Review Article Role Of NEK2A In Human Cancer And Its . - Hindawi

1y ago
4 Views
2 Downloads
1.34 MB
13 Pages
Last View : 1m ago
Last Download : 3m ago
Upload by : Jenson Heredia
Transcription

Hindawi Publishing CorporationBioMed Research InternationalVolume 2015, Article ID 862461, 12 pageshttp://dx.doi.org/10.1155/2015/862461Review ArticleRole of NEK2A in Human Cancer and Its Therapeutic PotentialsJiliang Xia,1 Reinaldo Franqui Machin,2 Zhimin Gu,2 and Fenghuang Zhan21Cancer Research Institute, Southern Medical University, Guangzhou, Guangdong 510515, ChinaDepartment of Internal Medicine, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA2Correspondence should be addressed to Fenghuang Zhan; fenghuang-zhan@uiowa.eduReceived 19 September 2014; Accepted 14 November 2014Academic Editor: Stefan KnappCopyright 2015 Jiliang Xia et al. This is an open access article distributed under the Creative Commons Attribution License,which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.Chromosome instability (CIN) has been identified as a common feature of most human cancers. A number of centrosomal kinasesare thought to cause CIN in cancer cells. Part of those centrosomal kinases exhibit elevated expression in a wide variety of tumoursand cancer cell lines. Additionally, critical roles in many aspects of cancer cell growth, proliferation, metastasis, and drug resistancehave been assigned to some of these centrosomal kinases, such as polo-like kinase 1 (PLk1) and Aurora-A kinase. Recent studiesfrom our group and others revealed that a centrosomal kinase, Never in Mitosis (NIMA) Related Kinase 2A (NEK2A), is frequentlyupregulated in multiple types of human cancers. Uncontrolled activity of NEK2A activates several oncogenic pathways and ABCtransporters, thereby leading to CIN, cancer cell proliferation, metastasis, and enhanced drug resistance. In this paper, we highlightrecent findings on the aberrant expression and functional significance of NEK2A in human cancers and emphasize their significancefor therapeutic potentials.1. IntroductionCancer cells tend to show some degree of genetic instability.It is now clear that high genetic change or instability playsa major role in cancer development [1]. Genetic instabilitycan trigger tumorigenesis mainly through the activation ofoncogenes and/or the inactivation of tumor suppressor genes.Chromosome instability (CIN), a phenotype characterized bya high rate of gain and/or loss of whole or large portions ofchromosomes at each cell division, has been implicated in theinitiation of genetic instability [2]. CIN generates a disparityin chromosome number (aneuploidy) and an enhanced rateof loss of heterozygosity, which is frequently seen in cancercells [3–5]. Theodor Boveri observed abnormal chromosomequantities in cancer cells as early as a century ago [6].However it was only in the recent years that CIN has beenpositively correlated with tumorigenesis, cancer progression,and therapeutic resistance [3–5].Former studies have indicated that defects in cell division,telomere stability, and the DNA damage response all contribute to CIN in cancer [7]. Numerous cell division relatedproteins, which are highly expressed in multiple cancers,are involved in the initiation of CIN in cancer cells [8, 9].Centrosomal kinases are important regulators of cell division.Uncontrolled activity of centrosomal kinases can lead tospindle abnormalities, centrosome fragmentation, prematurecentriole splitting, multiple nucleuses, supernumerary centrosomes, and chromosome segregation errors. All thoseabnormal phenotype are important risk factors for CIN,indicating that overexpression of centrosomal kinases mightdrive tumor progression by promoting CIN [10, 11]. Studiesfrom our group and others have demonstrated that elevatedNever in Mitosis (NIMA) Related Kinase 2A (NEK2A), amember of the NIMA-related serine/threonine kinase familyand a core component of centrosome, results in CIN in cancercells [12, 13]. Importantly, our previous studies indicated thathigh expression of NEK2A is associated with poor survivalin various cancers [12]. In recent years, a larger number ofstudies focused on the roles of NEK2A in tumorigenesis,cancer progression, and drug resistance have been published.In view of previous studies, we speculated that NEK2A maybe a novel potential biomarker for diagnosis and a possibletherapeutic target for human cancers.

2BioMed Research InternationalCC1 (302–368)271 3021Ser/Thr kinasePP1 binding site(383–386)368D-box(436–445)404439CC2LZKEN box (391–399)304340Centrosome and microtubulelocalization (335–370)445Nucleolar localization(399–445)Figure 1: NEK2A protein structure. The relative positions of the catalytic domain (serine/threonine kinase), leucine zipper (LZ), coiled coil(CC), PP1 binding site, centrosome localization microtubule site, nucleolar localization, KEN-box, and D-box are indicated. Numbers aboveand below the structures indicate amino acid positions.2. Basic Biology of NEK2A and ValidatedFunctions of NEK2A in Normal CellsThe NEK2 gene in humans is located in chromosome 1 andit is comprised of 8 exons. There are three isoforms thatresult from the alternate splicing of this gene, termed NEK2A,NEK2B, and NEK2C. NEK2A is the most studied isoformand it is a cell cycle-regulated kinase structurally related tothe mitotic regulator NIMA of Aspergillus nidulans, being47% identical within the catalytic domains [14]. NEK2Ais also 31% structurally identical to Aurora-A, a humanserine/threonine kinase involved in proper cell division [15].Other structural studies demonstrated that human NEK2A isa 445 amino acid (48 kDa) protein comprising an N-terminalkinase domain and a C-terminal noncatalytic regulatorydomain [15]. The NEK2A N-terminal kinase domain hasall the motifs typical of a serine/threonine kinase. The Cterminal region possesses multiple regulatory motifs, whichregulate the activity, location, and stability of NEK2A. Theseinclude leucine zipper (LZ), coiled coil (CC), centrosome,microtubule and nucleolar localization sites, PP1 binding site(KVHF), and APC binding site KEN-box and extended cyclinA-type destruction box (D-box) (Figure 1) [15].Subcellular localization analysis shows that NEK2Aresides in both the nucleus and cytoplasm throughout thecell cycle [16]. More detailed localization studies on thecytoplasmic NEK2A concurred to show that it is a corecomponent of the centrosome [17]. In addition, NEK2A hasbeen detected at nucleoli in interphase cells, on condensedchromatin in meiotic and mitotic cells, and at the kinetochores and midbody of dividing cells [16–20]. Westernblot analysis demonstrates that NEK2A displays a cell cycledependent expression pattern, being low in G1, increasingthrough S and G2 to reach peak in late G2/M, and decreasingupon entry into mitosis [14, 21]. Several studies have shownthat a key mechanism that maintains NEK2A suppressedduring M phase is the ubiquitin proteasomal system (UPS)[22]. NEK2A degradation through the UPS depends on directbinding of NEK2A to the Anaphase Promoting Complex(APC/C) via two C-terminal motifs including the D-boxand the KEN-box [22, 23]. This interaction leads to theubiquitination of NEK2A and its degradation by the 26Sproteasome. No protein, to our knowledge, has yet been identified to stabilize NEK2A through deubiquitination; howeverthis could also represent another aspect of NEK2A regulation.Posttranslational modifications are not the only mechanismthat keeps NEK2A regulated in a cell cycle-dependentmanner. Negative transcriptional regulators, like E2F4, andthe epigenetic modulators, p107 and p130, negatively affectNEK2A levels directly and indirectly, respectively [24].Similar to its expression pattern, the activity of NEK2Ais cell cycle-regulated, with maximum activity in S andG2 phases and low activity upon mitotic entry. NEK2Adimerization via the leucine zipper motif is essential forfull activation, both in vitro and in vivo, most likely as aresult of its promoting of transautophosphorylation [25]. Thiswas shown by deleting the leucine zipper motif, which prevented the transautophosphorylation of NEK2A and reducedNEK2A activity. Many possible autophosphorylation sites ofNEK2A were first identified by mass spectrometry in boththe N-terminal catalytic domain and C-terminal regulatorydomain [15]. Some of these have been confirmed with in vitrokinase assays and their physiological relevance with variouscell lines. Of the most important autophosphorylation sitesdescribed thus far are T175 and T179, localized in thekinase domain, which allow activation of NEK2A [15]. Otherautophosphorylation sites outside the kinase domain havebeen described, some in the KEN-box and others in the coiled

BioMed Research International3Table 1: NEK2A interaction proteins and their functions.NEK2A interaction proteinDetection methodFunctionAPC/CCo-IPNEK2A degradationPP1Yeast two-hybrid, Co-IPNEK2A dephosphorylationReferencenumber[23][26, 27]C-Nap1Yeast two-hybridCentrosome separation[28]RootletinYeast two-hybridCentrosome separation[29]NLP2Yeast two-hybridMicrotubule organization[33][37]NumatrinCo-IP, pull-downCentrosome integrity and dynamicsHMGA2Co-IP, pull-downChromatin condensation[38]HEC1Co-IPSpindle assembly checkpoint, chromosome separation[42]MAD1Yeast two-hybrid, Co-IPSpindle assembly checkpoint, chromosome separation[40]TRF1Yeast two-hybrid, pull-downChromosome separation[41]MAD2Co-IPSpindle assembly checkpoint, chromosome separation[12]SGO1Pull-down, Co-IPChromosome congression[43]coil region, suggesting a role in kinase regulation and dimerization, respectively [15]. More biochemical studies must bedone to understand the role of these phospho-sites. NEK2Acan be negatively regulated through dephosphorylation byProtein Phosphatase 1 (PP1) that directly binds to a KVHFsequence within the C-terminal of NEK2A protein [26, 27].As expected, overexpression of PP1 suppresses NEK2A kinaseactivity, while depletion of PP1 by small interfering RNAshowed increased NEK2A activity.The subcellular localization, cell cycle-dependent expression, and activity together suggest that NEK2A may playan important role in cell division. Previous studies havedemonstrated that some cell division related proteins interactwith NEK2A (Table 1). Transfection of active, but not inactiveNEK2A, exhibited a premature separation of centrosomesin the cell cycle, while depletion of NEK2A interferes withcentrosome separation in G2 cells [17]. Subsequent studiesfurther suggested that NEK2A induces centrosome separation by phosphorylating centrosome cohesion proteins CNap1 and Rootletin [28–30]. Besides centrosome separation,NEK2A also regulates microtubule organization throughphosphorylation of ninein-like protein 2 (NLP2), resultingin its removal from the centrosome at the time of mitoticentry [31–35]. NEK2A can also help recruit numatrin to thecentrosome through its kinase activity. Previously consideredas noncentrosomal, recent data has surfaced that numatrin’srecruitment to the centrosome protects against centrosomehyperamplification and genomic instability [36, 37].Apart from its many functions in the centrosome, subcellular localization studies have found a fraction of endogenous NEK2A at condensed chromatin, particularly in cellsundergoing meiosis. According to previous studies, NEK2Ais activated by the MAPK pathway; it then phosphorylatesan architectural chromatin protein, HMGA2. This phosphorylation decreases the affinity of HMGA2 for DNAand might drive its release from the chromatin, therebypromoting chromatin condensation [38, 39]. NEK2A hasalso been reported to regulate chromosome separation bymodulating the spindle assembly checkpoint (SAC). NEK2Aachieves this through direct interaction or phosphorylationof mitotic arrest deficient-like 2 (MAD2), mitotic arrestdeficient-like 1 (MAD1), telomere repeat binding protein(TRF1), and highly expressed in cancer 1 (HEC1) [12, 40–42]. Some studies show that NEK2A regulates the alignmentof chromosomes to the spindle (chromosome congression)through phosphorylating SGO1 at Ser14 and Ser507 [43].Expression of nonphosphorylatable mutant SGO1 perturbedchromosome congression and resulted in a dramatic increasein microtubule attachment errors, including syntelic andmonotelic attachments. In addition to participating in celldivision, NEK2A was identified as a novel mRNA splicingfactor kinase. NEK2A colocalizes in splicing speckles withSRSF1 and SRSF2, important splicing factors, and has beenshown to phosphorylate the former [44]. Overexpression ofNEK2A induces phosphorylation of endogenous SR proteins,a kind of proteins containing a protein domain with longrepeats of serine and arginine amino acid residues, and affectsthe splicing activity of SRSF1 toward reporter minigenes andendogenous targets, independently of SRPK1. Conversely,knockdown of NEK2A, like that of SRSF1, induces expressionof proapoptotic variants from SRSF1-target genes and sensitizes cells to apoptosis.Although much progress has been made in our understanding of NEK2A in the past decades, several setbackshave slowed the progress in the study of this kinase. One ofthese is the lack of isoform-specific antibodies. Because of thesimilarity between NEK2A, NEK2B, and NEK2C, antibodiesfor each one are not available, making functional studiesof endogenous NEK2A somewhat troublesome. Anotherproblem involving the study of NEK2A is the toxicity ofthe wild type active protein in bacteria [66]. This does notallow the production of bulk NEK2A for crystallographyexperiments. The recent data uncovered by crystallographyis based on unphosphorylatable mutants [15]. Another issue

4with NEK2A research is the lack of mouse models. Thedocumented functional research of NEK2A is thus restrictedto in vitro studies so far. To better characterize the invivo role of NEK2A, mouse models of NEK2A are highlydemanded. Our group has generated a NEK2A transgenicmodel, in which expression of NEK2A is turned on uponbreeding with Cre mice in a tissue specific manner. AsNEK2A is highly expressed in B cell lymphoma and multiplemyeloma [12, 67], the NEK2A transgenic mice were bredwith CD19 promoter driving cre to overexpress NEK2A in Bcell lineage. Instead of developing B cell malignancies, thesemice had altered B-cell development by increasing immatureB-cells in the bone marrow and decreasing B-1 B-cellsin peritoneal cavity. Furthermore, transgenic expression ofNEK2A induced formation of spontaneous germinal centersand exhibits enhanced T-cell dependent immune response(unpublished data). All these provided the novel evidence ofNEK2A’s function in vivo. In addition, we are also developingNEK2A knockout mice using a gene trap strategy to betterexplore NEK2A’s role in pathophysiological conditions.3. Aberrant Expression of NEK2A inHuman CancersProtein kinases that regulate the centrosome cycle are oftenaberrantly controlled in tumor cells. Changes in their expression can lead to CIN and aneuploidy, potentially triggeringtumorigenesis. Increased expression of NEK2A has beenreported in various cancer cells (Table 1). Reports implicatinghigh expression of NEK2A in human cancer first appearedfollowing microarray analysis of mRNA abundance in cancercell line derived from Ewing tumors (ETs) (Table 2) [45].Subsequent RT-PCR or Western blot analysis showed thatmultiple types of human cancer cell lines, including cholangiocarcinoma [46], testicular seminomas [47], human breastcancer [48–50], cervical cancer [50], prostate cancer [50],and colorectal Cancer [53, 54], expressed higher NEK2A inthe level of mRNA or protein than normal human fibroblastcells. Consistently, analysis by Western blot, real-time PCR,DNA microarray, and immunohistochemistry indicated thatincreased NEK2A is found in various cancer tissues, such ashuman testicular seminomas [47], human breast carcinoma[49, 51, 52], colorectal cancer [53, 54], malignant peripheralnerve sheath tumors [55], nonsmall cell lung cancer [56],renal cell carcinoma [57], and pancreatic ductal adenocarcinoma [58]. Our previous gene expression profiling (GEP)analysis showed that NEK2A expression was significantlyupregulated in several types of human cancer samples compared to normal cells, including multiple myeloma, myeloidleukemia, breast cancer, lung adenocarcinoma, mantle celllymphoma, mesothelioma, head and neck squamous cellcarcinoma, bladder carcinoma, glioblastoma, T-cell acutelymphoblastic leukemia, colon carcinoma, hepatocellularcarcinoma, melanoma, and ovarian adenocarcinoma [12].Though we know NEK2A is highly expressed in variouscancer cells, the underling mechanisms of increased NEK2Ain cancer cells still remain poorly understood. Since bothmRNA and protein of NEK2A are increased in cancer cells,BioMed Research Internationalseveral tumor-associated transcription factors and posttranslational modifications may be involved in the high expressionof NEK2A in cancer cells. MicroRNA-128, a tumor suppressor, is thought to target NEK2A in colorectal cancer cell [68].Colorectal cancer patients with high miR-128 expression hadsignificantly lower NEK2A expression and lower recurrencerates than those with low miR-128 expression. Consistentwith other tumor suppressor microRNAs, microRNA-128is silenced by DNA methylation in colorectal cancer cells.A two- to threefold recovery of miR-128 expression wasfound after 5-aza-2-deoxycytidine (5aza-dC) treatment, aDNA-demethylating agent. Moreover, NEK2A expressionlevels were significantly reduced after 5aza-dC treatment.In addition to being indirectly inhibited by demethylation,NEK2A transcript levels are reduced by direct demethylationin HCT116 colon cancer cells, which is restricted to thedistal region of the NEK2A promoter, but not in isogenicp53 / cells [69]. Chromatin immunoprecipitation analysisdemonstrated that p53 directly and specifically binds tothe distal NEK2A promoter. Stabilization of endogenousp53 by doxorubicin or ectopic expression of p53, but nota p53 DNA-binding mutant, decreased NEK2A expression[69]. This study suggests that demethylation of the distalNEK2A promoter represses NEK2A expression in a p53dependent manner. As mentioned previously, in G1 and Mphase normal cells, NEK2A expression is downregulated bytumor suppressors including the retinoblastoma (Rb) familymembers p107 and p130 [24] and APC [22, 23]. Chromatinimmunoprecipitation (ChIP) assays demonstrated that thepromoter of NEK2A is bound by E2F4 transcription factor inearly G1 [24]. E2F4, a member of the E2F transcription factorfamily, interacts with Rb family members p107 and p130 andacts as a transcriptional repressor in G0 and G1 throughrecruitment of histone deacetylase which suppressed geneexpression. In p107 / and p130 / mouse embryo fibroblasts(MEFs), the expression of NEK2A is significantly increasedeven in the absence of serum suggesting that tumours lackingp107 or p130 are likely to have elevated levels of NEK2A [24].Moreover, overexpression of E7, a human papillomavirusencoded protein which represses the function of Rb familymembers, leads to increased NEK2A expression in humankeratinocytes [70]. Forkhead transcription factor FOXM1regulates the expression of many G2-specific genes includingNEK2A and is essential for proper mitotic progression [71].Overexpression of recombinant FOXM1 increases NEK2Aexpression; conversely, FOXM1 depletion reduces NEK2Aexpression. So far, very few reports about the relationshipbetween NEK2A expression and tumor suppressors andoncoproteins in cancer cells have been published. Lowexpression of p130 and p107 or inactivated APC frequentlyoccurs in the carcinogenic processes of multiple types ofcancers [72, 73]. Both high expressions of FOXM1 andE7 are important risk factors for tumorigenesis [74, 75].Thus elevated NEK2A in cancer cells may be induced bythose abnormal conditions. Studies about the mechanisms ofNEK2A expression regulation in cancers may contribute toclinical application of NEK2A-based anticancer therapeutics.

BioMed Research International5Table 2: Aberrant expression of NEK2A in different cancers.ReferencenumberCancer typeNEK2A upregulationDetect methodEwing tumorCholangiocarcinomaTesticular seminomasCancer cell lineCancer cell lineCancer cell line and tumor tissueBreast cancerCancer cell line and tumor tissueCervical cancerProstate cancerCancer cell lineCancer cell lineColorectal cancerCancer cell line and tumor tissueMalignant peripheral nervesheath tumorsTumor tissueLung adenocarcinomaTumor tissueRenal cell carcinomaPancreatic ductaladenocarcinomaMultiple myelomaMyeloid leukemiaMantle cell lymphomaMesotheliomaHead and neck squamouscell carcinomaBladder carcinomaGlioblastomaT cell acute lymphoblasticleukemiaHepatocellular carcinomaMelanomaOvarian adenocarcinomaTumor tissueCancer cell line and tumor tissueTumor tissueTumor tissueTumor tissueDNA microarray analysisRT-PCR, Western blotImmunohistochemistry, Western blotRT-PCR, Western blot,immunohistochemistry, and DNAmicroarray analysisWestern blotWestern blotWestern blot, DNA microarrayanalysis, and immunohistochemistryDNA microarray analysis,immunohistochemistryDNA microarray A microarray analysisReal-time PCR,immunohistochemistryDNA microarray analysisDNA microarray analysisDNA microarray analysisDNA microarray analysisTumor tissueDNA microarray analysis[12]Tumor tissueTumor tissueDNA microarray analysisDNA microarray analysis[12][12]Tumor tissueDNA microarray analysis[12]Tumor tissueTumor tissueTumor tissueDNA microarray analysisDNA microarray analysisDNA microarray analysis[12][12][12]Tumor tissue4. Roles of NEK2A in Tumorigenesis, TumorProgression, and Drug ResistanceFormer studies have demonstrated that NEK2A involvesvarious signaling in a broad range of cancers (Table 3).4.1. Tumorigenesis. As discussed above, studies have implicated NEK2A in the regulation of centrosome separation,microtubule organization, chromatin condensation, SAC,and chromosome congression during cell division. Overexpression of NEK2A in cancer cells may result in prematurecentriole splitting, spindle abnormalities, multinucleation,centrosome amplification (CA), and chromosome segregation errors. These cellular phenotypes ultimately lead to CINand aneuploidy, which is frequently observed in transformedcells with overexpressed NEK2A. This suggests that overexpression of NEK2A triggers tumorigenesis by promotingCIN and aneuploidy. Consistent with this idea, our previousstudies show that overexpression of NEK2A in multiplemyeloma cell results in CIN [12].[45][46][47][48–52][50][50][53, 54][55][12, 56][57][58][12][12][12][12]Several cell division proteins and signaling pathwaysare involved in NEK2A mediated CIN and aneuploidy.The MAPK pathway is required for maintaining chromatincondensed during the two meiotic divisions and uncontrolledactivity of MAPK pathway has been implicated in CIN[76]. Previous studies show that NEK2A is phosphorylatedby the MAPK effector P90Rsk2 , thus placing these twoproteins in the same pathway. Moreover, the induction ofchromatin condensation requires the MAPK pathway andP90Rsk2 . Interestingly, inhibiting MAPK in the presence ofokadaic acid prevents not only chromatin condensation,but also the activation of NEK2A [39]. So NEK2A may beinvolved in the MAPK induced CIN. The Hippo pathwaycomponents, MST2 and HSAV1, also have a direct interactionwith NEK2A, thereby regulating its ability to localize tocentrosome and phosphorylate C-Nap1 and Rootletin [59].Polo-like kinase 1 (PLK1), a serine/threonine kinase identifiedas a potential drug target in cancer therapy, may also affectNEK2A activity in cancer cells, albeit indirectly. Tumors with

6BioMed Research InternationalTable 3: Signaling involved in the tumorigenic function of NEK2A.Proteins or signaling pathwaysinteract with NEK2AUpstream of NEK2AMAPK pathwayMST2PLK1ReferencenumberThe relationship with NEK2AFunctionPhosphorylate NEK2ARegulate NEK2A’s ability to localize to centrosome andphosphorylate C-Nap1 and RootletinRegulate MST2-NEK2A/NEK2A-𝛽-catenin-inducedcentrosome separation and NEK2A-NIP-induced microtubuleorganizationRegulate NEK2A genesis[32, 60, rigenesisTumorigenesisTumor progression anddrug resistanceTumor progression anddrug resistanceDrug resistanceDrug resistanceDrug resistanceDrug resistanceDrug resistanceDrug resistanceDrug resistanceDrug resistanceDrug resistanceDrug resistanceDrug resistanceDrug resistanceDrug resistance[63][63][42][64]CDK4Downstream of NEK2ACDC20MAD2HEC1TRF1Phosphorylated by NEK2APhosphorylated by NEK2APhosphorylated by NEK2APhosphorylated by NEK2AAKTPhosphorylated by NEK2A𝛽-CateninNEK2A induces nuclear accumulation of aspase-3Caspase-8Caspase-9RBHistone H3 (p-Ser10)Upregulated by NEK2AUpregulated by NEK2AUpregulated by NEK2ADownregulated by NEK2ADownregulated by NEK2AUpregulated by NEK2AUpregulated by NEK2AActivated in NEK2A silenced cancer cellActivated in NEK2A silenced cancer cellActivated in NEK2A silenced cancer cellActivated in NEK2A silenced cancer cellActivated in NEK2A silenced cancer cellInactivated in NEK2A silenced cancer cellPLK1 overexpression were associated more frequently withCIN (𝑃 0.0001), DNA aneuploidy (𝑃 0.0007), and CA(𝑃 0.0013) than those without PLK1 overexpression [77].Functional studies have demonstrated that PLK1 can phosphorylate MST2, and this happens upstream of the MST2NEK2A-induced centrosome separation [60]. The absenceof PLK1 phosphorylation of MST2 promotes assembly ofNEK2A-PP1𝛾-MST2 complexes, in which PP1𝛾 counteractsNEK2A kinase activity. In contrast, PLK1 phosphorylationof MST2 prevents PP1𝛾 binding to MST2-NEK2A, allowing NEK2A activity to promote centrosome separation. Inaddition to regulating MST2-NEK2A-induced centrosomeseparation, PLK1 was shown to promote the NEK2A-𝛽catenin-induced centrosome separation [61] and NEK2ANIP-induced microtubule organization [32]. This suggeststhat PLK1 is an essential regulator of NEK2A in cancer cells.In summary, NEK2A has roles downstream of the MAPKpathway and PLK1; hence NEK2A may be involved in MAPKand PLK1-induced CIN and [12][12][12][12][65][65]Abnormal expression of SAC proteins can cause cell aneuploidy, an important factor in tumorigenesis. High expressions of cell division cycle 20 homolog (CDC20) and MAD2,key components of SAC, have been reported in variouscarcinomas. Previous studies have demonstrated that NEK2Acan phosphorylate MAD2 and CDC20. Moreover, overexpression of NEK2A acts upon the MAD2-CDC20 complexand induces a delay in mitosis, promoting aneuploidy incancer [63]. HEC1, a Ndc80 complex protein localized atkinetochores and highly expressed in cancer, is phosphorylated by NEK2A at 165-serine [42]. Overexpression of HEC1in an inducible mouse model results in mitotic checkpointhyperactivation and is sufficient to generate tumors thatharbor significant levels of aneuploidy in vivo [78]. Formerstudies have demonstrated that the phosphorylation of HEC1by NEK2A is essential for MAD1 and MAD2 to localize to thekinetochores, which is involved in HEC1 induced tumorigenesis. Their studies suggest that HEC1, MAD2, and CDC20may be involved in NEK2A induced CIN in cancer cells.

BioMed Research InternationalIn Her2 breast cancer cells, knockdown of NEK2Areduces CA and binucleation while its overexpressionenhances CA [62]. Moreover, ectopic expression of NEK2Ain immortalized HBL100 breast epithelial cells leads to accumulation of multinucleated cells with supernumerary centrosomes [50]. NEK2A expression is regulated by CDK4,which is a major regulator of CA in Her2 breast cancercells [62], suggesting that NEK2A may be a downstreamtarget of CDK4, and is involved in CDK4 induced CA.Additionally, TRF1 was shown to be involved in NEK2Ainduced aneuploidy. It has been discovered that TRF1 interacts directly with and is phosphorylated by NEK2A. NEK2Aoverexpression in the breast cancer cell lines, MDA-MB231 and MCF7, results in CA and multinucleation, whichleads to aneuploidy; however TRF1 depletion by siRNAprevents this phenomenon [64]. Moreover, when exogenousTRF1 was added back in NEK2A-overexpressed cells with noendogenous TRF1, cells had reinduced cytokinetic failure.As summarized above, the expression and activity ofNEK2A are regulated by many tumor suppressors and oncoproteins that show aberrant behavior in cancer. This, coupledwith the abundant evidence on the effects of NEK2A on cellphysiology, strongly suggests that NEK2A is an oncoproteincapable of being deregulated by several pathways. On theother hand, NEK2A regulates the activity of some cancerrelated proteins by interacting and phosphorylating them;hence NEK2A may be involved in the process of tumorigenesis.4.2. Tumor Progression. Studies in multiple types of cancers have demonstrated that elevated NEK2A promotes cellproliferation, while its suppression with siRNA inhibitedthis proliferation and induced cell death [12, 46, 48–50].Moreover, cancer cells overexpressing NEK2A showed asignificant increase in colony formation compared withcontrol cells [12, 48]. In a xenograft nude mouse model,subcutaneous injection of NEK2A siRNA around the tumornodules resulted in reduction of tumor size compared withthose of control siRNA injection [46, 48]. In a peritonealdissemination model, NEK2A siRNA-treated mice showedstatistically longer survival periods in comparison with thoseof the control siRNA treated mice [46]. Former studiesshow that NEK2A expression was positively associated withKi-67 expression, a cell proliferation marker, in multiplemyeloma, human primary breast cancer tissue, and nonsmall cell lung cancer [12, 49, 56]. In addition, NEK2Acytoplasmic expression was positively associated with cancergrade and tumor size in breast invasive ductal carcinoma(IDC) [51]. These data all point to NEK2A supportingtumor progression both in vitro and in vivo. Interesti

of mitotic arrest de cient-like (MAD), mitotic arrest de cient-like (MAD) , telomere repeat binding protein (TRF), and highly expressed in cancer (HEC) [ , ]. Some studies show that NEKA regulates the alignment of chromosomes to the spindle (chromosome congression) through phosphorylating SGO at Ser and Ser [ ].

Related Documents:

Amendments to the Louisiana Constitution of 1974 Article I Article II Article III Article IV Article V Article VI Article VII Article VIII Article IX Article X Article XI Article XII Article XIII Article XIV Article I: Declaration of Rights Election Ballot # Author Bill/Act # Amendment Sec. Votes for % For Votes Against %

Article 27 Article 32 26 37 Journeyman Glazier Wages Article 32, Section A (2) 38 Jurisdiction of Work Article 32, Section L 43 Legality Article 2 3 Mechanical Equipment Article 15, Section B 16 Out-of-Area Employers Article 4, Section B 4 Out-of-Area Work Article 4, Section A 4 Overtime Article 32, Section G 41

Jefferson Starship article 83 Jethro Tull (Ian Anderson) article 78 Steve Marriott article 63, 64 Bill Nelson article 96 Iggy Pop article 81 Ramones article 74 Sparks article 79 Stranglers article 87 Steve Winwood article 61 Roy Wood art

1 ARTICLES CONTENTS Page Article 1 Competition Area. 2 Article 2 Equipment. 4 Article 3 Judo Uniform (Judogi). 6 Article 4 Hygiene. 9 Article 5 Referees and Officials. 9 Article 6 Position and Function of the Referee. 11 Article 7 Position and Function of the Judges. 12 Article 8 Gestures. 14 Article 9 Location (Valid Areas).

article 22, call time 41 article 23, standby time 42 article 24, life insurance 42 article 25, health benefits 43 article 26, work-related injuries 51 article 27, classification 55 article 28, discharge, demotion, suspension, and discipline 58 article 29, sen

Section I. Introductory provisions Chapter 1 General Provisions (Article 1 - Article 9) Chapter 2 Voting rights (Article 10 - Article 11) Chapter 3 Electoral Districts (Article 12 - Article 17) Chapter 4 The register of voters (Article 18 - Article 25) Ch

Article 9. Conditions of Operation Article 10. Disciplinary Actions Article 11. Penalties Article 12. Revenues Article 13. Local Governments Article 14. Miscellaneous Provisions Article 15. Additional Restrictions Related to Fair Elections and Corruption of Regulators Article 16. Additional Contracts: Proposition Players

The American Revolution Part One: The events leading up the Revolutionary War (1750 – 1775) Background Historically speaking, right now “we” are British. The Colonies are an extension of Britain, so we share their government, their identity, their pride, and also their enemies. There is NO United States of America. Taunton Flag, flown by colonists to show unity with the British crown .