RASSF2 (Ras association (RalGDS/AF-6) domain family member 2)

2009-08-01   Luke B Hesson , Farida Latif 

Identity

HGNC
LOCATION
20p13
LOCUSID
ALIAS
CENP-34,RASFADIN
FUSION GENES

DNA/RNA

Atlas Image
Figure 1: RASSF2 gene structure. Transcription of the RASSF2A isoforms begins within a CpG island that spans exons 1 and 2 (grey box) at -105 to +1745 bp relative to the transcription start site of NM_014737. RASSF2B transcription begins at exon 1beta in intron 5. Transcription of RASSF2C begins at exon 1gamma in intron 2. RASSF2B and RASSF2C do not have 5 CpG islands or predicted promoter regions.

Description

The RASSF2 gene occupies 43,621bp of genomic DNA (-ve strand). RASSF2A variant 1 [GenBank:NM_014737] contains 12 exons and is transcribed from a large (1,850bp) 5 CpG island encompassing the first two non-coding exons. There is evidence of multiple splice variants and transcription initiation sites for the RASSF2 gene. Additional isoforms of RASSF2 include RASSF2A variant 2 [GenBank:CR627436] that is predicted to produce an identical protein to RASSF2A variant 1, RASSF2B [GenBank:AY154471] and RASSF2C [GenBank:AY154472]. A further isoform [GenBank:CR620887] produces a non-coding mRNA. RASSF2B and RASSF2C are not associated with CpG islands (figure 1). Akino et al., (2005) investigated promoter activity of the region upstream of the transcription start site of NM_014737 (RASSF2A variant 1) and found promoter activity was dependent on a CACCC box and SP1 site just upstream of exon 1. The authors however did not investigate the CpG island region of the RASSF2 gene, which is largely located in intron 1 of the NM_014737 transcript.

Proteins

Atlas Image
Figure 2: RASSF2 transcript and protein structure. RASSF2A [GenBank:NP_055552] is a 326 aa protein containing a central bipartite nuclear localisation signal (NLS), a Ras-association (RA) domain of the RalGDS/AF-6 variety and acidic coiled-coil Sav/RASSF/Hpo (SARAH) domain. RASSF2B [GenBank:AAN59976] is a predicted 157 amino acid protein containing a truncated RA domain. The RASSF2C predicted protein is identical to RASSF2A. The mRNA transcript shown (red bar) represents RASSF2A variant 1 (NM_014737).

Description

As mentioned above multiple isoforms are expressed from the RASSF2 locus. However, RASSF2A variants 1 and 2, as well as RASSF2C (if expressed at all) contain identical open reading frames encoding a 326 amino acid protein, whilst RASSF2B mRNA is predicted to encode a truncated protein of 157 amino acids but is expressed at extremely low levels in all tissues analysed. Therefore, the protein is simply referred to as RASSF2 in the literature. The RASSF2 protein (figure 2) contains C-terminal Ras-association (RA) and Sav/RASSF/Hpo (SARAH) domains that define the classical RASSF family (RASSF1, RASSF2, RASSF3, RASSF4, RASSF5, RASSF6). In addition RASSF2 contains a central bipartite nuclear localisation signal (NLS) which has been shown to be essential for tumour suppressor function (Cooper et al., 2008). The C-terminus of RASSF2 also contains a sequence shown to be necessary for nuclear export (Kumari et al., 2009). Detection of endogenous RASSF2 protein has been described in a variety of cell lines using an in-house antibody (Vos et al., 2003) or a commercially available antibody from Santa Cruz (Cooper et al., 2009).
Atlas Image
Figure 3: RASSF2 is conserved with RASSF paralogues. Schematic representation of the 10 human members of the RASSF family showing the Ras-association, SARAH and predicted diacylglycerol binding domains. The longest isoform for each RASSF gene is represented (GenBank accession numbers: RASSF1A[NP_009113], RASSF2[NP_055552], RASSF3[NP_835463], RASSF4[NP_114412], RASSF5A[NP_872604], RASSF6[NP_958834], RASSF7[NP_003466], RASSF8[BAC98838], RASSF9[NP_005438], RASSF10[NP_001073990]). The RASSF family is subdivided into classical RASSF members and N-terminal RASSF members as indicated. Shown is the percentage amino acid identity of each RASSF members with RASSF2. Protein sequence identity of the classical RASSF members is greatest over the C-terminus. * RASSF10 protein sequence as described in Hesson et al., 2009 (which is N-terminally truncated with respect to GenBank accession number NP_001073990).

Expression

Northern blotting shows RASSF2 mRNA is highly expressed in many normal tissues including brain, thymus, spleen, liver, small intestines, placenta, lung and peripheral blood (Vos et al., 2003). The probe used for northern blotting did not discriminate between RASSF2 isoforms. The coding region of RASSF2 has been cloned from a brain-specific cDNA library (Hesson et al., 2005). Currently there has been limited analysis of expression patterns and distribution of the different RASSF2 isoforms. RASSF2A variants 1 and 2 are both ubiquitously expressed in a range of normal tissues including colon, stomach, heart, bone marrow, kidney, ovary, lung, liver, breast, testis and pancreas (Maruyama et al., 2008). However, expression of the RASSF2B and RASSF2C isoforms was virtually undetectable in a range of normal tissues (Maruyama et al., 2008; L Hesson and F Latif, unpublished observations). Expression of the RASSF2 gene is inactivated by DNA methylation of the 5 CpG island promoter region in a broad spectrum of cancers (see below).

Localisation

When over expressed RASSF2 is clearly predominantly nuclear, as demonstrated by immunofluorescence (Cooper et al., 2008; Kurnari et al., 2007). Some evidence suggests that the NLS of RASSF2 is an integral part of the ability of RASSF2 to act as a tumour suppressor. The localisation of RASSF2 is cell context specific. Two independent studies indicate that phosphorylation of RASSF2 appears to be critical for relocalisation to the cytoplasm, though the critical phosphorylation sites remain to be determined. Cooper et al., (2009) demonstrated that relocalisation of over expressed RASSF2 from the nucleus to the cytoplasm is dependent on active MST1 or MST2 and that either kinase was capable of phosphorylating RASSF2 in vitro. However, the work of Kumari et al., (2009) demonstrates that RASSF2 relocalisation is dependent on the activity of Extracellular signal-Related Kinase 2 (ERK2). Both MSTs and ERK2 can participate in Ras signalling therefore both studies may be observing the effects of activation of the same pathway. The presence of sequences essential for both nuclear import and export within RASSF2 seems to suggest that the protein may continuously cycle between cytoplasm and nucleus in a similar manner to MST1 and MST2 (Lee and Yonehara, 2002). Given the strong binding of RASSF2 with MST1 and MST2 (see below) it seems likely that this would occur in complex with MSTs. RASSF2 nuclear important may be dependent on importin-alpha interaction (Kumari et al., 2007), whilst nuclear export appears to involve the NES (nuclear export signal)-dependent transport protein CRM-1/XPO1 (Kumari et al., 2009). What remains to be determined is the exact conditions under which the kinetics of nuclear export predominates nuclear import and vice versa.

Function

Tumour suppressor function of RASSF2
Similar to several other RASSF members RASSF2 suppresses tumour growth when expressed. This has been demonstrated for colorectal, lung, breast, gastric, nasopharyngeal and oral squamous cell carcinoma (OSCC) cell lines in vitro using colony formation, growth curve and soft agar growth assays (Akino et al., 2005; Vos et al., 2003; Cooper et al., 2008; Maruyama et al., 2008; Imai et al., 2008; Zhang et al., 2006). Furthermore, RASSF2 re-expression in breast tumour cells inhibits in vivo tumour growth when cells are subcutaneously injected into severe combined immunodeficiency (SCID) mice (Cooper et al., 2008). Several studies demonstrate that these tumour suppressive properties are likely to arise from the ability of RASSF2 to regulate apoptosis and cell cycle progression (Vos et al., 2003; Maruyama et al., 2008; Imai et al., 2008; Akino et al., 2005). In breast cancer cells and Cos-7 cells growth suppression by RASSF2 is dependent on the nuclear localisation signal (NLS) located at amino acids 151-167 (Cooper et al., 2008; Kumari et al., 2009), whilst other reports have indicated that in OSCC and gastric cancer cells the C-terminal portion of RASSF2 (RASSF2 [163-326]), containing the RA domain, is critical for tumour suppressive function (Imai et al., 2008; Maruyama et al., 2008). Interestingly, in OSCC this C-terminal portion exhibited enhanced growth suppression relative to full length RASSF2 (Imai et al., 2008). In fact, RASSF2 [163-326] also disrupts the NLS yet leaves the sequence required for nuclear export intact. In a separate study of colorectal cancer cells both RASSF2 truncations (RASSF2 [1-163] and RASSF2 [163-326]) exhibited reduced growth suppression compared to full length RASSF2 (Akino et al., 2005). Whilst in gastric cancer transfection of RASSF2 with deletion of the NLS [RASSF2deltaNLS] actually increased the percentage of apoptotic cells relative to full length RASSF2 (Maruyama et al., 2008). These studies indicate the growth suppressive properties of RASSF2 are likely cell background specific but more importantly that nuclear import, nuclear export and the Ras-association domain are required for correctly regulated RASSF2 growth suppression.

RASSF2 interaction with Ras
RASSF2 contains a functional RA domain that displays a strong binding to K-Ras, but only weak binding to H-Ras (Vos et al., 2003). RASSF2 associates with the K-Ras effector domain in a GTP-dependent manner thus displaying the basic properties of a genuine Ras effector. RASSF2 growth inhibition is enhanced in the presence of K-RasG12V. Furthermore, siRNA-mediated knock-down of RASSF2 in K-Ras transformed cells enhanced anchorage-independent growth. However, in the absence of K-Ras transformation knock-down of RASSF2 inhibited growth (Akino et al., 2005). These data indicate that RASSF2 mediates some of the growth inhibitory properties of K-Ras and that inactivation of RASSF2 enhances K-Ras-induced transformation.

RASSF2 interaction with the proapoptotic kinases MST1 and MST2
How the interaction of RASSF proteins with Ras results in growth suppression has been the subject of intense investigation. The proapoptotic mammalian Serine/Threonine kinases MST1 and MST2 were identified as RASSF2 interacting partners by yeast two-hybrid (Y2-H) (Khokhlatchev et al., 2002). RASSF1 and RASSF5 were also identified as MST binding partners as part of a novel Ras-regulated signalling pathway. Recently the interaction of RASSF2 and MST1/2 was formally demonstrated in human cells at the endogenous level. Interaction occurs between the SARAH domains found within RASSF2 and MST1/2 (Cooper et al., 2009). RASSF2 appears to have a distinct role in regulating MST2 function. Activation of MST2 is followed by a rapid proteasome-dependent loss of MST2 stability (that is not associated with MST2 cleavage). Interestingly, over expression of RASSF2 results in increased levels of MST2 and provides protection of MST2 from degradation following its activation. In agreement with this loss of RASSF2 protein in colorectal tumours, or in colorectal tumour cell lines in which RASSF2 levels are decreased by shRNAi, also leads to decreases in MST2 levels (Cooper et al., 2009). RASSF2 appears to be a substrate for MST1 and MST2 and co-expression of either kinase with RASSF2 relocalises RASSF2 from the nucleus to the cytoplasm in a manner dependent on kinase activity (Cooper et al., 2009). Since MST2 remains in complex with RASSF2 following its activation these data collectively suggest RASSF2 stabilises active MST2 allowing (or perhaps even targeting) MST2 substrate phosphorylation. This RASSF2-mediated stabilisation also appears to be true for MST1. Thus loss of RASSF2, as is frequently observed in cancer, leads to loss of MST1 and MST2 leading to a decrease in apoptotic potential. That RASSF2 appears to be capable of influencing MST stability so drastically is likely to be due to the observation that the majority of both MST1 and MST2 are in complex with RASSF2 in at least some cell types (Cooper et al., 2009). The interaction of RASSF2 with MST1/2 poses an interesting question with regards to the regulation of RASSF2 localisation. Both RASSF2 and MST1/2 have been shown to contain sequences essential for nuclear import and export (Lee and Yonehara, 2002; Kumari et al., 2009). Both the NLS and NES sequences within MST1/2 and RASSF2 respectively are located very close to or within the SARAH domains and neither are canonical NLS/NES sequences. Mapping of both these sequences were determined by deletion mapping, which would most likely also affect RASSF2-MST1/2 interaction thus it now seems likely that the RASSF2-MST1/2 complex constantly cycles between the nucleus (by virtue of RASSF2 NLS) and cytoplasm (by virture of MST1/2 NES) and disruption of the interaction between RASSF2 and MST1/2 would likely affect the localisation of both proteins. Also, the fact that RASSF2 translocation to the cytoplasm is dependent on ERK2 activity (Kumari and Mahalingam, 2009) suggests the Ras-MEK-ERK pathway may serve to phosphorylate MST1/2, which then phosphorylates RASSF2, translocating it to the cytoplasm and allowing RASSF2 to interact with Ras (figure 4). That nuclear RASSF2 is required for full tumour suppressor activity (Cooper et al., 2008; Kumari et al., 2009) may be explained by the fact that ERK2 translocates to the nucleus upon its activation (Khokhlatchev et al., 1998).

Other functions of RASSF2
Other functions and interacting partners of RASSF2 are extremely likely. Y2-H using RASSF2 as bait implicates NORE1A and RASSF3 in RASSF2 function, although these have not yet been confirmed in mammalian cells (Hesson et al., 2005). These interactions may implicate other RASSF members in modulating RASSF2 function and suggests a complex network of cross-talk between signalling pathways involving RASSF proteins. Also, the exact mechanisms of apoptotic and cell cycle regulation of RASSF2 have yet to be completely defined. Microarray analysis of gene expression before and after exogenous expression of RASSF2 in gastric and OSCC cancer cell lines showed RASSF2 downregulates expression of several inflammatory response genes including the cytokines IL-8, LCN2, CXCL1, CXCL2, CXCL3, CXCL5 and CXCL6, CCL20 and CCL21 and genes involved in immune-cell chemotaxis (Maruyama et al., 2008; Imai et al., 2008). A possible pathway influenced by RASSF2 is the NF-kB pathway since over expression of RASSF2 significantly downregulated NF-kB transcriptional activity (Maruyama et al., 2008; Imai et al., 2008). Of note is the recent observation that pigs experimentally infected with Porcine Circovirus Type 2 (PCV2) show upregulation of several CXCL family cytokines as well as RASSF2 (Fernandes et al., 2009) therefore it is likely a role for RASSF2 in regulating immune response pathways remains to be discovered. There is also evidence that RASSF2 may regulate the actin cytoskeleton since re-expression of RASSF2 leads to loss of stress fibres, cell rounding and the suppression of RhoGTPase activation (Maruyama et al., 2008; Akino et al., 2005). Additionally, RASSF2 upregulation appears to be a cellular response to ionising radiation (Sakamoto-Hojo et al., 2003).

Atlas Image
Figure 4: One possible RASSF2 pathway. Recent evidence suggests a RASSF2-MST1/2 complex may continuously cycle through the nucleus and that nuclear localisation of RASSF2 is essential for tumour suppressor function. There are also reports demonstrating that the activity of the kinases ERK2 and MST1/2 is crucial for cytoplasmic relocalisation of RASSF2. Therefore activation of the RASSF2 tumour suppressor pathway may emanate from the nucleus following ERK2 and MST1/2 activation allowing RASSF2 to accumulate in the cytoplasm where it may encounter another interacting partner K-RasGTP.

Homology

Human RASSF2 has highly conserved orthologues across many species (table 1). The main features of the RASSF2 protein are conserved across these species including the RA and SARAH domains as well as the NLS and the sequence important for nuclear export. RASSF2 is one of 10 members of the Ras-association domain family (RASSF) comprising RASSF1-10. RASSF1-6 are termed the classical RASSF family and contain C-terminal RA and SARAH domains. Consequently, RASSF1-6 are most similar in sequence within their C-termini. RASSF7-10 (RASSF7, RASSF8, RASSF9, RASSF10) represent evolutionarily conserved but structurally distinct RASSF members that lack the SARAH domains and contain N-terminal RA domains. RASSF7-10 are termed the N-terminal RASSF family. RASSF2 is most similar to RASSF4 and RASSF6 (figure 3), both of which are also epigenetically inactivated in cancer, and participate in K-Ras signalling to inhibit tumour cell growth and induce apoptosis (Hesson et al., 2009; Ikeda et al., 2006; Allen et al., 2006; Eckfeld et al., 2005; Chow et al., 2004).
Atlas Image
Table 1: RASSF2 has highly conserved orthologues in several species.

Mutations

Note

Similar to all other RASSF members, perhaps with the exception of RASSF1A (Kashuba et al., 2009; Pan et al., 2005), mutation of RASSF2 is a rare event and to date no inactivating mutations have been described. Analysis of ovarian primary tumours failed to identify a single amino acid changing or truncating point mutation (Cooper et al., 2008). However, more thorough investigations of larger cohorts of different tumour types may be required to determine this definitively.

Implicated in

Entity name
Colorectal carcinoma (CRC)
Note
Colorectal carcinoma (CRC) including colon adenomas.
Prognosis
In early colorectal cancers RASSF2 methylation with oncogenic activation of either K-Ras, B-Raf or PIK3CA presented significantly more frequently in cases of venous invasion (Nosho et al., 2007).
Oncogenesis
Similar to several other RASSF members, RASSF2 is frequently inactivated by CpG island DNA hypermethylation (Hesson et al., 2007). RASSF2 inactivation has been most extensively investigated in colorectal cancer in which inactivation of other RASSF members is relatively rare. Several studies now strongly suggest that RASSF2 inactivation is a frequent and early event in colorectal cancer formation being present in colon adenomas, particularly those with a villous component (Kakar et al., 2008; Hesson et al., 2005; Akino et al., 2005; Harada et al., 2007). The frequencies of RASSF2 methylation in CRCs vary between 42% (Akino et al., 2005) and 70% (Hesson et al., 2005) whilst for adenomas methylation occurs between 25% (Harada et al., 2007) and 94% (Kakar et al., 2008). This most likely reflects the differences in histopathological subtypes and the CpG island region analysed as well differences in colorectal cancer aetiology in different populations. Some reports describe variable frequencies of RASSF2 methylation depending on tumour location within the colon (Harada et al., 2007). Interestingly, cells from apparently normal colonic epithelium from patients with hyperplastic polyposis (HPP) also frequently demonstrate methylation of several TSGs including RASSF2 (Minoo et al., 2006). This may indicate an early role for RASSF2 inactivation in colonic hyperplasia. RASSF2 methylation was found to be associated with K-Ras or BRAF mutation (Harada et al., 2007; Akino et al., 2005) however, in another study K-Ras mutation and RASSF2 methylation were mutually exclusive (Hesson et al., 2005). Methylation of the RASSF2 promoter and loss of expression occurred in conjunction with loss of histone H3 acetylation, a marker of transcriptional activity. Reduced RASSF2A expression also correlated with methylation in primary CRCs (Akino et al., 2005). In colorectal tumour cell lines re-expression of RASSF2 resulted in inhibition of anchorage-independent growth in soft agar, which was associated with morphological changes, cell detachment, disruption of the actin stress fibre network, decreased Rho activity, increased apoptosis and inhibition of cell cycle progression. Deregulation of the actin cytoskeletal network and morphological changes that result in cellular detachment may therefore be a mechanism by which RASSF2 induces anoikis, a form of suspension-dependent apoptosis.
Entity name
Non-small cell lung carcinoma (NSCLC)
Oncogenesis
Investigation of NSCLC primary tumours found methylation of the RASSF2 CpG island in 44% (22/50). Methylation was found at an equal frequency in all grades (I/II = 44% (7/16); IIIA = 44% (4/9) and IV = 44% (10/23)) suggesting RASSF2 became hypermethylated early in tumour formation and was not associated with development to higher grades (Cooper et al., 2008). The incidence of RASSF2 methylation appears much more frequent in NSCLC than SCLC (small cell lung cancer) as shown by an earlier study by Kaira et al., (2007), in which only 18% (4/22) SCLC but 62% (16/26) NSCLC cell lines demonstrated RASSF2 methylation with concomitant loss of RASSF2A expression. RASSF2A expression was restored following treatment with the DNA demethylating agent 5-aza-2deoxycytidine and/or trichostatin A. In primary NSCLC tumours 31% (33/106) were methylated and methylation was more frequent in specimens from non-smokers than from smokers (45%, 18/40 vs 23%, 15/66 respectively; p=0.014).
Entity name
Nasopharyngeal carcinoma
Prognosis
RASSF2 methylation correlated with lymph node metastasis in nasopharyngeal carcinoma (Zhang et al., 2006).
Oncogenesis
Fifty one percent (27/53) of primary nasopharyngeal carcinomas (NPCs) showed cancer-specific RASSF2 methylation, which correlated with loss of RASSF2A expression in both NPC cell lines and primary tumours (Zhang et al., 2006). This study also provided evidence that RASSF2 re-expression suppressed colony formation ability in NPC cell lines (with concomitant inhibition of cell cycle progression) and decreased cell motility and migration as determined by wound healing assay.
Entity name
Gastric cancer
Prognosis
Both Maruyama et al., (2008) and Endoh et al., (2005) found DNA methylation of the region around the transcription start site of RASSF2A variant 1 significantly correlated with an absence of lymphatic invasion. Whilst Maruyama et al., (2009) found further associations with methylation and the absence of venous invasion or lymph node metastasis, less advanced stage, presence of EBV infection, the absence of TP53 mutations and the presence of a CpG-island methylator phenotype (CIMP).
Oncogenesis
The RASSF2 gene contains a 1.8kb CpG island that encompasses the first two non-coding exons (figure 1). The majority of this CpG island was interrogated for hypermethylation in a series of gastric cancers (Endoh et al., 2005). The study found varying frequencies of methylation throughout the CpG island ranging from 29% (23/78) at the region encompassing the transcription start site, to 79% (62/78) at a region of the CpG island within intron 1 (though this intronic region also exhibited a frequency of 60% (47/78) methylation in corresponding normal gastric epithelia). Methylation was mostly cancer specific at and around the transcription start site and silencing of RASSF2A expression most closely correlated with methylation at this region (Endoh et al., 2005). Similar findings were described by Maruyama et al., (2008) who examined the methylation status of the CpG island regions encompassing the transcriptional start sites of RASSF2A variants 1 and 2. Methylation of RASSF2A variant 1 was detected in 29.5% (23/78), whilst RASSF2A variant 2 was methylated in 25.6% (23/78) of gastric cancer cases. This may be significant since the expression of RASSF2A variants 1 and 2 appear to be differentially regulated by DNA methylation from within the same CpG island indicating multiple promoters and possibly accounting for the differences in methylation densities throughout the region. RASSF2A re-expression inhibited the growth of gastric cancer cell lines as shown by reduced colony formation ability. This was a result of inhibition of cell cycle progression and induction of apoptosis (Maruyama et al., 2008).
Entity name
Prostate cancer
Prognosis
A prospective study of a large cohort of patients referred for prostate biopsy determined that detection of RASSF2 methylation in patient urine shows promising clinical utility as an early detection biomarker for prostate cancer (Payne et al., 2009). Though primary prostate tumour tissues were not investigated the study nevertheless provided information independent of the extensively used pre-existing prostate cancer biomarker PSA (prostate specific antigen). Detection of RASSF2 methylation was significantly more frequent in patients with non-organ-confined prostate cancer (Payne et al., 2009). Thus RASSF2 methylation may represent an interesting biomarker for early prostate cancer detection and predicting invasive potential but requires further validation.
Entity name
Breast cancer
Oncogenesis
RASSF2 was frequently hypermethylated in primary breast cancers (38%, 15/40) and re-expression in breast cancer cell lines inhibited colony formation ability, anchorage-independent growth in soft agar and in vivo tumour formation in SCID mice (Cooper et al., 2008). RASSF2 growth suppression was dependent on a functional NLS (located at amino acids 151-167) since its mutation prevented anchorage-independent growth inhibition.
Entity name
Hepatocellular carcinoma (HCC)
Oncogenesis
In an extensive DNA hypermethylation analysis of cancerous and non-cancerous liver tissues (including normal liver tissues from non-cancer patients) Nishida et al., (2008) assessed the methylation status of 19 gene loci in hepatitis B virus (HBV) and hepatitis C virus (HCV)-related HCCs. This study found that normal ageing within liver tissues is associated with a gradual increase in aberrant methylation and that HCV infection in particular may accelerate age-related methylation. RASSF2 methylation was identified as a cancer-specific event that is completely absent in normal liver (0/22), infrequent in non-cancerous liver from HCC patients (2.6%, 2/77), yet frequent in HCV-related HCC (48%, 21/44) vs virus-negative HCC (5.6%, 1/18; p=0.0029). Though this suggests loss of RASSF2 expression may play a role in HCC the potential importance and clinical implications of these findings require further investigation.
Entity name
Oral squamous cell carcinoma (OSCC)
Oncogenesis
Analysis of the expression of RASSF1-6 in OSCC cell lines identified RASSF2 as the most frequently downregulated RASSF gene analysed. This loss of expression was caused by RASSF2 CpG island methylation, which was found in 26% (12/46) of primary OSCCs (Imai et al., 2008). In OSCC cell lines re-expression of RASSF2 inhibited colony formation ability by inducing apoptosis and inhibiting cell cycle progression. Investigation of 482 OSCCs identified RASSF2 methylation in 28% (134/482) cases. The combination of RASSF1A and RASSF2 methylation was significantly associated with poor disease-free survival (p=0.009, Huang et al., 2009). Furthermore, methylation of RASSF1A and RASSF2 increased in patients undergoing post-surgical radiotherapy when compared with surgery-only patients possibly indicating that hypermethylation of RASSF1A and RASSF2 is associated with the radioresistance commonly observed in OSCC patients. It also indicates the potential of using combined epigenetic and radiotherapy as an adjuvant to surgery.
Entity name
Endometrial carcinoma
Oncogenesis
Liao et al., (2008) investigated endometrial carcinomas for RASSF2 CpG island methylation and found 25/76 (33%) were methylated. RASSF2 methylation was found more frequently in tumour samples from older patients. Similar findings were observed in colorectal carcinomas (Hesson et al., 2005) and in OSCC (Imai et al., 2008) suggesting loss of RASSF2 expression may be a gradual age-related process.
Entity name
Ovarian cancer
Oncogenesis
Although methylation of the RASSF2 promoter is not an event associated with ovarian cancer (Cooper et al., 2008) the gene does localise to one of the regions deleted in ovarian tumour cell lines as indicated by array-based genomic hybridisation (Lambros et al., 2005).
Entity name
Leukaemia
Oncogenesis
Recent evidence suggests that RASSF2 expression may be downregulated in leukaemias with MLL rearrangement by overexpression of one or more of the miR-17-92 polycistronic miRNA oncogene cluster (Li et al., 2009) that may target a region within the 3UTR of the RASSF2 mRNA.

Bibliography

Pubmed IDLast YearTitleAuthors
160129452005The Ras effector RASSF2 is a novel tumor-suppressor gene in human colorectal cancer.Akino K et al
174045712007RASSF6 is a novel member of the RASSF family of tumor suppressors.Allen NP et al
164609082006Comparative genomic organization of the human and bovine PRNP locus.Choi SH et al
153755002004Aberrant methylation of RASSF4/AD037 in nasopharyngeal carcinoma.Chow LS et al
178911782008Epigenetic regulation of the ras effector/tumour suppressor RASSF2 in breast and lung cancer.Cooper WN et al
195259782009RASSF2 associates with and stabilizes the proapoptotic kinase MST2.Cooper WN et al
155747782004RASSF4/AD037 is a potential ras effector/tumor suppressor of the RASSF family.Eckfeld K et al
162653492005RASSF2, a potential tumour suppressor, is silenced by CpG island hypermethylation in gastric cancer.Endoh M et al
195442062009Exploratory study on the transcriptional profile of pigs subclinically infected with porcine circovirus type 2.Fernandes LT et al
179238752007Genetic and epigenetic alterations of Ras signalling pathway in colorectal neoplasia: analysis based on tumour clinicopathological features.Harada K et al
173254272007The role of RASSF1A methylation in cancer.Hesson LB et al
195702202009The novel RASSF6 and RASSF10 candidate tumour suppressor genes are frequently epigenetically inactivated in childhood leukaemias.Hesson LB et al
158061692005CpG island promoter hypermethylation of a novel Ras-effector gene RASSF2A is an early event in colon carcinogenesis and correlates inversely with K-ras mutations.Hesson LB et al
195091632009Methylation of RASSF1A, RASSF2A, and HIN-1 is associated with poor outcome after radiotherapy, but not surgery, in oral squamous cell carcinoma.Huang KH et al
173677792007Ras-association domain family protein 6 induces apoptosis via both caspase-dependent and caspase-independent pathways.Ikeda M et al
182942752008Epigenetic inactivation of RASSF2 in oral squamous cell carcinoma.Imai T et al
175494182007Epigenetic inactivation of the RAS-effector gene RASSF2 in lung cancers.Kaira K et al
179507802008CpG island methylation is frequently present in tubulovillous and villous adenomas and correlates with size, site, and villous component.Kakar S et al
194789412009High mutability of the tumor suppressor genes RASSF1 and RBSP3 (CTDSPL) in cancer.Kashuba VI et al
118645652002Identification of a novel Ras-regulated proapoptotic pathway.Khokhlatchev A et al
195556842009Extracellular signal-regulated kinase 2 (ERK-2) mediated phosphorylation regulates nucleo-cytoplasmic shuttling and cell growth control of Ras-associated tumor suppressor protein, RASSF2.Kumari G et al
173201102007Nuclear transport of Ras-associated tumor suppressor proteins: different transport receptor binding specificities for arginine-rich nuclear targeting signals.Kumari G et al
155863662005Analysis of ovarian cancer cell lines using array-based comparative genomic hybridization.Lambros MB et al
118050892002Phosphorylation and dimerization regulate nucleocytoplasmic shuttling of mammalian STE20-like kinase (MST).Lee KK et al
191552942009Consistent deregulation of gene expression between human and murine MLL rearrangement leukemias.Li Z et al
184046742008Hypermethylation of RAS effector related genes and DNA methyltransferase 1 expression in endometrial carcinogenesis.Liao X et al
183106592008Cytoplasmic RASSF2A is a proapoptotic mediator whose expression is epigenetically silenced in gastric cancer.Maruyama R et al
164697932006Extensive DNA methylation in normal colorectal mucosa in hyperplastic polyposis.Minoo P et al
181610482008Aberrant methylation of multiple tumor suppressor genes in aging liver, chronic hepatitis, and hepatocellular carcinoma.Nishida N et al
171830692007Genetic and epigenetic profiling in early colorectal tumors and prediction of invasive potential in pT1 (early invasive) colorectal cancers.Nosho K et al
160963692005High frequency somatic mutations in RASSF1A in nasopharyngeal carcinoma.Pan ZG et al
194591762009DNA methylation biomarkers of prostate cancer: confirmation of candidates and evidence urine is the most sensitive body fluid for non-invasive detection.Payne SR et al
146443432003Gene expression profiles in human cells submitted to genotoxic stress.Sakamoto-Hojo ET et al
163445482006The RASSF1A tumor suppressor activates Bax via MOAP-1.Vos MD et al
127326442003RASSF2 is a novel K-Ras-specific effector and potential tumor suppressor.Vos MD et al
170138962007Inactivation of RASSF2A by promoter methylation correlates with lymph node metastasis in nasopharyngeal carcinoma.Zhang Z et al

Other Information

Locus ID:

NCBI: 9770
MIM: 609492
HGNC: 9883
Ensembl: ENSG00000101265

Variants:

dbSNP: 9770
ClinVar: 9770
TCGA: ENSG00000101265
COSMIC: RASSF2

RNA/Proteins

Gene IDTranscript IDUniprot
ENSG00000101265ENST00000379376P50749
ENSG00000101265ENST00000379400P50749

Expression (GTEx)

0
50
100
150
200
250
300
350
400

Pathways

PathwaySourceExternal ID
Hippo signaling pathway -multiple speciesKEGGko04392
Hippo signaling pathway -multiple speciesKEGGhsa04392

Protein levels (Protein atlas)

Not detected
Low
Medium
High

References

Pubmed IDYearTitleCitations
203796142010Personalized smoking cessation: interactions between nicotine dose, dependence and quit-success genotype score.62
127326442003RASSF2 is a novel K-Ras-specific effector and potential tumor suppressor.61
197733812009Aberrant methylation of APC, MGMT, RASSF2A, and Wif-1 genes in plasma as a biomarker for early detection of colorectal cancer.60
160129452005The Ras effector RASSF2 is a novel tumor-suppressor gene in human colorectal cancer.34
170138962007Inactivation of RASSF2A by promoter methylation correlates with lymph node metastasis in nasopharyngeal carcinoma.33
195259782009RASSF2 associates with and stabilizes the proapoptotic kinase MST2.30
158061692005CpG island promoter hypermethylation of a novel Ras-effector gene RASSF2A is an early event in colon carcinogenesis and correlates inversely with K-ras mutations.27
195091632009Methylation of RASSF1A, RASSF2A, and HIN-1 is associated with poor outcome after radiotherapy, but not surgery, in oral squamous cell carcinoma.27
162653492005RASSF2, a potential tumour suppressor, is silenced by CpG island hypermethylation in gastric cancer.24
199629602010Role of the tumor suppressor RASSF2 in regulation of MST1 kinase activity.23

Citation

Luke B Hesson ; Farida Latif

RASSF2 (Ras association (RalGDS/AF-6) domain family member 2)

Atlas Genet Cytogenet Oncol Haematol. 2009-08-01

Online version: http://atlasgeneticsoncology.org/gene/43461/rassf2