share more than 90% amino acid sequence identity with the human protein, and Xenopus EVI1 is 77% identical to its human counterpart. MDS1-EVI1 shares an overall homology with the C. elegans Egl 43 protein that includes the PR domain at the N-terminus and the two zinc-finger domains. An MDS1/EVI1 ortholog, hamlet, is also present in Drosophila.

Implicated in

Note
3q21q26 syndrome. Chromosomal rearrangements located either 5 or 3 of the EVI1 gene can activate its transcription in haematopoietic cells. t(3;3)(q21;q26) breakpoints are often located 5 of EVI1, and inv(3)(q21q26) breakpoints 3 of it. In either case, transcription of EVI1 is activated by the distal enhancer of the GATA2 gene that is located in 3q21 (Gröschel et al, 2014; Yamazaki et al, 2014).
Prognosis
Patients with EVI1 rearrangements have elevated platelet counts, marked hyperplasia with dysplasia of megakaryocytes, and a poor prognosis.
Cytogenetics
Rearrangements at 3q26 may occur as a sole anomaly, but are often associated with monosomy 7 or deletion of the long arm of chromosome 7, and, less frequently, deletion in chromosome 5.
Atlas Image
Normal and leukemia-associated EVI1 protein variants.
Oncogenesis
Inappropriate expression of EVI1 in haematopoietic cells alters differentiation into granulocytes, erythrocytes and megakaryocytes. EVI1 promotes the proliferation of certain cell types, but inhibits the growth of others. It interferes with growth inhibition by TGF-b and with apoptosis elicited by a variety of stimuli. In a murine bone marrow transduction/transplantation model, experimental expression of EVI1 caused a disease resembling human myelodysplastic syndrome (Buonamici et al, 2004), while its coexpression with Hoxa9 and Meis 1, a mutated RUNX1 gene, or LIP led to overt leukemia (Jin et al, 2007; Watanabe-Okochi et al, 2008; Watanabe-Okochi et al, 2013).
Entity name
Disease
CML during the blast crisis and MDS in transformation.
Prognosis
Poor.
Cytogenetics
Complex.
Fusion protein
Overexpression of a fusion protein between the amino terminus of ETV6, which does not contain any functional domains, and the entire MDS1/EVI1 protein is driven by the ETV6 promoter.
Entity name
Disease
Therapy-related MDS/AML and CML during the blast crisis.
Prognosis
Poor.
Cytogenetics
Complex.
Fusion protein
RUNX1 /MDS1/EVI1
Oncogenesis
RUNX1/MDS1/EVI1 is a chimeric transcription factor that interferes with RUNX1 functions in a dominant negative manner, but shares some biological effects with EVI1.
Entity name
Various cancers with known fusion genes
Cytogenetics
Other translocations involving MECOM at 3q26 have been found:
t(2;3)(p16;q26) BCL11A/MECOM in acute myeloid leukemia
t(3;21)(q26;q21) NRIP1/MECOM in acute myeloid leukemia (Haferlach et al., 2012) t(3;3)(q21;q26) LINC01565/MECOM in acute myeloid leukemia (Pekarsky et al., 1997)
t(3;7)(q26;q21) MECOM/CDK6 in myeloid leukemias
t(3;7)(q26;q34) TRB/MECOM in acute myeloid leukemia (M0-AML) (Suzukawa et al., 1999)
t(3;15)(q26;p13) MECOM/FRMD5 in lung adenocarcinoma (Yoshihara et al., 2014)
Entity name
AML without 3q26 rearrangements.
Disease
EVI1 may also be overexpressed in AML, MDS, or CML in blast crisis in the absence of any cytogenetically detectable 3q26 rearrangements.
Prognosis
Poor (AML).
Entity name
Acute lymphoblastic leukemia
Note
Overexpression of EVI1 reported in some cases (Konantz et al, 2013; Su et al, 2015)
Entity name
Chronic lymphoblastic leukemia
Prognosis
Low EVI1 expression levels are associated with shorter survival (Vasyutina et al, 2015).
Entity name
Fanconi anemia
Note
Overexpression of EVI1 (Meyer et al, 2007; Meyer et al, 2011)
Entity name
Ovarian cancer
Note
Overexpression of EVI1
Prognosis
Poor (Nanjundan et al, 2007)
Entity name
Estrogen receptor negative breast cancer
Note
Overexpression of EVI1
Prognosis
Poor (Patel et al, 2010)
Entity name
Infratentorial ependymoma
Note
Overexpression of EVI1
Prognosis
Poor (Koos et al, 2011)
Entity name
Pancreatic cancer
Note
Overexpression of EVI1 (Tanaka et al, 2014)
Entity name
Colon cancer
Note
Overexpression of EVI1 (Deng et al, 2013)

Breakpoints

Atlas Image

Bibliography

Pubmed IDLast YearTitleAuthors

Other Information

Locus ID:

NCBI: 2122
MIM: 165215
HGNC: 3498
Ensembl: ENSG00000085276

Variants:

dbSNP: 2122
ClinVar: 2122
TCGA: ENSG00000085276
COSMIC: MECOM

RNA/Proteins

Gene IDTranscript IDUniprot
ENSG00000085276ENST00000264674Q03112
ENSG00000085276ENST00000433243A0A0C3SFZ7
ENSG00000085276ENST00000460814E7EUL6
ENSG00000085276ENST00000460890E7EPY2
ENSG00000085276ENST00000464456Q03112
ENSG00000085276ENST00000466623A0A1B0GXI8
ENSG00000085276ENST00000468789Q03112
ENSG00000085276ENST00000472280A0A0C3SFZ7
ENSG00000085276ENST00000475754E7EU48
ENSG00000085276ENST00000481315E9PGE9
ENSG00000085276ENST00000484519E7ERX0
ENSG00000085276ENST00000486748C9JU02
ENSG00000085276ENST00000487503E7EPY2
ENSG00000085276ENST00000492586H9KVD4
ENSG00000085276ENST00000494292Q03112
ENSG00000085276ENST00000494597E7EPY2
ENSG00000085276ENST00000628990Q03112
ENSG00000085276ENST00000651503Q03112

Expression (GTEx)

0
5
10
15
20
25
30
35

Pathways

PathwaySourceExternal ID
MAPK signaling pathwayKEGGko04010
Chronic myeloid leukemiaKEGGko05220
MAPK signaling pathwayKEGGhsa04010
Pathways in cancerKEGGhsa05200
Chronic myeloid leukemiaKEGGhsa05220
Chromatin organizationREACTOMER-HSA-4839726
Chromatin modifying enzymesREACTOMER-HSA-3247509
PKMTs methylate histone lysinesREACTOMER-HSA-3214841

Protein levels (Protein atlas)

Not detected
Low
Medium
High

References

Pubmed IDYearTitleCitations
165829162006Correction of X-linked chronic granulomatous disease by gene therapy, augmented by insertional activation of MDS1-EVI1, PRDM16 or SETBP1.404
200984312010Genomic instability and myelodysplasia with monosomy 7 consequent to EVI1 activation after gene therapy for chronic granulomatous disease.279
247037112014A single oncogenic enhancer rearrangement causes concomitant EVI1 and GATA2 deregulation in leukemia.173
205121452010A genome-wide association study of nasopharyngeal carcinoma identifies three new susceptibility loci.133
229396222012Prdm3 and Prdm16 are H3K9me1 methyltransferases required for mammalian heterochromatin integrity.102
182728132008High EVI1 levels predict adverse outcome in acute myeloid leukemia: prevalence of EVI1 overexpression and chromosome 3q26 abnormalities underestimated.73
123933832003High EVI1 expression predicts poor survival in acute myeloid leukemia: a study of 319 de novo AML patients.65
203796142010Personalized smoking cessation: interactions between nicotine dose, dependence and quit-success genotype score.62
203086562010High EVI1 expression predicts outcome in younger adult patients with acute myeloid leukemia and is associated with distinct cytogenetic abnormalities.61
247039062014A remote GATA2 hematopoietic enhancer drives leukemogenesis in inv(3)(q21;q26) by activating EVI1 expression.60

Citation

Rotraud Wieser

MECOM (Ecotropic Viral Integration Site 1 (EVI1) and Myelodysplastic Syndrome 1 (MDS1-EVI1)

Atlas Genet Cytogenet Oncol Haematol. 2015-05-01

Online version: http://atlasgeneticsoncology.org/gene/19/css/lib/deep-insight-explorer/

Historical Card

2007-12-01 MECOM (Ecotropic Viral Integration Site 1 (EVI1) and Myelodysplastic Syndrome 1 (MDS1-EVI1) by  Rotraud Wieser 

2003-05-01 MECOM (Ecotropic Viral Integration Site 1 (EVI1) and Myelodysplastic Syndrome 1 (MDS1-EVI1) by  Soumen Chakraborty,Silvia Buonamici,Vitalyi Senyuk,Giuseppina Nucifora 

Dept. of Pathology(Rm.3314), Molecular Biology, Research Building University Of Illinois At Chicago 900 South Ashland Avenue Chicago, IL-60607, USA