G Model

ARTICLE IN PRESS

YSCBI-1117; No. of Pages 8

Seminars in Cancer Biology xxx (2014) xxx–xxx

Contents lists available at ScienceDirect

Seminars in Cancer Biology journal homepage: www.elsevier.com/locate/semcancer

Review

Nucleoporins and nucleocytoplasmic transport in hematologic malignancies Akiko Takeda, Nabeel R. Yaseen ∗ Department of Pathology and Immunology, Washington University in St. Louis, United States

a r t i c l e

i n f o

Keywords: Nucleoporin NUP98 NUP214 CRM1 XPO1 Leukemia Acute myeloid leukemia Acute lymphoblastic leukemia Nuclear export Nucleocytoplasmic transport

a b s t r a c t Hematologic malignancies are often associated with chromosomal rearrangements that lead to the expression of chimeric fusion proteins. Rearrangements of the genes encoding two nucleoporins, NUP98 and NUP214, have been implicated in the pathogenesis of several types of hematologic malignancies, particularly acute myeloid leukemia. NUP98 rearrangements result in fusion of an N-terminal portion of NUP98 to one of numerous proteins. These rearrangements often follow treatment with topoisomerase II inhibitors and tend to occur in younger patients. They have been shown to induce leukemia in mice and to enhance proliferation and disrupt differentiation in primary human hematopoietic precursors. NUP214 has only a few fusion partners. DEK-NUP214 is the most common NUP214 fusion in AML; it tends to occur in younger patients and is usually associated with FLT3 internal tandem duplications. The leukemogenic activity of NUP214 fusions is less well characterized. Normal nucleoporins, including NUP98 and NUP214, have important functions in nucleocytoplasmic transport, transcription, and mitosis. These functions and their disruptions by oncogenic nucleoporin fusions are discussed. © 2014 Elsevier Ltd. All rights reserved.

1. Introduction Hematologic malignancies are often associated with chromosomal rearrangements that lead to the expression of chimeric fusion proteins [1] (Fig. 1). Among the proteins that are known to be part of such oncogenic fusions, are two nucleoporins, NUP98 and NUP214. Nucleoporins are protein components of the nuclear pore complex (NPC). In conjunction with soluble shuttling carrier proteins (karyopherins), they play important roles in the nucleocytoplasmic transport of macromolecules [2–6]. Components of the nucleocytoplasmic transport machinery also play pivotal roles in other cellular processes such as mitosis and transcription [2–6]. Oncogenic nucleoporin fusions have been described in several types of hematologic malignancies, most commonly acute myeloid leukemia (AML), but also including myelodysplastic syndromes (MDS) and T-cell acute lymphoblastic leukemia (T-ALL). AML is a malignant proliferation of myeloid precursors characterized by failure of myeloid differentiation with accumulation of primitive cells (blasts) in the bone marrow and blood [7]. Myeloid malignancies

∗ Corresponding author at: Department of Pathology and Immunology, Washington University School of Medicine, 660 S. Euclid Avenue, Campus Box 8118, St. Louis, MO 63110, United States. Tel.: +1 314 362 0306. E-mail addresses: [email protected] (A. Takeda), [email protected] (N.R. Yaseen).

associated with NUP98 fusions often arise as a complication of prior chemotherapy with topoisomerase II inhibitors [8]. Nucleoporinassociated malignancies tend to occur at a younger age and have a poor clinical outcome [8–27].

2. Oncogenic NUP98 fusions At least 29 NUP98 fusions have been reported in hematopoietic malignancies, mostly AML, but also including T-ALL and MDS [8–22,28–30] (Table 1). In most cases the breakpoints occur in introns 11–13 of NUP98, resulting in fusion of the N-terminal region of NUP98 that is rich in phenylalanine–glycine (FG) repeats to one of 29 different proteins [30]. Many of the NUP98 fusion partners are transcription factors of the homeobox family; the prototype of such fusions is NUP98-HOXA9 [31,32]. The overall incidence of NUP98 fusions is not clear and there is evidence that it may vary by geographical region [30]. For example, the vast majority of NUP98HOXA9 fusions have been reported in patients from the Far East [8,26,33,34]. NUP98-NSD1 has been reported in 16.1% of pediatric and 2.3% of adult cytogenetically normal AML [35]. NUP98-JARID1A was identified in 10.5% of cases of pediatric acute megakaryoblastic leukemia (a subset of AML) [36]. A study from Taiwan identified NUP98-HOXA9 in 2.23% of adult AML patients [26]. NUP98 fusions cause aberrant differentiation and increased proliferation when expressed in primary human hematopoietic

http://dx.doi.org/10.1016/j.semcancer.2014.02.009 1044-579X/© 2014 Elsevier Ltd. All rights reserved.

Please cite this article in press as: Takeda A, Yaseen NR. Nucleoporins and nucleocytoplasmic transport in hematologic malignancies. Semin Cancer Biol (2014), http://dx.doi.org/10.1016/j.semcancer.2014.02.009

G Model

ARTICLE IN PRESS

YSCBI-1117; No. of Pages 8

A. Takeda, N.R. Yaseen / Seminars in Cancer Biology xxx (2014) xxx–xxx

2

t(6;9)(p23;q34)

Normal

cells [37–40]. When introduced into mice by transplantation or transgenically, they cause myeloproliferation, MDS and AML with relatively long latency and variable penetrance, suggesting that additional cooperating oncogenic events are needed for the development of full-blown leukemia [16,41–49]. This notion is supported by clinical data as well as in vivo studies in mice. NUP98 rearrangements have been described in patients with BCR-ABL1positive chronic myelogenous leukemia who developed blast crisis, which is a form of AML, suggesting that NUP98 fusions can cooperate with BCR-ABL1 in causing AML [50]. Further, patients with hematologic malignancies associated with NUP98 fusions have an increased incidence of additional mutations such as FLT3 internal tandem duplications (FLT3-ITD), KIT, WT1 and KRAS [26,35,51]. Several mouse studies have shown that oncogenes, including Meis1, FLT3, and BCR-ABL1, can cooperate with NUP98 fusions and enhance their leukemogenic potential [41,42,47,52–58]. 3. Oncogenic NUP214 fusions

Chr. 6

Chr. 9

der(6)

der(9)

Fig. 1. Schematic of the t(6;9)(p23;q34) chromosomal rearrangement that results in the DEK-NUP214 oncogenic fusion. On the left are normal chromosomes 6 and 9; the Giemsa banding pattern is shown in brown and blue, respectively. The centromeres are shown in red and the non-centromeric heterochromatin of chromosome 9 is shown in pink. On the right are shown the derivative chromosomes 6 and 9 that result from the t(6;9)(p23;q34) chromosomal rearrangement. The arrowheads indicate the chromosomal breakpoints. Table 1 Nucleoporin gene rearrangements in hematologic malignancies. AML = acute myeloid leukemia, CML-BC = chronic myelogenous leukemia in blast crisis, MDS = myelodysplastic syndrome, t-AML = therapy-related acute myeloid leukemia, t-MDS = therapy-related myelodysplastic syndrome, APL = acute promyelocytic leukemia, T-ALL = T-cell Acute lymphoblastic leukemia, B-ALL = B-cell acute lymphoblastic leukemia, and AUL = acute undifferentiated leukemia. Rearrangement

Fusion transcript

Disease

NUP98 t(7;11)(p15;p15)

NUP98-HOXA9

t(7;11)(p15;p15) t(7;11)(p15;p15) t(11;12)(p15;q13) t(11;12)(p15;q13) t(2;11)(q35;p15) t(2;11)(q31;p15) t(1;11)(q23;p15) t(9;11)(q34;p15) t(10;11)(q23;p15) inv(11)(p15;q22) t(11;20)(p15;q11) t(9;11)(p22;p15) t(5;11)(q31;p15) t(8;11)(p11.2;p15) t(3;11)(p24;p15) Complex (12p13) t(11;17)(p15;p13) Complex (3p25) t(6;11)(q24.1;p15.5)

NUP98-HOXA11 NUP98-HOXA13 NUP98-HOXC11 NUP98-HOXC13 NUP98-HOXD11 NUP98-HOXD13 NUP98-PMX1 NUP98-PRRX2 NUP98-HHEX NUP98-DDX10 NUP98-TOP1 NUP98-PSIP1 NUP98-NSD1 NUP98-NSD3 NUP98-TOP2B NUP98-JARID1A NUP98-PHF23 NUP98-ANKRD28 NUP98-CCDC28A

Complex (3q29) t(11;18)(p15;q12) t(4;11)(q21;p15) t(10;11)(q25;p15) t(X;11)(q28;p15) t(3;11)(q12;p15) inv(11)(p15q23) t(11;12)(p15;q13) t(3;11)(p11;p15)

NUP98-IQCG NUP98-SETBP1 NUP98-RAP1GDS1 NUP98-Adducin 3 NUP98-HMGB3 NUP98-LOC348801 NUP98-MLL NUP98-RARG NUP98-POU1F1

AML/MDS, t-AML/MDS, CML CML-BC AML, MDS AML AML Pediatric AML AML, t-AML AML, t-MDS/AML t-AML AML AML, MDS, CML AML, t-MDS AML, MDS Pediatric AML AML AML (Monoblastic) AML (Megakaryoblastic) AML MDS/AML AML (Megakaryoblastic), T-ALL Biphenotypic T-ALL/AML Pediatric T-ALL Adult T-ALL T-ALL t-AML AML AML APL t-AML

DEK-NUP214 SET-NUP214 SQSTM1-NUP214 NUP214-ABL1

AML T-ALL, AML, AUL T-ALL T-ALL, B-ALL

NUP214 t(6;9)(p23;q34) del(9)(q34) der(5)t(5;9)(q35;q34) Amplified episomes

NUP214 was the first nucleoporin to be implicated in the pathogenesis of hematologic malignancies [59]. The translocation t(6;9)(p23;q34) (Fig. 1) results in a DEK-NUP214 fusion and defines a specific subcategory of AML under the most recent World Health Organization classification of AML [23]. This subtype of AML is characterized by basophilia, poor clinical outcome, and a high incidence of FLT3-ITD [23,60–62]. The breakpoints always occur within the same introns of both DEK and NUP214, resulting in an invariant fusion protein that includes the C-terminal FG repeat region of NUP214 [25,63]. SET-NUP214 is a fusion between exon 7 of SET and exon 18 of NUP214 that results from del(9)(q34). It is associated with T-ALL and less frequently with AML and acute undifferentiated leukemia [64–70]. Most reported cases in the literature are from the Far East [64–70]. Fusion SQSTM1-NUP214 has been reported in a patient with refractory T-ALL [71]. Recently, a NUP214-ABL1 fusion was described in a series of patients with T-ALL [72,73]. This fusion is cytogenetically cryptic and is often located on amplified episomes. In contrast to other NUP214 fusions, NUP214-ABL1 fusions contain an N-terminal portion of NUP214 that includes some but not all of the FG repeats. This fusion has also been described in B cell acute lymphoblastic leukemia [74,75]. In T-ALL, NUP214-ABL1 is usually associated with rearrangement and/or overexpression of TLX1 or TLX3 and/or deletion of the tumor suppressor CDKN2a [72,76]. In some patients there are more cells with abnormalities of these genes than with NUP214-ABL1, suggesting that the latter is a secondary mutation [76]. In transduction/transplantation studies, DEK-NUP214 induced AML in mice with low penetrance and long latency; the penetrance was enhanced and the latency shortened by enriching the transduced population for long-term hematopoietic stem cells [77]. Conflicting data have been reported on the effects of DEK-NUP214 in human myeloid cell lines. In one study DEK-NUP214 caused mild growth inhibition in the myeloid cell line U937 [78], whereas in a subsequent study DEK-NUP214 was found to enhance proliferation of U937 and another myeloid cell line, PL-21, through upregulation of the mTOR pathway [79]. SET-NUP214 transgenic mice showed some abnormalities in hematopoietic differentiation and expansion of early precursors but no clear-cut leukemic phenotype [80,81]. 4. Subcellular localization of nucleoporin fusions Normally, NUP214 is present primarily on the cytoplasmic face of the NPC [59], whereas NUP98 is present on both the nuclear and cytoplasmic sides of the NPC as well as within the nucleus [82–84].

Please cite this article in press as: Takeda A, Yaseen NR. Nucleoporins and nucleocytoplasmic transport in hematologic malignancies. Semin Cancer Biol (2014), http://dx.doi.org/10.1016/j.semcancer.2014.02.009

G Model

ARTICLE IN PRESS

YSCBI-1117; No. of Pages 8

A. Takeda, N.R. Yaseen / Seminars in Cancer Biology xxx (2014) xxx–xxx

A

CRM1

Cytoplasm

NES

Ran GDP

3

B Cytoplasm

Txn Factor

NUP214 CRM1

NES RanGTP

NPC

NPC

NPC

NPC

Txn Factor NUP98 CRM1

NES RanGTP

Txn Factor

NUP98 CRM1

NES RanGTP

Txn Factor DNA

?

NUP98 CRM1

NES RanGTP

Txn Factor DNA

HOXA9

?

Fig. 2. CRM1-mediated nuclear export and hypothetical role of CRM1 in transcription. (A) CRM1 binds to NES-bearing cargo, such as transcription factors, in the presence of RanGTP. NUP98 facilitates the transport of the export complex by interacting with the NPC through its C-terminal portion. The export complex also interacts with NUP214 on its way toward the cytoplasm. On the cytoplasmic side of the NPC, GTP hydrolysis results in conversion of RanGTP to RanGDP, which results in dissociation of the export complex and release of the cargo into the cytoplasm. In the pink oval is shown a hypothetical model for a role of the export complex containing a transcription factor, CRM1, NUP98, and RanGTP in transcription. Under normal conditions, this complex would be in equilibrium between DNA binding and export. (B) In leukemias associated with a NUP98 fusion, the C-terminal portion of NUP98 is lost, precluding its interaction with the NPC and resulting in sequestration of the export complex inside the nucleus. According to the suggested model, the equilibrium is shifted in favor of the export complex remaining bound to DNA, resulting in increased transcriptional activity.

In both DEK-NUP214 and SET-NUP214, the NPC-targeting coiled-coil domains of NUP214 are lost, resulting intranuclear localization of the fusion protein [85]. In contrast, NUP214-ABL1 retains the coiled-coil domains resulting in its localization to the NPC [86]. NPC localization appears necessary for the transforming ability of NUP214-ABL1 [86]. The NPC-targeting sequences of NUP98 reside in its C-terminal portion [87–89], which is lost in oncogenic NUP98 fusions. As a result, NUP98 fusions do not localize to the nuclear rim and are instead localized inside the nucleus in a punctate distribution that has been observed both in transduced primary human cells [40,90] and in cells from patients with AML [35,91].

including transcription factors [90]. Similarly, SET-NUP214 binds to CRM1, causes aberrant punctate intranuclear localization of CRM1, and inhibits CRM1-mediated nuclear protein export [104]. Further, our unpublished data show that the oncogenic fusion DEK-NUP214 similarly interacts with, and mislocalizes, CRM1. These findings raise the possibility that nuclear retention of transcriptional regulators that are normally exported from the nucleus by CRM1 may contribute to the dysregulation of transcription by nucleoporin fusions. There is evidence that NUP98 and NUP214 are involved in RNA export and protein import [105–118]. However there is no evidence to date that nucleoporin fusions interfere with these processes.

5. Nucleoporins and nucleocytoplasmic transport

6. Nucleoporins in mitosis

The mammalian NPC is a very large structure with 8-fold symmetry that consists of approximately 30 nucleoporins, each present in multiple copies. It is the gateway for the exchange of macromolecules between the nucleus and the cytoplasm [2,92–95]. In addition to nucleoporins, nucleocytoplasmic transport generally requires carrier proteins that bind to cargo and facilitate its transport through the NPC. Different carriers mediate the import and export of different subsets of proteins and RNAs [6,93,96]. A number of nucleoporins, including NUP98 and NUP214, contain FG repeats and function in nucleocytoplasmic transport by interacting with these carriers [93]. Both NUP98 and NUP214 play a role in the nuclear export of proteins by binding to the nuclear export carrier CRM1 (XPO1) [6,90,97–101] (Fig. 2A). CRM1 recognizes leucine-rich nuclear export sequences (NESs) on cargo proteins and binds to them in the presence of the GTP-bound form of the small GTPase Ran (Ran-GTP). The export complex passes through the NPC out to the cytoplasm where GTP hydrolysis results in dissociation of the cargo from CRM1 [102,103] (Fig. 2A). NUP98 fusions bind to CRM1 resulting in its mislocalization to punctate intranuclear domains. This is associated with a block of CRM1-mediated nuclear export of proteins

Recent evidence indicates that some nucleoporins, in addition to their roles in nucleocytoplasmic transport, play significant roles during mitosis. An extensive review of the roles of nucleoporins in mitosis has been published recently [4]. Here, we will focus on recent data pertaining to the roles of NUP98, NUP214, and oncogenic nucleoporin fusions in mitosis. Phosphorylation of NUP98 is an important rate-limiting step in NPC disassembly during mitosis [119]. Nup98 regulates bipolar spindle assembly through association of its C-terminal domain with microtubules [120]. Nup98, in complex with the mRNA export factor Rae1, is one of the modulators of mitotic spindle assembly checkpoint (SAC). The Nup98/Rae1 complex regulates timely destruction of securin by the anaphase-promoting complex (APC), and combined Rae1 and Nup98 haploinsufficiency in mice results in premature separation of sister chromatids, severe aneuploidy and untimely degradation of securin [121]. RNAi-mediated knockdown of NUP98 similarly causes severe chromosome segregation defects as well as disrupted RAE1 expression and localization [122]. Nup214 and Nup88, that normally form a complex at the NPC, are localized at the spindle during mitosis [123]. Other nucleoporins also play critical roles in mitosis through interactions with a large

Please cite this article in press as: Takeda A, Yaseen NR. Nucleoporins and nucleocytoplasmic transport in hematologic malignancies. Semin Cancer Biol (2014), http://dx.doi.org/10.1016/j.semcancer.2014.02.009

G Model YSCBI-1117; No. of Pages 8

ARTICLE IN PRESS A. Takeda, N.R. Yaseen / Seminars in Cancer Biology xxx (2014) xxx–xxx

4

network of proteins at both the kinetochore and the centrosome [124,125]. There is some evidence that NUP98 fusions may play an aberrant role in mitosis. Expression of NUP98-HOXA9 causes downregulation and mislocalization of RAE1 protein [122]. NUP98homeodomain fusions interact with endogenous NUP98 during interphase but during mitosis they localize to kinetochores and chromosome arms without endogenous NUP98 [126]. The network that connects nucleoporins to kinetochores and centrosomes includes the cytoplasmic dynein motor complex [124,125,127]. Interestingly, one dynein light chain, DYNLT1, has been found to specifically associate with NUP98-HOXA9 as well as wildtype NUP98 and other FG repeat-containing nucleoporins, and to play a role in the dysregulation of gene expression and induction of hematopoietic cell proliferation by NUP98-HOXA9 [128]. These data raise the possibility that, in patients with NUP98 rearrangements, oncogenic NUP98 fusions and deficiency of wild-type NUP98 may contribute to oncogenic transformation by causing defective chromosomal segregation. However, it remains to be determined whether patient cells with NUP98 fusions express less endogenous NUP98 and whether they show abnormalities in chromosomal segregation. In addition to nucleoporins, soluble carrier proteins involved in nucleocytoplasmic transport, including karyopherin ␤1 (importin ␤1) and CRM1, as well as the small GTPase Ran, play an important role in mitosis [124]. CRM1, independently from its well-known role in nuclear export, appears to be involved in maintenance of centrosome integrity and microtubule nucleation from kinetochores. It is of particular interest because one of its inhibitors, KPT-330 [129], is currently in clinical trials for the treatment of hematopoietic malignancies [130]. A fraction of CRM1 localizes to the centrosomes and is involved in maintaining centrosome integrity [131]. The mechanism by which CRM1 plays this role is not clearly understood. It is possible that CRM1 cargoes such as nucleophosmin (NPM1), by binding to CRM1 via their NES, play a role in maintaining proper centrosome duplication [132,133]. CRM1 is also found at the kinetochore during mitosis and it recruits NUP358 (RanBP2) and RanGAP1 to mitotic kinetochores in the presence of RanGTP [134]. CRM1 is phosphorylated by CDK1/Cyclin B at serine 391 during mitosis, which enhances its RanGTP-dependent interaction with RanGAP1-RanBP2 and promotes their recruitment to the mitotic spindle for proper assembly [135]. Another important role of CRM1 in mitosis is to tether the chromosomal passenger complex (CPC) to the centromere by interacting with the NES of survivin in early prophase [136]. The CPC is required at various stages during mitosis and cytokinesis [137]. As discussed above, NUP98 and NUP214 oncogenic fusions interact with CRM1 resulting in its mislocalization. These interactions may play a role in oncogenesis by perturbing the normal mitotic process.

7. Nucleoporins in transcription NUP98 is a mobile nucleoporin whose mobility is linked to ongoing transcriptional activity [83]. The first evidence for a direct transcriptional role of nucleoporins was that the FG repeat regions of NUP98 and NUP214 can recruit CBP/p300 and act as transactivating domains when linked to a GAL4 DNA-binding domain [138]. The FG repeat region of NUP98 can recruit CBP/p300 to activate transcription or HDAC1 to repress transcription [138,139]. Data from both human and Drosophila cells showed that wild-type nucleoporins, including NUP98, bind to chromatin at transcriptionally active genes and regulate transcription [140–144]. Binding of NUP98 to gene regulatory elements was demonstrated in human HeLa cells where it is required for IFN␥-mediated transcriptional memory through dimethylation of H3K4 [145]. At least part of the

chromatin binding activity of NUP98 resides in its FG repeat region, and this binding appears to preferentially correlate with increased gene expression [143]. Similarly, the FG repeat-containing nucleoporin NUP153 and another nucleoporin, Mtor/TPR, have been shown to interact with the dosage compensation complex (DCC) containing MSL proteins and the MOF histone acetyl transferase and are required for the targeting of MSL proteins to the X chromosome [140]. The DCC is responsible for acetylation of H4K16 on the male X chromosome resulting in chromatin decondensation and increased gene expression [146]. Depletion of NUP153 resulted in downregulation of dosage-compensated genes [140]. These findings were later expanded to show genome-wide association of NUP153 and Mtor with chromatin at regions that are enriched for active genes, RNA polymerase II binding, and H4K16 acetylation [144]. The mechanisms by which nucleoporins are targeted to chromatin, however, remain unclear. All NUP98 fusions, and most NUP214 fusions include the FG repeats that can regulate transcription by recruiting coactivators and corepressors as discussed above. In a number of NUP98 fusions, the fusion partner supplies a DNA-binding domain. At least 10 NUP98 rearrangements result in fusion of NUP98 to a homeobox transcription factor, and the fusion product always includes the DNA-binding homeodomain [30]. There is evidence that in these cases the homeodomain is required for DNA binding, dysregulated transcription, and leukemic transformation [38,41,42,44,47,48,138,139,147–150]. Other NUP98 fusion partners contain chromatin-interacting motifs. NUP98-JARID1A and NUP98-PHF23 use their PHD fingers to bind to H3K4me3 and upregulate expression of HOXA genes [45]. Similarly, NUP98-NSD1 was shown to bind to the HOXA locus and upregulate the expression of HOX transcription factors through maintenance of H3K36 methylation and histone acetylation [44]. The region of NSD1 encompassing the 5th PHD finger (PHD5) and the adjacent Cys-His-rich domain (C5HCH) is required for binding to the HOXA locus [44]. While the chromatin component bound by the PHD5-C5HCH motif of NSD1 is not known, the corresponding PHD5-C5HCH motif of NSD3 binds to unmodified H3K4 and trimethylated H3K9 through the PHD5 domain [151]. Of note, a NUP98-NSD3 fusion has been reported in AML and MDS [152,153]. On the other hand, there is no evidence that the remaining nucleoporin fusions (Table 1) contain a DNA-binding domain. This raises the question of how these fusions, such as NUP98-DDX10 [40], can dysregulate transcription. One possibility, discussed above, is interference with the nuclear export of transcription factors such as NF␬B [90]. Another possibility is that the oncogenic fusion can be targeted to chromatin through the nucleoporin moiety, regardless of the fusion partner. This possibility is supported by the findings described above showing that wild type nucleoporins associate with chromatin. However, the mechanism by which NUP98 and other nucleoporins are targeted to chromatin remains unclear. One possible model is illustrated in Fig. 2A. NUP98 binds to CRM1, which in turn is known to bind numerous transcription factors in its capacity as a nuclear export carrier [98,154–161]. It is possible that these interactions also occur in the context of chromatin, with CRM1 and its associated transcription factors cooperating with NUP98 in binding to chromatin and transcriptional regulation (Fig. 2A). With NUP98 fusions, on the other hand, the C-terminal portion of NUP98, which is responsible for interaction with the NPC, is lost resulting in a block of CRM1-mediated nuclear export [90]. Under the proposed model, the export complex would remain in the nucleus, favoring its association with chromatin, leading to increased transcriptional activity (Fig. 2B). As discussed above, NUP214 and its fusions also bind to CRM1 and inhibit CRM1-mediated nuclear export. It is possible that NUP214 regulates transcription and that its fusions dysregulate transcription by mechanisms similar to those proposed in Fig. 2.

Please cite this article in press as: Takeda A, Yaseen NR. Nucleoporins and nucleocytoplasmic transport in hematologic malignancies. Semin Cancer Biol (2014), http://dx.doi.org/10.1016/j.semcancer.2014.02.009

G Model YSCBI-1117; No. of Pages 8

ARTICLE IN PRESS A. Takeda, N.R. Yaseen / Seminars in Cancer Biology xxx (2014) xxx–xxx

8. Inhibitors of nuclear export Several inhibitors of CRM1-mediated nuclear export are under development for the treatment of cancer [102,130,162,163]. These inhibitors generally act by binding to the cysteine residue 528 at the active site of CRM1, which disrupts the interaction of CRM1 with its cargo and results in a block of CRM1-mediated nuclear export [102]. Of the many CRM1 inhibitors under development, KPT-330 is currently in Phase 1 trials in solid tumors as well as in hematologic malignancies, including AML [130,163]. Inhibition of the nuclear export of tumor suppressors such as p53, APC, and retinoblastoma has been suggested as a possible mechanism for the antitumor activity of CRM1 inhibitors [102,164]. The roles of CRM1 in mitosis may provide additional targets for CRM1 inhibitors in cancer therapy. For example, the CRM1 inhibitor leptomycin B (LMB) prevents the recruitment of NUP358/RanBP2 to the kinetochore and disrupts mitotic progression and chromosome segregation [134]. On the other hand, if CRM1 indeed cooperates with nucleoporin fusions in dysregulating transcription as proposed in the model shown in Fig. 2B, patients with nucleoporin fusions may respond preferentially to treatment with CRM1 inhibitors. In addition, a subset of AML is characterized by mutations in the nucleolar protein nucleophosmin (NPM1) that result in the formation of an extra NES recognized by CRM1 [165]. This results in aberrant cytoplasmic localization of NPM1 [166] that is thought to contribute to leukemogenesis [167]. It has been shown that CRM1 inhibition can reverse the aberrant cytoplasmic localization of mutant NPM1 [90]. It would be of great interest to determine whether CRM1 inhibitors are effective in treating patients with nucleoporin fusions and NPM1 mutations. Conflict of interest Our laboratory has received research funding from Karyopharm Therapeutics, which develops nuclear export inhibitors for clinical use. References [1] Scandura JM, Boccuni P, Cammenga J, Nimer SD. Transcription factor fusions in acute leukemia: variations on a theme. Oncogene 2002;21:3422–44. [2] Raices M, D’Angelo MA. Nuclear pore complex composition: a new regulator of tissue-specific and developmental functions. Nat Rev Mol Cell Biol 2012;13:687–99. [3] Chatel G, Fahrenkrog B. Dynamics and diverse functions of nuclear pore complex proteins. Nucleus 2012;3:162–71. [4] Chatel G, Fahrenkrog B. Nucleoporins: leaving the nuclear pore complex for a successful mitosis. Cell Signal 2011;23:1555–62. [5] Capelson M, Doucet C, Hetzer MW. Nuclear pore complexes: guardians of the nuclear genome. Cold Spring Harb Symp Quant Biol 2010;75:585–97. [6] Hutten S, Kehlenbach RH. CRM1-mediated nuclear export: to the pore and beyond. Trends Cell Biol 2007;17:193–201. [7] Bain BJ. Leukaemia diagnosis. 2nd ed. London: Blackwell Science; 1999. [8] Romana SP, Radford-Weiss I, Ben Abdelali R, Schluth C, Petit A, Dastugue N, et al. NUP98 rearrangements in hematopoietic malignancies: a study of the Groupe Francophone de Cytogenetique Hematologique. Leukemia 2006;20:696–706. [9] Tosi S, Ballabio E, Teigler-Schlegel A, Boultwood J, Bruch J, Harbott J. Characterization of 6q abnormalities in childhood acute myeloid leukemia and identification of a novel t(6;11)(q24.1;p15.5) resulting in a NUP98-C6orf80 fusion in a case of acute megakaryoblastic leukemia. Genes Chromosomes Cancer 2005;44:225–32. [10] Nebral K, Schmidt HH, Haas OA, Strehl S. NUP98 Is Fused to Topoisomerase (DNA) II{beta} 180 kDa (TOP2B) in a patient with acute myeloid leukemia with a new t(3;11)(p24;p15). Clin Cancer Res 2005;11:6489–94. [11] van Zutven LJ, Onen E, Velthuizen SC, van Drunen E, von Bergh AR, van den Heuvel-Eibrink MM, et al. Identification of NUP98 abnormalities in acute leukemia: JARID1A (12p13) as a new partner gene. Genes Chromosomes Cancer 2006;45:437–46. [12] Panagopoulos I, Kerndrup G, Carlsen N, Strombeck B, Isaksson M, Johansson B. Fusion of NUP98 and the SET binding protein 1 (SETBP1) gene in a paediatric acute T cell lymphoblastic leukaemia with t(11;18)(p15;q12). Br J Haematol 2007;136:294–6.

5

[13] Reader JC, Meekins JS, Gojo I, Ning Y. A novel NUP98-PHF23 fusion resulting from a cryptic translocation t(11;17)(p15;p13) in acute myeloid leukemia. Leukemia 2007;21:842–4. [14] Pan Q, Zhu YJ, Gu BW, Cai X, Bai XT, Yun HY, et al. A new fusion gene NUP98-IQCG identified in an acute T-lymphoid/myeloid leukemia with a t(3;11)(q29q13;p15)del(3)(q29) translocation. Oncogene 2008;27:3414–23. [15] Ishikawa M, Yagasaki F, Okamura D, Maeda T, Sugahara Y, Jinnai I, et al. A novel gene, ANKRD28 on 3p25, is fused with NUP98 on 11p15 in a cryptic 3-way translocation of t(3;5;11)(p25;q35;p15) in an adult patient with myelodysplastic syndrome/acute myelogenous leukemia. Int J Hematol 2007;86:238–45. [16] Jankovic D, Gorello P, Liu T, Ehret S, La Starza R, Desjobert C, et al. Leukemogenic mechanisms and targets of a NUP98/HHEX fusion in acute myeloid leukemia. Blood 2008;111:5672–82. [17] Yamamoto M, Kakihana K, Kurosu T, Murakami N, Miura O. Clonal evolution with inv(11)(p15q22) and NUP98/DDX10 fusion gene in imatinib-resistant chronic myelogenous leukemia. Cancer Genet Cytogenet 2005;157:104–8. [18] Gorello P, Brandimarte L, La Starza R, Pierini V, Bury L, Rosati R, et al. t(3;11)(q12;p15)/NUP98-LOC348801 fusion transcript in acute myeloid leukemia. Haematologica 2008;93:1398–401. [19] Kaltenbach S, Soler G, Barin C, Gervais C, Bernard OA, Penard-Lacronique V, et al. A NUP98-MLL fusion in human acute myeloblastic leukemia. Blood 2010;116:2332–5. [20] Petit A, Ragu C, Della-Valle V, Mozziconacci MJ, Lafage-Pochitaloff M, Soler G, et al. NUP98-HMGB3: a novel oncogenic fusion. Leukemia 2010;24:654–8. [21] Slape C, Lin YW, Hartung H, Zhang Z, Wolff L, Aplan PD. NUP98-HOX translocations lead to myelodysplastic syndrome in mice and men. J Natl Cancer Inst Monogr 2008;2008:64–8. [22] Such E, Cervera J, Valencia A, Barragan E, Ibanez M, Luna I, et al. A novel NUP98/RARG gene fusion in acute myeloid leukemia resembling acute promyelocytic leukemia. Blood 2011;117:242–5. [23] Arber DA, Brunning RD, Le Beau MM, Falini B, Vardiman JW, Porwit A, et al. Acute myeloid leukemia with recurrent genetic abnormalities. In: Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H, et al, editors. WHO classification of tumours of haematopoietic and lymphoid tissues. 4th ed. Lyon: International Agency for Research on Cancer; 2008. p. 110–23. [24] Chi Y, Lindgren V, Quigley S, Gaitonde S. Acute myelogenous leukemia with t(6;9)(p23;q34) and marrow basophilia: an overview. Arch Pathol Lab Med 2008;132:1835–7. [25] Garcon L, Libura M, Delabesse E, Valensi F, Asnafi V, Berger C, et al. DEKCAN molecular monitoring of myeloid malignancies could aid therapeutic stratification. Leukemia 2005;19:1338–44. [26] Chou WC, Chen CY, Hou HA, Lin LI, Tang JL, Yao M, et al. Acute myeloid leukemia bearing t(7;11)(p15;p15) is a distinct cytogenetic entity with poor outcome and a distinct mutation profile: comparative analysis of 493 adult patients. Leukemia 2009;23:1303–10. [27] Grimwade D, Hills RK, Moorman AV, Walker H, Chatters S, Goldstone AH, et al. Refinement of cytogenetic classification in acute myeloid leukemia: determination of prognostic significance of rare recurring chromosomal abnormalities among 5876 younger adult patients treated in the United Kingdom Medical Research Council trials. Blood 2010;116:354–65. [28] Yamamoto K, Nakamachi Y, Yakushijin K, Funakoshi Y, Okamura A, Kawano S, et al. Expression of the novel NUP98/PSIP1 fusion transcripts in myelodysplastic syndrome with t(9;11)(p22;p15). Eur J Haematol 2012;88: 244–8. [29] Lisboa S, Cerveira N, Bizarro S, Correia C, Vieira J, Torres L, et al. POU1F1 is a novel fusion partner of NUP98 in acute myeloid leukemia with t(3;11)(p11;p15). Mol Cancer 2013;12:5. [30] Gough SM, Slape CI, Aplan PD. NUP98 gene fusions and hematopoietic malignancies: common themes and new biologic insights. Blood 2011;118:6247–57. [31] Borrow J, Shearman AM, Stanton Jr VP, Becher R, Collins T, Williams AJ, et al. The t(7;11)(p15;p15) translocation in acute myeloid leukaemia fuses the genes for nucleoporin NUP98 and class I homeoprotein HOXA9. Nat Genet 1996;12:159–67. [32] Nakamura T, Largaespada DA, Lee MP, Johnson LA, Ohyashiki K, Toyama K, et al. Fusion of the nucleoporin gene NUP98 to HOXA9 by the chromosome translocation t(7;11)(p15;p15) in human myeloid leukaemia. Nat Genet 1996;12:154–8. [33] Huang SY, Tang JL, Liang YJ, Wang CH, Chen YC, Tien HF. Clinical, haematological and molecular studies in patients with chromosome translocation t(7;11): a study of four Chinese patients in Taiwan. Br J Haematol 1997;96:682–7. [34] Kwong YL, Pang A. Low frequency of rearrangements of the homeobox gene HOXA9/t(7;11) in adult acute myeloid leukemia. Genes Chromosomes Cancer 1999;25:70–4. [35] Hollink IH, van den Heuvel-Eibrink MM, Arentsen-Peters ST, Pratcorona M, Abbas S, Kuipers JE, et al. NUP98/NSD1 characterizes a novel poor prognostic group in acute myeloid leukemia with a distinct HOX gene expression pattern. Blood 2011;118:3645–56. [36] de Rooij JD, Hollink IH, Arentsen-Peters ST, van Galen JF, Berna Beverloo H, Baruchel A, et al. NUP98/JARID1A is a novel recurrent abnormality in pediatric acute megakaryoblastic leukemia with a distinct HOX gene expression pattern. Leukemia 2013;27:2280–8. [37] Chung KY, Morrone G, Schuringa JJ, Plasilova M, Shieh JH, Zhang Y, et al. Enforced expression of NUP98-HOXA9 in human CD34(+) cells enhances stem cell proliferation. Cancer Res 2006;66:11781–91.

Please cite this article in press as: Takeda A, Yaseen NR. Nucleoporins and nucleocytoplasmic transport in hematologic malignancies. Semin Cancer Biol (2014), http://dx.doi.org/10.1016/j.semcancer.2014.02.009

G Model YSCBI-1117; No. of Pages 8 6

ARTICLE IN PRESS A. Takeda, N.R. Yaseen / Seminars in Cancer Biology xxx (2014) xxx–xxx

[38] Yassin ER, Sarma NJ, Abdul-Nabi AM, Dombrowski J, Han Y, Takeda A, et al. Dissection of the transformation of primary human hematopoietic cells by the oncogene NUP98-HOXA9. PLoS ONE 2009;4:e6719. [39] Takeda A, Goolsby C, Yaseen NR. NUP98-HOXA9 induces long-term proliferation and blocks differentiation of primary human CD34+ hematopoietic cells. Cancer Res 2006;66:6628–37. [40] Yassin ER, Abdul-Nabi AM, Takeda A, Yaseen NR. Effects of the NUP98-DDX10 oncogene on primary human CD34+ cells: role of a conserved helicase motif. Leukemia 2010;24:1001–11. [41] Pineault N, Buske C, Feuring-Buske M, Abramovich C, Rosten P, Hogge DE, et al. Induction of acute myeloid leukemia in mice by the human leukemia-specific fusion gene NUP98-HOXD13 in concert with Meis1. Blood 2003;101:4529–38. [42] Hirose K, Abramovich C, Argiropoulos B, Humphries RK. Leukemogenic properties of NUP98-PMX1 are linked to NUP98 and homeodomain sequence functions but not to binding properties of PMX1 to serum response factor. Oncogene 2008;27:6056–67. [43] Gurevich RM, Aplan PD, Humphries RK. NUP98-topoisomerase I acute myeloid leukemia-associated fusion gene has potent leukemogenic activities independent of an engineered catalytic site mutation. Blood 2004;104:1127–36. [44] Wang GG, Cai L, Pasillas MP, Kamps MP. NUP98-NSD1 links H3K36 methylation to Hox-A gene activation and leukaemogenesis. Nat Cell Biol 2007;9:804–12. [45] Wang GG, Song J, Wang Z, Dormann HL, Casadio F, Li H, et al. Haematopoietic malignancies caused by dysregulation of a chromatin-binding PHD finger. Nature 2009;459:847–51. [46] Lin YW, Slape C, Zhang Z, Aplan PD. NUP98-HOXD13 transgenic mice develop a highly penetrant, severe myelodysplastic syndrome that progresses to acute leukemia. Blood 2005;106:287–95. [47] Kroon E, Thorsteinsdottir U, Mayotte N, Nakamura T, Sauvageau G. NUP98HOXA9 expression in hemopoietic stem cells induces chronic and acute myeloid leukemias in mice. EMBO J 2001;20:350–61. [48] Calvo KR, Sykes DB, Pasillas MP, Kamps MP. NUP98-HOXA9 immortalizes myeloid progenitors, enforces expression of Hoxa9, Hoxa7 and Meis1, and alters cytokine-specific responses in a manner similar to that induced by retroviral co-expression of Hoxa9 and Meis1. Oncogene 2002;21:4247–56. [49] Slape C, Chung YJ, Soloway PD, Tessarollo L, Aplan PD. Mouse embryonic stem cells that express a NUP98-HOXD13 fusion protein are impaired in their ability to differentiate and can be complemented by BCR-ABL. Leukemia 2007;21:1239–48. [50] Ahuja HG, Popplewell L, Tcheurekdjian L, Slovak ML. NUP98 gene rearrangements and the clonal evolution of chronic myelogenous leukemia. Genes Chromosomes Cancer 2001;30:410–5. [51] Taketani T, Taki T, Nakamura T, Kobayashi Y, Ito E, Fukuda S, et al. High frequencies of simultaneous FLT3-ITD, WT1 and KIT mutations in hematological malignancies with NUP98-fusion genes. Leukemia 2010;24:1975–7. [52] Iwasaki M, Kuwata T, Yamazaki Y, Jenkins NA, Copeland NG, Osato M, et al. Identification of cooperative genes for NUP98-HOXA9 in myeloid leukemogenesis using a mouse model. Blood 2005;105:784–93. [53] Slape C, Hartung H, Lin YW, Bies J, Wolff L, Aplan PD. Retroviral insertional mutagenesis identifies genes that collaborate with NUP98-HOXD13 during leukemic transformation. Cancer Res 2007;67:5148–55. [54] Pineault N, Abramovich C, Ohta H, Humphries RK. Differential and common leukemogenic potentials of multiple NUP98-Hox fusion proteins alone or with Meis1. Mol Cell Biol 2004;24:1907–17. [55] Palmqvist L, Argiropoulos B, Pineault N, Abramovich C, Sly LM, Krystal G, et al. The Flt3 receptor tyrosine kinase collaborates with NUP98-HOX fusions in acute myeloid leukemia. Blood 2006;108:1030–6. [56] Dash AB, Williams IR, Kutok JL, Tomasson MH, Anastasiadou E, Lindahl K, et al. A murine model of CML blast crisis induced by cooperation between BCR/ABL and NUP98/HOXA9. Proc Natl Acad Sci USA 2002;99:7622–7. [57] Mayotte N, Roy DC, Yao J, Kroon E, Sauvageau G. Oncogenic interaction between BCR-ABL and NUP98-HOXA9 demonstrated by the use of an in vitro purging culture system. Blood 2002;100:4177–84. [58] Greenblatt S, Li L, Slape C, Nguyen B, Novak R, Duffield A, et al. Knock-in of a FLT3/ITD mutation cooperates with a NUP98-HOXD13 fusion to generate acute myeloid leukemia in a mouse model. Blood 2012;119:2883–94. [59] Kraemer D, Wozniak RW, Blobel G, Radu A. The human CAN protein, a putative oncogene product associated with myeloid leukemogenesis, is a nuclear pore complex protein that faces the cytoplasm. Proc Natl Acad Sci USA 1994;91:1519–23. [60] Alsabeh R, Brynes RK, Slovak ML, Arber DA. Acute myeloid leukemia with t(6;9) (p23;q34): association with myelodysplasia, basophilia, and initial CD34 negative immunophenotype. Am J Clin Pathol 1997;107:430–7. [61] Oyarzo MP, Lin P, Glassman A, Bueso-Ramos CE, Luthra R, Medeiros LJ. Acute myeloid leukemia with t(6;9)(p23;q34) is associated with dysplasia and a high frequency of flt3 gene mutations. Am J Clin Pathol 2004;122:348–58. [62] Slovak ML, Gundacker H, Bloomfield CD, Dewald G, Appelbaum FR, Larson RA, et al. A retrospective study of 69 patients with t(6;9)(p23;q34) AML emphasizes the need for a prospective, multicenter initiative for rare ‘poor prognosis’ myeloid malignancies. Leukemia 2006;20:1295–7. [63] von Lindern M, Breems D, van Baal S, Adriaansen H, Grosveld G. Characterization of the translocation breakpoint sequences of two DEK-CAN fusion genes present in t(6;9) acute myeloid leukemia and a SET-CAN fusion gene found

[64]

[65]

[66]

[67]

[68]

[69]

[70]

[71]

[72]

[73] [74]

[75]

[76]

[77]

[78]

[79]

[80]

[81]

[82]

[83]

[84]

[85]

[86]

[87]

[88]

[89]

in a case of acute undifferentiated leukemia. Genes Chromosomes Cancer 1992;5:227–34. Liu F, Gao L, Jing Y, Xu YY, Ding Y, Zhou MH, et al. Detection and clinical significance of gene rearrangements in Chinese patients with adult acute lymphoblastic leukemia. Leuk Lymphoma 2013;54:1521–6. Lee EY, Park TS, Kim MJ, Chang MH, Cho EH, Park SJ, et al. Detection of SETNUP214 rearrangement using multiplex reverse transcriptase-polymerase chain reaction (RT-PCR) in acute leukemias: a case report and literature review on a Korean case series. Ann Hematol 2012;91:1135–8. Lee SG, Park TS, Cho SY, Lim G, Park GJ, Oh SH, et al. T-cell acute lymphoblastic leukemia associated with complex karyotype and SET-NUP214 rearrangement: a case study and review of the literature. Ann Clin Lab Sci 2011;41:267–72. Chae H, Lim J, Kim M, Park J, Kim Y, Han K, et al. Phenotypic and genetic characterization of adult T-cell acute lymphoblastic leukemia with del(9)(q34);SET-NUP214 rearrangement. Ann Hematol 2012;91:193–201. Li WJ, Cui L, Gao C, Zhao XX, Liu SG, Xing YP, et al. MRD analysis and treatment outcome in three children with SET-NUP214-positive hematological malignancies. Int J Lab Hematol 2011;33:e25–7. von Lindern M, Fornerod M, Soekarman N, van Baal S, Jaegle M, Hagemeijer A, et al. Translocation t(6;9) in acute non-lymphocytic leukaemia results in the formation of a DEK-CAN fusion gene. Baillieres Clin Haematol 1992;5:857–79. Kim J, Lee SG, Song J, Kim SJ, Rha SY, Lee KA, et al. Molecular characterization of alternative SET-NUP214 fusion transcripts in a case of acute undifferentiated leukemia. Cancer Genet Cytogenet 2010;201:73–80. Gorello P, La Starza R, Di Giacomo D, Messina M, Puzzolo MC, Crescenzi B, et al. SQSTM1-NUP214: a new gene fusion in adult T-cell acute lymphoblastic leukemia. Haematologica 2010;95:2161–3. Graux C, Cools J, Melotte C, Quentmeier H, Ferrando A, Levine R, et al. Fusion of NUP214 to ABL1 on amplified episomes in T-cell acute lymphoblastic leukemia. Nat Genet 2004;36:1084–9. Hagemeijer A, Graux C. ABL1 rearrangements in T-cell acute lymphoblastic leukemia. Genes Chromosomes Cancer 2010;49:299–308. Roberts KG, Morin RD, Zhang J, Hirst M, Zhao Y, Su X, et al. Genetic alterations activating kinase and cytokine receptor signaling in high-risk acute lymphoblastic leukemia. Cancer Cell 2012;22:153–66. Eyre T, Schwab CJ, Kinstrie R, McGuire AK, Strefford J, Peniket A, et al. Episomal amplification of NUP214-ABL1 fusion gene in B-cell acute lymphoblastic leukemia. Blood 2012;120:4441–3. Graux C, Stevens-Kroef M, Lafage M, Dastugue N, Harrison CJ, Mugneret F, et al. Heterogeneous patterns of amplification of the NUP214-ABL1 fusion gene in T-cell acute lymphoblastic leukemia. Leukemia 2009;23:125–33. Oancea C, Ruster B, Henschler R, Puccetti E, Ruthardt M. The t(6;9) associated DEK/CAN fusion protein targets a population of long-term repopulating hematopoietic stem cells for leukemogenic transformation. Leukemia 2010;24:1910–9. Boer J, Bonten-Surtel J, Grosveld G. Overexpression of the nucleoporin CAN/NUP214 induces growth arrest, nucleocytoplasmic transport defects, and apoptosis. Mol Cell Biol 1998;18:1236–47. Sanden C, Ageberg M, Petersson J, Lennartsson A, Gullberg U. Forced expression of the DEK-NUP214 fusion protein promotes proliferation dependent on upregulation of mTOR. BMC Cancer 2013;13:440. Saito S, Nouno K, Shimizu R, Yamamoto M, Nagata K. Impairment of erythroid and megakaryocytic differentiation by a leukemia-associated and t(9;9)-derived fusion gene product, SET/TAF-Ibeta-CAN/Nup214. J Cell Physiol 2008;214:322–33. Ozbek U, Kandilci A, van Baal S, Bonten J, Boyd K, Franken P, et al. SET-CAN, the product of the t(9;9) in acute undifferentiated leukemia, causes expansion of early hematopoietic progenitors and hyperproliferation of stomach mucosa in transgenic mice. Am J Pathol 2007;171:654–66. Radu A, Moore MS, Blobel G. The peptide repeat domain of nucleoporin Nup98 functions as a docking site in transport across the nuclear pore complex. Cell 1995;81:215–22. Griffis ER, Altan N, Lippincott-Schwartz J, Powers MA. Nup98 is a mobile nucleoporin with transcription-dependent dynamics. Mol Biol Cell 2002;13:1282–97. Fontoura BM, Dales S, Blobel G, Zhong H. The nucleoporin Nup98 associates with the intranuclear filamentous protein network of TPR. Proc Natl Acad Sci USA 2001;98:3208–13. Fornerod M, Boer J, van Baal S, Jaegle M, von Lindern M, Murti KG, et al. Relocation of the carboxyterminal part of CAN from the nuclear envelope to the nucleus as a result of leukemia-specific chromosome rearrangements. Oncogene 1995;10:1739–48. De Keersmaecker K, Rocnik JL, Bernad R, Lee BH, Leeman D, Gielen O, et al. Kinase activation and transformation by NUP214-ABL1 is dependent on the context of the nuclear pore. Mol Cell 2008;31:134–42. Fontoura BM, Blobel G, Matunis MJ. A conserved biogenesis pathway for nucleoporins: proteolytic processing of a 186-kilodalton precursor generates Nup98 and the novel nucleoporin, Nup96. J Cell Biol 1999;144:1097–112. Vasu S, Shah S, Orjalo A, Park M, Fischer WH, Forbes DJ. Novel vertebrate nucleoporins Nup133 and Nup160 play a role in mRNA export. J Cell Biol 2001;155:339–54. Griffis ER, Xu S, Powers MA. Nup98 localizes to both nuclear and cytoplasmic sides of the nuclear pore and binds to two distinct nucleoporin subcomplexes. Mol Biol Cell 2003;14:600–10.

Please cite this article in press as: Takeda A, Yaseen NR. Nucleoporins and nucleocytoplasmic transport in hematologic malignancies. Semin Cancer Biol (2014), http://dx.doi.org/10.1016/j.semcancer.2014.02.009

G Model YSCBI-1117; No. of Pages 8

ARTICLE IN PRESS A. Takeda, N.R. Yaseen / Seminars in Cancer Biology xxx (2014) xxx–xxx

[90] Takeda A, Sarma NJ, Abdul-Nabi AM, Yaseen NR. Inhibition of CRM1-mediated nuclear export of transcription factors by leukemogenic NUP98 fusion proteins. J Biol Chem 2010;285:16248–57. [91] Moore MA, Chung KY, Plasilova M, Schuringa JJ, Shieh JH, Zhou P, et al. NUP98 dysregulation in myeloid leukemogenesis. Ann NY Acad Sci 2007;1106:114–42. [92] Cronshaw JM, Krutchinsky AN, Zhang W, Chait BT, Matunis MJ. Proteomic analysis of the mammalian nuclear pore complex. J Cell Biol 2002;158:915–27. [93] Walde S, Kehlenbach RH. The part and the whole: functions of nucleoporins in nucleocytoplasmic transport. Trends Cell Biol 2010;20:461–9. [94] Adams RL, Wente SR. Uncovering nuclear pore complexity with innovation. Cell 2013;152:1218–21. [95] Hoelz A, Debler EW, Blobel G. The structure of the nuclear pore complex. Annu Rev Biochem 2011;80:613–43. [96] Grunwald D, Singer RH, Rout M. Nuclear export dynamics of RNA-protein complexes. Nature 2011;475:333–41. [97] Oka M, Asally M, Yasuda Y, Ogawa Y, Tachibana T, Yoneda Y. The mobile FG nucleoporin Nup98 is a cofactor for Crm1-dependent protein export. Mol Biol Cell 2010;21:1885–96. [98] Roth P, Xylourgidis N, Sabri N, Uv A, Fornerod M, Samakovlis C. The Drosophila nucleoporin DNup88 localizes DNup214 and CRM1 on the nuclear envelope and attenuates NES-mediated nuclear export. J Cell Biol 2003;163:701–6. [99] Fornerod M, van Deursen J, van Baal S, Reynolds A, Davis D, Murti KG, et al. The human homologue of yeast CRM1 is in a dynamic subcomplex with CAN/Nup214 and a novel nuclear pore component Nup88. EMBO J 1997;16:807–16. [100] Bogerd HP, Echarri A, Ross TM, Cullen BR. Inhibition of human immunodeficiency virus Rev and human T-cell leukemia virus Rex function, but not Mason-Pfizer monkey virus constitutive transport element activity, by a mutant human nucleoporin targeted to Crm1. J Virol 1998;72:8627–35. [101] Hutten S, Kehlenbach RH. Nup214 is required for CRM1-dependent nuclear protein export in vivo. Mol Cell Biol 2006;26:6772–85. [102] Turner JG, Dawson J, Sullivan DM. Nuclear export of proteins and drug resistance in cancer. Biochem Pharmacol 2012;83:1021–32. [103] Kutay U, Guttinger S. Leucine-rich nuclear-export signals: born to be weak. Trends Cell Biol 2005;15:121–4. [104] Saito S, Miyaji-Yamaguchi M, Nagata K. Aberrant intracellular localization of SET-CAN fusion protein, associated with a leukemia, disorganizes nuclear export. Int J Cancer 2004;111:501–7. [105] Bachi A, Braun IC, Rodrigues JP, Pante N, Ribbeck K, von Kobbe C, et al. The C-terminal domain of TAP interacts with the nuclear pore complex and promotes export of specific CTE-bearing RNA substrates. RNA 2000;6:136–58. [106] Blevins MB, Smith AM, Phillips EM, Powers MA. Complex formation among the RNA export proteins Nup98, Rae1/Gle2 and TAP. J Biol Chem 2003;278:20979–88. [107] Levesque L, Bor YC, Matzat LH, Jin L, Berberoglu S, Rekosh D, et al. Mutations in tap uncouple RNA export activity from translocation through the nuclear pore complex. Mol Biol Cell 2006;17:931–43. [108] Powers MA, Forbes DJ, Dahlberg JE, Lund E. The vertebrate GLFG nucleoporin, Nup98, is an essential component of multiple RNA export pathways. J Cell Biol 1997;136:241–50. [109] Wang X, Babu JR, Harden JM, Jablonski SA, Gazi MH, Lingle WL, et al. The mitotic checkpoint protein hBUB3 and the mRNA export factor hRAE1 interact with GLE2p-binding sequence (GLEBS)-containing proteins. J Biol Chem 2001;276:26559–67. [110] Fontoura BM, Blobel G, Yaseen NR. The nucleoporin nup98 is a site for GDP/GTP exchange on ran and termination of karyopherin beta 2-mediated nuclear import. J Biol Chem 2000;275:31289–96. [111] van Deursen J, Boer J, Kasper L, Grosveld G. G2 arrest and impaired nucleocytoplasmic transport in mouse embryos lacking the proto-oncogene CAN/Nup214. EMBO J 1996;15:5574–83. [112] Trotman LC, Mosberger N, Fornerod M, Stidwill RP, Greber UF. Import of adenovirus DNA involves the nuclear pore complex receptor CAN/Nup214 and histone H1. Nat Cell Biol 2001;3:1092–100. [113] Xu L, Alarcon C, Col S, Massague J. Distinct domain utilization by Smad3 and Smad4 for nucleoporin interaction and nuclear import. J Biol Chem 2003;278:42569–77. [114] von Moeller H, Basquin C, Conti E. The mRNA export protein DBP5 binds RNA and the cytoplasmic nucleoporin NUP214 in a mutually exclusive manner. Nat Struct Mol Biol 2009;16:247–54. [115] Katahira J, Strasser K, Podtelejnikov A, Mann M, Jung JU, Hurt E. The Mex67pmediated nuclear mRNA export pathway is conserved from yeast to human. EMBO J 1999;18:2593–609. [116] Napetschnig J, Kassube SA, Debler EW, Wong RW, Blobel G, Hoelz A. Structural and functional analysis of the interaction between the nucleoporin Nup214 and the DEAD-box helicase Ddx19. Proc Natl Acad Sci USA 2009;106: 3089–94. [117] Browne SK, Roesser JR, Zhu SZ, Ginder GD. Differential IFN-gamma stimulation of HLA-A gene expression through CRM-1-dependent nuclear RNA export. J Immunol 2006;177:8612–9. [118] Forler D, Rabut G, Ciccarelli FD, Herold A, Kocher T, Niggeweg R, et al. RanBP2/Nup358 provides a major binding site for NXF1-p15 dimers at the nuclear pore complex and functions in nuclear mRNA export. Mol Cell Biol 2004;24:1155–67.

7

[119] Laurell E, Beck K, Krupina K, Theerthagiri G, Bodenmiller B, Horvath P, et al. Phosphorylation of Nup98 by multiple kinases is crucial for NPC disassembly during mitotic entry. Cell 2011;144:539–50. [120] Cross MK, Powers MA. Nup98 regulates bipolar spindle assembly through association with microtubules and opposition of MCAK. Mol Biol Cell 2011;22:661–72. [121] Jeganathan KB, Malureanu L, van Deursen JM. The Rae1-Nup98 complex prevents aneuploidy by inhibiting securin degradation. Nature 2005;438:1036–9. [122] Funasaka T, Nakano H, Wu Y, Hashizume C, Gu L, Nakamura T, et al. RNA export factor RAE1 contributes to NUP98-HOXA9-mediated leukemogenesis. Cell Cycle 2011;10:1456–67. [123] Hashizume C, Nakano H, Yoshida K, Wong RW. Characterization of the role of the tumor marker Nup88 in mitosis. Mol Cancer 2010;9:119. [124] Guttinger S, Laurell E, Kutay U. Orchestrating nuclear envelope disassembly and reassembly during mitosis. Nat Rev Mol Cell Biol 2009;10:178–91. [125] Bolhy S, Bouhlel I, Dultz E, Nayak T, Zuccolo M, Gatti X, et al. A Nup133dependent NPC-anchored network tethers centrosomes to the nuclear envelope in prophase. J Cell Biol 2011;192:855–71. [126] Xu S, Powers MA. Nup98-homeodomain fusions interact with endogenous Nup98 during interphase and localize to kinetochores and chromosome arms during mitosis. Mol Biol Cell 2010;21:1585–96. [127] Nakano H, Funasaka T, Hashizume C, Wong RW. Nucleoporin translocated promoter region (Tpr) associates with dynein complex, preventing chromosome lagging formation during mitosis. J Biol Chem 2010;285:10841–9. [128] Sarma NJ, Yaseen NR. Dynein light chain 1 (DYNLT1) interacts with normal and oncogenic nucleoporins. PLoS ONE 2013;8:e67032. [129] Etchin J, Sun Q, Kentsis A, Farmer A, Zhang ZC, Sanda T, et al. Antileukemic activity of nuclear export inhibitors that spare normal hematopoietic cells. Leukemia 2013;27:66–74. [130] Cheng Y, Holloway MP, Nguyen K, McCauley D, Landesman Y, Kauffman MG, et al. XPO1 (CRM1) inhibition represses STAT3 activation to drive a survivindependent oncogenic switch in triple negative breast cancer. Mol Cancer Ther 2014;13:675–86. [131] Forgues M, Difilippantonio MJ, Linke SP, Ried T, Nagashima K, Feden J, et al. Involvement of Crm1 in hepatitis B virus X protein-induced aberrant centriole replication and abnormal mitotic spindles. Mol Cell Biol 2003;23:5282–92. [132] Budhu AS, Wang XW. Loading and unloading: orchestrating centrosome duplication and spindle assembly by Ran/Crm1. Cell Cycle 2005;4:1510–4. [133] Grisendi S, Mecucci C, Falini B, Pandolfi PP. Nucleophosmin and cancer. Nat Rev Cancer 2006;6:493–505. [134] Arnaoutov A, Azuma Y, Ribbeck K, Joseph J, Boyarchuk Y, Karpova T, et al. Crm1 is a mitotic effector of Ran-GTP in somatic cells. Nat Cell Biol 2005;7:626–32. [135] Wu Z, Jiang Q, Clarke PR, Zhang C. Phosphorylation of Crm1 by CDK1cyclin-B promotes Ran-dependent mitotic spindle assembly. J Cell Sci 2013;126:3417–28. [136] Knauer SK, Mann W, Stauber RH. Survivin’s dual role: an export’s view. Cell Cycle 2007;6:518–21. [137] Ruchaud S, Carmena M, Earnshaw WC. Chromosomal passengers: conducting cell division. Nat Rev Mol Cell Biol 2007;8:798–812. [138] Kasper LH, Brindle PK, Schnabel CA, Pritchard CE, Cleary ML, van Deursen JM. CREB binding protein interacts with nucleoporin-specific FG repeats that activate transcription and mediate NUP98-HOXA9 oncogenicity. Mol Cell Biol 1999;19:764–76. [139] Bai XT, Gu BW, Yin T, Niu C, Xi XD, Zhang J, et al. Trans-repressive effect of NUP98-PMX1 on PMX1-regulated c-FOS gene through recruitment of histone deacetylase 1 by FG repeats. Cancer Res 2006;66:4584–90. [140] Mendjan S, Taipale M, Kind J, Holz H, Gebhardt P, Schelder M, et al. Nuclear pore components are involved in the transcriptional regulation of dosage compensation in Drosophila. Mol Cell 2006;21:811–23. [141] Brown CR, Kennedy CJ, Delmar VA, Forbes DJ, Silver PA. Global histone acetylation induces functional genomic reorganization at mammalian nuclear pore complexes. Genes Dev 2008;22:627–39. [142] Capelson M, Liang Y, Schulte R, Mair W, Wagner U, Hetzer MW. Chromatinbound nuclear pore components regulate gene expression in higher eukaryotes. Cell 2010;140:372–83. [143] Kalverda B, Pickersgill H, Shloma VV, Fornerod M. Nucleoporins directly stimulate expression of developmental and cell-cycle genes inside the nucleoplasm. Cell 2010;140:360–71. [144] Vaquerizas JM, Suyama R, Kind J, Miura K, Luscombe NM, Akhtar A. Nuclear pore proteins nup153 and megator define transcriptionally active regions in the Drosophila genome. PLoS Genet 2010;6:e1000846. [145] Light WH, Freaney J, Sood V, Thompson A, D’Urso A, Horvath CM, et al. A conserved role for human Nup98 in altering chromatin structure and promoting epigenetic transcriptional memory. PLoS Biol 2013;11:e1001524. [146] Lucchesi JC, Kelly WG, Panning B. Chromatin remodeling in dosage compensation. Ann Rev Genet 2005;39:615–51. [147] Bei L, Lu Y, Eklund EA. HOXA9 activates transcription of the gene encoding gp91Phox during myeloid differentiation. J Biol Chem 2005;280:12359–70. [148] Palmqvist L, Pineault N, Wasslavik C, Humphries RK. Candidate genes for expansion and transformation of hematopoietic stem cells by NUP98-HOX fusion genes. PLoS ONE 2007;2:e768. [149] Nakamura T, Yamazaki Y, Hatano Y, Miura I. NUP98 is fused to PMX1 homeobox gene in human acute myelogenous leukemia with chromosome translocation t(1;11)(q23;p15). Blood 1999;94:741–7.

Please cite this article in press as: Takeda A, Yaseen NR. Nucleoporins and nucleocytoplasmic transport in hematologic malignancies. Semin Cancer Biol (2014), http://dx.doi.org/10.1016/j.semcancer.2014.02.009

G Model YSCBI-1117; No. of Pages 8 8

ARTICLE IN PRESS A. Takeda, N.R. Yaseen / Seminars in Cancer Biology xxx (2014) xxx–xxx

[150] Ghannam G, Takeda A, Camarata T, Moore MA, Viale A, Yaseen NR. The oncogene NUP98-HOXA9 induces gene transcription in myeloid cells. J Biol Chem 2004;279:866–75. [151] He C, Li F, Zhang J, Wu J, Shi Y. The methyltransferase NSD3 has chromatinbinding motifs, PHD5-C5HCH, that are distinct from other NSD (nuclear receptor SET domain) family members in their histone H3 recognition. J Biol Chem 2013;288:4692–703. [152] Rosati R, La Starza R, Veronese A, Aventin A, Schwienbacher C, Vallespi T, et al. NUP98 is fused to the NSD3 gene in acute myeloid leukemia associated with t(8;11)(p11.2;p15). Blood 2002;99:3857–60. [153] Taketani T, Taki T, Nakamura H, Taniwaki M, Masuda J, Hayashi Y. NUP98NSD3 fusion gene in radiation-associated myelodysplastic syndrome with t(8;11)(p11;p15) and expression pattern of NSD family genes. Cancer Genet Cytogenet 2009;190:108–12. [154] Banninger G, Reich NC. STAT2 nuclear trafficking. J Biol Chem 2004;279:39199–206. [155] Kehlenbach RH, Assheuer R, Kehlenbach A, Becker J, Gerace L. Stimulation of nuclear export and inhibition of nuclear import by a Ran mutant deficient in binding to Ran-binding protein 1. J Biol Chem 2001;276:14524–31. [156] McBride KM, McDonald C, Reich NC. Nuclear export signal located within the DNA-binding domain of the STAT1transcription factor. EMBO J 2000;19:6196–206. [157] Tam WF, Lee LH, Davis L, Sen R. Cytoplasmic sequestration of rel proteins by IkappaBalpha requires CRM1-dependent nuclear export. Mol Cell Biol 2000;20:2269–84. [158] Ivanova IA, Dagnino L. Activation of p38- and CRM1-dependent nuclear export promotes E2F1 degradation during keratinocyte differentiation. Oncogene 2007;26:1147–54.

[159] Mylonis I, Chachami G, Samiotaki M, Panayotou G, Paraskeva E, Kalousi A, et al. Identification of MAPK phosphorylation sites and their role in the localization and activity of hypoxia-inducible factor-1alpha. J Biol Chem 2006;281:33095–106. [160] Harhaj EW, Sun SC. Regulation of RelA subcellular localization by a putative nuclear export signal and p50. Mol Cell Biol 1999;19:7088–95. [161] Kehlenbach RH, Dickmanns A, Gerace L. Nucleocytoplasmic shuttling factors including Ran and CRM1 mediate nuclear export of NFAT In vitro. J Cell Biol 1998;141:863–74. [162] London CA, Bernabe LF, Barnard S, Kisseberth WC, Borgatti A, Henson M, et al. Preclinical evaluation of the novel, orally bioavailable selective inhibitor of nuclear export (SINE) KPT-335 in spontaneous canine cancer: results of a phase I study. PLoS ONE 2014;9:e87585. [163] Walker CJ, Oaks JJ, Santhanam R, Neviani P, Harb JG, Ferenchak G, et al. Preclinical and clinical efficacy of XPO1/CRM1 inhibition by the karyopherin inhibitor KPT-330 in Ph+ leukemias. Blood 2013;122:3034–44. [164] Ranganathan P, Yu X, Na C, Santhanam R, Shacham S, Kauffman M, et al. Preclinical activity of a novel CRM1 inhibitor in acute myeloid leukemia. Blood 2012;120:1765–73. [165] Falini B, Bolli N, Shan J, Martelli MP, Liso A, Pucciarini A, et al. Both carboxyterminus NES motif and mutated tryptophan(s) are crucial for aberrant nuclear export of nucleophosmin leukemic mutants in NPMc+ AML. Blood 2006;107:4514–23. [166] Falini B, Mecucci C, Tiacci E, Alcalay M, Rosati R, Pasqualucci L, et al. Cytoplasmic nucleophosmin in acute myelogenous leukemia with a normal karyotype. N Engl J Med 2005;352:254–66. [167] Chou SH, Ko BS, Chiou JS, Hsu YC, Tsai MH, Chiu YC, et al. A knock-in Npm1 mutation in mice results in myeloproliferation and implies a perturbation in hematopoietic microenvironment. PLoS ONE 2012;7:e49769.

Please cite this article in press as: Takeda A, Yaseen NR. Nucleoporins and nucleocytoplasmic transport in hematologic malignancies. Semin Cancer Biol (2014), http://dx.doi.org/10.1016/j.semcancer.2014.02.009

Nucleoporins and nucleocytoplasmic transport in hematologic malignancies.

Hematologic malignancies are often associated with chromosomal rearrangements that lead to the expression of chimeric fusion proteins. Rearrangements ...
2MB Sizes 6 Downloads 3 Views