Oncotarget

Reviews:

IκBζ: an emerging player in cancer

PDF |  HTML  |  How to cite

Oncotarget. 2016; 7:66310-66322. https://doi.org/10.18632/oncotarget.11624

Metrics: PDF 2047 views  |   HTML 5629 views  |   ?  

Marie Willems, Nadège Dubois, Lucia Musumeci, Vincent Bours and Pierre A. Robe _

Abstract

Marie Willems1, Nadège Dubois1, Lucia Musumeci1, Vincent Bours1 and Pierre A. Robe1,2

1 Department of Human Genetics and GIGA Research Center, University of Liège, Liege, Belgium

2 Department of Neurology and Neurosurgery, T&P Bohnenn Laboratory for Neuro-Oncology, Brain Center Rudolf Magnus, University Medical Center of Utrecht, Heidelberglaan, Utrecht, The Netherlands

Correspondence to:

Pierre A. Robe, email:

Keywords: IκBζ, nuclear IκB protein, NF-κB pathway, cancer, perspectives

Received: January 19, 2016 Accepted: August 23, 2016 Published: August 26, 2016

Abstract

IκBζ, an atypical member of the nuclear IκB family of proteins, is expressed at low levels in most resting cells, but is induced upon stimulation of Toll-like/IL-1 receptors through an IRAK1/IRAK4/NFκB-dependent pathway. Like its homolog Bcl3, IκBζ can regulate the transcription of a set of inflamatory genes through its association with the p50 or p52 subunits of NF-κB. Long studied as a key component of the immune response, IκBζ emerges as an important regulator of inflammation, cell proliferation and survival. As a result, growing evidence support the role of this transcription factor in the pathogenesis number of human hematological and solid malignancies.


Introduction

The NF-κB family of proteins

NF-κB (Nuclear Factor kappa B) is a ubiquitous family of transcription factors involved in biological processes such as inflammation, immunity, proliferation and apoptosis [1-3]. This family of proteins comprises two subfamilies that share a DNA-binding and dimerization domain called the Rel homology domain (RHD) [4] and form homo- or hetero- dimers. The first subfamily of proteins (c-Rel, RelB, p65/RelA) contains a C-terminal transactivation domain. The second subfamily of proteins (p105 and p100) has a C-terminal region that contains multiple copies of ankyrin repeats, instead of a transactivation domain, and can bind to and inhibit Rel proteins. p100 and p105 can however undergo limited proteolysis to generate p52 and p50, respectively, which can form heterodimers with Rel proteins to form transcriptional activators [5].

The involvement of NF-κB in the development, the progression and the therapeutic resistance of many human cancers is well established. Constitutive p50/p65 activity is observed in a large variety of hematological as well as solid tumors [6-8], as a result of an aberrant expression of p50/p65, deletions of the IκBα inhibitor gene or an increased IKK activity [9-13]. Through this constitutive activity, NF-κB p50/p65 acts in tumors mainly as an inhibitor of apoptosis [8, 14]. In addition, anti-cancerous agents, such as TNFα, ionizing radiation and chemotherapeutic drugs activate p50/p65 [15, 16] leading to cell survival and consequently to drug resistance.

Several clinical trials using inhibitors of NF-κB activation have been performed, and have shown variable results in a few types of cancers [17-21]. To date, the most significant clinical results have been obtained with bortezomib, an inhibitor of the proteasome, for the treatment of multiple myeloma [22].

The IκB family of proteins

NF-κB protein dimers are kept in the cytoplasm by interaction with proteins of the IκB family (IκB -α, -β and -ε), or by their p100 or p105 component that masks their nuclear localization sequences (NLS, Figure 1, panel A). Upon phosphorylation of specific serine residues, these ankyrin-repeat proteins undergo proteasome- or calpain-dependent complete or limited degradation, allowing the nuclear translocation of the NF-κB protein dimers [23]. The activation of NF-κB occurs via either the classical, the alternative, the atypical or the p105-dependent pathways according to the stimuli and the kinases implicated. IκBα, -β and -ε can be phosphorylated by IKKβ (classical pathway), inducing their proteasome degradation. Following UV-irradiation, CK2 can also phosphorylate IκBα, leading to its calpain-dependent degradation (atypical pathway). p100 and p105 phosphorylations respectively depend upon IKKα and IKKβ, themselves activated by NIK. These alternative pathways lead to the activation of RelB/p52 and RelB/p50 pathways, respectively [24, 25].

The IκB family of proteins also comprises additional members (Figure 1, panel B) named nuclear IκB proteins due to the presence of a conserved nuclear localization signal. Unlike the cytoplasmic IκB proteins, the nuclear IκB proteins also harbor a trancriptional activity. Bcl3, which is predominantly expressed in the nucleus, acts as a nuclear transcriptional co-activator or co-repressor that can activate or repress a set of NF-κB target genes through the formation of heterocomplexes with p50 or p52 dimers [26]. Another nuclear IκB protein, called IκBNS, was also shown to be a nuclear transcription factor. IκBNS is a short-lived protein induced by NF-κB activation and its degradation depends upon the proteasome and is regulated by ubiquitin-independent post-traductional modifications of its PEST-domain [27].

Schematic representation of the IκB family of proteins.

Figure 1: Schematic representation of the IκB family of proteins. A. The cytoplasmic IκB proteins. Notes: PEST: domain rich in proline, glutamic acid, serine and threonine; AR: ankyrin-repeat; NES: nuclear export signal; NIS: nuclear import signal; RHD: Rel homology domain; GRR: glycine-rich region. b. The nuclear IκB proteins. Notes: AR: ankyrin-repeat; NLS: nuclear localization signal; TAD: transactivating domain.

IκBζ

IκBζ, a third member of the nuclear IκB family that shares a strong functional and structural homology with Bcl3 and IκBNS, was discovered in 2000 by Kitamura and collaborators as a new ankyrin repeats-containing protein of unknown function that is induced in the mouse brain in response to LPS and that shares homology with IκB protein [28]. Almost at the same time, Haruta identified the same gene in OP9 stromal cells stimulated with interleukin-1 [29].

IκBζ is encoded by NFKBIZ, Nuclear Factor Of Kappa Light Polypeptide Gene Enhancer In B-Cells Inhibitor Zeta. Southern hybridization showed that NFKBIZ is a single-copy gene and is conserved in human, chimpanzee, Rhesus monkey, dog, cow, mouse, rat, chicken and zebrafish. Using fluorescence in situ hybridization analysis, human NFKBIZ gene was mapped to chromosome 3q12.3 [30].

Transcription of NFKBIZ produces fifteen alternative mRNA splice and truncated variants, but only three of these mRNA code for a protein. The long IκBζ(L) mRNA variant contains the sequence from 14 exons while the short IκBζ(S) lacks exon 3 which contains the initiation codon of IκBζ(L), and thus encodes from a downstream initiation site a shorter protein lacking the N-terminal 99 amino acids of IκBζ(L). Further investigations are needed to be able to functionnally distinguish these two variants. The third variant, called IκBζ(D), has a large deletion in the central region and results from an additional splicing in the seventh exon. Present as a minor form in cells [31], IκBζ(D) does not possess the TAD (Transactivating domain) and consequently does not have any transcriptional activity (Figure 1, panel B).

Regulation of IκBζ protein

The IκBζ protein is barely detectable in most resting cells, with the exception of keratinocytes and several mucosal tissues [32, 33]. Its expression is however readily induced in most tissues upon stimulation of Toll-like receptors (TLR) 2, 4,5, 7 and 9 by their exogenic ligands peptidoglycan, bacterial and mycoplasmal lipopeptides, flagellin, CpG oligonucleotides or LPS [28, 34, 35]. Proinflammatory cyokines, such as IL-1β also strongly induces IκBζ via its receptor IL1-R [36, 37].

The TLR -with the exception of TLR-3- and IL1-R share similar cytoplasmic domains called TIR (Toll/IL1Receptors) and bind the adaptor protein MyD88. Upon stimulation, MyD88 recruits the serine-threonine kinases IRAK 1 and 4 to the receptor [38]. Activated IRAK4 then phosphorylates IRAK1, inducing its dissociation from the receptor complex and allowing its interaction with TRAF-6. TRAF-6 in turn activates MAP3K7/TAK-1 which activates the NIK/IKK/IκB/NF-κB as well as the MAPK pathways [39, 40]. The induction of IκBζ is completely abolished in MyD88-/- embryonic fibroblasts [35], by several NF-κB drug inhibitors, or by the overexpression of IκB-α [34]. MAP kinase inhibitors on the contrary do not prevent the induction of IκBζ, indicating that the three MAP kinases, Erk, JNK and p38 kinases are dispensable in this process.

While necessary, the activation of NF-κB is however not sufficient for the activation of IκBζ, and an additional step of mRNA stabilization is required. Indeed, the overexpression of p65 or the activation of NF-κB and MAPK by TNFα barely increase IκBζ protein expression [34, 37] and the short half-life of the IκBζ mRNA (30 min) increases after stimulation with LPS or IL-1β, but not after TNFα receptor activation [41].

This mRNA stabilization depends on the recruitment of IRAK-1 and TRAF-6 to the TIR domain of IL1-R and TLR receptors [42] (Figure 2) and on a 165-nucleotide cis-element present in the 3’-UTR of the IκBζ mRNA (Untranslated region) [43]. This cis-element contains four AU-rich elements (AREs) that are the recognition signals for an mRNA processing pathway restricted to certain lymphokines, cytokines and proto-oncogenes [44]. The stabilization of IκBζ mRNA does however not respond to the same stimuli as that of cytokines, and the overexpression of HuR [45] or Apobec-1 [46], the transacting factors that bind ARE to stabilize the mRNA of these cytokines, does not affect the stability of the IκBζ mRNA. The exact post-trascriptional regulatory mechanism that leads to IκBζ mRNA stabilization via its cis-element remains thus largely unknown, although some recent findings may provide some clues.

Stable induction of IκBζ.

Figure 2: Stable induction of IκBζ. Barely detectable in resting cells, IκBζ is induced by lipopolysaccharide (LPS) and IL-1β. Both Toll-like receptor (TLR) and IL-1R share a similar cytoplasmic TIR domain that binds the MyD88 adaptator protein. Under stimulation, MyD88 recruits IRAK1 and IRAK4 leading to the dissociation of IRAK1 and its binding to TRAF6. The complex IRAK1/TRAF6 activates then TAK1 which in turn induces NF-κB translocation. The mRNA stabilization of IκBζ depends upon the recruitment of IRAK1 to the TIR domain of the IL-1R and TLR receptors as well as on a 165-nucleotides sequence present in the 3’-UTR of the IκBζ mRNA. Notes: LPS: lipopolysaccharides; IL-1β: interleukin 1β; TLR: toll-like receptor; IL-1R: IL-1 receptor; IRAK1/4: interleukin-1 receptor-associated kinase 1/4; TRAF6: TNF receptor-associated factor 6; TAK1: transforming growth factor beta-activated kinase 1; IκB: inhibitor of κB ; IKK: IκB kinase.

Recently for instance, the micro-RNA miR-124a was found to directly target IκBζ mRNA by base pairing to a partially complementary sequence in the 3’UTR, called 7mer (7 nt sites that match the seed region of the miRNA). As a result, miR-124a can suppress IκBζ expression through translational repression [47]. Likewise, in silico data suggest that other miRNAs could regulate the stability of IκBζ mRNA as well [48].

Little is known about the post-translational regulation of IκBζ activity. Immunoprecipitation experiments indicate that transfected IκBζ strongly associates with p50/p50 and p50/p65 complexes. IκBζ preferentially binds the p50 subunits of these complexes and its association with the p65 subunit has to date exclusively been detected after overexpression of both proteins [37]. This preferential binding to the p50 subunit is reminiscent of that of Bcl3 [49] and IκBNS [50, 51]. IκBζ, like Bcl3, was also recently shown to associate with p52 in ABC DLBCL (activated B-cell-like subtype of diffuse large B-cell lymphoma) [52]. Like other nuclear IκB proteins, IκBζ regulates the transcriptional activity of NF-κB by forming a stable ternary complex with the subunits of NF-κB and κB sites in the nucleus [53]. The details of the formation of these ternary complexes between IκBζ, NF-κB and the DNA is not yet completely understood. This interaction however appears to be independent from the DNA sequences flanking the NF-κB binding site but involves both the C-terminal extremity of IκBζ, which interacts with the subunits of NF-κB linked to the DNA, and its N-terminal NLS [54, 55]. Of note, experimental IκBζ mutants defective for their NLS localize in the cytosol and inhibit NF-κB like conventional IκB proteins [37, 56]. Whether such a phenomenon also occurs in physiological conditions is to date unknown.

It is currently unknown whether IκBζ phosphorylation, ubiquitination or other post-translational protein modifications alter its interactions with NF-κB nuclear or cytoplasmic complexes. In silico analyses, however, reveals the presence of several serine/threonine or tyrosine- containing motives for casein kinase 2, EGFR, Chck2, ATR and MAP kinases in functional domains of the protein (Figure 3).

In silico analysis of serine/thréonine and tyrosine- containing motives in IκBζ functional domains.

Figure 3: In silico analysis of serine/thréonine and tyrosine- containing motives in IκBζ functional domains. Notes: JNK1: c-Jun N-terminal kinase 1; Pim1: serine/threonine-protein kinase pim-1; NeK10: NIMA-related kinase 10; ATR: ataxia telangiectasia and Rad3 related; ERK2: extracellular signal-regulated kinase 2; LRRK2: leucine-rich repeat kinase 2; EGFR: epidermal growth factor receptor; ErbB3: erb-b2 receptor tyrosine kinase 3; CHK2: checkpoint kinase 2; MEK4: mitogen-activated protein kinase kinase 4; PKC: protein kinase C; CKII: casein kinase 2.

IκBζ and gene transcription

Like its homolog Bcl3 that can either induce or repress gene transcription depending on the cellular context and through its association with the p50 or p52 subunit of NF-κB [57], IκBζ can both promote or inhibit gene expression [56, 58] (Figure 4).

Under transient stimulation, IκBζ inhibits the activity of NF-κB by preventing the binding of this transcription factor to the DNA in the nucleus. Detailed electrophoretic mobility shift assays using a probe harboring a canonical NF-κB binding sequence showed that the DNA-binding activity of the NF-κB p65/p50 heterodimer or p50/p50 homodimer was inhibited by the C-terminal ankyrin-repeats of a IκBζ [37]. As such, IκBζ can participate in the control of NF-κB through a negative feedback loop [59]. Likewise, IκBζ can inhibit the DNA binding of, STAT3, another key transcription factor which acts downstream of the JAK-STAT (Janus kinase/signal transducer and activator of transcription) pathway to regulate cell proliferation and apoptosis [60].

IκBζ can however also activate the transcription of a set of genes (Table 1, [28, 35, 53, 58, 61-66]). Since IκBζ has no obvious DNA binding motif, and since no consensus structural feature has been found among the promoter sequences of IκBζ-regulated genes, it is unlikely that IκBζ directly associates with DNA to activate gene transcription. It more likely stabilizes or assists the promoter binding of other transcription regulators.

Reporter gene and chromatin immunoprecipitation assays have indeed shown that the NF-κB and C/EBP(CCAAT/enhancer-binding protein) DNA binding sites are minimal elements essential for the IκBζ mediated transcriptional activation of IκBζ-responsive genes [67]. Yamazaki and collaborators also found that the activation of NF-κB, besides being required for IκBζ induction, is also substantially involved in the transcriptional up-regulation of the IκBζ target genes [68]. Gene knockdown experiment using specific siRNAs indicated that p50, which is known to be constitutively bound to NF-κB-driven promoters, and IκBζ could form a core element for transcriptional activation of target genes while p65 transcriptional activity might be required for the full activation of some of them [69].

A study performed on HEK293 cells using a GAL4 fusion protein technique also suggests that an internal fragment of IκBζ rich in glutamines and prolines (amino acids 329 to 402) possesses an intrinsic transcriptional activity. This transcriptional function would mainly proceed via the interaction of IκBζ with the NF-κB p50 subunit. The binding of p50 to IκBζ via the ankyrin repeats of IκBζ would in this model prevent the C-terminal region of IκBζ from inhibiting the activity of its own N-terminal region [58].

Finally, IκBζ was identified as a selective regulator of H3K4 trimethylation after nucleosome remodeling. H3K4 trimethylation is an histone-modifying reaction that alters the N-terminal tails and core domains of histones to regulate transcription. This epigenetic mechanism has a well known physiological role in the molecular cascades that regulates transcription of genes involved in primary and secondary inflammatory responses [69] and in cancer [70, 71].

Table 1: Confirmed IκBζ target genes

Regulation

Partners

Cell types

References

IL-6

+

p50; p65

Swiss 3T3 cells; Monocytes

28; 53; 61

hBD2

+

p50

HBE1

62

NGAL

+

NF-κB

A549

63

CCL2

+

NF-κB

Raw264.7

64

IFNγ

+

p50; p65

Lymphocytes; NK cells;

HEK 293;

KG-1; Monocytes

65; 66

GM-CSF

+

?

Macrophages

35

M-CSF

+

?

Macrophages

35

TNFα

-

p50

HeLa; COS-7; HEK 293

58

IL-12

+

?

Macrophages

35

Notes: IL-6/12: interleukin 6/12; hBD2: human beta-defensin 2; NGAL: neutrophil gelatinase-associated lipocalin; CCL2: chemokine ligand 2; IFNγ: interferon gamma; GM/M-CSF: granulocyte-macrophage/macrophage colony-stimulating factor; TNFα: tumor necrosis factor alpha. Positive (+) or negative (-) transcriptionnal regulation of targeted genes by IκBζ.

IκBζ function in gene régulation.

Figure 4: IκBζ function in gene régulation. A. After NF-κB activation through diverse stimuli, IκBζ is transcribed and transiently induced. Under these conditions, IκBζ acts as a inhibitor of the homodimer p50/p50 or the heterodimer p50/p65 through a negative feedback loop B. Upon specific stimulation with LPS or IL-1β leading to IκBζ mRNA stabilization and protein expression, IκBζ forms a ternary complex with p50 and p65 on the promoter of target genes and activates or repress their transcription. Notes: IKK: IκB kinase; IκB: inhibitor of κB; NF-κB: nuclear factor of κB.

IκBζ, inflammation and immunity

In line with its transcriptional targets (Table 1), the most important known physiological role of IκBζ was demonstrated in innate immunity against common pathogens, through the modulation of genes of the secondary inflammatory response [35]. Besides its main function in innate immunity, some studies suggested a role for IκBζ in adaptative immunity [36, 72]. As an example, it was showed that IκBζ is induced upon stimulation of B cell antigen receptor (BCR) [73, 74].

As a corollary, IκBζ is involved in diseases related to the response of physical and chemical barriers against infectious agents. NFKBIZ gene-invalidated mice show atopic dermatitis-like lesions [32]. Likewise, IκBζ is involved in the epithelial cell cytokine responses observed in asthma due to house dust mite, where allergens induce monocyte IL-1β production triggering an IκBζ-dependent GM-CSF release from human lung epithelial cells [75]. In patients with ulcerative colitis, the expression of lipocalin-2, an essential marker of activity of the disease, is regulated synergically by IL17-A, IL22 and TNFα in an IκBζ-dependent manner [76, 77]. Likewise, an important role of IκBζ was highlighted in various autoimmune diseases, for example in Sjögren’s syndrome-like disease [78], Crohn’s disease [79], rhumatoid arthritis [80] as well as in psoriasis. For this last disease, a new susceptibility DNA polymorphism (rs7637230, G→A) was found at a locus adjacent to NFKBIZ [81].

IκBζ in cancer

A strong relation exists between inflammation and cancer, as inflammation plays a critical role in tumor initiation and progression but also influences the response to the treatment [82, 83]. The tumor microenvironment contains innate and adaptative immune cells [84] that interact with cancer cells by direct contact or cytokine and chemokine production. The expression of immune mediators as well as the abundance and activation state of infiltrating immune cells therefore influence tumor growth, anti-tumor immune response, tissue invasion and metastasis, as well as the clinical response to chemotherapy or immunotherapy [85, 86]. The well-known role of IκBζ in cytokine production [62, 64, 65] and its expression in various immune cells suggests a possible role of IκBζ in the tumor microenvironment. In support of this hypothesis, chemically-induced skin carninogenesis was found to associate with both a significant inflammatory response and a major induction of NFKBIZ in mice [87].

Bcl3, which is highly homologous to IκBζ, is also directly involved in lymphoproliferative disorders [88-91] and in solid tumors [92]. Bcl3 was for instance found to promote metastasis in ERBB2-driven mammary tumors [93] and to attenuate the efficacy of Temozolomide in glioma cells [94]. High levels of Bcl3 expression have been observed in various solid tumors where it is involved in the control of cell death and proliferation [95-97]. Likewise, IκBζ is activated and overexpressed in ATL (Adult T cell Leukemia) induced by HTLV1 (Human T cell leukemia virus type I) via the oncoprotein Tax [98], suggesting a role in these lymphoid cancers. NFKBIZ was also recently identified in a molecular signature characteristic of mycosis fungoides, the most common type of primary cutaneous T-cell lymphoma (CTCL) [99]. A recent study showed that activated B-cell-like subtype of diffuse large B-cell lymphoma overexpress IκBζ as compared to control B cells and that its downregulation is selectively toxic to these tumor cells [52] through an activation of the the caspase 3 pathway [78]. More recently, both mutations and amplification of the NFKBIZ gene are associated with the occurrence of primary testicular and primary central nervous system lymphomas [100, 101].

High levels of IκBζ expression have also been observed in solid tumors. For instance, Görranson and colleagues described a role for the interaction of IκBζ with the FUS-DDIT3 fusion oncoprotein in the initiation of myxoïd/round cell liposarcomas (MLS/RCLS) through the transcription of NF-κB dependent genes [102]. The tumor-suppressor miR-124a, and miR-223, which target the NFKBIZ mRNA, were recently shown to be silenced in glioblastomas [47, 48, 103], and we have observed that IκBζ is expressed in these glial tumors where it prevents spontaneous cell death (unpublished data).

The biological role of IκBζ in human cancers might however be more intricate than seems at first. This protein indeed modulates altogether cell death, survival and proliferation, and might even work as an oncosupressor in certain tumor types (Figure 5). IκBζ is for instance a regulator of the senescence associated secretory phenotype (SASP) constituted by various growth factors and cytokines secreted by senescent cells, and transgenic IκBζ expression results in enhanced SASP cytokine expression [104]. Wu and collaborators showed that IκBζ physically interacts with and inhibits the transcriptional activity of the oncoprotein Bcl3, leading to apoptosis induction [60]. As mentioned above, IκBζ can also inhibit the activity of the transcription factor STAT3 [60], a transcription factor that is itself frequently overexpressed in tumors, and that regulates the expression of numerous oncogenic genes controlling cell growth and metastasis [105]. Finally, in human fibrosarcoma cells (HT-1080) and breast carcinoma cells (MCF-7/casp-3), the repression of IκBζ with interferent RNA render the cells more resistant to apoptosis, while its overexpression is sufficient to induce cell death [106].

Figure 5 : IκBζ and its involvment in cancer.

Figure 5 : IκBζ and its involvment in cancer. Schematic representation of the potential upstream regulators of IκBζ as well as the IκBζ targets and their relative biological effects. Notes: Tax: transactivator of pX; FUS: fused in sarcoma; DDIT3: DNA damage-inducible transcript 3; Bcl3: B cell lymphoma 3; NF-κB: nuclear factor of κB; STAT3: signal transducer and activator of transcription 3.

ConclusionS, therapeutic potential, ongoing research and unexplored aspects

IκBζ emerges as an important regulator of inflammation, cell proliferation and survival through its modulation of NF-κB and STAT3 signalings. As such, growing evidence points to the physiopathological role of this transcription factor in a number of hematological and solid malignancies.

Additional tumor specific knowledge is mandatory prior to translating current experimental data to the bedside, given the potentially dual role of IκBζ in cell proliferation and survival. Indeed, IκBζ inhibition can lead either to cell death, in most of cell types, or to cell survival in a few experimental settings. These findings should stimulate further research on the cell-type specific mechanisms regulating IκBζ protein-protein and protein-DNA interactions and pave the way to innovative anti-cancer therapies.

Conflicts of interest

The authors have no conflict of interest with respect to the topic of this manuscript.

Funding

This work was supported by from: the Belgian Federal Cancer Plan, FNRS (National Fund for Scientific Research, Belgium), the StopHersenTumoren.nl Foundation, the T&P Bohnenn Fund for Neuro-Oncological Research, the Zabawas Foundation, , the Lyons Club, the ‘Vrienden van het UMC Utrecht’ Foundation and the Anticancer Center of the University of Liège.

REFERENCES

1. Forsyth PA, Wong H, Laing TD, Rewcastle NB, Morris DG, Muzik H, Leco KJ, Johnston RN, Brasher PM, Sutherland G, Edwards DR. Gelatinase-A (MMP-2), gelatinase-B (MMP-9) and membrane type matrix metalloproteinase-1 (MT1-MMP) are involved in different aspects of the pathophysiology of malignant gliomas. Br J Cancer. 1999 Apr;79(11-12):1828-35.

2. Baeuerle PA, Henkel T. Function and activation of NF-kappa B in the immune system. Annu Rev Immunol. 1994;12:141-79.

3. Munaut C, Noël A, Hougrand O, Foidart J-M, Boniver J, Deprez M. Vascular endothelial growth factor expression correlates with matrix metalloproteinases MT1-MMP, MMP-2 and MMP-9 in human glioblastomas. Int J Cancer. 2003 Oct 10;106(6):848-55.

4. Gilmore TD. NF-kappa B, KBF1, dorsal, and related matters. Cell. 1990 Sep 7;62(5):841-3.

5. Gilmore TD. Introduction to NF-kappaB: players, pathways, perspectives. Oncogene. 2006 Oct 30;25(51):6680-4.

6. Loercher A, Lee TL, Ricker JL, Howard A, Geoghegen J, Chen Z, Sunwoo JB, Sitcheran R, Chuang EY, Mitchell JB, Baldwin AS, Van Waes C. Nuclear factor-kappaB is an important modulator of the altered gene expression profile and malignant phenotype in squamous cell carcinoma. Cancer Res. 2004 Sep 15;64(18):6511-23.

7. Robe PA, Bentires-Alj M, Bonif M, Rogister B, Deprez M, Haddada H, Khac M-TN, Jolois O, Erkmen K, Merville M-P, Black PM, Bours V. In vitro and in vivo activity of the nuclear factor-kappaB inhibitor sulfasalazine in human glioblastomas. Clin Cancer Res. 2004 Aug 15;10(16):5595-603.

8. Sovak MA, Bellas RE, Kim DW, Zanieski GJ, Rogers AE, Traish AM, Sonenshein GE. Aberrant nuclear factor-kappaB/Rel expression and the pathogenesis of breast cancer. J Clin Invest. 1997 Dec 15;100(12):2952-60.

9. Pikarsky E, Ben-Neriah Y. NF-kappaB inhibition: a double-edged sword in cancer? Eur J Cancer. 2006 Apr;42(6):779-84.

10. Rayet B, Gélinas C. Aberrant rel/nfkb genes and activity in human cancer. Oncogene. 1999 Nov 22;18(49):6938-47.

11. Greten FR, Eckmann L, Greten TF, Park JM, Li Z-W, Egan LJ, Kagnoff MF, Karin M. IKKbeta links inflammation and tumorigenesis in a mouse model of colitis-associated cancer. Cell. 2004 Aug 6;118(3):285-96.

12. Hoffmann A, Levchenko A, Scott ML, Baltimore D. The IkappaB-NF-kappaB signaling module: temporal control and selective gene activation. Science. 2002 Nov 8;298(5596):1241-5.

13. Pikarsky E, Porat RM, Stein I, Abramovitch R, Amit S, Kasem S, Gutkovich-Pyest E, Urieli-Shoval S, Galun E, Ben-Neriah Y. NF-kappaB functions as a tumour promoter in inflammation-associated cancer. Nature. 2004 Sep 23;431(7007):461-6.

14. Bentires-Alj M, Barbu V, Fillet M, Chariot A, Relic B, Jacobs N, Gielen J, Merville M-P, Bours V. NF-kappaB transcription factor induces drug resistance through MDR1 expression in cancer cells. Oncogene. 2003 Jan 9;22(1):90-7.

15. Bours V, Bonizzi G, Bentires-Alj M, Bureau F, Piette J, Lekeux P, Merville M. NF-kappaB activation in response to toxical and therapeutical agents: role in inflammation and cancer treatment. Toxicology [Internet]. 2000 Nov 16;153(1-3):27-38.

16. Pahl HL. Activators and target genes of Rel/NF-kappaB transcription factors. Oncogene. 1999 Nov 22;18(49):6853-66.

17. Olivier S, Robe P, Bours V. Can NF-kappaB be a target for novel and efficient anti-cancer agents? Biochemical Pharmacology. 2006 Oct 30;72(9):1054-68.

18. Stark LA, Din FV, Zwacka RM, Dunlop MG. Aspirin-induced activation of the NF-kappaB signaling pathway: a novel mechanism for aspirin-mediated apoptosis in colon cancer cells. FASEB J. 2001 May;15(7):1273-5.

19. Scheinman RI, Gualberto A, Jewell CM, Cidlowski JA, Baldwin AS. Characterization of mechanisms involved in transrepression of NF-kappa B by activated glucocorticoid receptors.MCB. 1995 Feb;15(2):943-53.

20. Murgo AJ. Clinical trials of arsenic trioxide in hematologic and solid tumors: overview of the National Cancer Institute Cooperative Research and Development Studies. Oncologist. 2001;6 Suppl 2:22-8.

21. Robe PA, Martin DH, Nguyen-Khac MT, Artesi M, Deprez M, Albert A, Vanbelle S, Califice S, Bredel M, Bours V. Early termination of ISRCTN45828668, a phase 1/2 prospective, randomized study of Sulfasalazine for the treatment of progressing malignant gliomas in adults. BMC Cancer. 2009;9(1):372.

22. Broijl A, Kersten M-J, Alemayehu WG, Levin M-D, de Weerdt O, Vellenga E, Meijer E, Wittebol S, Tanis BC, Cornelisse PB, Stevens-Kroef M, Bos GMJ, Wijermans PW et al. Phase I/II trial of Weekly Bortezomib with Lenalidomide and Dexamethasone in First Relapse or Primary Refractory Myeloma. Haematologica. 2015 Dec 11.

23. Karin M, Ben-Neriah Y. Phosphorylation meets ubiquitination: the control of NF- [kappa]B activity. Annu Rev Immunol. 2000;18:621-63.

24. Gasparini C, Celeghini C, Monasta L, Zauli G. NF-kappaB pathways in hematological malignancies. Cell Mol Life Sci. 2014 Jun;71(11):2083-102.

25. Viatour P, Merville M-P, Bours V, Chariot A. Phosphorylation of NF-kappaB and IκB proteins: implications in cancer and inflammation. Trends in Biochemical Sciences. 2005 Jan;30(1):43-52.

26. Viatour P. GSK3-Mediated BCL-3 Phosphorylation Modulates Its Degradation and Its Oncogenicity. 2004 Sep 29;:1-11.

27. Park KC, Jeong J, Kim KI. Regulation of mIκBNS stability through PEST-mediated degradation by proteasome.BBRC. 2014 Jan 24;443(4):1291-5.

28. Kitamura H, Kanehira K, Okita K, Morimatsu M, Saito M. MAIL, a novel nuclear I kappa B protein that potentiates LPS-induced IL-6 production. FEBS Letters. 2000 Nov 17;485(1):53-6.

29. Haruta H. Isolation of a Novel Interleukin-1-inducible Nuclear Protein Bearing Ankyrin-repeat Motifs.J. Biol. Chem.. 2001 Mar 2;276(16):12485-8.

30. Shiina T, Morimatsu M, Kitamura H, Ito T, Kidou S, Matsubara K, Matsuda Y, Saito M, Syuto B. Genomic organization, chromosomal localization, and promoter analysis of the mouse Mail gene. Immunogenetics. 2001 Oct;53(8):649-55.

31. Chapman SJ, Khor CC, Vannberg FO, Rautanen A, Segal S, Moore CE, Davies RJO, Day NP, Peshu N, Crook DW, Berkley JA, Williams TN et al. NFKBIZ polymorphisms and susceptibility to pneumococcal disease in European and African populations. Genes Immun. 2009 Oct 1;11(4):319-25.

32. Shiina T. Targeted Disruption of MAIL, a Nuclear I B Protein, Leads to Severe Atopic Dermatitis-like Disease.J. Biol. Chem.. 2004 Oct 8;279(53):55493-8.

33. Ueta M. Spontaneous Ocular Surface Inflammation and Goblet Cell Disappearance in I B Gene-Disrupted Mice. Investigative Ophthalmology & Visual Science. 2005 Feb 1;46(2):579-88.

34. Eto A, Muta T, Yamazaki S, Takeshige K. Essential roles for NF-kappaB and a Toll/IL-1 receptor domain-specific signal(s) in the induction of IκB-ζ. BBRC. 2003 Feb;301(2):495-501.

35. Yamamoto M, Yamazaki S, Uematsu S, Sato S, Hemmi H, Hoshino K, Kaisho T, Kuwata H, Takeuchi O, Takeshige K, Saitoh T, Yamaoka S, Yamamoto N et al. Regulation of Toll/IL-1-receptor-mediated gene expression by the inducible nuclear protein IkappaBzeta. Nature. 2004 Jul 8;430(6996):218-22.

36. Kitamura H, Matsushita Y, Iwanaga T, Mori K, Kanehira K, Fujikura D, Morimatsu M, Saito M. Bacterial lipopolysaccharide-induced expression of the IkappaB protein MAIL in B-lymphocytes and macrophages. Arch Histol Cytol. 2003 Mar;66(1):53-62.

37. Yamazaki S. A Novel Ikappa B Protein, Ikappa B-zeta , Induced by Proinflammatory Stimuli, Negatively Regulates Nuclear Factor-kappa B in the Nuclei.J. Biol. Chem. 2001 May 16;276(29):27657-62.

38. Wesche H, Henzel WJ, Shillinglaw W, Li S, Cao Z. MyD88: an adapter that recruits IRAK to the IL-1 receptor complex. Immunity. 1997 Dec;7(6):837-47.

39. Irie T, Muta T, Takeshige K. TAK1 mediates an activation signal from toll-like receptor(s) to nuclear factor-kappaB in lipopolysaccharide-stimulated macrophages. FEBS Letters. 2000 Feb 11;467(2-3):160-4.

40. Ninomiya-Tsuji J, Kishimoto K, Hiyama A, Inoue J, Cao Z, Matsumoto K. The kinase TAK1 can activate the NIK-I kappaB as well as the MAP kinase cascade in the IL-1 signalling pathway. Nature. 1999 Mar 18;398(6724):252-6.

41. Yamazaki S. Stimulus-specific Induction of a Novel Nuclear Factor- B Regulator, I B- , via Toll/Interleukin-1 Receptor Is Mediated by mRNA Stabilization.J. Biol. Chem. 2004 Nov 1;280(2):1678-87.

42. Ohba T, Ariga Y, Maruyama T, Truong NK, Inoue J-I, Muta T. Identification of interleukin-1 receptor-associated kinase 1 as a critical component that induces post-transcriptional activation of IkappaB-ζ. FEBS Journal. 2011 Nov 30;279(2):211-22.

43. Watanabe S, Takeshige K, Muta T. A cis-element in the 3′-untranslated region of IkappaB-ζ mRNA governs its stimulus-specific expression. BBRC. 2007 May;356(3):785-91.

44. Shaw G, Kamen R. A conserved AU sequence from the 3’ untranslated region of GM-CSF mRNA mediates selective mRNA degradation. Cell. 1986 Aug 29;46(5):659-67.

45. Myer VE, Fan XC, Steitz JA. Identification of HuR as a protein implicated in AUUUA-mediated mRNA decay. EMBO J. 1997 Apr 15;16(8):2130-9.

46. Anant S, Davidson NO. An AU-rich sequence element (UUUN [A/U]U) downstream of the edited C in apolipoprotein B mRNA is a high-affinity binding site for Apobec-1: binding of Apobec-1 to this motif in the 3’ untranslated region of c-myc increases mRNA stability. MCB. 2000 Mar;20(6):1982-92.

47. Lindenblatt C, Schulze-Osthoff K, Totzke G. IkappaBzeta expression is regulated by miR-124a. Cell Cycle. 2009 Jul 1;8(13):2019-23.

48. Huang Y-T, Hsu T, Kelsey KT, Lin C-L. Integrative analysis of micro-RNA, gene expression, and survival of glioblastoma multiforme. Genet Epidemiol. 2015 Feb;39(2):134-43.

49. Wulczyn FG, Naumann M, Scheidereit C. Candidate proto-oncogene bcl-3 encodes a subunit-specific inhibitor of transcription factor NF-kappa B. Nature. 1992 Aug 13;358(6387):597-9.

50. Hirotani T, Lee PY, Kuwata H, Yamamoto M, Matsumoto M, Kawase I, Akira S, Takeda K. The nuclear IkappaB protein IkappaBNS selectively inhibits lipopolysaccharide-induced IL-6 production in macrophages of the colonic lamina propria. J Immunol. 2005 Mar 15;174(6):3650-7.

51. Fiorini E, Schmitz I, Marissen WE, Osborn SL, Touma M, Sasada T, Reche PA, Tibaldi EV, Hussey RE, Kruisbeek AM, Reinherz EL, Clayton LK. Peptide-induced negative selection of thymocytes activates transcription of an NF-kappa B inhibitor. Molecular Cell. 2002 Mar;9(3):637-48.

52. Nogai H, Wenzel S-S, Hailfinger S, Grau M, Kaergel E, Seitz V, Wollert-Wulf B, Pfeifer M, Wolf A, Frick M, Dietze K, Madle H, Tzankov A et al. IkappaB-ζ controls the constitutive NF-kappaB target gene network and survival of ABC DLBCL. Blood. 2013 Jul 18.

53. Trinh DV, Zhu N, Farhang G, Kim BJ, Huxford T. The Nuclear IκB Protein IkappaBζ Specifically Binds NF-kappaB p50 Homodimers and Forms a Ternary Complex on κB DNA. J. Mol. Biol. 2008 May 23;379(1):122-35.

54. HUXFORD T, Huang DB, Malek S, Ghosh G. The crystal structure of the IkappaBalpha/NF-kappaB complex reveals mechanisms of NF-kappaB inactivation. Cell. 1998 Dec 11;95(6):759-70.

55. Jacobs MD, Harrison SC. Structure of an IkappaBalpha/NF-kappaB complex. Cell. 1998 Dec 11;95(6):749-58.

56. Muta T, Yamazaki S, Eto A, Motoyama M, Takeshige K. IkappaB-zeta, a new anti-inflammatory nuclear protein induced by lipopolysaccharide, is a negative regulator for nuclear factor-kappaB. Journal of Endotoxin Research. 2003;9(3):187-91.

57. Palmer S, Chen YH. Bcl-3, a multifaceted modulator of NF-kappaB-mediated gene transcription. Immunol Res. 2008;42(1-3):210-8.

58. Motoyama M, Yamazaki S, Eto-Kimura A, Takeshige K and Muta A. Positive and Negative Regulation of Nuclear Factor-kappa B-mediated Transcription by IkappaB-zeta , an Inducible Nuclear Protein. J. Biol. Chem. 2005 Dec 16;280(9):7444-51.

59. Totzke. A novel member of the IkappaB family, human IkappaB-zeta, inhibits transactivation of p65 and its DNA binding. J. Biol. Chem. 2010 Aug 18;281:12645-54.

60. Wu Z, Zhang X, Yang J, Wu G, Zhang Y, Yuan Y, Jin C, Chang Z, Wang J, Yang X, He F. Nuclear protein IkappaB-ζ inhibits the activity of STAT3. BBRC. Elsevier Inc; 2009 Sep 18;387(2):348-52.

61. Seshadri S, Kannan Y, Mitra S, Parker-Barnes J, Wewers MD. MAIL Regulates Human Monocyte IL-6 Production. JI. 2009 Oct 2;183(8):5358-68.

62. Karlsen JR, Borregaard N, Cowland JB. Induction of neutrophil gelatinase-associated lipocalin expression by co-stimulation with interleukin-17 and tumor necrosis factor-alpha is controlled by IkappaB-zeta but neither by C/EBP-beta nor C/EBP-delta. J. Biol. Chem. 2010 May 7;285(19):14088-100.

63. Kao C-Y, Kim C, Huang F, Wu R. Requirements for two proximal NF-kappaB binding sites and IkappaB-zeta in IL-17A-induced human beta-defensin 2 expression by conducting airway epithelium. J Biol Chem. 2008 May 30;283(22):15309-18.

64. Hildebrand DG, Alexander E, Hörber S, Lehle S, Obermayer K, Münck N-A, Rothfuss O, Frick J-S, Morimatsu M, Schmitz I, Roth J, Ehrchen JM, Essmann F, Schulze-Osthoff K. IkappaBζ is a transcriptional key regulator of CCL2/MCP-1. JI. 2013 May 1;190(9):4812-20.

65. Kannan Y, Yu J, Raices RM, Seshadri S, Wei M, Caligiuri MA, Wewers MD. IκBζ augments IL-12- and IL-18-mediated IFN-γ production in human NK cells. Blood. 2011 Mar 10;117(10):2855-63.

66. Raices RM, Kannan Y, Bellamkonda-Athmaram V, Seshadri S, Wang H, Guttridge DC, Wewers MD. A Novel Role for IκBζ in the Regulation of IFNγ Production. Unutmaz D, editor. PLoS ONE. 2009 Aug 26;4(8):e6776.

67. Matsuo S, Yamazaki S, Takeshige K, Muta T. Crucial roles of binding sites for NF-kappaB and C/EBPs in IkappaB-ζ-mediated transcriptional activation. Biochem J. 2007 Aug 1;405(3):605.

68. Yamazaki S, Matsuo S, Muta T, Yamamoto M, Akira S, Takeshige K. Gene-specific Requirement of a Nuclear Protein, I B- , for Promoter Association of Inflammatory Transcription Regulators. J. Biol. Chem. 2008 Sep 10;283(47):32404-11.

69. Kayama H, Ramirez-Carrozzi VR, Yamamoto M, Mizutani T, Kuwata H, Iba H, Matsumoto M, Honda K, Smale ST, Takeda K. Class-specific regulation of pro-inflammatory genes by MyD88 pathways and IkappaBzeta. J. Biol. Chem. 2008 May 2;283(18):12468-77.

70. Schwartzentruber J, Korshunov A, Liu X-Y, Jones DTW, Pfaff E, Jacob K, Sturm D, Fontebasso AM, Quang D-AK, Tönjes M, Hovestadt V, Albrecht S, Kool M, Nantel A et al. Driver mutations in histone H3.3 and chromatin remodelling genes in paediatric glioblastoma. Nature. 2012 Feb 9;482(7384):226-31.

71. Lim S, Metzger E, Schüle R, Kirfel J, Buettner R. Epigenetic regulation of cancer growth by histone demethylases. Int J Cancer. 2010 Nov 1;127(9):1991-8.

72. Hanihara-Tatsuzawa F, Miura H, Kobayashi S, Isagawa T, Okuma A, Manabe I, Maruyama T. Control of Toll-like receptor-mediated T cell-independent type 1 antibody responses by the inducible nuclear protein IkappaB-ζ. J. Biol. Chem. 2014 Nov 7;289(45):30925-36.

73. Hijioka K, Matsuo S, Eto-Kimura A, Takeshige K, Muta T. Induction of the nuclear IkappaB protein IkappaB-ζ upon stimulation of B cell antigen receptor. BBRC. 2007 May;356(2):476-80.

74. Hanihara F, Takahashi Y, Okuma A, Ohba T, Muta T. Transcriptional and post-transcriptional regulation of IkappaB-ζ upon engagement of the BCR, TLRs and FcγR. Int Immunol. 2013 Sep;25(9):531-44.

75. Sundaram K, Mitra S, Gavrilin MA, Wewers MD. House Dust Mite Allergens Induce Monocyte IL-1β Production Triggering an IkappaBζ Dependent GMCSF Release from Human Lung Epithelial Cells. Am J Respir Cell Mol Biol. 2015 Jan 28.

76. Stallhofer J, Friedrich M, Konrad-Zerna A, Wetzke M, Lohse P, Glas J, Tillack-Schreiber C, Schnitzler F, Beigel F, Brand S. Lipocalin-2 Is a Disease Activity Marker in Inflammatory Bowel Disease Regulated by IL-17A, IL-22, and TNF-α and Modulated by IL23R Genotype Status. Inflamm Bowel Dis. 2015 Aug 7.

77. Kohda A, Yamazaki S, Sumimoto H. DNA element downstream of the κB site in the Lcn2 promoter is required for transcriptional activation by IkappaBζ and NF-kappaB p50. Genes Cells. 2014 Aug;19(8):620-8.

78. Okuma A, Hoshino K, Ohba T, Fukushi S, Aiba S, Akira S, Ono M, Kaisho T, Muta T. Enhanced apoptosis by disruption of the STAT3-IkappaB-ζ signaling pathway in epithelial cells induces Sjögren’s syndrome-like autoimmune disease. Immunity. 2013 Mar 21;38(3):450-60.

79. Kerami Z, Duijvis NW, Vogels EW, van Dooren FH, Moerland PD, Velde Te AA. Effect of interleukin-17 on gene expression profile of fibroblasts from Crohn’s disease patients. J Crohns Colitis. 2014 Oct 1;8(10):1208-16.

80. Ikeda S, Saijo S, Murayama MA, Shimizu K, Akitsu A, Iwakura Y. Excess IL-1 signaling enhances the development of Th17 cells by downregulating TGF-β-induced Foxp3 expression. JI. 2014 Feb 15;192(4):1449-58.

81. Tsoi LC, Spain SL, Ellinghaus E, Stuart PE, Capon F, Knight J, Tejasvi T, Kang HM, Allen MH, Lambert S, Stoll SW, Weidinger S, Gudjonsson JE et al. Enhanced meta-analysis and replication studies identify five new psoriasis susceptibility loci. Nat Commun. 2015;6:7001.

82. Karin M. Nuclear factor-kappaB in cancer development and progression. Nature. 2006 May 25;441(7092):431-6.

83. Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell. 2010 Mar 19;140(6):883-99.

84. de Visser KE, Eichten A, Coussens LM. Paradoxical roles of the immune system during cancer development. Nat Rev Cancer. 2006 Jan;6(1):24-37.

85. Lin W-W, Karin M. A cytokine-mediated link between innate immunity, inflammation, and cancer. J Clin Invest. 2007 May;117(5):1175-83.

86. Smyth MJ, Dunn GP, Schreiber RD. Cancer immunosurveillance and immunoediting: the roles of immunity in suppressing tumor development and shaping tumor immunogenicity. Adv Immunol. 2006;90:1-50.

87. Martin-Olivia D, Aguilar-Quesada R, O’valle F, Muñoz-Gámez JA, Martínez-Romero R, García Del Moral R, Ruiz de Almodóvar JM, Villuendas R, Piris MA, Oliver FJ. Inhibition of poly(ADP-ribose) polymerase modulates tumor-related gene expression, including hypoxia-inducible factor-1 activation, during skin carcinogenesis. Cancer Res. 2006 Jun 1;66(11):5744-56.

88. Au WY, Horsman DE, Ohno H, Klasa RJ, Gascoyne RD. Bcl-3/IgH translocation (14;19)(q32;q13) in non-Hodgkin’s lymphomas. Leuk Lymphoma. 2002 Apr;43(4):813-6.

89. Canoz O, Rassidakis GZ, Admirand JH, Medeiros LJ. Immunohistochemical detection of BCL-3 in lymphoid neoplasms: a survey of 353 cases. Mod Pathol. 2004 Aug;17(8):911-7.

90. Schlette E, Rassidakis GZ, Canoz O, Medeiros LJ. Expression of bcl-3 in chronic lymphocytic leukemia correlates with trisomy 12 and abnormalities of chromosome 19. Am J Clin Pathol. 2005 Mar;123(3):465-71.

91. van Krieken JH, McKeithan TW, Raghoebier S, Medeiros LJ, Kluin PM, Raffeld M. Chromosomal translocation t(14;19) as indicated by bcl-3 rearrangement is a rare phenomenon in non-Hodgkin’s lymphoma and chronic lymphocytic leukemia: a molecular genetic analysis of 176 cases. Leukemia. 1990 Dec;4(12):811-2.

92. Maldonado V, Melendez-Zajgla J. Role of Bcl-3 in solid tumors. Mol Cancer. 2011;10:152.

93. Wakefield A, Soukupova J, Montagne A, Ranger J, French R, Muller WJ, Clarkson RWE. Bcl3 selectively promotes metastasis of ERBB2-driven mammary tumors. Cancer Research. 2013 Jan 15;73(2):745-55.

94. Mansour NM, Bernal GM, Wu L, Crawley CD, Cahill KE, Voce DJ, Balyasnikova IV, Zhang W, Spretz R, Nunez L, Larsen GF, Weichselbaum RR, Yamini B. Decoy Receptor DcR1 Is Induced in a p50/Bcl3-Dependent Manner and Attenuates the Efficacy of Temozolomide. Cancer Research. 2015 May 15;75(10):2039-48.

95. Puvvada SD, Funkhouser WK, Greene K, Deal A, Chu H, Baldwin AS, Tepper JE, O’Neil BH. NF-kappaB and Bcl-3 activation are prognostic in metastatic colorectal cancer. Oncology. 2010;78(3-4):181-8.

96. Cogswell PC, Guttridge DC, Funkhouser WK, Baldwin AS. Selective activation of NF-kappa B subunits in human breast cancer: potential roles for NF-kappa B2/p52 and for Bcl-3. Oncogene. 2000 Feb 24;19(9):1123-31.

97. Thornburg NJ, Pathmanathan R, Raab-Traub N. Activation of nuclear factor-kappaB p50 homodimer/Bcl-3 complexes in nasopharyngeal carcinoma. Cancer Res. 2003 Dec 1;63(23):8293-301.

98. Kimura R, Senba M, Cutler SJ, Ralph SJ, Xiao G, Mori N. Human T Cell Leukemia Virus Type I Tax-Induced IkappaB-ζ Modulates Tax-Dependent and Tax-Independent Gene Expression in T Cells. Neoplasia. 2013 Sep;15(9):1110-24.

99. van Kester MS, Borg MK, Zoutman WH, Out-Luiting JJ, Jansen PM, Dreef EJ, Vermeer MH, van Doorn R, Willemze R, Tensen CP. A meta-analysis of gene expression data identifies a molecular signature characteristic for tumor-stage mycosis fungoides. J Invest Dermatol. 2012 Aug;132(8):2050-9.

100. Chapuy B, Roemer MG, Stewart C, Tan Y, Abo RP, Zhang L, Dunford AJ, Meredith DM, Thorner AR, Jordanova ES, Liu G, Feuerhake F, Ducar MD et al. Targetable genetic features of primary testicular and primary central nervous system lymphomas. Blood. 2016 Feb 18;127(7):869-81.

101. Morin RD, Assouline S, Alcaide M, Mohajeri A, Johnston RL, Chong L, Grewal J, Yu S, Fornika D, Bushell K, Nielsen TH, Petrogiannis-Haliotis T, Crump M et al. Genetic Landscapes of Relapsed and Refractory Diffuse Large B-Cell Lymphomas. Clin Cancer Res. 2016 May 1;22(9):2290-300.

102. Göransson M, Andersson MK, Forni C, Ståhlberg A, Andersson C, Olofsson A, Mantovani R, Åman P. The myxoid liposarcoma FUS-DDIT3 fusion oncoprotein deregulates NF-kappaB target genes by interaction with NFKBIZ. Oncogene. 2008 Oct 13;28(2):270-8.

103. Tivnan A, Zhao J, Johns TG, Day BW, Stringer BW, Boyd AW, Tiwari S, Giles KM, Teo C, McDonald KL. The tumor suppressor microRNA, miR-124a, is regulated by epigenetic silencing and by the transcriptional factor, REST in glioblastoma. Tumour Biol. 2014 Feb;35(2):1459-65.

104. Alexander E, Hildebrand DG, Kriebs A, Obermayer K, Manz M, Rothfuss O, Schulze-Osthoff K, Essmann F. IκBζ is a regulator of the senescence-associated secretory phenotype in DNA damage- and oncogene-induced senescence. J Cell Sci. 2013 Aug 15;126(Pt 16):3738-45.

105. Chai EZP, Shanmugam MK, Arfuso F, Dharmarajan A, Wang C, Kumar AP, Samy RP, Lim LHK, Wang L, Goh BC, Ahn KS, Hui KM, Sethi G. Targeting transcription factor STAT3 for cancer prevention and therapy. Pharmacol Ther. 2015 Oct 20.

106. Totzke G, Essmann F, Pohlmann S, Lindenblatt C, Jänicke RU, Schulze-Osthoff K. A novel member of the IkappaB family, human IkappaB-zeta, inhibits transactivation of p65 and its DNA binding. J Biol Chem. 2006 May 5;281(18):12645-54.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 11624