RANBP2 (RAN binding protein 2)

2014-08-01   Erica Di Cesare , Patrizia Lavia 

Institute of Biology, Molecular Medicine, NanoBiotechnology (IBMN), National Research Council (CNR), c\\\/o La Sapienza University, via degli Apuli 4, 00185 Rome, Italy

Identity

HGNC
LOCATION
2q12.3
LOCUSID
ALIAS
ADANE,ANE1,IIAE3,NUP358,TRP1,TRP2
FUSION GENES

DNA/RNA

Note

RANBP2 is an essential gene and RANBP2-null mice display early embryonic lethality (Aslanukov et al., 2006; Dawlaty et al., 2008). A single RANBP2 hypomorphic allele is, however, sufficient for viability.

Description

The human RANBP2 gene comprises 31 exons and gives rise to one major mRNA encoding the RANBP2 protein, with at least 8 less represented alternative splicing variants (AceView; NCBI; GeneCards).

Transcription

RANBP2 mRNA transcription is widespread in many though not all tissues (Fauser et al., 2001). In the mouse genome, the Ranbp2 promoter region lies in a CpG island, typical of "housekeeping" gene promoters and potentially subjected to epigenetic regulation. In silico analysis of the human RANBP2 gene promoter has identified potential binding sites for cell cycle- and cell proliferation-dependent transcription factors, some validated in chromatin immunoprecipitation (ChIP) assays (e.g., c-Fos, AP1 and others) (GeneCards).
Binding sites for tissue-specific factors are also present, and RANBP2 mRNA transcript and protein product are highly expressed in certain tissues and cell types, e.g. neuronal cells (Fauser et al., 2001).
Serial analysis of gene expression (SAGE) depicted aberrant up-regulation of RANBP2 in certain cancers, e.g. multiple myeloma (Felix et al., 2009).

Proteins

Note

Biological overview: The RAN-binding protein 2 (RANBP2) or Nucleoporin 358 (NUP358, nucleoporin of 358 kDa) is the largest component of nuclear pore complexes (NPCs). The latter are highly complex structures composed of several orderly assembled proteins, called nucleoporins (NUPs), that represent gateways across the nuclear envelope (NE) for the exchange of macromolecules between the nucleus and the cytoplasm. This exchange is critical to many essential processes, e.g., DNA replication, DNA repair, DNA damage response, establishment of functional chromatin domains, replication checkpoint, transcriptional and epigenetic regulation of genes and genome function, mitotic entry.
The RANBP2 nucleoporin is specific of higher eukaryotes and has a multimodular structure (Wu et al., 1995; Yokoyama et al., 1995; Wilken et al., 1995). It is unique among NUPs in that it is endowed with E3-type ligase activity for SUMO (small ubiquitin-related modifier) peptides (Pichler et al., 2002). This will be discussed in more depth below.
RANBP2 operates in two major groups of cellular processes:
- NPC- and NE-dependent processes ensuring nuclear functions in interphase (e.g., nuclear positioning, recruitment of motor proteins at the NE, centrosome anchoring to the NE, import and export of macromolecules in and out of the nucleus, including transcription and regulatory factors governing genome function); and
- cell division events (NE breakdown, centrosome migration, assembly of the mitotic apparatus, chromosome segregation).
In many of these processes, RANBP2 stimulates the conjugation of SUMO peptides (SUMO-1 to -4) to various target proteins at specific intracellular sites: the nuclear rim in interphase, and microtubules (MTs) as well as kinetochores (KTs) in mitosis. SUMO conjugation is emerging as a protein post-translational modification that modulates the localization and interactions of several proteins (reviewed by Lomelí and Vázquez, 2011; Flotho and Melchior, 2013).
The protein-modifying and transport-regulating activities of RANBP2 target specific substrates in many tissues and cell types. As a result, RANBP2 acts as a cell context-dependent pleiotropic protein in a variety of physiological and pathological processes, including tumor suppression, neuroprotection and familial necrotic encephalopathy.
Atlas Image
Figure 1. A Schematic of RANBP2 domains. Boxes 1-4 identify four RAN-binding domains, Cy indicates a cyclophilin-like domain, vertical dashes mark the position of FG-repeats that interact with transport receptors (modified from Werner et al., 2012).

Description

The human RANBP2 protein is composed of 3224 aminoacidic residues, with a molecular weight of 358 KDa, hence its name (Wu et al., 1995; Yokoyama et al., 1995). The alternative name RANBP2 derives from the presence of four RAN-binding domains (RBD), through which it binds the GTPase RAN. RANBP2 contains several more structural domains:
- An N-terminal leucine-rich region anchors RANBP2 to the NPC. This region is also implicated in binding interphase microtubules (MTs) and regulating their dynamics (Joseph and Dasso, 2008). The structure of this region reveals an alpha-helical domain harboring three central tetratricopeptide repeats (TPRs) capable to bind single-stranded RNA in solution and thought to contribute to messenger ribonucleoprotein (mRNP) remodeling at the cytoplasmic face of the NPC (Kassube et al., 2012).
- Four RAN binding domains (RBD1-4) (Yokoyama et al., 1995), 46-60% identical to the prototype RAN-binding domain (Pfam) in the first cloned RAN-binding partner, RANBP1 (Bressan et al., 1991; Coutavas et al., 1993). The RBDs act as coactivators of GTP hydrolysis on RAN with a dual purpose: a) to assist nuclear protein import, by preventing the accumulation of RANGTP at the NPC cytoplasmic side and avoid that RANGTP prematurely dissociates import complexes while traversing the NPC to reach the nucleus (Yaseen and Blobel, 1999a); b) to facilitate the export of nuclear cargos by assisting the dissociation of RANGTP from exportin-cargo complexes (Bernad et al., 2004).
- Eight zinc-finger motifs (Cys2-Cys2 type) in the central portion of RANBP2; they provide a binding platform for exportin-1/CRM1 (Singh et al., 1999) and help CRM1 recycling into the nucleus (Bernad et al., 2004). These motifs can also interact with RanGDP (Yaseen and Blobel, 1999a).
- Phenylalanine-glycine (FG) and FxFG repeats (the nucleoporin "signature" motif; x is any aminoacid) present on the fibril-like structures projecting from the NPC into the cytoplasm. These repeats provide multiple binding sites for nuclear transport receptors (karyopherin beta/ importin beta and exportin-1/CRM1. The interaction of FG-rich fibrils with transport vectors facilitates their passage across the NPC.
- A domain endowed with SUMO E3 ligase activity, the first enzymatic activity identified for RANBP2 (Pichler et al., 2002), residing between RBDs 3 and 4, that regulates sumoylation of target proteins (more detail below).
- A C-terminal domain with peptidyl-prolyl isomerase activity, the second enzymatic activity ascribed to RANBP2 (Lin et al., 2013).
- A cyclophilin A-like domain, harbouring an active site cavity that facilitates the binding to the HIV-1 capsid proteins during viral infection (Lin et al., 2013).
Thus far, the SUMO E3 ligase domain, two RBDs, the N-terminal TPR domain and the C-terminal domain have been crystallized and structurally characterized (Vetter et al., 1999; Reverter and Lima, 2005; Geyer et al., 2005; Kassube et al., 2012; Lin et al., 2013).

The SUMO E3 ligase activity of RANBP2 and the RANBP2/RANGAP1*SUMO1/Ubc9 (RRSU) complex
In addition to binding RAN, RANBP2 stably associates with the RAN GTPase-activating protein 1 (RANGAP1) (Mahajan et al., 1997; Saitoh et al., 1997; Matunis et al., 1998; Swaminathan et al., 2004) throughout the cell cycle. The interaction requires SUMO-1 conjugation to RANGAP1 (Matunis et al., 1996; Mahajan et al., 1997) and the presence of the SUMO ubiquitin-like-conjugating Ubc9 enzyme (Zhang et al., 2002; Zhu et al., 2006), an E2 enzyme that transfers SUMO peptides to SUMO chains, analogous to enzymes acting in the ubiquitination cascade.
RANBP2 is possibly the most abundant SUMO E3 ligase in the cell, and as such has a prominent role in SUMO modification of proteins. Most RANBP2-dependent functions are likely mediated by its activity in SUMO conjugation of target proteins.
The RANBP2 SUMO E3 ligase domain lacks homology to other known SUMO or ubiquitin E3 ligases (Pichler et al., 2004). It is characterized by two 50 aminoacid-long internal repeats, IR1 and IR2 (43% identical), separated by a 20 aminoacid-long linker. Both IR1 and IR2 can bind to Ubc9 and catalyze sumoylation of substrates in vitro, though IR2 has lower affinity for Ubc9 than IR1 (Pichler et al., 2004; Tatham et al., 2005).
RANBP2 is quantitatively engaged in complexes with sumoylated RANGAP1 and Ubc9. This binding requires RANBP2s IR1 and the SUMO-interaction motif SIM1, as well as Ubc9. Once IR1 and SIM1 are occupied by RanGAP1*SUMO1 and Ubc9, the E3 ligase activity depends on IR2 (Werner et al., 2012). In that sense, the entire RANBP2/RANGAP1-SUMO complex (called RRSU complex) is viewed as a multisubunit SUMO ligase. Indeed, RRSU effectively sumoylates the physiological substrate Borealin in vitro (Klein et al., 2009), whereas free RANBP2 does not (Werner et al., 2012).
After NPC disassembly at NEB, RANBP2 remains associated with RANGAP1-SUMO1 and Ubc9 (Swaminathan et al., 2004); RRSU associates with the mitotic spindle and a fraction is recruited to KTs after MTs attach to them (Joseph et al., 2002; Joseph et al., 2004).

Atlas Image
Figure 2. The localization of RANBP2 (red) in human Hela cells. Top row: note the punctuate red staining around the nucleus, which identifies the regular distribution of NPCs. Bottom row: a metaphase cell with aligned chromosomes (left panel). RANBP2 co-localizes with mitotic MTs with an accumulation at poles and at the kinetochore level, appearing as red spots proximal to the MT growing ends.

Localisation

Intracellular localization: In interphase cells, RANBP2 localizes at the cytoplasmic face of the NPCs (Wu et al., 1995; Yokoyama et al., 1995; Wilken et al., 1995; Walther et al., 2002): RANBP2-containing filaments are anchored to the NPC via interaction with a complex of nucleoporins containing Nup214 and Nup88 (Bernad et al., 2004) and project into the cytoplasm.
Joseph et al. (2002 and 2004) first reported that, at the onset of mitosis, when the NE breaks down and NPCs disassemble, RANBP2 localizes to the microtubules (MTs) of the forming mitotic spindle, with an accumulation at poles; a fraction is recruited to chromosomal KTs when the latter become attached to MTs (Figure 2, bottom row). This localization underlies RANBP2s mitotic functions (see below). In early telophase RANBP2 is recruited back around chromatin of the reforming nuclei as the NE and NPCs reorganize.

Function

RANBP2 in interphase nucleocytoplasmic transport
As anticipated above, RANBP2 localization at cytoplasmic fibrils emanating from the NPC underlies its function in nucleocytoplasmic transport.
- Nuclear protein import
RANBP2 serves as a docking site for import complexes (the latter are of two main types: either composed of importin vectors interacting with proteins marked by a nuclear localization signal, NLS, or composed of transportin bound to ribonucleoproteins marked by the so-called M9 signal sequence, originally described in hnRNPs).
The docking of import complex at RANBP2 cytoplasmic fibrils of the NPC aids the earliest step in nuclear import (Melchior et al., 1995; Delphin et al., 1997; Mahajan et al., 1997; Yaseen and Blobel, 1999b). RANBP2 itself does not directly participate in import, but facilitates it. In RANBP2-depleted HeLa cells, in vivo nuclear import by either Importin alpha/beta (Hutten et al., 2008) or transportin (Hutten et al., 2009) still occurs, but at substantially reduced rates.
In a screening for nuclear proteins that accumulate in the cytoplasm upon RANBP2 depletion, Wälde and coworkers (2012) have also identified direct RANBP2 interactors: they found that an N-terminal fragment of RANBP2, harboring the NPC-binding domain, three FG motifs and RBD1, was sufficient to promote protein import, while neither the interaction with RANGAP1 nor the SUMO E3 ligase activity were required (Wälde et al., 2012). This is consistent with functional mapping data from Hamada and coworkers (2011) using various RANBP2-derived regions to complement RANBP2 knockout MEF cells, in which the RANBP2 N-terminal fragment restored import to RANBP2-null cells; the authors demonstrated a crucial role of this domain in aiding the recycling of RAN and importin beta complexes for nuclear import (Hamada et al., 2011).
In summary, RANBP2 aids nuclear import by at least two mechanisms: i) by "capturing" transport receptors through the FG-repeats, it conveys them towards the NPC and reduces the effective concentration of import receptors required for efficient transport, while ii) by interacting with selected cargos in a receptor-independent manner, through the RANBP2 N-ter domain, it increases the overall efficiency of nuclear import.
Interestingly, RANBP2 is also implicated in the nuclear delivery and integration of certain human viruses, including Herpes simplex (Copeland et al., 2009) and immunodeficiency virus-1 (HIV-1) (Zhang et al., 2010; Ocwieja et al., 2011; Schaller et al., 2011).
- Nuclear export
RANBP2 also plays roles in mRNA export. Poly(A)+ mRNA accumulates in nuclei of RANBP2-null MEFs (Hamada et al., 2011), although the intracellular distribution of poly(A)+ mRNA is not affected in RANBP2 hypomorphic mice-derived MEF cells (Dawlaty et al., 2008): thus, mRNA export requires RANBP2, but can proceed, albeit being impaired, in the presence of significantly decreased abundance. These data suggest that RANBP2 facilitates the export pathway, yet is not an indispensable component.
Overall, RANBP2 affects the rate of nucleo-cytoplasmic transport of many proteins, including transcriptional and epigenetic factors. The latter are often mislocalized in tumor cells and in other cellular contexts in which RANBP2 expression is altered, with a global impact on genome functions. An emerging concept is that tumor cells exploit specific properties of NUPs to deregulate gene transcription, chromatin boundaries and essential transport-dependent regulatory circuits (Xu and Powers, 2009; Köhler and Hurt, 2010).

Structural functions at the nuclear rim and NPCs
RANBP2 has structural roles at the NE besides nuclear transport proper. The development of in situ SUMOylation assays has revealed that both the nuclear rim and PML nuclear bodies are major sites of SUMOylation; RANBP2 inactivation abolished SUMOylation processes along the nuclear rim and reduced the number of PML bodies, while not affecting the nuclear lamina (Saitoh et al., 2006). The loss of nuclear PML bodies has been observed in tumorigenesis, particularly in colon cancer. Satow et al. (2012) reported that β-catenin overexpression disrupts PML bodies in colon cancer cell lines and inhibits RANBP2-dependent SUMOylation of specific PML-associated proteins. The data suggest that RANBP2 is required for SUMOylation of proteins associated with the formation of particular subnuclear structures, the loss of which impinges on nuclear functions in cancer cells.
RANBP2 also recruits motor proteins at the NE to regulate NE breakdown at the onset of mitosis. Through its zinc finger domain, RANBP2 binds the COPI coatomer complex, which coats the Golgi vesicles and contributes to membrane remodelling at the Golgi; the RANBP2-derived zinc finger domain alone dominantly interferes with COPI recruitment to the nuclear rim and inhibits NE breakdown (Prunuske et al., 2006). Interestingly, RANBP2 acts cooperatively with Nup153, the most nuclear of the NUPs, which contains a distinct zinc finger domain, in coordinating NE breakdown.
RANBP2 also binds to BICD2 (homologous to Drosophila Bicaudal D), an adaptor between motor proteins and their cargo, and recruits BICD2 to NPCs in the G2 phase of the cell cycle (Splinter et al., 2010). BICD2 in turn regulates dynein-dynactin motor complexes at NPCs, and thus centrosome tethering to the NE prior to mitotic entry. BIC2 is also required for the antagonistic activity of kinesin-1, which pushes centrosomes apart. The balance between dynein and kinesin-1 opposite activities governs centrosomal positioning, and hence sites where centrosomes will nucleate the spindle MTs to form asters and later spindle poles; RANBP2 recruitment of BICD2 to the NPCs just before NE breakdown represents a most upstream step in this cascade of events.
A specialized version of this process takes place in radial glial progenitors (RGPs), from which neurons, glia, and brain adult stem cells originate. RGP nuclei migrate basally during G1, then apically during G2 via dynein, and eventually divide at the ventricular surface. Hu et al. (2013) discovered that apical nuclear migration requires dynein recruitment at NPCs by two cooperating G2-specific mechanisms: the "RanBP2-BicD2" pathway acts first, and "Nup133-CENP-F" operates sequentially. This work identifies spatially regulated mechanisms, implying that only restricted regions of neurogenic tissues are permissive for mitosis: in this context, RANBP2 is essential for dynein control of apical nuclear migration, nuclear membrane remodelling and centrosome dynamics prior to mitosis.

Cell differentiation-associated functions
The NPC is not a static transport gate and undergoes dynamic remodelling during differentiation. In myogenic differentiation, myoblasts fuse to form syncithial myotubes. By atomic force microscopy, NPCs have been found to undergo structural differences during C2C12 myogenic cell differentiation from myoblasts to myotubes, parallel to an increased amount of RanBP2 at NPCs (Asally et al., 2011). siRNA-mediated depletion of RanBP2 in myoblasts suppresses differentiation to myotubes. Thus, RanBP2 is required for NPC remodelling in myogenesis, suggesting that a re-adaptation of transport mechanisms, and of the gateways through which these take place, is required as myotubes fuse and many nuclei become immersed in a common cytoplasm.
RANBP2 can carry out more specialized functions in a tissue-specific manner. RANBP2 is highly abundantly expressed in the vertebrate retina. Its cyclophilin domain (which led Ferreira et al., (1996) to classify RANBP2 as Type-II cyclophilin), and the RB4 domain, interacts with opsin, a retinal transmembrane protein; the RBD4 and cyclophilin domains are therefore proposed to act as a functional "supradomain" with a chaperone function for opsin in the retina. Along with this chaperone function, RANBP2 associates through its cyclophilin-like domain with subunits of the 19S regulatory complex of the 26S proteasome in the neuroretina (Ferreira et al., 1998), and thus contributes to control the stability of proteins that it "chaperones" in the retina.
Related to RANBP2 transport functions but independent from them, Cai et al., (2001) identified a novel domain between RBD2 and RBD3 capable of direct association with two MT-based kinesin motors, KIF5B and KIF5C, in neuronal cells. Preventing the interaction of the RANBP2 kinesin-binding domain (KBD) with KIF5B / KIF5C in neuronal cells caused perinuclear clustering of mitochondria, deficits in mitochondrial membrane potential and cell shrinkage (Cho et al., 2007): thus RANBP2 modulates kinesin-dependent mitochondria transport and function. The RBD2, KBD and RBD3 domains of RANBP2 are proposed to constitute a tripartite domain (R2KR3), modulating mitochondrial transport via kinesin subtypes in subsets of neuroretinal cells (Patil et al., 2013).
Aslanukov et al. (2006) discovered yet another association of RANBP2, via its leucine-rich domain, with Cox11, a mitochondrial metallochaperone, and HKI (hexokinase type I), defined as the "pacemaker" of glycolysis. Cox11 inhibits HKI activity, but RANBP2 suppresses this inhibition. Consequently, RANBP2 haploinsufficient mice show markedly decreased HKI and ATP levels in the central nervous system, with deficits in growth rates and glucose catabolism (Aslanukov et al., 2006). These mice also show absent or severely reduced cell death response to light-induced oxidative stress in the retina (Cho et al., 2010). RANBP2 cell type-conditional mice models, selectively lacking RANBP2 either in rod or in cone photoreceptors (Cho et al., 2013), showed that RANBP2 ablation in cone photoreceptors promoted their non-apoptotic death, while rod photoreceptors underwent cone-dependent non-autonomous apoptosis. Thus, RANBP2 modulates cell type-specific and distinct pathways of cell death - a key feature of neurodegenerative diseases.

Mitosis
- Mitotic spindle organization
Chromosome segregation at mitosis is crucial to the maintenance of genomic stability, a process often disrupted in cancer. A role of RANBP2 in chromosome segregation was first suggested by the finding that RANBP2 accumulates at the mitotic spindle in prometaphase, and in part at KTs upon MT attachment (Joseph et al., 2002; see figure 2); at these sites RANBP2 remains associated in complex with RANGAP1-SUMO1, suggesting that some of its functions entail RANGTP hydrolysis at specific mitotic sites.
In RNAi-based studies in human cells, RANBP2 down-regulation caused multipolar spindles, with supernumerary poles lacking centrioles (hence suggestive of MT dysfunction) as well as defects in chromosome congression and segregation (Salina et al., 2003; Joseph et al., 2004; Klein et al., 2009; Hashizume et al., 2013). Consistent findings in Caenorhabditis elegans embryos (Askjaer et al., 2002) suggest that mitotic functions of RANBP2 are conserved across species in which this protein is present.
- Mitotic microtubule-kinetochore interactions
Further studies showed that RANBP2 depletion resulted in aberrant KT morphology, associated with mis-localization of RANGAP1 and other KT proteins, e.g. Mad1, Mad2, Zw10, Mis12, CENP-A, CENP-E, CENP-F and dynein; RANBP2 depletion also caused lengthened prometaphase duration and chromosome misalignment at metaphase, but the simultaneous depletion of RANBP2 and either Mad1 (Salina et al., 2003) or Mad2 (Joseph et al., 2004), two major spindle assembly checkpoint (SAC) regulators, restored normal prometaphase duration; these findings suggest that RANBP2 depletion-dependent abnormalities activate the SAC. Interestingly, RANBP2 depletion yields unstable KT-MT interactions, suggesting that the concentration of RSSU complex at MT-attached KTs contributes to the functional connections between the spindle and chromosomes prior to chromosome segregation (Joseph et al., 2004). In conditions under which RSSU targeting to KTs was prevented, discrete attachments between MTs and KTs were not maintained, yielding high rates of chromosome mis-segregation (Salina et al., 2003; Joseph et al., 2004; Arnaoutov et al., 2005). Indeed, RANBP2 hypomorphic mice develop severe aneuploidy (Dawlaty et al., 2008).
RSSU targeting to KTs is highly regulated in human cells and requires i) MT attachment to KTs, and ii) proteins that stabilize MT interaction with KTs, e.g. Hec1/Ndc80 and Nuf2 (Joseph et al., 2004). There is therefore a functional cross-talk between proteins that regulate MT/KT interactions, and RSSU recruitment to KTs, which in turn reinforces these interactions. Interestingly, CRM1 is required for the RSSU complex recruitment to KTs (Arnaoutov et al., 2005) while importin beta overexpression inhibits it (Roscioli et al., 2012).
However, neither endogenous RANBP2, nor GFP-tagged RANBP2 constructs, localize to KTs in MEFs (Hamada et al., 2011). Some cell cycle checkpoints are not yet fully proficient in embryonic cell cycles and some of their regulatory mechanisms may diverge from those operating in somatic cells.
It is worth noting that cells subjected to extended RANBP2 RNAi for longer times eventually escape the mitotic arrest with defective MT/KT connections, originating multinucleated cells and/or citokynesis defects (intracellular bridges) followed by cell death (Salina et al., 2003; Joseph et al., 2004). In terms of cancer prognosis, these data suggest that a very narrow threshold, probably modulated by the genetic background of the cells, defines whether defective RANBP2 expression is pro-tumorigenic (by inducing genetic instability in cells that remain viable) or anti-tumorigenic (by preventing normal cell division altogether and inducing the death of the severely aberrant cell products).

Regulation of the SUMO conjugation pathway in mitosis
MEFs with reduced RANBP2 levels are viable and display no overt nuclear transport abnormalities compared to wild-type, yet develop severe aneuploidy associated with chromosome segregation defects, including anaphase bridges (Dawlaty et al., 2008). Chromatin bridges in anaphase are typical of cells in which DNA decatenation is impaired by mutation or inhibition of topoisomerase II alpha (Topo IIa) (Bhat et al., 1996).
Studies in S. cerevisiae (Takahashi et al., 2006), Xenopus egg extracts (Azuma et al., 2003) and human cells have shown that Topo IIa is subjected to sumoylation (Azuma et al., 2003; Azuma et al., 2005; Mao et al., 2000). Indeed, Dawlaty and coworkers observed that i) RANBP2 hypomorphic MEFs fail to accumulate Topo IIa at inner centromeres in mitosis, and ii) RANBP2 SUMO E3 ligase activity is required for Topo IIa SUMO conjugation and inner centromere targeting, to enable decatenation of sister centromeres prior to anaphase onset (Dawlaty et al., 2008).
RANBP2 also associates with the chromosomal passenger complex (CPC) during mitosis and stimulates sumoylation of Borealin (Klein et al., 2009); this, however, affects neither CPC assembly nor its localization; RANBP2-dependent Borealin SUMOylation might be required for CPC interaction with an as yet unidentified protein(s) at centromeres (Klein et al., 2009).

RANBP2 in cell viability
As remarked, RANBP2 inactivation causes early embryonic lethality (Aslanukov et al., 2006; Dawlaty et al., 2008). Hamada and coworkers (2011) studied mitotic cell viability using a Cre-mediated RANBP2 conditional knockout approach. The incidence of chromosome missegregation was 100% for RANBP2-null MEFs, yet these cells did not die during faulty mitosis and rarely died during the next 12 hours after mitotic exit, suggesting that the mitotic errors caused RANBP2 knock-out are not the primary cause of cell death (Hamada et al., 2011). Rescue experiments, expressing various RANBP2 portions in a RANBP2-null background, revealed that a short N-terminal fragment corrected transport defects and restored cell viability, suggesting prominent NPC dysfunction, rather than mitotic failure, as the cause of cell death (Hamada et al., 2011). By contrast, RANBP2 siRNA-silenced HeLa cells underwent prolonged metaphase followed by mitotic catastrophe in live cell imaging (Hashizume et al., 2013); the use of a fluorescently-tagged import reporter demonstrated that, under these conditions, RANBP2-depletion-induced mitotic death is not a side effect of failed nuclear import. The discrepancy between these models remains to be explained.

Homology

RANBP2 is conserved among metazoa but absent in Saccharomyces cerevisiae.

Mutations

Note

An autosomal dominant mutation of RANBP2 (1880C-->T, yielding the Thr585Met missense mutation in the leucine-rich domain required for binding to both the NPC and to MTs) has been identified in the familial predisposition to acute necrotizing encephalopathy (ANE), arising in otherwise healthy children after common viral infections, such as influenza (Neilson et al., 2009; Loh and Appleton, 2010).
Fusions of the RANBP2 gene with the gene encoding anaplastic lymphoma kinase (ALK) are associated with inflammatory myofibroblastic tumors (see below).
RANBP2 mutants (i.e. point mutations or deletion mutants) have been engineered in several laboratories to study the role of different domains in various cellular processes.

Implicated in

Entity name
Various cancers
Note
RANBP2 is implicated in many cancer types. It is difficult to draw a single unifying mechanism, yet two recurrent features are worth noting:
a) the SUMO ligase and SUMO-stabilizing activity of RANBP2 targets many mitotic factors, as explained above, which can contribute to genetic instability and tumorigenesis when dysregulated (e.g., RANGAP1 and hence the functional state of RAN at KTs, Topo II, Borealin). Mouse models created by crossing RANBP2 hypomorphic (RANBP2H) and null (RANBP2-) alleles, displaying gradual degrees of RANBP2 insufficiency, are prone to carcinogen-induced and spontaneous tumors: the incidence of skin tumors dramatically increased in mice with reduced compared to wild-type RANBP2 expression and lung adenocarcinomas developed in virtually all insufficient mice (Dawlaty et al., 2008).
b) generally, a striking link exists between some NUPs, their propensity to undergo translocation and fusion with other gene partners and neoplastic diseases (reviewed in Köhler and Hurt, 2010). RANBP2 shares this tendency with some other NUPs: residing in a chromosomal recombination "hot spot", is involved in several instances of translocation; signalling molecules involved in the resulting fusion protein become aberrantly concentrated at the NE, with tumorigenic consequences (see below).
Entity name
Inflammatory myofibroblastic tumors
Note
RANBP2 is implicated in a subset of inflammatory myofibroblastic tumors (IMT), rare soft tissue tumors involving mesenchymal cell types, with a prominent inflammatory component. IMTs rarely metastasize, yet often recur rapidly with fatal outcomes in some cases. Some 50% of IMTs harbor rearrangements of the ALK gene (encoding the anaplastic lymphoma kinase ALK), located at 2p23, with diverse partners, and overexpress the ALK protein, mostly in the cytoplasm. In several IMT cases, ALK is fused to RANBP2 and acquires a perinuclear localization. These cases generally have a more aggressive clinical course, suggesting that the RANBP2-dependent ALK perinuclear localization may be prognostic of malignant behavior.
The first two IMT cases with a RANBP2-ALK fusion were described by Ma et al. (2003). By sequence analysis, the N-terminal 867 residues of RANBP2 were fused to the cytoplasmic segment of ALK, originating an 1430-amino acid chimeric protein. In both cases, the RANBP2-ALK fusion was present in myofibroblasts and was nuclear membrane-associated, attributable to the presence of the NPC-binding domain of RANBP2 in the fusion. Patel et al. (2007) reported on an IMT in a young boy (karyotype 45,XY,der(2)inv(2)(p23q12)del(2)(p11.1p11.2),-22) with an ALK-RANBP2 fusion, identified by FISH and confirmed by cloning and sequencing of the breakpoints.
Chen and Lee (2008) described a hepatic IMT with a RANBP2-ALK rearrangement. PCR product sequencing revealed the presence of exon 18 from RANBP2 and exon 20 from ALK. Tumor cells showed a round cell phenotype with nuclear membrane accumulation of ALK protein. Mariño-Enríquez et al. (2011) characterized 11 cases of intra-abdominal IMT with epithelioid morphology. Nine showed perinuclear ALK staining, three of which harbored a RANBP2-ALK fusion. These patients experienced rapid recurrence. The authors suggest that the epithelioid variant of IMT with nuclear membrane or perinuclear ALK represents an aggressive form of sarcoma, with rapid recurrences and frequently fatal. Li et al. (2013) reported two more cases of IMT with RANBP2-ALK fusions, with epithelioid and rounded tumor cell morphology, from the pelvic and peritoneal cavities respectively, both associated with quick recurrence and poor prognosis. In 2014 the first case of a large tumor appearing in the pleural cavity was described (Kozu et al., 2014) in a patient with massive pleural effusion. The tumor showed the presence of a RANBP2-ALK fusion, rounded cells with an epithelioid shape, and a prominent inflammatory infiltrate, which led the authors to diagnose an epithelioid inflammatory myofibroblastic sarcoma (EIMS) and recognize it as an IMT variant. An EIMS case arising in the pelvic cavity was also described by Kimbara et al. (2014) as an aggressive variant of IMT. The tumor cells displayed epithelioid morphology and ALK staining on the nuclear membrane, associated with RANBP2-ALK fusion identified by RT-PCR. The patient experienced rapid local recurrence after surgery. The tumor was resistant to doxorubicin, but underwent shrinkage after treatment with the ALK inhibitor crizotinib.
Entity name
Note
Felix et al. (2009) generated SAGE libraries from normal and neoplastic plasma cells to identify differentially expressed genes in multiple myeloma (MM). They identified 46 upregulated genes in the MM library and validated them by qRT-PCR. RANBP2 belongs to a group of upregulated genes in >50% of tested MM cases and in meta-analyses (ONCOMINE database) of MM compared to normal plasma cells. The authors proposed that RANBP2 might be a potential therapeutic target in myeloma.
Note
Maesako et al. (2014) identified a RANBP2-ALK fusion mRNA transcript in a case of myeloid leukemia, associated with the chromosomal inversion inv(2)(p23q13), and resulting in nuclear membrane association of ALK. Another rearrangement involving RANBP2 and ALK was reported by Lim et al. (2014) in an acute myelomonocytic leukemia (AML) in a 31-year-old woman with a karyotype of 45,XX,inv(2)(p23q21),-7[20], associated with a RANBP2-ALK fusion transcript and strong staining of the fusion protein around the nuclear membrane in leukemic cells. The patient had an unfavorable clinical course.
Entity name
Colorectal cancer
Note
Gylfe et al. (2013) highlighted another type of recombination tumorigenic events involving RANBP2. Because microsatellite instability occurs in some 15% of all colorectal cancers, the authors sequenced the exomes of 25 colorectal tumors and respective healthy colon tissue. They confirmed potential mutation hot spots in 15 genes, among which RANBP2; these were validated in tumors with microsatellite instability and showed that RANBP2 also contains hot spot mutations in the validation set.
Entity name
Proposed tumor-promoting mechanisms of RANBP2 via SUMO-conjugation and stimulation of tumorigenic signaling
Note
The data discussed above indicate some major routes through which RANBP2 can contribute to cancer onset and progression: increasing their genetic instability during mitosis and impairing global nuclear functions in interphase. An increasing implication of SUMO conjugation in the function of proteins relevant to cancer is emerging, particularly in DNA damage and repair. Among the growing instances of RANBP2-dependent protein SUMOylation, some proteins have established roles in tumorigenic signaling pathways.
Miyauchi et al. (2012) demonstrated a role of RANBP2 in SUMOylation and localization of MDM2, a major regulator of p53 stability, suggesting therefore a possible indirect implication of RANBP2 in p53 functions.
Packham et al. (2014) showed that RANBP2 is implicated in the pro-tumorigenic activity of the insulin-like growth factor-1 receptor (IGF-1R), an activator of the PI3K/Akt pathway with key roles in tumorigenesis. The biological activity of IGF-1R depends on its nuclear translocation, which in turn depends on SUMOylation. Packham et al. (2014) characterized spatially regulated interactions of IGF-1R, first with dynactin, which transports IGF-1R to NPCs, and therein with importin-β and RANBP2. RANBP2 interacts with and stabilizes sumoylated IGF-1R, enabling its nuclear accumulation and hence the activation of tumorigenic pathways that depend on it.
Interestingly, RANBP2 levels are abnormally elevated in transgenic mouse models of prostate cancer constitutively expressing a PI3K catalytic subunit (PIK3CA), and treating the animals with a PI3K inhibitor decreases RANBP2 protein abundance (Renner et al., 2007).
These data converge to suggest functional cross-talks between RANBP2 and tumorigenic pathways.

Bibliography

Pubmed IDLast YearTitleAuthors
159089462005Crm1 is a mitotic effector of Ran-GTP in somatic cells.Arnaoutov A et al
212051962011Nup358, a nucleoporin, functions as a key determinant of the nuclear pore complex structure remodeling during skeletal myogenesis.Asally M et al
124759582002Ran GTPase cycle and importins alpha and beta are essential for spindle formation and nuclear envelope assembly in living Caenorhabditis elegans embryos.Askjaer P et al
170694632006RanBP2 modulates Cox11 and hexokinase I activities and haploinsufficiency of RanBP2 causes deficits in glucose metabolism.Aslanukov A et al
159337172005PIASy mediates SUMO-2 conjugation of Topoisomerase-II on mitotic chromosomes.Azuma Y et al
149932772004Nup358/RanBP2 attaches to the nuclear pore complex via association with Nup88 and Nup214/CAN and plays a supporting role in CRM1-mediated nuclear protein export.Bernad R et al
89786141996Chromatid segregation at anaphase requires the barren product, a novel chromosome-associated protein that interacts with Topoisomerase II.Bhat MA et al
18897461991Characterization of the opposite-strand genes from the mouse bidirectionally transcribed HTF9 locus.Bressan A et al
115536122001The docking of kinesins, KIF5B and KIF5C, to Ran-binding protein 2 (RanBP2) is mediated via a novel RanBP2 domain.Cai Y et al
190008322008Nucleoporin levels regulate cell cycle progression and phase-specific gene expression.Chakraborty P et al
187011322008An inflammatory myofibroblastic tumor in liver with ALK and RANBP2 gene rearrangement: combination of distinct morphologic, immunohistochemical, and genetic features.Chen ST et al
178879602007Association of the kinesin-binding domain of RanBP2 to KIF5B and KIF5C determines mitochondria localization and function.Cho KI et al
238188612013Distinct and atypical intrinsic and extrinsic cell death pathways between photoreceptor cell types upon specific ablation of Ranbp2 in cone photoreceptors.Cho KI et al
206827512010Neuroprotection resulting from insufficiency of RANBP2 is associated with the modulation of protein and lipid homeostasis of functionally diverse but linked pathways in response to oxidative stress.Cho KI et al
190737272009Herpes simplex virus replication: roles of viral proteins and nucleoporins in capsid-nucleus attachment.Copeland AM et al
82552971993Characterization of proteins that interact with the cell-cycle regulatory protein Ran/TC4.Coutavas E et al
183949932008Resolution of sister centromeres requires RanBP2-mediated SUMOylation of topoisomerase IIalpha.Dawlaty MM et al
93986621997RanGTP targets p97 to RanBP2, a filamentous protein localized at the cytoplasmic periphery of the nuclear pore complex.Delphin C et al
113533872001Genomic organization, expression, and localization of murine Ran-binding protein 2 (RanBP2) gene.Fauser S et al
191714222009SAGE analysis highlights the importance of p53csv, ddx5, mapkapk2 and ranbp2 to multiple myeloma tumorigenesis.Felix RS et al
97337661998The cyclophilin-like domain mediates the association of Ran-binding protein 2 with subunits of the 19 S regulatory complex of the proteasome.Ferreira PA et al
237462582013Sumoylation: a regulatory protein modification in health and disease.Flotho A et al
147299612004RanBP2/Nup358 provides a major binding site for NXF1-p15 dimers at the nuclear pore complex and functions in nuclear mRNA export.Forler D et al
158266662005Solution structure of the Ran-binding domain 2 of RanBP2 and its interaction with the C terminus of Ran.Geyer JP et al
236847492013Identification of candidate oncogenes in human colorectal cancers with microsatellite instability.Gylfe AE et al
218598632011Ran-dependent docking of importin-beta to RanBP2/Nup358 filaments is essential for protein import and cell viability.Hamada M et al
241131882013Down-modulation of nucleoporin RanBP2/Nup358 impaired chromosomal alignment and induced mitotic catastrophe.Hashizume C et al
240342522013Dynein recruitment to nuclear pores activates apical nuclear migration and mitotic entry in brain progenitor cells.Hu DJ et al
183051002008The Nup358-RanGAP complex is required for efficient importin alpha/beta-dependent nuclear import.Hutten S et al
192994632009The nuclear pore component Nup358 promotes transportin-dependent nuclear import.Hutten S et al
180706022008The nucleoporin Nup358 associates with and regulates interphase microtubules.Joseph J et al
150621032004The RanGAP1-RanBP2 complex is essential for microtubule-kinetochore interactions in vivo.Joseph J et al
118543052002SUMO-1 targets RanGAP1 to kinetochores and mitotic spindles.Joseph J et al
229599722012Crystal structure of the N-terminal domain of Nup358/RanBP2.Kassube SA et al
175459952007SUMO junction-what's your function? New insights through SUMO-interacting motifs.Kerscher O et al
250286982014A case report of epithelioid inflammatory myofibroblastic sarcoma with RANBP2-ALK fusion gene treated with the ALK inhibitor, crizotinib.Kimbara S et al
189460852009RanBP2 and SENP3 function in a mitotic SUMO2/3 conjugation-deconjugation cycle on Borealin.Klein UR et al
203850852010Gene regulation by nucleoporins and links to cancer.Köhler A et al
233451542014Epithelioid inflammatory myofibroblastic sarcoma arising in the pleural cavity.Kozu Y et al
92444461997Localization of the gene encoding the Ran-binding protein RanBP2 to human chromosome 2q11-q13 by fluorescence in situ hybridization.Krebber H et al
240348962013Inflammatory myofibroblastic tumor with RANBP2 and ALK gene rearrangement: a report of two cases and literature review.Li J et al
246132772014RANBP2-ALK fusion combined with monosomy 7 in acute myelomonocytic leukemia.Lim JH et al
233538302013Structural and functional analysis of the C-terminal domain of Nup358/RanBP2.Lin DH et al
204735212010Untreated recurrent acute necrotising encephalopathy associated with RANBP2 mutation, and normal outcome in a Caucasian boy.Loh NR et al
218927722011Emerging roles of the SUMO pathway in development.Lomelí H et al
126610112003Fusion of ALK to the Ran-binding protein 2 (RANBP2) gene in inflammatory myofibroblastic tumor.Ma Z et al
243075152014inv(2)(p23q13)/RAN-binding protein 2 (RANBP2)-ALK fusion gene in myeloid leukemia that developed in an elderly woman.Maesako Y et al
90194111997A small ubiquitin-related polypeptide involved in targeting RanGAP1 to nuclear pore complex protein RanBP2.Mahajan R et al
108626132000SUMO-1 conjugation to human DNA topoisomerase II isozymes.Mao Y et al
211642972011Epithelioid inflammatory myofibroblastic sarcoma: An aggressive intra-abdominal variant of inflammatory myofibroblastic tumor with nuclear membrane or perinuclear ALK.Mariño-Enríquez A et al
94563121998SUMO-1 modification and its role in targeting the Ran GTPase-activating protein, RanGAP1, to the nuclear pore complex.Matunis MJ et al
75931801995GTP hydrolysis by Ran occurs at the nuclear pore complex in an early step of protein import.Melchior F et al
123939062002Sumoylation of Mdm2 by protein inhibitor of activated STAT (PIAS) and RanBP2 enzymes.Miyauchi Y et al
191188152009Infection-triggered familial or recurrent cases of acute necrotizing encephalopathy caused by mutations in a component of the nuclear pore, RANBP2.Neilson DE et al
94807521998Identification of a novel Ran binding protein 2 related gene (RANBP2L1) and detection of a gene cluster on human chromosome 2q11-q12.Nothwang HG et al
214236732011HIV integration targeting: a pathway involving Transportin-3 and the nuclear pore protein RanBP2.Ocwieja KE et al
249091652015Nuclear translocation of IGF-1R via p150(Glued) and an importin-β/RanBP2-dependent pathway in cancer cells.Packham S et al
176562522007RANBP2 and CLTC are involved in ALK rearrangements in inflammatory myofibroblastic tumors.Patel AS et al
235365492013Kinesin-1 and mitochondrial motility control by discrimination of structurally equivalent but distinct subdomains in Ran-GTP-binding domains of Ran-binding protein 2.Patil H et al
117923252002The nucleoporin RanBP2 has SUMO1 E3 ligase activity.Pichler A et al
153780332004The RanBP2 SUMO E3 ligase is neither HECT- nor RING-type.Pichler A et al
163143932006Nuclear envelope breakdown is coordinated by both Nup358/RanBP2 and Nup153, two nucleoporins with zinc finger modules.Prunuske AJ et al
173722722007Mst1, RanBP2 and eIF4G are new markers for in vivo PI3K activation in murine and human prostate.Renner O et al
159312242005Insights into E3 ligase activity revealed by a SUMO-RanGAP1-Ubc9-Nup358 complex.Reverter D et al
223318472012Importin-β negatively regulates multiple aspects of mitosis including RANGAP1 recruitment to kinetochores.Roscioli E et al
91080471997RanBP2 associates with Ubc9p and a modified form of RanGAP1.Saitoh H et al
166888582006In situ SUMOylation analysis reveals a modulatory role of RanBP2 in the nuclear rim and PML bodies.Saitoh N et al
129637082003Nup358 integrates nuclear envelope breakdown with kinetochore assembly.Salina D et al
221551842012β-catenin inhibits promyelocytic leukemia protein tumor suppressor function in colorectal cancer cells.Satow R et al
221746922011HIV-1 capsid-cyclophilin interactions determine nuclear import pathway, integration targeting and replication efficiency.Schaller T et al
106013071999The zinc finger cluster domain of RanBP2 is a specific docking site for the nuclear export factor, exportin-1.Singh BB et al
203867262010Bicaudal D2, dynein, and kinesin-1 associate with nuclear pore complexes and regulate centrosome and nuclear positioning during mitotic entry.Splinter D et al
150376022004RanGAP1*SUMO1 is phosphorylated at the onset of mitosis and remains associated with RanBP2 upon NPC disassembly.Swaminathan S et al
162042162006SIZ1/SIZ2 control of chromosome transmission fidelity is mediated by the sumoylation of topoisomerase II.Takahashi Y et al
156086512005Unique binding interactions among Ubc9, SUMO and RanBP2 reveal a mechanism for SUMO paralog selection.Tatham MH et al
100785291999Structure of a Ran-binding domain complexed with Ran bound to a GTP analogue: implications for nuclear transport.Vetter IR et al
219957242012The nucleoporin Nup358/RanBP2 promotes nuclear import in a cargo- and transport receptor-specific manner.Wälde S et al
121051822002The cytoplasmic filaments of the nuclear pore complex are dispensable for selective nuclear protein import.Walther TC et al
224647302012The RanBP2/RanGAP1*SUMO1/Ubc9 complex is a multisubunit SUMO E3 ligase.Werner A et al
86036731995Localization of the Ran-GTP binding protein RanBP2 at the cytoplasmic side of the nuclear pore complex.Wilken N et al
77754811995Nup358, a cytoplasmically exposed nucleoporin with peptide repeats, Ran-GTP binding sites, zinc fingers, a cyclophilin A homologous domain, and a leucine-rich region.Wu J et al
195777362009Nuclear pore proteins and cancer.Xu S et al
104736101999GTP hydrolysis links initiation and termination of nuclear import on the nucleoporin nup358.Yaseen NR et al
76035721995A giant nucleopore protein that binds Ran/TC4.Yokoyama N et al
121920482002Enzymes of the SUMO modification pathway localize to filaments of the nuclear pore complex.Zhang H et al
211794832010Perturbation of host nuclear membrane component RanBP2 impairs the nuclear import of human immunodeficiency virus -1 preintegration complex (DNA).Zhang R et al
164693112006SUMO modification through rapamycin-mediated heterodimerization reveals a dual role for Ubc9 in targeting RanGAP1 to nuclear pore complexes.Zhu S et al

Other Information

Locus ID:

NCBI: 5903
MIM: 601181
HGNC: 9848
Ensembl: ENSG00000153201

Variants:

dbSNP: 5903
ClinVar: 5903
TCGA: ENSG00000153201
COSMIC: RANBP2

RNA/Proteins

Gene IDTranscript IDUniprot
ENSG00000153201ENST00000283195P49792
ENSG00000153201ENST00000425282F8WBP7
ENSG00000153201ENST00000629728F8WBP7

Expression (GTEx)

0
5
10
15
20
25
30
35
40

Pathways

PathwaySourceExternal ID
RNA transportKEGGko03013
RNA transportKEGGhsa03013
Nuclear pore complexKEGGhsa_M00427
Nuclear pore complexKEGGM00427
Metabolism of proteinsREACTOMER-HSA-392499
Post-translational protein modificationREACTOMER-HSA-597592
SUMOylationREACTOMER-HSA-2990846
SUMO E3 ligases SUMOylate target proteinsREACTOMER-HSA-3108232
SUMOylation of DNA damage response and repair proteinsREACTOMER-HSA-3108214
DiseaseREACTOMER-HSA-1643685
Infectious diseaseREACTOMER-HSA-5663205
HIV InfectionREACTOMER-HSA-162906
HIV Life CycleREACTOMER-HSA-162587
Late Phase of HIV Life CycleREACTOMER-HSA-162599
Rev-mediated nuclear export of HIV RNAREACTOMER-HSA-165054
Host Interactions of HIV factorsREACTOMER-HSA-162909
Interactions of Vpr with host cellular proteinsREACTOMER-HSA-176033
Vpr-mediated nuclear import of PICsREACTOMER-HSA-180910
Interactions of Rev with host cellular proteinsREACTOMER-HSA-177243
Nuclear import of Rev proteinREACTOMER-HSA-180746
Influenza InfectionREACTOMER-HSA-168254
Influenza Life CycleREACTOMER-HSA-168255
Transport of Ribonucleoproteins into the Host NucleusREACTOMER-HSA-168271
Influenza Viral RNA Transcription and ReplicationREACTOMER-HSA-168273
Viral Messenger RNA SynthesisREACTOMER-HSA-168325
Export of Viral Ribonucleoproteins from NucleusREACTOMER-HSA-168274
NEP/NS2 Interacts with the Cellular Export MachineryREACTOMER-HSA-168333
Host Interactions with Influenza FactorsREACTOMER-HSA-168253
NS1 Mediated Effects on Host PathwaysREACTOMER-HSA-168276
Immune SystemREACTOMER-HSA-168256
Cytokine Signaling in Immune systemREACTOMER-HSA-1280215
Interferon SignalingREACTOMER-HSA-913531
Antiviral mechanism by IFN-stimulated genesREACTOMER-HSA-1169410
ISG15 antiviral mechanismREACTOMER-HSA-1169408
Signal TransductionREACTOMER-HSA-162582
Signaling by Rho GTPasesREACTOMER-HSA-194315
RHO GTPase EffectorsREACTOMER-HSA-195258
RHO GTPases Activate ForminsREACTOMER-HSA-5663220
Gene ExpressionREACTOMER-HSA-74160
Processing of Capped Intron-Containing Pre-mRNAREACTOMER-HSA-72203
Transport of Mature Transcript to CytoplasmREACTOMER-HSA-72202
Transport of Mature mRNA derived from an Intron-Containing TranscriptREACTOMER-HSA-159236
Transport of Mature mRNAs Derived from Intronless TranscriptsREACTOMER-HSA-159234
Transport of the SLBP Dependant Mature mRNAREACTOMER-HSA-159230
Transport of the SLBP independent Mature mRNAREACTOMER-HSA-159227
Transport of Mature mRNA Derived from an Intronless TranscriptREACTOMER-HSA-159231
Gene Silencing by RNAREACTOMER-HSA-211000
Transcriptional regulation by small RNAsREACTOMER-HSA-5578749
Metabolism of non-coding RNAREACTOMER-HSA-194441
snRNP AssemblyREACTOMER-HSA-191859
Cell CycleREACTOMER-HSA-1640170
Cell Cycle, MitoticREACTOMER-HSA-69278
M PhaseREACTOMER-HSA-68886
Mitotic ProphaseREACTOMER-HSA-68875
Nuclear Envelope BreakdownREACTOMER-HSA-2980766
Nuclear Pore Complex (NPC) DisassemblyREACTOMER-HSA-3301854
Mitotic PrometaphaseREACTOMER-HSA-68877
Resolution of Sister Chromatid CohesionREACTOMER-HSA-2500257
Mitotic Metaphase and AnaphaseREACTOMER-HSA-2555396
Mitotic AnaphaseREACTOMER-HSA-68882
Separation of Sister ChromatidsREACTOMER-HSA-2467813
Transmembrane transport of small moleculesREACTOMER-HSA-382551
SLC-mediated transmembrane transportREACTOMER-HSA-425407
Hexose transportREACTOMER-HSA-189200
Glucose transportREACTOMER-HSA-70153
Regulation of Glucokinase by Glucokinase Regulatory ProteinREACTOMER-HSA-170822
MetabolismREACTOMER-HSA-1430728
Metabolism of carbohydratesREACTOMER-HSA-71387
Cellular responses to stressREACTOMER-HSA-2262752
Cellular response to heat stressREACTOMER-HSA-3371556
Regulation of HSF1-mediated heat shock responseREACTOMER-HSA-3371453
tRNA processingREACTOMER-HSA-72306
tRNA processing in the nucleusREACTOMER-HSA-6784531
SUMOylation of RNA binding proteinsREACTOMER-HSA-4570464
SUMOylation of DNA replication proteinsREACTOMER-HSA-4615885

Protein levels (Protein atlas)

Not detected
Low
Medium
High

References

Pubmed IDYearTitleCitations
159312242005Insights into E3 ligase activity revealed by a SUMO-RanGAP1-Ubc9-Nup358 complex.191
120320812002The SUMO E3 ligase RanBP2 promotes modification of the HDAC4 deacetylase.107
129637082003Nup358 integrates nuclear envelope breakdown with kinetochore assembly.81
88575421996Cyclophilin-related protein RanBP2 acts as chaperone for red/green opsin.62
156086512005Unique binding interactions among Ubc9, SUMO and RanBP2 reveal a mechanism for SUMO paralog selection.59
191188152009Infection-triggered familial or recurrent cases of acute necrotizing encephalopathy caused by mutations in a component of the nuclear pore, RANBP2.58
183051002008The Nup358-RanGAP complex is required for efficient importin alpha/beta-dependent nuclear import.55
178879602007Association of the kinesin-binding domain of RanBP2 to KIF5B and KIF5C determines mitochondria localization and function.50
211794832010Perturbation of host nuclear membrane component RanBP2 impairs the nuclear import of human immunodeficiency virus -1 preintegration complex (DNA).47
218598632011Ran-dependent docking of importin-beta to RanBP2/Nup358 filaments is essential for protein import and cell viability.46

Citation

Erica Di Cesare ; Patrizia Lavia

RANBP2 (RAN binding protein 2)

Atlas Genet Cytogenet Oncol Haematol. 2014-08-01

Online version: http://atlasgeneticsoncology.org/gene/483/ranbp2