Issue
4open
Volume 2, 2019
Disruption of homeostasis-induced signaling and crosstalk in the carcinogenesis paradigm “Epistemology of the origin of cancer”
Article Number 13
Number of page(s) 35
Section Life Sciences - Medicine
DOI https://doi.org/10.1051/fopen/2019010
Published online 10 May 2019

© B.L.D.M. Brücher et al., Published by EDP Sciences, 2019

Licence Creative CommonsThis is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB)

DNA transcription factors

Transcription factors (TFs) are proteins that control the transfer of genetic information from deoxyribonucleic acid (DNA) to messenger ribonucleic acid (mRNA) by binding to specific DNA sequences. TFs are effective by themselves or in conjunction with other proteins as a complex. TFs stimulate or suppress the recruitment of the corresponding ribonucleic acid (RNA) polymerase. In general, TFs are classified by structure or function [1]. The complexity of transcription is reflected by the fact that a tumor suppressor protein, such as protein 53 (p53), can act as an intracellular ligand (autocrine)-dependent functional TF and can be activated by small intracellular molecules. Other TFs are inactive and become activated only after translocation into the nucleus e.g., nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB).

NF-κB discovery and structure

NF-κB is a TF which occurs in all cell types and tissues [2]. NF-κB was discovered at the Salk Institute by Ranjan and Harinder Singh in David Baltimore’s lab in 1986 [3]. The hidden form of NF-κB within the cytoplasm of unstimulated cells was discovered by Patrick Baeuerle in 1988 [4] who also reported the purification of an inhibitor [5] as previously reviewed [6, 7].

The NF-κB protein superfamily family [8] has a DNA-binding/dimerization termed the Rel homology domain (RHD) [9] and consists of NF-κB1 protein 50 (p50) and its progenitor protein 105 (p105), NF-κB2, protein 52 (p52) and its progenitor protein 100 (p100)), transcription factor p65 encoded by RELA gene (RelA, protein 65, p65), transcription factor encoded by the RELB gene (RelB), and proto-oncogene, c-Rel, encoded by REL gene (cRel, Rel) and Drosophila Dorsal and Dif. NF-κB inhibiting proteins are IκB proteins (nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor protein) such as IκBα, IκBβ, IκBγ, IκBε, and Drosophila Cactus.

The NF-κB-IκB complex is located in the cytoplasm in its inactive latent form. The IκB kinase complex (IKK) with its regulating subunits nuclear factor kappa-B kinase 1 (IKK1, inhibitor of nuclear factor kappa-B kinase subunit alpha, IKK-α), inhibitor of nuclear factor kappa-B kinase 2 (IKK2, inhibitor of nuclear factor kappa-B kinase subunit beta, IKK-β) and the framework protein NF-κB essential modulator (NEMO, inhibitor of nuclear factor kappa-B kinase subunit gamma, IKK-γ) phosphorylates and degrades IκB resulting in NF-κB dimers with their translocation into the nucleus. The major dimer in cells is the specific p50-RelA heterodimer.

NF-κB controls many genes involved in inflammation and in cancer and directly influences cell proliferation, cell survival, and can decrease apoptosis via tumor necrosis factor alpha (TNFα) receptor-associated factor 1 and 2 (TRAF1, TRAF2).

NF-κB inhibiting IκB proteins

NF-κB is present in the cytoplasm as “a heterotrimer consisting of p50, p65, and inhibitory subunit of NF-κB (IκB) α subunits” (reviewed in [10]). NF-κB builds a complex with inhibitory IκB proteins such as IκBα, IκBβ, IκBɛ, IκBζ, p100, p105, B-cell lymphoma 3 (Bcl-3), or the Toll-like receptor (TLR)- inducible nuclear IκB protein IκBNS, and this complex is maintained within the cytoplasm in its inactive form [11, 12]. Activating signaling pathways promote the degradation of IκB, mediated by the IKK complex consisting of the kinases IKKα (IKK1) and IKKβ (IKK2) and a regulatory scaffolding protein, NEMO (IKKγ). In this manner, IκB is phosphorylated resulting in its degradation with translocation of NF-κB dimers into the nucleus to affect the target gene expression. After phosphorylation of the subunit of IκBα and its degradation on the p50–p65 heterodimer, phosphorylation of the p65 molecule occurs with binding to a specific DNA-sequence, resulting in gene transcription. IκBn can regulate inflammation by inhibiting the induction of TLRs-dependent genes through modulation of NF-κB [13].

Canonical and non-canonical NF-κB signaling

Canonical pathways are “idealized or generalized pathways that represent common properties of a particular signaling module or pathway, and accordingly categorizes the genes in specific canonical pathways and networks” [14]. The canonical (Fig. 1) and non-canonical NF-κB (Fig. 3) pathways have been extensively reviewed [11, 12]. Both, canonical (classical) and non-canonical (alternative) pathways in IKK/NF-κB signaling influence whether a cell lives or dies [11].

thumbnail Figure 1

Canonical NF-κB signaling pathway. Simplified scheme of the canonical NF-κB pathway. Nomenclature: TNFα: tumor necrosis factor α; TNFR1: tumor necrosis factor receptor 1; IL-1: interleukin 1; ILR1: interleukin 1 receptor; LPS: lipopolysacharide; TLR4: toll-like receptor 4; IKK complex: IκB kinase enzyme complex to upregulate the NF-κB signaling; IκBα: IκB kinase 1 (IKK1); IκBβ: IκB kinase 2 (IKK2); IκBγ: IκB kinase gamma; NEMO: NF-kappa-B essential modulator, regulatory scaffolding protein; P: phosphorylated; Akt: protein kinase B (PBK); p65: NF-κB3 (RelA); c-Rel: proto-oncogene NF-κB transcription factor; p50: NF-κB1 (progenitor is p105). Canonical NF-κB-IKK-complex is the inactive cytoplasmic form, consisting out of IκBα, p65 and p50. The canonical NF-κB heterodimer is the active form entering into the nucleus consisting out of p65 and p50

thumbnail Figure 2

Atypical (p105) NF-κB signaling pathway. Simplified scheme of the atypical (p105) NF-κB signaling pathway. Nomenclature: TNFα: tumor necrosis factor α; TNFR1: tumor necrosis factor receptor 1; IL-1: interleukin 1; ILR1: interleukin 1 receptor; LPS: lipopolysacharide; TLR4: toll-like receptor 4; MAPK3: mitogen-activated protein kinase 3 (ERK1); IKK complex: IκB kinase enzyme complex to upregulate the NF-κB signaling; IκBα: IκB kinase 1 (IKK1); IκBβ: IκB kinase 2 (IKK2); IκBγ: IκB kinase gamma, NEMO: NF-kappa-B essential modulator, regulatory scaffolding protein; P: phosphorylated; p105: p50 (NF-κB1) progenitor. Atypical (p105) NF-κB-complex is the inactive cytoplasmic form, consisting out of p105 and two p50. The NF-κB (p50-p50) homodimer is the active form entering into the nucleus; it can act as a transcriptional repressor and as an activator together with B-cell lymphoma 3-encoded protein (BCL3).

thumbnail Figure 3

Non-canonical NF-κB signaling pathway. Simplified scheme of the non-canonica NF-κB signaling pathway. Nomenclature: RANKL: receptor activator of nuclear factor kappa-Β ligand; RANK: receptor activator of nuclear factor kappa-Β; TNFβ: tumor necrosis factor-beta, lymphotoxin; LTβR: lymphotoxin β-receptor; CD40L: cluster of differentiation 40 ligand, CD154, protein expressed on T-cells; CD40: cluster of differentiation 40, costimulatory protein on T-cells; BAFF: B-cell activating factor, tumor necrosis factor ligand superfamily member 13B; BAFFR: B-cell activating factor receptor; NIK: NF-κB inducing kinase; IKK complex: IκB kinase enzyme complex to upregulate the NF-κB signaling; IκBα: IκB kinase 1 (IKK1); IκBβ: IκB kinase 2 (IKK2); IκBγ: IκB kinase gamma; NEMO: NF-kappa-B essential modulator, regulatory scaffolding protein; P: phosphorylated; p100: NF-κB2 (p52) precursor protein; p52: NF-κB2; RelB: transcription factor RelB. Non-canonical NF-κB-IKK-complex is the inactive cytoplasmic form, consisting out of IκBα, p52 and RelB. The non-canonical NF-κB heterodimer is the active form entering into the nucleus consisting out of p52 and RelB.

In the canonical pathway, various cytokines such as TNFα, interleukin 1 beta (IL-β1), and viruses or TLRs are involved in inflammatory and immune-mediated responses. These include increases in TNFα, IL-β1, interleukin 6 (IL-6), granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin 8 (IL-8, chemokine (C-X-C motif) ligand 8, CXCL8), macrophage inflammatory protein 1 alpha (MIP1α, chemokine (C–C motif) ligand 3, CCL3), vascular cell adhesion molecule 1 (VCAM-1, cluster of differentiation 106, CD106), intercellular adhesion molecule 1 (ICAM-1, cluster of differentiation 54, CD54), E-selectin (endothelial-leukocyte adhesion molecule 1, ELAM-1, CD62 antigen-like family member E, CD62E, leukocyte-endothelial cell adhesion molecule 2, LECAM2), or nitric oxidase synthase (iNOS), cyclooxygenase 2 (Cox-2) and phospholipase A2 (PLA2).

The non-canonical NF-κB signaling is dependent on IKKα homodimers. It is activated by lymphotoxin beta receptor (LTβR), B cell–activating factor belonging to the TNF family (BAFF), cluster of differentiation 40 (CD40) ligand (CD40L), and cluster of differentiation 154 (CD154)-induced expression of interferon regulatory factor 3 (Irf3) or retinoid X receptor alpha (Rxra, nuclear receptor subfamily 2, group B, member 1, NR2B1) [11]. LTβR can induce apoptosis in both the canonical and non-canonical pathways [15]. Knocking down Irf3 or the stimulator of interferon genes (STING) results in reduced inflammation and apoptosis modulated by NF-κB [16], and Irf3 inhibition results in decreases of the transforming growth factor beta 1 (TGF-β1)-induced proliferation of hepatic stellate cells (HSC) [17].

Recently, a novel mechanism of NF-κB activation B-cell receptor (BCR) was reported which could be relevant in B-lymphoproliferative disorders: NF-κB p50/p65 was rapidly activated (within 30 s) by anti-IgM stimulation of BCR through a Bruton’s tyrosine kinase (Btk)-dependent and IKK-independent mechanism [18]. Btk expression is increased and required for EGFR-induced NF-κB activation with poor prognosis in glioma [19]. Furthermore, Btk membrane translocation is observed in multiple meloma [20] as it regulates Toll-like receptor 7 and 8 (TLR7/8) induced TNF transcription through NF-κB [21]. Inhibiting Btk by the small molecule and inhibitor of tubulin polymerization, KS99, results in the inhibition of tumor growth in multiple myeloma and osteoclastogenesis in vivo [22].

NF-κB polymorphism

Blood samples from 565 healthy volunteers in a Turkish cohort were tested to determine the frequency of polymorphisms. Polymerase chain reaction (PCR) amplification was performed followed by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). This revealed that NF-κB1-94ins/delATTG and NF-κBIA 3′ UTR polymorphisms were, in general, quite similar to other populations in Germany, Sweden, Czechoslovakia, and Australia [23]. Wang et al. tested 564 gastric cancer patients and 566 healthy controls to see if the polymorphism rs2233408 T/C genotype in the promoter region of IκBα was associated with increased risk for gastric cancer [24], and found that “IκBα rs2233408 T heterozygotes were associated with reduced gastric cancer risk”. Stable expressions of Runt-related transcription factor 3 (RUNX3)/protein 33 (p33) were associated with a 1.9-fold elevation in NF-κB transcriptional activity [25]. In 1010 gastric cancer patients compared to 1500 healthy controls in Guangdong, China, IκBα rs17103265 deletion homozygote was identified as a novel risk factor for gastric cancer [26]. This was followed by the association of homozygous rs4648068 GG with an increased risk of gastric cancer in the Han Chinese population [27]. Lo et al. correlated polymorphisms of NFΚB1 promoter with susceptibility to gastric cancer in aged patients [28].

Ubiquitination and degradation

The process of adding ubiquitin known as “ubiquitination” is used to mark proteins or protein complexes for degradation through covalent binding by monoubuiquitin or polyubiquitin chains with different enzymes such as ubiquitin-activating enzyme (E1), ubiquitin-conjugating enzyme (E2), and ubiquitin ligase (E3). Adding a monoubiquitin is a regulatory modification compared to adding polychains by a polyubquitin which is involved in distinct cellular functions, e.g., signaling a molecule for degradation by the proteasome [2935].

Because of their discovery of protein-regulating systems, the 2004 Nobel Prize for chemistry was awarded to Aaron Ciechanover, Avram Hershko, and Irwin Rose [3639].

Ubiquitin-mediated proteolysis of IκBs is irreversible, and ubiquitination and degradation of the inhibitors of NF-κB is an important component of transcriptional activation (reviewed in [40]).

NF-κB and cancer

NF-κB association with precancerous lesions and cancer

Increased constitutive NF-κB activity has been reported in precancerous lesions of the skin [41, 42], breast [43], head and neck [44, 45], Barrett’s esophagus [46], colon adenoma [47], chronic pancreatitis [48], colitis [49, 50], in premetastatic lung [51] and in the stroma of precancerous lesions of colon adenoma [52].

Furthermore, NF-κB has been reported in a number of adenocarcinoma cell lines [53] and NF-κB activity was associated with cancers of the breast [5460], ovaries [61, 62], endometrium [63], prostate [6466], thyroid [6770], pancreas [7174], squamous cell carcinoma (SCC) [75, 76], esophagus [7779], stomach [80], colorectum [61, 8183], liver [8486], kidneys [8789], bladder [90], lymphoma [9195], leukemia [9698], multiple myeloma [99102], brain [103105], melanoma [106109], and sarcoma [110112].

However, NF‐κB signaling in carcinogenesis is complex and depends on which subunits are involved: NF‐κB2/p52 seems to be required for colitis-associated adenoma while c‐Rel‐induced signaling is involved in colonic epithelial cell turnover [113]. Furthermore, there is a difference between acute versus chronic inflammation. NF‐κB can have anti-inflammatory effects in an acute, chemically induced colitis model with IL-β1 suppression and IKKβ inhibitors could potentially serve as a therapeutic option in such cases [49], but IKKβ is needed for healing after colitis [114] and “NF-κB2/p52 is necessary for the development of colitis, whilst c-Rel-mediated signalling regulates colonic epithelial cell turnover” [113].

In vitro NF-κB cancer model

For about 25 years, increased NF-κB activity has been recognized to be associated with cancer development in transgenic mice [115]. For example, the chemotherapeutic efficacy of cisplatin can be enhanced by inhibiting NF-κB in vitro and in vivo [116]. In 2014, an in vitro model of NF-κB driven carcinogenesis was published [117]. The authors used a cell-based phenotypic readout and isolated 12 genetic elements that induced NF-κB activity (NF-κB–activating genetic elements, NASPs) of lentiviral libraries encoding 20 or 50 amino acid-long polypeptides “none of which was previously associated with NF-κB activation, were isolated from libraries of 200 000 peptides derived from 500 human extracellular proteins”. By selective knockdown experiments, it was shown that isolated NASPs “act either via or upstream of TNF receptor-associated factor 6” (TRAF6). Growth in mice or rat embryo fibroblasts was unaffected after NASP transduction but co-expression with Ras (protein superfamily of small guanosine triphosphate hydrolase enzymes (GTPases) by GTP hydrolase enzyme, transforming protein p21 (H-ras, H-RasV12) resulted in cell transformation. Constitutive activation of NF-κB attenuated p53 and promoted carcinogenesis. In contrast, activated K-Ras, but not H-Ras or N-RRas was assumed to imitate tumors of endodermal origin via stem cell expansion [118] though this could be related to the model used. Buchanan et al. showed that phospholipase D1 (PLD1) activity in H-RasV12 is required for transformation [119].

NF-κB and cancer aggressiveness

Tumor aggressiveness in gastric cancer was shown by investigating 90 human cancer tissues versus 50 nonmalignant specimens. A higher NF-κB expression in cancer tissue versus normal mucosa (31% vs. 4%, p < 0.0001) was found along with activation of metalloproteinase 9 (MMP-9), IL-β1, and IL-8 in AGS cells [120]. Another example of NF-κB signaling and chronic inflammation was provided by Kwon et al. [121] who demonstrated that the Vitamin D(3) upregulated protein 1 (VDUP1) with its tumor suppressive effect was shown in VDUP1 knockout (KO) revealing that VDUP1 negatively regulates Helicobacter pylori (H. pylori)-associated gastric cancer by inhibiting the induction of TNFα, NF-κB, and Cox-2, and by disrupting cell growth.

Combining non-steroidal anti-inflammatory drugs (NSAIDs) with NF-κB inhibitors increased apoptosis in different ovarian cancer cell lines SKOV-3, CAOV-3, SW626 and 36M2 [122].

NF-κB signaling and crosstalk in carcinogenesis and pathogenic stimulus

The importance of NF-κB signaling in carcinogenesis was emphasized in the proposed new paradigm for the origin of the majority of cancers [123, 124].

Viruses

Increased NF-κB activity has been associated with pathogenic stimuli such as Epstein-Barr virus (EBV) [125], Hepatitis B virus (HBV) [126], and Hepatitis C Virus (HCV) [127].

Hepatitis B viral protein (HBx) is a small transcriptional transactivator essential for infectivity and which activates NF-κB signaling in the cytoplasm [128] via deactivation of two NF-κB inhibitors [129], phosphorylation of IκBa inhibitor and NF-κB1 (p50) precursor inhibitor protein p105 (Fig. 2) with reduction of IκBa stability, and decreased NF-κB1 (p50), resulting in the accumulation of NF-κB within the nucleus.

HBV and HCV trigger chronic inflammation with NF-ΚB activation, which in turn, is associated with hepatocellular carcinoma (HCC) [130132]. Despite HBV suppression, HCC development is still observed in patients with residual hepatitis B surface antigen (HBsAg) titers ([133, 134] reviewed in [135]). Patients with spontaneous HBV DNA clearance and residual high HBsAg titers also show increased HCC risk ([136] reviewed in [135]).

HBV induces an unfolded protein response (UPR), NF-κB activation [136], signal transducer and activator of transcription 3 (STAT3) [137], and IL-8 [138]. HBsAg, together with inhibition of NF-κB, decreases UPR, binding immunoglobulin protein (BiP, 78 kDa glucose-regulated protein, GRP-78, heat shock 70 kDa protein 5, HSPA5), Cyclin E, cyclin-dependent kinase 2 (cdk2), and increases cytosine-cytosine-adenosine-adenosine-thymidine (CCAAT)/enhancer binding protein homologous protein (CHOP), activating transcription factor 3 (AFT3), Cyclin D1 which results in a 100% incidence of HCC [135].

Human papilloma virus (HPV) infection with HPV Type 16 (HPV16) appears to be responsible for some 18% of oral cancers [139] and has been shown to modulate NF-κB signaling [140]. This is associated with the p65 NF-κB complex formation and consequent heterodimerization of p50/p65 which is thought to be one reason why disease outcome is better in patients with HPV-positive oral cancers than in patients with HPV-negative oral cancers [141]. HPV16 E6 protein activates NF-κB signaling via p50, NF-kappa-B-inducing kinase (NIK), and TRAF-interacting protein [142] while human fibroblasts expressing E7 protein decreases pro-caspase 8 activation, and can partially protect from apoptosis by activating the TNF receptor 1-related cytokine receptor, first apoptosis signal receptor (Fas, FasR, apoptosis antigen 1, APT, cluster of differentiation 95, CD95) [143].

HPV-negative tumors are associated with STAT3/pSTAT3 together with NF-κB “irrespective of the presence or absence of activator protein 1 (AP1)” while “AP1 and NF-κB lacking involvement of STAT3” are associated in HPV-positive tumors [144].

Inhibition of NF-κB and AP1 transcription factors by the T-cell–specific inhibitor, 2-chloro-4-(trifluoromethyl)-pyrimidine-5-N-(39,59-bis[trifluoromethyl]phenyl)-carboxamide (SP100030), suppressed fibrosis in the lung by decreasing coagulation, and decreasing collagen deposition in the lung [145].

Another important example is the first apoptosis signal receptor (Fas, FasR, apoptosis antigene, APO-1, APT, cluster of differentiation 95 CD95), TNF receptor superfamily member 6 (TNFRSF6). Fas is a cell surface receptor whose activation results in apoptosis [146]. NF-κB can directly regulate Fas-mediated apoptosis by acting as a Fas transcriptor activator in human colon carcinoma cells and in mouse embryonic cells (MEFs). Thus, an anti-NF-κB therapy might suppress Fas-mediated apoptosis and impair natural immune anti-cancer cell suppression [147] or act via the regulation of caspase-4 [148].

Stromal fibroblasts in HPV infection acts via IL6/CCAAT/enhancer-binding protein β (C/EBPβ) signaling to recruit T helper 17 cells (Th17) with consequent chronic inflammation during carcinogenesis [149]. C/EBPβ signaling together with adenovirus early region 1A (E1A) binding protein (p300, EP300) induces IL-8/CXCL8 expression ín lung cancer [150] and promotes NF-κB triggered invasion and cell migration in renal cancer [151]. Metastasis by migration via C/EBPβ also occurs by TNFα-induced matrix metalloproteinase 1 (MMP-1)/matrix metalloproteinase 3 (MMP-3) expressions in a p38 MAPK-dependent manner [152]. Interestingly, knockdown of C/EBPβ inhibits p65-NF-κB signaling resulting in protection against fibrosis in cardiac myocytes [153] and in adipocyte-mediated chronic inflammation via mitogen-activated protein kinase kinase (MEK, MAPK2, MAPKK), C/EBPβ with NF-κB/RelA inducing 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) [154].

Human papilloma virus type 8 (HPV8) induces matrix metalloproteinase 14 (MMP-14, MT1-MMP) in the stroma and cells of cutaneous tumors [155], but the exact role of HPV-induced fibrosis in HPV-triggered cancers remains to be elucidated. Human ovarian carcinoma cell line, SK-OV-3, transfected with adenovirus early region E1A protein revealed increased sensitivity to radiation-induced cell death for which NF-κB modulation and/or inactivation is required [156].

NF-κB signaling is associated with G2/mitotic-specific cyclin-B1 (Cyclin B1), Cyclin D1, human inhibitor of apoptosis protein (HIAP), BAG family molecular chaperone regulator 1 (BAG-1), transcription termination factor (TTF), and fibronectin in radioresistance of p53-inactive human keratinocytes [157].

Bacteria

Enteroinvasive bacteria induce chronic inflammation [158160] through the activation of cytokines such as IL-8, iNOS, and Cox-2 [161]. Enteroinvasive bacteria can also suppress NF-κB activation and expression of IL-6 and IL-8 [162, 163].

NF-κB activation via DNA and RNA from bacteria and viruses likely occurs through the promotion of inflammatory cytokines such as TNF, IL-6, inactive IL-β1 precursor (pro-IL-1 β), intracellular NOD-like receptor pyrin domain containing 3 (NIrp3), IFN, TLRs [164167], lipopolysaccharide (LPS) [168] mediated by cluster of differentiation 14 (CD14) [169].

Helicobacter pylori was shown to be actively involved in stimulating IL-8 in gastric cancer [170]. Increased NF-κB binding activity is observed with elevated IL-8 levels from the more virulent H. pylori strains [171, 172]. In a series of 289 biopsies, an association between increased NF-κB activity was reported in H. pylori virulence factor (cagA) positive metaplasia, dysplasia, and in gastric carcinoma [173]. H. pylori infected human gastric epithelial cells induce matrix metalloproteinase 7 (MMP-7) through activation of the Ras homolog gene family (Rho) and the subfamily of the Rho family of GTPases (Rac). Here Ras homolog gene family member A (RhoA) activates NF-κB and AP1, while Rac activates NF-κB, but not AP1 [174] so the effects are selective.

The H. pylori virulence factor CagA promotes chronic inflammation and proliferation through phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) signaling with consequent activation of beta-catenin and NF-κB [175]. The Helicobacter species, H. bilis, in the human bile duct cancer cell line, HuCCT-1, activated NF-κB activity independent of the tumor stage. This was associated with production of vascular endothelial growth factor (VEGF) [176] and was later shown in extrahepatic cholangiocellular carcinoma (CCC) specimens to occur especially within the common bile duct [177].

Mycoplasm

Mycoplasma infection was shown to induce NF-κB activation, p53 suppression, and promotion of Ras-induced cell transformation [178, 179]. Infecting the gastric mucosa of immunodeficient mice with the prokaryotic intracellular parasite, Mycoplasma sp., induced chronic inflammation and facilitated malignant cell transformation with increased expression of p53, p21, B-cell lymphoma 2 (Bcl-2)-associated X protein (Bax), transforming protein p21 (GTPase HRas, H-ras), B-cell lymphoma 2 (Bcl-2), transcription factor p65 (p65, nuclear factor NF-kappa-B p65 subunit) and TNFα together with upregulated NF-κB signaling, which by itself elicits resistance to apoptosis [180].

Mycoplasma hyorhinis (M. hyorhinis) expresses the M. hyorhinis membrane protein p37 and binds to gastric cancer cells, MGC803 and AGS, in a time- and dose- dependent manner as shown in the cell ELISA, which is p37-dependent mediating the p37/annexin II (ANXA2) interaction and important for the host-receptor mediating M. hyorhinis infection.

EGFR increases ANXA2 phosphorylation and facilitates M. hyorhinis infection. This increases expression of NF-κB target genes IκBα, Cox-2, MMP-1, PR domain zinc finger protein 1 (PRDM1), suppressor of cytokine signaling 2 (SOCS2), dual specificity mitogen-activated protein kinase kinase 1 (MAP2K1) contributing to cell invasiveness [181]. Furthermore, the direct induction of the LTβR and non-canonical NF-κB signaling has recently been demonstrated [182].

Tsai et al. found unusually high frequencies of Mycoplasma fermentans (M. fermentans) and Mycoplasma penetrans (M. penetrans) in acquired immune deficiency syndrome (AIDS) patients that could not be associated with immediate (acute) transformation of cells into cancer cells [183]. Investigating the mouse fibroblast cell line, C3H cells, a persistent infection with M. fermentans resulted in cell phenotypes with malignant characteristics, which were more pronounced with prolonged infection. Furthermore, the authors reported that this phenomenon was dependent on the number of passages: mycoplasma needed one week per passage, at least six passages were necessary and until the 11th passage the malignant transformation was reversible if anti-mycoplasma therapy was implemented. In contrast, the malignant state was irreversible if mycoplasma infection persisted until the 18th passage. Importantly, no chromosomal loss or translocations were observed during the reversibility window. In regards to our knowledge today, the authors drew the incorrect conclusions when they assumed that genetic instability was most likely caused by somatic mutations. The important take away from this investigation was that despite the long latency, a mycoplasma infection triggered development of malignant cells, that there was a reversible stage which could be eradicated, and which speaks against hypothesizing the need for mutations or other genetic changes at least within a window of time. Nonetheless, after another long period of persistent infection, signs of genetic instability were reported.

This also explains why locally advanced cancers reveal mutations or other genetic changes and why recurrent cancers show chromosomal aberrations [184] and even why reports of mycoplasma-infected cancer cells were correctly observed to show chromosomal aberrations [185, 186] but incorrectly interpreted in assigning genetic changes as being causal to the cancer.

Tsai et al. showed almost 50 years ago that latency, as well as multiple cell passages, were mandatory events in carcinogenesis [183].

In 194 Nigerian women tested for high-risk human papillomavirus (hrHPV), a significant association with Mycoplasma hominis (M. hominis) was observed with an OR of 8.78 (95% Confidence Interval, 1.49–51.6, P = 0.01) and human immunodeficiency virus (HIV) positive females had a three-fold increase in OR in the presence of persistent hrHPV infection [187].

As multiple biological stimuli may be involved during carcinogenesis, it seems that we are at the beginning of creating a focus in cancer research by exploring the role played by physiological and chemical stimuli [123, 124, 188191].

Schistosomiasis

Schistosomula are the parasitic stage of Schistosomiasis residing in the lungs and retained in capillaries responsible for initiating inflammatory response and leukocyte recruitment [192]. Endothelial cells (ECs) are activated by Schistosoma mansoni (S. mansoni) with schistosomula gaining an anti-inflammatory phenotype with reduction of VCAM-1 and E-selectin in TNFα stimulated ECs mediated by cyclic 5′ adenosine monophosphate (cAMP)/protein kinase A (PKA) signaling [193] which, in turn, is affected in reducing NF-κB transcriptional activity (and not by blocking NF-κB) [194] through PKA-induced cAMP response element-binding protein (CREB) phosphorylation. This results in the removal of the transcriptional co-activator CREB binding protein (CBP) from the NF-κB complex ([195] reviewed in [194]).

RNAse resistant double-stranded RNAs (dsRNAs) from Schistosomia mansoni activates toll like receptor 3 (TLR3) with consequent phsophorylation of signaling transducer activator of transcription 1 (STAT1) together with interferon (IFN) stimulated gene expression [196].

The association of Schistosomiasis-induced cancers and fibrosis has long been reported in liver cancers such as HCC and CCC [197, 198] but under-appreciated in the literature. Anti-Schistosomiasis therapy improves and ameliorates fibrosis via NF-κB signaling [199, 200]. The application of Boswellic acid (BA)-containing extracts attenuated Schistosoma japonicum (S. japonicum)-induced fibrosis via decreases in NF-κB signaling and subsequent decreases in VEGF, TNFα, and monocyte chemoattractant protein 1 (MCP-1, chemokine (C–C motif) ligand 2, CCL2) [201]. Other anti-helminthic drugs, such as niclosamide, target NF-κB, Wnt/β-catenin, Notch, reactive oxygen species (ROS), mammalian target of rapamycin complex 1 (mTORc1), and STAT3 [202]. Paeoniflorin (PAE) decreases interleukin 13 (IL-13), NF-κB, TGF-β1 and alpha-smooth muscle actin (α-SMA, alpha-actin 2) expression resulting in a decrease of fibrosis [199].

Although increases of lysyl oxidase (LOX) were shown in murine Schistosomiasis some 25 years ago [203], an anti-LOX approach was not investigated. LOX is expressed in Escherichia coli (E. coli) [204]. Lysyl oxidase homolog 2 (LOXL2) [205] was found in human lung fibrosis, bronchiolo-alveolar carcinomas, and in situ ductal breast tumors [206]. In S. mansoni-infected mice and early stages of liver granuloma, the LOX gene and LOX-like gene (LOXL) were upregulated. LOXL2 promotes zinc finger protein SNAI1 (Snail) and decreases E-cadherin, both important for metastasis. Investigations of the extracellular matrix (ECM) by atomic force microscopy showed that LOXL2 does not affect ECM properties [207]. Incubating oral epithelial cells with the natural di-thiol α-Lipoic acid was shown “to modulate periodontal bacterial induced NF-κB activation, pro-inflammatory gene expression and cytokine production” [208].

Barrett’s, reflux and bile acid

The development of Barrett’s intestinal metaplasia is multifactorial in etiology [209]. The incidence rates vary between 0.19 and 0.33% ([210, 211] reviewed in [212]) while in 1997 the prevalence was reported as being 19.8% in patients with heartburn [213]. Although it was previously assumed that Barrett’s originates from intestinal metaplasia, Takubo et al. showed that small lesions of Barrett’s are surrounded by non-intestinalised epithelium and not by intestinal metaplasia [214], a finding that was subsequently reproduced [215, 216]. On the one hand, intestinal metaplasia shows higher rates of molecular aberrations compared to non-intestinal metaplasia [217] while on the other, even The Cancer Genome Atlas (TCGA) which consists of approximately 10 000 specimens representing 33 types of cancer [218] and the most recent Mutational Assessment of iClusters [219] failed to provide evidence of one unique genetic signature for the majority of Barrett’s cancers. Despite this, the contributors to Barrett’s are assumed to be gastro-esophageal reflux disease (GERD), hiatal hernia, and alcohol [212], all of which have just one condition in common: chronic inflammation.

Babar et al. investigated Barrett’s patients who underwent anti-reflux surgery versus proton pump inhibitor (PPI) treatment and investigated endoscopic biopsy specimen from 2 cm below the squamo-columnar junction for NF-κB, cytokines, and growth factors [220]. “Mean activated NF-kappaB p50 and p65 subunits, interleukin (IL)-1alpha, IL-1beta, and interleukin-8 levels, were significantly (P < 0.05) lower in the surgically treated group” and led the authors to conclude that anti-reflux surgery may create a less inflammatory environment.

Bile acids activate farnesoid X receptor (FXR)/nerve growth factor (NGF)/transient receptor potential vanilloid 1 (TRPV1) axis through mucosal mast cell-to-nociceptor signaling inducing visceral hypersensitivity in a rat model investigating diarrhea-predominant irritable bowel syndrome (IBS-D) [221].

In obesity, FXR additionally promotes inflammation in diet-induced obese mice through a dysregulation of homeostasis of pro- and antiflammatory signaling [222]. Otherwise, FXR activation occurred in high-fat diet (HFD) fed mice with increased proinflammatory leukotrienes B4 (LTB4) and lower (∼3-fold) anti-inflammatory epoxyeicosatrienoic acids (EETs) [223]. FXR induced inflammation can be suppressed by the novel FXR-agonist, dioscin, with consequent suppression of NF-κB and High mobility group box 1 protein (HMGB-1), and subsequently decreased the mRNA levels of IL-β1, IL-6, and TNFα [224].

NGF attenuates apoptosis and inflammation in diabetic keratinopathy decreasing ROS activation and levels of cleaved caspase-3, BAX, IL-β1, and TNFα [225]. These contradictory findings are explained as the NGF effect is dose-dependent in that it appears to be dependent on how strongly the homeostasis is disrupted [226, 227] as it was shown, that NGF stimulates T-lymphocyte-dependent basophilic cell differentiation in vivo at sites of allergic tissue inflammation [228] as NGF is also often a secondary upregulated endogenous factor [229].

The induction of NF-κB through NGF works through the p75 receptor with an anti-apoptotic effect in Schwannoma cells [230] contributing to survival of NGF-dependent sympathetic neurons [231]. Here, NGF is contrary to TNFα, as the Bcl-x survival gene expression requires tyrosine phosphorylation of IkappaBalpha [232] explaining the disruption of homeostasis of the NGF-TNFα axis is of importance. TNFα regulates the response to NGF in neuroblastoma cells [233].

Upregulation of TRPV1 is mediated by the p38 mitogen-activated protein kinase (MAPK) and NF-κB signaling pathways by urban particulate matter (UPM) resulting into chronic inflammation [234]. Local acidodic microenvironment incudes TRPV1 activation with consequent NF-κB activation with observed lymphangionesis in lymphatic endothelial cells (LECs) [235].

Bile acid is associated with homeobox protein CDX-2 (Cdx2) expression, a transcription factor for the intestine-specific tumor suppressor guanyl cyclase (GC-C), and mediated by NF-κB. The increase in Cdx2 by deoxycholate is associated with NF-κB nuclear translocation. This was interpreted as bile acid-induced intestinal metaplasia involving Cdx2 and NF-κB [236]. Deoxycholic acid shows a non-linear dose response relationship for DNA damage only above doses of 100 mM and even higher in esophageal OE33 cells with a similar dose-response association to NF-κB activation but it was not possible to replicate the applied deoxycholic acid doses in the gene expression part of the study [237]. However, the close association of IL-8 and IκB gene expression levels, together with NF-κB activity was shown earlier (Figure 2 in [237] – not shown here).

The bile acid-induced chronic inflammation involves the suppression of EGFR and Akt, resulting in activation of the NF-κB- and the Cdx2 axis [238]. Human telomerase reverse transcriptase (hTERT) which upregulates Cdx2 through NF-κB signaling promoting intestinal metaplasia [239]. Wang et al. found increased activity of NF-κB and hTERT when comparing normal gastric mucosa with dysplasia, intestinal metaplasia, and gastric cancer [240].

Morbid obesity

The association of morbid obesity and the role played by its different signaling pathways during carcinogenesis have recently been published [191] and the disruption of NF-κB signaling could present a therapeutic approach in the future [241].

The gastric peptide, Ghrelin, associated with food intake and energy balance was shown to be able to induce cell migration via its receptor growth hormone secretagogue receptor (GHS-R), Ca2+/calmodulin-dependent protein kinase II (CaMKII), 5′ adenosine monophosphate-activated protein kinase (AMP)-activated protein kinase (AMPK), and the NF-κB signaling pathway in rat C6 and human U251 glioma cells [242].

NF-κB signaling and crosstalk in carcinogenesis and chronic inflammation

Chronic inflammation is extensively reviewed within this Special Issue (Fig. 4) [189, 190]. In summary, the inclusion of chronic inflammation within a multistep sequence to explain carcinogenesis revealed that continuously induced inflammatory cells such as monocytes, lymphocytes, plasma cells, fibroblasts, and mast cells (MCs), together with cell-communication and the crosstalk of two major components, TGF-β and LOX play important roles in the overall process [123, 124].

thumbnail Figure 4

NF-κB signaling and crosstalk during carcinogenesis – Special Issue: Disruption of homeostasis-induced signaling and crosstalk in the carcinogenesis paradigm “Epistemology of the origin of cancer”. Simplified scheme of NF-κB signaling and crosstalk during carcinogenesis in the Disruption of signaling homeostasis induced crosstalk in the carcinogenesis paradigm “Epistemology of the origin of cancer” consisting of a six-step sequence: (1) a pathogenic stimulus followed by (2) chronic inflammation from which develops (3) fibrosis with associated remodeling of the cellular microenvironment; and from these changes a (4) precancerous niche (PCN), a product of fibrosis, with remodeling by persistent inflammation, develops which triggers the deployment of (5) a chronic stress escape strategy and when this fails resolve it by (6) normal cell to cancerous cell transition (NCCCT) by PCN-induced cell matrix stress occurs. This figure was published in paper 2 of this Special Issue [189] and modified in accordance to NF-κB signaling and crosstalk. Nomenclature: Common abbreviations are bold, followed by the common trivial names (if available) and (if available) by the name in accordance to the International Union of Pure and Applied Chemistry (IUPAC): PCN: precancerous niche; CSES: chronic stress escape strategy; NCCCT: normal cell to cancerous cell transition; SphK: sphingosine kinase isoform; S1P: sphingosine-1-phosphate; IL-6: interleukin 6; IL-8: interleukin 8; TNFα: tumor necrosis factor alpha; IFNγ: interferon gamma; ALOX: lipoxygenase, arachidonate lipoxygenase; ALOX12: 12-lipoxygenase, 12-LOX, 12S-LOX, arachidonate 12-lipoxygenase 12S type; ALOX5: 5- lipoxygenase, 5-LOX, arachidonate 5-lipoxygenase; 12-HETE: 12-hydroxyeicosatetraenoic acid; LTA4: leukotriene A4, 4-[(2S,3S)-3-[(1E,3E,5Z,8Z)-tetradeca-1,3,5,8-tetraenyl]oxiran-2-yl]butanoic acid; LTB4: leukotriene B4, (5S,6Z,8E,10E,12R,14Z)-5,12-dihydroxyicosa-6,8,10,14-tetraenoic acid; LTC4: leukotriene C4, (5S,6R,7E,9E,11Z,14Z)-6-[(2R)-2-[[(4S)-4-amino-4-carboxybutanoyl]amino]-3-(carboxymethylamino)-3-oxopropyl]sulfanyl-5-hydroxyicosa-7,9,11,14-tetraenoic acid; LTD4: leukotriene D4, (5S,6R,7E,9E,11Z,14Z)-6-[(2R)-2-amino-3-(carboxymethylamino)-3-oxopropyl]sulfanyl-5-hydroxyicosa-7,9,11,14-tetraenoic acid; LTE4: leukotriene E4, (5S,6R,7E,9E,11Z,14Z)-6-[(2R)-2-amino-2-carboxyethyl]sulfanyl-5-hydroxyicosa-7,9,11,14-tetraenoic acid; 5-oxo-ETE: (6E,8Z,11Z,14Z)-5-oxoicosa-6,8,11,14-tetraenoic acid; Cox: cyclooxygenase; Cox-1: cyclooxygenase 1; Cox-2: cyclooxygenase 2; Cox-3: isoform of Cox-2 (therefore in brakes); PGG2: prostaglandin G2, (Z)-7-[(1S,4R,5R,6R)-5-[(E,3S)-3-hydroperoxyoct-1-enyl]-2,3-dioxabicyclo[2.2.1]heptan-6-yl]hept-5-enoic acid; PGH2: prostaglandin H2, (Z)-7-[(1S,4R,5R,6R)-5-[(E,3S)-3-hydroxyoct-1-enyl]-2,3-dioxabicyclo[2.2.1]heptan-6-yl]hept-5-enoic acid; PGFF2α: prostaglandine F2 alpha, (Z)-7-[(1R,2R,3R,5S)-3,5-dihydroxy-2-[(E,3S)-3-hydroxyoct-1-enyl]cyclopentyl]hept-5-enoic acid; PGD2: prostaglandin D2, (Z)-7-[(1R,2R,5S)-5-hydroxy-2-[(E,3S)-3-hydroxyoct-1-enyl]-3-oxocyclopentyl]hept-5-enoic acid; PGE2: prostaglandin E2, (Z)-7-[(1R,2R,3R)-3-hydroxy-2-[(E,3S)-3-hydroxyoct-1-enyl]-5-oxocyclopentyl]hept-5-enoic acid; MDA: malondialdehyde, propanedial; TXA2: thromboxane A2, (Z)-7-[(1S,2S,3R,5S)-3-[(E,3S)-3-hydroxyoct-1-enyl]-4,6-dioxabicyclo[3.1.1]heptan-2-yl]hept-5-enoic acid; CYP*: cytochrome P450 isoforms; 20-OH-PGE2: 20-hydroxy prostaglandin E2; 20-HETE: 20-hydroxyeicosatetraenoic acid, (5Z,8Z,11Z,14Z)-20-hydroxyicosa-5,8,11,14-tetraenoic acid; SOX: [sex-determining region Y (Sry) box-containing] transcription factor family; IL-β1: interleukin beta 1; IL-33: interleukin 33; ROS: reactive oxygen species; CXC CC: chemokine receptors; αSMAD: alpha-smooth muscle actin; miR21: micro RNA-21; p300: protein 300 (p300-CBP coactivator family); SP1: specificity protein 1; AP1: activator protein 1; E2F4/5: cytoplasmic complex of Smad3, retinoblastoma-like protein 1 (P107, RBL1), E2F4/5 and D-prostanoid (DP1); p107: retinoblastoma-like protein 1, RBL1; TGFβ: transforming growth factor beta; Pro-MMP-9: pro-matrix metalloproteinase 9; Pro-MMP-1: pro-matrix metalloproteinase 1; Pro-MMP-7: pro matrix metalloproteinase 7; SNAIL: zinc finger protein SNAI1; MMP-1: matrix metalloproteinase 1; MMP-7: matrix metalloproteinase 7; MMP-2: matrix metalloproteinase 2; E-Cadherin: CAM 120/80 or epithelial cadherin, cadherin-1, epithelial cadherin; CXCL1: chemokine (C-X-C motif) ligand 1; Osm: oncostatin-M; PI3K: phosphatidylinositide 3-kinase; FOXO3a: forkhead box protein O3a; p120: catenin delta-1, protein 120; Rho: Ras homolog gene family, member A; Rac1: Ras-related C3 botulinum toxin substrate 1; cdc42: cell division control protein 42 homolog; BIM: Bcl-2 interacting mediator of cell death; PUMA: BH3-only protein; CXCR4: C-X-C motif of chemokine receptor 4; cdk2: cyclin-dependent kinase 2; LOXL3: lysyl oxidase homolog 3; mTORc1: rapamycin complex 1; PAI1: Plasminogen activator inhibitor-1; NF-κB: nuclear factor kappa-light-chain-enhancer of activated B cells.

TGF-β1 activates Akt through PI3K followed by phosphorylation of glycogen synthase kinase-3β (GSK3β) promoting stability and activity of Snail. Furthermore, TGF-β1 activates the mechanistic targets of mTORc1 and mTORc2. Together with various inflammatory cytokines, such as TNFα, NF-κB is activated along with other transcriptional factors, such as hypoxia-inducible factor alpha (HIF1α) and STAT3, resulting in decreases of E-cadherin and apoptosis. Nuclear protein 120 (p120) accumulation stimulated by TGF-β and LOX promotes cell division control protein 42 homolog (cdc42) activating Ras-related C3 botulinum toxin substrate 1 (Rac1), further decreasing E-cadherin and increasing matrix metalloproteinases (MMPs), fibronectin, vimentin, and twist-related protein 1 (TWIST), zinc finger E-box-binding homeobox 1 (ZEB1), and ZEB2 [123].

The proinflammatory cytokine, interleukin 32 (IL-32), induces NF-κB. Gastric cancer patients were more often IL-32 positive than negative for IL-32 expression (p < 0.01) and the five-year survival rate of the IL-32 positive group was 56%, significantly higher than the IL-32 negative group (p < 0.01). Multivariate analysis showed that IL-32 expression was an independent prognostic marker for gastric cancer (p < 0.05) after lymph node stage and metastasis [243].

NF-κB is constitutively found within the nucleus in B-lymphocytes and dendritic cells and inactive NF-κB is usually found in the cytoplasm where it can be triggered by inhibitory proteins, such as nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha (IκBα). Activation can also occur by cytokines, bacteria, viruses, or chemical agents, which result in phosphorylation (pNF-κB). Activation of NF-κB was also shown to be triggered by Cox-2 or IL-6 with increases in TNFα, IL-β1 as well as production of prostaglandin-E2 (PGE2, (Z)-7-[(1R,2R,3R)-3-hydroxy-2-[(E,3S)-3-hydroxyoct-1-enyl]-5-oxocyclopentyl]hept-5-enoic acid) [97, 244247].

Caspase-associated recruitment domain 6 (CARD6)

CARDs are important in virus infections and in interactions with mitochondria and innate immunity [248]. CARDs exist as inactive zymogens and are activated with adaptor molecules containing CARD and involved in apoptosis and inflammation through NF-κB signaling [249]. The microtubule-associated protein, CARD6, increases receptor-interacting protein1 and 2 (RIP1 and 2) triggering mitogen-activated protein kinase kinase kinase 3 (MEKK3) which induces NF-κB. CARD6 acts as a NF-κB modulator [250].

Kim et al. analyzed 100 gastric carcinoma (GC) and 58 esophageal squamous cell carcinoma (ESCC) tissues and 103 colorectal cancer (CRC) specimens and found increased CARD6 expression in ESCC (70.7%), GC (45%) and CRC (78.6%) compared to adjacent normal epithelium used as controls [251]. The GC expression was higher in intestinal type GC (77.8%) according to the Lauren classification compared to diffuse GC (20%).

Chronic inflammation by H. pylori increases Cox-2 [252] resulting in an increase of NF-κB, which also can be activated by Akt. This results in an increase of Snail and a decrease of E-cadherin resulting in compromised tissue integrity, increased cell detachment, and greater invasive potential, which serves as a precursor to metastasis [253].

Activation of NF-κB occurs by cysteine-rich 61 (Cyr61, CCN family member 1, CCN1) which promotes the expression of Cox-2 mRNA as suppression of Cyr61-mediated NF-κB activation, Cox-2 gene expression, and invasiveness can be achieved by applying function-neutralizing antibodies to alphavbeta3 (but not to alphavbeta5) [254, 255]. NF-κB activation signaling also occurs through the ubiquitin-proteasome pathway [256].

Lipopolysaccharide (LPS) of bacteria was previously shown to induce NF-κB in pre-B cells ([3, 168] reviewed in [257]).

There is extensive cytokine involvement in inflammation. An example is the sequential events in interleukin 1 (IL-1) binding to its receptor, thereby demonstrating that tyrosine kinase signaling is essential [258]: IL-1 triggers tyrosine kinase activity and NF-κB protein activation followed by NF-κB protein binding to NF-κB1 site in gro-promoter regions and simulating the growth-related oncogenes (gro).

The previous term “gro-oncogenes” is synonymous with gro1 oncogene, groα, KC, neutrophil-activating protein 3 (NAP-3), and melanoma growth stimulating activity alpha (MSGA-α) all of which are now summarized by the chemokine (C-X-C motif) ligand 1-3: CXCL1 (gro-α), CXCL2 (gro-β), and CXCL3 (gro-γ). The isolation and characterizing of the CXCL/gro-oncogenes occurred within the last three decades (reviewed in [259]): gro-α [260], gro-β and gro-γ [261, 262].

Increased gro-α expression in melanoma enhances “colony-forming activity and tumorigenicity” in nude mice ([263] reviewed in [28]). The inhibition of NF-κB by sodium salicylate or nuclear factor NF-kappa-B protein 65 (p65) subunit (p65) RNA results in decreases of gro-α and gro-β expression [28].

The constitutive expression of gro-α and its receptor CXCR2 (gro-α receptor) was shown to be associated with metastatic potential, modulation of cancer cell proliferation, and with an invasive phenotype [264].

The chemokine family members “are divided into four main classes based on their cysteine (C) residue sequence: the CXC chemokines, the CC chemokines, the C chemokines, and the CX3C chemokines, in which X represents any amino acid” ([265] reviewed in [266]). Their roles are complex with the immunology in different cell compartments versus when carcinogenesis is completed and cancer cells result. Chemokines are therapeutically promising in inhibiting angiogenic CXC chemokine ligands and/or receptors as options to decrease cancer cell development or to decrease metastasis [267, 268].

Another ubiquitous protein is valosin-containing protein (VCP, CDC48) which can be stimulated by IL-6 and results in the progression of prostate cancer LNCaP cells through proto-oncogene serine/threonine-protein kinase (Pim‐1) via signal transducer and STAT3 signaling [269]. Transfecting LNCaP cells for VCP overexpression resulted in an increase in cell proliferation, migration, and invasive behavior. VCP expression is also involved in the regulation of NF-κB activation [270, 271]. Yamamoto et al. found VCP to be an independent factor in multivariate analysis in GC patients for disease-free progression and overall survival, for lymph node metastasis (P < 0.01), and depth of invasion (P < 0.01) [272, 273].

A member of the IL-1 family, interleukin 33 (IL-33), is involved in carcinogenesis and high doses result in inflammation, mucosal atrophy and metaplasia in the gastric fundus in mice, with concurrent increases in IL-6 and interleukin 9 (IL-9) expression. IL-33 also binds to interleukin 1 receptor-like 1 (IL1RL1, ST2) and IL-1 receptor protein IL1-RAP activating NF-κB and MAPK signaling ([274] reviewed in [275]).

Exosomes derived from GC cells have the ability to sustain chronic inflammation by activating macrophages through phosphorylation of NF-κB in macrophages [276].

Knockdown of the cytoskeleton protein, Radixin, together with the NF-κB/Snail signaling resulted in an increase of E-cadherin with suppression of metastasis in human gastric carcinoma SGC-7901 cells [277]. Radixin is also involved in the transition of a normal cell to a cancer cell: Akt phosphorylates Ezrin (pEzrin), which is a member of the ERM (Ezrin/Radixin/Moesin) proteins. High levels of pEzrin together with increased vimentin, and decreased E-Cadherin levels, were seen in tongue squamous cell carcinoma patients with poor prognosis and metastasis [278]. Vimentin is induced by NF-κB and TGF-β1 and is an independent predictor of recurrence after radical prostatectomy [279].

NF-κB signaling and crosstalk during carcinogenesis in fibrosis and remodeling

The association of NF-κB signaling and fibrosis due to various pathogenic stimuli have been previously reviewed. The involvement of NF-κB and fibrosis in carcinogenesis has been recognized for some 20 years [280, 281]. IL-1 induces NF-κB with consequent c-myc expression in fibroblasts [282]. Yokoo et al. showed that NF-κB together with c-Jun/AP1 activation is needed to induce MMP-9 in glomerular mesangial cells and IL-β1 [283]. In human gingival fibroblasts, NF-κB is induced by IL-1 [284].

MMP-9 induction in malignant glioma cells needs actin polymerization with consequent MMP-9 modulation [285] LOX, and especially LOXL2, are major players in remodeling the tumor microenvironment to create the PCN for the transition of a normal cell to a cancer cell [123, 286]. LOXL2 promotes cell proliferation and inhibits apoptosis via a regulator of cellular homeostasis: myristoylated alanine-rich C kinase substrate-like 1 (MARCKSL1) [287].

Keratin 8 (K8) is involved in the development of papillomas to malignant tumors in transgenic mice [288] and has a regulatory signaling effect on target of methylation-mediated silencing (TMS1), MARCKSL1, Ran-specific binding protein 1 (RanBP1), tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein gamma (14-3-3γ), Rho GDP-dissociation inhibitor 2 (Rho-GDI2) and K8 loss results into alteration of TMS1 NF-κB signaling [289].

NF-κB signaling is increased in the precancerous lesion, oral submucousal fibrosis (OSF) compared to human buccal mucosal fibroblasts (BMFs) [44]. Another regulator of transcription interacting with NF-κB is the DNA repair protein, Ku [290]. The double-strand repair of DNA with its DNA repair protein, Ku activity, can be mediated by NF-κB activity affecting cell growth and proliferation [291, 292].

Inflammatory signaling induces chronic stress with consequent fibrosis and a remodeled tumor microenvironment which leads to the PCN [293]. Even for stress-induced lymphatic remodeling, chronic inflammatory signaling is necessary [294]. Such chronic stress results in the disruption of homeostasis, which uncorrected, leads to carcinogenesis [286]. With regard to NF-κB, protein kinase RNA-like endoplasmic reticulum kinase (PERK) appears to be important since it functions as a sensor of UPR [295] – next to inositol-requiring enzyme 1 (Ire1) and activating transcription factor 6 (ATF6) [296].

The transcription factor STAT3 induces IL-6 and modulates PERK “independently of the classic canonical IκBα signaling pathway” [295]. Fan et al. showed that inhibition of PERK interfered with “DNA binding of both STAT3 and NF-κB, thereby preventing induction of NF-κB-dependent genes and E2-induced apoptosis” in breast cancer cells.

The PCN consists of an altered tumor microenvironment that also induces chemoresistance by fibroblast-derived IL-6 upregulating the cell membrane receptor C-X-C motif of chemokine receptor 7 (CXCR7, G-protein coupled receptor 159, GPR159, atypical chemokine receptor 3) via NF-κB [297].

Thelen M and Thelen S investigated 45 ESCC tissues, normal esophageal cell line Het-1a and human ESCC lines TE1, TE7, EC109, KYSE70 and KYSE450 for chemoresistance mechanism and CXCR7 knockdown decreased gene expression associated with cell transition. Until recently it was assumed that CXCR7 “does not mediate typical chemokine receptor responses” [298] but Peng et al. demonstrated that inhibiting CXCR7 decreases chemotaxis of adipose tissue macrophages [299].

C–C chemokine receptor type 7 (CCR7) promotes invasion via activation of PI3K/Akt/mTOR signaling [300]. The PI3K/Akt/mTOR axis is associated with cell proliferation, migration and invasion in cancer [301] and activation through TGF-β1 resulting in EMT [302].

HIF-1α activation by inflammation induces cell transition via PI3K/Akt/mTOR [303] and inhibition results in apoptosis and autophagy [304]. The crosstalk between NF-κB and PI3K/Akt/mTOR has been shown in lymphoma [305] and there is hope that targeting this pathway might be effective in anticancer therapy [306].

NF-κB activation by CCR7 via PI3K/Akt/mTOR signaling is relevant as both NF-κB and CCR7 are upregulated in head and neck cancers and promote cell invasion [300]. C–C chemokine receptor (CCR) together with chemokine (C–C motif) ligand 19 (CCL19) results in phosphorylation of IκBα, the inhibitor of NF-κB promoting NF-κB translocation to the nucleus while inhibiting NF-κB and CCR7/PI3K signaling increases apoptosis, cell arrest, and attenuated survival of SCCHN cells.

The novel PEST-containing nuclear protein (PCNP) along with the ubiquitin ligase ubiquitin-like, containing PHD and RING finger domains 1 (UHRF1, Np95)/ICBP90-like RING finger protein (NIRF), both located in the nucleus, were recently associated with increased NIRF expression in a human fibrosarcoma and hepatocellular carcinoma cell line (HT-1080, HepG2 cells) [307, 308]. PCNP mediates cell proliferation, migration and invasion in neuroblastoma through activating PI3K/Akt/mTOR together with MAPK [309].

NF-κB regulates C-X-C chemokine receptor 4 (CXCR4) while the O-GlcNAc modification of p65-NF-κB stimulates its activity [310]. Prostate cancer cells showed dysregulation of NF-κB and STAT1 signaling, a property dependent upon the cell line under investigation and the production of angiostatic CXC chemokines [311]: PZ-HPV-7 cells produce a high amount of CXC chemokines whereas a lower amount were observed in CA-HPV-10 and PC-3 cell lines.

NF-κB signaling and crosstalk during carcinogenesis in cell transition

Transformation of a normal cell to a cancer cell is dependent on the microenvironment, the cell matrix, and transcription factors ([312] reviewed in [188]). NF-κB is important for cell transition [313, 314].

Chronic application of TGF-β1 to HeLa cells resulted in chronic inflammation with increased TNFα, with TGF-β1 induced increase of epithelial-to-mesenchymal cell transition, and self-renewal [315]. This effect was dependent on NF-κB and Twist1 and “Overexpression of NF-κBp65 upregulated Twist and promoted EMT and cancer stem cell-like (CSCL) properties in HeLa cells exposed to inflammatory cytokines”. The inhibition of TGF-β1 by Disulfiram resulted in the inhibition of cell transition and stem cell features via extracellular signal–regulated kinase (ERK)/NF-κB/Snail signaling [316].

Using an in vivo breast cancer model it was shown that the IKK-2/IkappaBalpha/ NF-κB pathway is required for introduction, maintenance, and cell transition, and that NF-κB inhibition in mesenchymal cells reversed cell transition [317].

Inhibiting NF-κB or TGF-β1 signaling in prostate cancer PC-3 cell line decreased Vimentin expression as well as invasive cell capability. Cell transition was shown to be TGF-β1 induced and mediated by NF-κB. Investigating a large number of prostatectomy specimen revealed that high expressions of TGF-β1, NF-κB and vimentin together with low levels of cytokeratin 18 resulted in recurrence [279].

TGF-β1 induces IL-6 through mothers against decapentaplegic homolog 2 (Smad2), p38-NF-kappaB, c-Jun N-terminal kinase (JNK), and the protein superfamily of small guanosine triphosphate hydrolase enzymes (Ras, GTPases) in prostate cancer cell lines [318]. However, it seemed to matter as to which NF-κB pathway and/or subunit was activated [319]. The inhibition of Akt and NF-κB suppressed epidermal growth factor (EGF)-induced cell transition in a squamous cell carcinoma cell line of the tongue and facilitated both cell migration and invasion [278].

In a tetracycline model, activation of proto-oncogene c-Rel (cRel, transcription factor containing Rel homolog domain, RHD) activation with consequent nuclear expression, arrested cell proliferation in the G1/S-phase together with accumulation of hypophosphorylated retinoblastoma protein (Rb), increase of the cyclin-dependent kinase (CDK) inhibitor 1 (p21, CIP1/WAF1/CAP20/SDI1, p21WAF1), reduced cdk2 activity, increased p53 protein stability [318]. cRel knockdown abolished these effects. This is concordant to cRel expression found in all hematopoietic tissues, but cRel is abundant in mature lymphocytes [320] and induces cell proliferation and survival in mature B-Cells [321]. Furthermore, cRel induces apoptosis in avian fibroblasts [322] but this apoptosis can be inhibited by cRel activating the Bcl-2 homolog Bfl-1/A1 [323, 324]. However, it is not just the transcription factor itself, but that NF-κB activation is dependent on which way NF-κB acetylation/deacetylation occurs with its effect on other pathways such as HIF-1 [325].

TNF-like weak inducer of apoptosis (TWEAK), together with its fibroblast growth factor-inducible 14 (Fn14), has pro-inflammatory and tissue remodeling effects ([326] reviewed in [327]) and increases TGF-β1 induced epithelial-mesenchymal transition in human bronchial epithelial cells and downregulates E-cadherin which requires p38 MAPK and NF-κB [327]. The fibroblast growth factor-inducible 14 (Fn14) gene was located and isolated in 1999 and shown to modulate fibroblast adhesion [328]. The member of the TWEAK superfamily is a ligand to Fn14 meaning Fn14 serves as a receptor at the cell surface and TWEAK increases during cancer progression [329]. The absence of Fn14 is associated with carcinogenesis in colitis-associated cancer but Fn14 can be protective in an acute inflammatory situation [330]. Thus, Fn14 action is necessary in homeostasis and acts aberrantly when homeostasis is disrupted.

The Fn14 protein mediates NF-κB activation [331] and contains NF-κB binding sites [332]. TWEAK/Fn14 activation is also involved in the development of another chronic inflammatory disease, atherosclerosis, where it results in increases of MMP-9 through NF-κB and involved in modulation of the ECM [333]. Fn14 is increased in various cancers such as glioblastoma [332], breast cancer [334], ovarian cancer [335], melanoma [336], non-small lung cancer [337], hepatocellular cancer (HCC) [338], pancreatic cancer [339], colon cancer [340], as well as in esophageal [341] and gastric cancer [342].

Fn14 knockdown showed that Fn14 affects cell growth through NF-κB and B-cell lymphoma-extra large (Bcl-xL) in gastric cancer [342] and in small-cell lung cancer (SCLC) [343]. Microarray analysis comparing Barrett’s biopsies to surgically resected esophageal adenocarcinoma showed only Fn14 over expression when the biopsied Barrett’s epithelium was directly adjacent to esophageal adenocarcinoma [341] which was in contrast to a prior investigation [344]. We contend that this is related to the difference in the degree of development of the PCN.

The TWEAK/Fn14 axis is reported in biological pathogenic stimulus, such as HPV infection, triggering the switch of keratinocytes from apoptosis into proliferation [345]. TWEAK also regulates mesenchymal cells [346] and the switch to proliferation was also observed in endothelial cells [347]. Intraabdominal TWEAK application induced peritoneal inflammation with “increased Fn14, MCP-1 and chemokine (C–C motif) ligand 21 (CCL21) expression and submesothelial tissue macrophage recruitment” [348]. This might be relevant to a future understanding how peritoneal carcinomatosis develops and progresses. TWEAK induces next to changes of phenotype a decrease of cell-cell interaction and anchoring junctions such as E-cadherin, Cadherin-16, β-catenin and adherens, and the tight junction protein Zonula occludens-1 (ZO-1) resulting in weaker epithelial integrity as well as epithelial-mesenchymal transition (EMT) which occur through Fn14 together with NF-κB ERK activation and the vitamin D receptor modulation independent of TGF-β1 [349].

TRADDs, TRAFs, RIPs, FADD, RANK signaling with NF-κB

The TNF superfamily contains multiple transmembrane proteins that interact with the TNF receptor superfamily (TNFRSF) and various cytokines, named TNF-related activation-induced cytokines (TRANCE), which are expressed on cell surfaces, such as activated T-cells and osteoblasts [350]. TNF receptors need an adaptor protein such as TNF receptor type 1-associated DEATH domain proteins (TRADDs), TNF receptor associated factors (TRAFs), receptor-interacting protein kinases (RIPs) and/or Fas-associated protein with death domain (FADD) [351]. TNFs, TNFRSFs as well as TRANCE, TRADDs, TRAFs and RIPSs and FADDs are of importance in the signaling and crosstalk with NF-κB [352, 353].

The disruption of bone homeostasis affecting osteoclastogenesis and osteoblastogenesis is important in the elderly who may be osteoporotic, and apparently with cancers connected by multiple signaling pathways of cytokines, hormones, and growth factors [354356]. Bone remodeling is of importance in prostate cancers. In a routine autopsy study among 19,316 individuals from 1967 to 1995, 8.2% (1589) had prostate cancers and, of these, hematogeneous metastasis were most frequently observed (90%) in the bone of men older than 40 years [357]. The NF-κB signaling pathway is involved here as well.

The receptor of nuclear factor kappaB (RANK) was discovered in 1997 [358] and is of importance together with its receptor activator of NF-κB ligand (RANKL), and decoy receptor osteo NF-κB protegerin (OPG), as expression analysis of the RANKL/RANK/OPG axis correlates with aggressive advanced and metastatic prostate cancers [359].

Esophageal high-grade dysplasia tissue revealed a weaker RANK immunoreactivity compared to 23 esophagectomy cancer specimens [360], and investigating 309 ESCC showed that RANK over expression was associated with a poor prognosis [361]. Increased RANK expression was found in breast cancer [358, 362364]. As reviewed recently, RANK signaling targets various pathways, such as the PI3K/Akt axis, MAPK signaling (JNK, ERK and p38), and NF-κB [365]. RANK promotes IL-1, IL-6 and interleukin 12 (IL-12) and the RANK signaling pathway is mediated by the adaptor protein TRAF6 to induce osteoclast differentiation [366].

TGF-β1 mediated NF-κB activation induces expression of metalloproteinase 12 (ADAM-12) in a dose-dependent manner in MDA-MB-231 breast cancer cells [367]. The gene expression signature studied by Ooi et al. identified proliferation/stem cell- and Wnt/β-catenin- signaling as well as deregulation of the NF-κB pathway in more than 70% of gastric cancers [368].

NF-κB influences transcription-dependent genes by binding to specific sections of DNA (κB-motif) [369] and thus has numerous target genes which mediate its varied effects [370]. The κB-motif, by itself, has a certain degree of variability which suggests that it has the ability to fine tune its effects [371]. NF-κB counteracts senescence induced by oncogenes and thus drives pro-carcinogenic inflammation [117, 179]. KO experiments of the NF-κB-pathway revealed that NASPs occur upstream of TNF receptor-associated factor 6. After transduction of NASPs into the fibroblasts of mice and rats resulted in the co-expression of Ras (H-Ras V12) and revealed that rodent fibroblasts mastered the p53-dependent senescence which is mediated by H-Ras V12 and this in turn resulted in a transformed carcinogenic phenotype [117]. These observations underly the different functions of NF-κB and its cooperation with Ras as an oncogene by the attenuation of p53 as well its effects on the inflammatory cascade.

NF-κB Inhibition

In 1992, p65 antisense oligonucleotide treatment in mice gave hope that anti-NF-κB therapy could be useful against cancer [115]. 7,12-dimethylbenz-(a)anthracene (DMBA) treatment in rats showed NF-κB activation with the development of mammary gland cancer [54]. NF-κB activation in fibroblasts in hormone-independent ER negative cancers occurs through IL-6 and urokinase plasminogen activator (uPA) dependent on interleukin 1 alpha (IL-1α) [56].

There is potential to using anti-NF-κB therapy in gastric cancer patients because Cyclosporin A increased docetaxel (Taxotere)-induced apoptosis through the activation of NF-κB in human gastric cancer cells and thereby prevented the anti-apoptotic NF-κB effect [372]. This may explain why NF-κB data were generated in upper GI cancers. However, the reality of immunosuppressed patients who have undergone organ transplants is different as such patients have an increased risk of developing cancer later [373]. Furthermore, this might serve as an example that inhibition of a single signaling pathway is not enough in diseases such as cancer as cancer occurs from the broader disruption of homeostasis [189191, 286, 293]. It has been reported that targeting NF-κB might have a therapeutic effect in cancer treatment [374376].

Preventing NF-κB activation does not alter deoxycholate-induced apoptosis, suggesting that NF-κB may not be essential for apoptosis and likely represents just one of many signaling pathways. However, aspirin prevents the deoxycholate-induced apoptosis although it has not been shown to have a direct anti- NF-κB effect [377]. This illustrates the complexity of the various pathways involved.

Urokinase-type plasminogen activator receptor (uPAR) together with uPA forms a complex that is associated with tumor cell invasion and that affects cell motility and integrin function ([378, 379] reviewed in [380]). The uPAR expression through NF-κB suppression was demonstrated by investigating the diterpenoid triepoxide, triptolide from the Chinese herb Tripterygium wilfordii Hook F.

NF-κB was also implicated in cancer with regard to 5-fluorouracil (5-FU). Scientists showed an induction of NF-κB by 5-FU application within human gastric adenocarcinoma cell line, NUGC3 cells (5-fluorouracil sensitive), but not within NUGC3/5FU/L cells, which are 5-fluorouracil resistant. The inhibition of NF-κB reduced chemoresistance and increased apoptosis again lends credence to the likelihood of addressing a huge problem in chemotherapy, namely the development of chemoresistance [381]. Inhibition of NF-κB-dependant signaling might overcome bromodomain and extra terminal protein inhibitors (BETi) resistance in uveal melanoma [382]. Additionally, paeoniflorin from Paeonia lactiflora, can inhibit NF-κB activity in gastric SGC-7901 cancer cells with consequent increases of 5-FU induced apoptosis [383].

Non-steroidal anti-inflammatory drugs (NSAID)

The NSAID drug aspirin and sodium salicylate suppresses NF-κB [384] and prostaglandin production by inhibiting cyclooxygenase 1 (Cox-1, prostaglandin G/H synthase 1) and Cox-2 and is effective in suppressing colon cancer growth but only at high concentrations [385]. The investigation of the mechanism of ibuprofen showed that it activates IkappaB kinase alpha with consequent inhibition of the activation of NF-κB and IKKα in human prostate cell lines (hormone-independent cell lines, PC-3 and DU-14)5 and it may be noted that ionizing PC-3 cells did not result in NF-κB modulated DNA-binding activity [386].

Metformin

Metformin inhibits inflammatory response and malignant cell transformation [387]. It also inhibits NF-κB in a dose-dependent manner [388]. Metformin decreases both pro-inflammatory cytokines and NF-κB and improves the immune response to cancer cells in colorectal, prostate, pancreatic, renal, cervical, endometrial, gastric, lung, breast, and ovarian cancer (reviewed in [389]). These findings, and the fact that NF-κB signaling triggers E-cadherin downregulation, and enhanced by connective tissue growth factor (CTGF, CCN2): it is upregulated in gastric cancer tissues promoting cell proliferation and metastasis [390] which explain recent signaling pathways identified during carcinogenesis as affecting the disruption of homeostasis [189192, 286, 293, 388, 391]. Furthermore, Metformin decreases NF-κB in in the soleus muscle of diabetic rats and increases its inhibitor, IκB [392]. The transcriptional factors NF-κB and STAT3 are ubiquitously expressed and operate in concert to promote cancer development and progression of colon, gastric and liver cancers [393].

Silibinin

The polyphenolic flavonoid, Silibinin, is an extract from milk thistle (Silybum marianium) and was earlier reported to have an anticancer effect [394397]. Silibinin is a direct STAT3 inhibitor [398] with no direct effect on apoptosis or changes in p53 and bcl2 [395]. Silibinin inhibits NF-κB p50 translocation via the upregulation of IκB and downregulates ZEB1 and Zinc finger protein SNAI2 (SLUG) transcription factors, and can reverse cell transition [399]. In colon carcinoma, protein levels of Bcl-2, Cox-2, iNOS, VEGF and MMPs, which are also NF-κB-regulated molecules, can be decreased in cell culture and xenograft analyses by the application of Silibinin due to inhibition of nuclear p50 (NF-κB1) and 65 translocation [400].

NF-kB signaling balances between apoptosis versus necrosis both at host and tumor interfaces through various pathways, which reveal that the weight of disruption of homeostasis in this regard determine which effect will prevail.

Caffeic acid phenethyl ester (CAPE)

Another inhibitor of NF-κB activity and chronic inflammation with reduced expression of mediators such as TNFα, interferon gamma (IFNγ), IL-2, IL-6, KC (IL-8 homologue), and inducible iNOS is the anti-inflammatory caffeic acid phenethyl ester (CAPE) in H. pylori-induced gastritis in Mongolian gerbils [401]. NF-κB activity was also shown to be suppressed by the novel oligosaccharide, JG3, derived from marine oligomannurarate which results in tumor growth in xenograft models [402].

Furthermore, more detailed information of inhibitory effects of potential anti NF-κB compounds were confirmed in TNFα induced NF-κB bla assay investigating 2800 clinically approved drugs and bioactive compounds from the NIH Chemical Genomics Center Pharmaceutical Collection (NPC) [403]. Nineteen drugs inhibiting NF-κB with potencies as low as 20 nM were identified: bithionol, bortezombi, cantharidin, chromomycin A3, daunorubicinum, digitoxin, ectinascidin 743, emetine, flurosalan, manidipine hydrochloride, narasin, lestaurtinib, ouabain, sorafenib tosylate, suitib malate, tioconazole, tribromsalan, triclabendazolum and zafirlukast. Some induce NF-κB inhibition through inhibition of IkappaBalpha phosphorylation (emetine, fluorosalan, sunitinib malate, bithionol, narasin, tribromsalan, and lestaurtinib).

However, as a disruption of homeostasis in NF-κB signaling also results in the consequent various activation/deactivation of pathways, the expectations may have to be toned down, especially as it has been shown that some classical drugs may act as NF-κB modulators or IKKβ inhibitors [404]. For example, the compound salvianolic acid C (SalC) isolated from the plant, Salvia miltiorrhiza Bunge, can inhibit “LPS-induced inflammatory response and NF‑κB activation through the activation of AMPK/Nrf2 signaling both in vivo and in vitro” [405].

Otherwise, having an anti-NF-κB therapeutic modality might result into unwanted effects. Nrf2 is responsible for migration and invasion in cancers of the cervix [406], and aggressiveness in various cancers, such as breast cancer [407, 408], gastric cancer [409] and colorectal cancer [410], or responsible for drug resistance in glioma and melanoma [411].

This reveals how carefully any anti-NF-κB approach needs to be investigated prior to drawing conclusions about its role in a specific anticancer regimen.

Summary

The available information on NF-κB expression in tissues provides a perspective on understanding signaling and crosstalk during pathogenic stimuli and carcinogenesis. This provides a rational basis to investigate NF-κB and to implement an anti-NF-κB therapy into existing anti-cancer treatment regimens, when appropriate. For this, the NF-κB interplay in sequences that contribute to carcinogenesis such as chronic inflammation, remodeled fibrosis with the precancerous niche (PCN), and the transition of a normal cell to a cancer cell is essential. However, inhibition of a single signaling pathway is not enough in diseases such as cancer as cancer occurs from the broader disruption of homeostasis.

Nomenclature of abbreviations

5-FU: 5-fluorouracil

5-oxo-ETE: (6E,8Z,11Z,14Z)-5-oxoicosa-6,8,11,14-tetraenoic acid

12-HETE: 12-hydroxyeicosatetraenoic acid

14-3-3γ: Tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein gamma

20-HETE: 20-hydroxyeicosatetraenoic acid, (5Z,8Z,11Z,14Z)-20-hydroxyicosa-5,8,11,14-tetraenoic acid

20-OH-PGE2: 20-hydroxy prostaglandin E2; 20-HETE 20-hydroxyeicosatetraenoic acid, (5Z,8Z,11Z,14Z)-20-hydroxyicosa-5,8,11,14-tetraenoic acid

α-SMA: Alpha smooth muscle actin, alpha-actin-2

11β-HSD1: 11β-hydroxysteroid dehydrogenase type 1

ADAM-12: Matrix metalloproteinase 12 (MMP-12)

AFT3: Activating transcription factor 3

AIDS: Acquired immune deficiency syndrome

Akt: Protein kinase B

ALOX: Lipoxygenase, arachidonate lipoxygenase

ALOX5: 5-lipoxygenase, 5-LOX, arachidonate 5-lipoxygenase

ALOX12: 12-lipoxygenase, 12-LOX, 12S-LOX, arachidonate 12-lipoxygenase 12S type

AMPK: 5′ adenosine monophosphate-activated protein kinase (AMP)-activated protein kinase

ANXA2: Annexin II

AP1: Activator protein 1

APO-1: Apoptosis antigen 1 (APT), cluster of differentiation 95, CD95, first apoptosis signal receptor (Fas, FasR)

APT: Apoptosis antigen 1 (APO-1), cluster of differentiation 95, CD95, first apoptosis signal receptor (Fas, FasR)

ATF6: Activating transcription factor 6

BAFF: B cell–activating factor belonging to the tumor necrosis factor (TNF) family

BAFFR: B-cell activating factor receptor

BAG-1: BAG family molecular chaperone regulator 1

Bax: B-cell lymphoma 2 (Bcl-2)-associated X protein

Bcl-2: B-cell lymphoma 2

Bcl-3: B-cell lymphoma 3

BCR: B-cell receptor

BETi: Bromodomain and extra terminal protein inhibitor

BIM: Bcl-2 interacting mediator of cell death

BiP: Binding immunoglobulin protein, 78 kDa glucose-regulated protein (GRP-78), heat shock 70 kDa protein 5 (HSPA5)

BMF: Buccal mucosal fibrosis

Btk: Bruton’s tyrosine kinase

C: Cysteine

CaMKII: Ca2+/calmodulin-dependent protein kinase II

cAMP: Cyclic 5′ adenosine monophosphate (AMP)

CAPE: Caffeic acid phenethyl ester

CARD6: Caspase-associated recruitment domain 6

CBP: Co-activator CREB binding protein

CCAAT: Cytosine-cytosine-adenosine-adenosine-thymidine

CCC: Cholangiocellular carcinoma

CCL2: Chemokine (C–C motif) ligand 2, monocyte chemoattractant protein 1, MCP-1

CCL3: Chemokine (C–C motif) ligand 3, macrophage inflammatory protein 1 alpha (MIP1α)

CD14: Cluster of differentiation 14

CD40: Cluster of differentiation 40, costimulatory protein on T-cells

CD40L: Cluster of differentiation 40 (CD40) ligand, CD154, protein expressed on T-cells

CD54: Cluster of differentiation 54, intercellular adhesion molecule 1 (ICAM-1)

CD62: Cluster of differentiation 62

CD62E: Cluster of differentiation (CD62, endothelial-leukocyte adhesion molecule 1, E-selectin) antigen-like family member E (leukocyte-endothelial cell adhesion, molecule 2, LECAM2)

CD95: Cluster of differentiation 95, first apoptosis signal receptor (Fas, FasR), apoptosis antigen 1 (APO-1, APT)

CD106: Cluster of differentiation 106, vascular cell adhesion protein 1, vascular cell adhesion molecule 1 (VCAM-1)

CD154: Cluster of differentiation 154

cdc42: Cell division control protein 42 homolog

CDK: Cyclin-dependent kinase

cdk2: Cyclin-dependent kinase 2

CDX2: Homeobox protein CDX-2

C/EBPβ: CCAAT/enhancer-binding protein β

cagA: Helicobacter pylori virulence factor CagA, cytotoxin-associated gene A

CCL19: Chemokine (C–C motif) ligand 19

CCL21: Chemokine (C–C motif) ligand 21

CCN1: CCN family member 1, cysteine-rich 61 (Cyr61)

CCN2: Connective tissue growth factor (CTGF)

CCR: C–C chemokine receptor

CCR7: C–C chemokine receptor type 7

CHOP: Cytosine-cytosine-adenosine-adenosine-thymidine (CCAAT)/enhancer binding protein homologous protein

Cox: Cyclooxygenase

Cox-1: Cyclooxygenase 1, prostaglandin G/H synthase 1

Cox-2: Cyclooxygenase 2

Cox-3: Isoform of Cox-2 (therefore in brakes)

CRC: Colorectal carcinoma

CREB: cAMP response element-binding protein

cRel: Proto-oncogene c-Rel, transcription factor containing Rel homolog domain (RHD)

CSES: Chronic stress escape strategy

CTGF: Connective tissue growth factor (CCN2)

Cyclin B1: G2/mitotic-specific cyclin-B1

Cyclin D1: G1/S phase transition specific cyclin-D1

Cyclin E: G1-to-S phase transition specific cyclin-E

CXC CC: Chemokine receptors

CXCL1: Chemokine (C-X-C motif) ligand 1

CXCL8: Chemokine (C-X-C motif) ligand 8, interleukin 8 (IL-8)

CXCR2: Gro-α receptor

CXCR4: C-X-C chemokine receptor 4

CXCR7: C-X-C motif of chemokine receptor 7 (G-protein coupled receptor 159, GPR159, atypical chemokine receptor 3)

CYP*: Cytochrome P450 isoforms

Cyr61: Cysteine-rich 61, CCN family member 1 (CCN1)

DMBA: 7,12-dimethylbenz-(a)anthracene

DNA: Deoxyribonucleic acid

dsRNAs: Double-stranded RNAs

E2F4/5: Cytoplasmic complex of Smad3, retinoblastoma-like protein 1 (P107, RBL1), E2F4/5 and D-prostanoid (DP1)

E. coli : Escherichia coli

E-Cadherin: CAM 120/80 or epithelial cadherin, cadherin-1, epithelial cadherin

ECM: Extracellular matrix

EMT: Epithelial-mesenchymal transition

E1: Ubiquitin-activating enzyme

E1A: Adenovirus early region E1A protein

E2: Ubiquitin-conjugating enzyme

E3: Ubiquitin ligase

EBV: Epstein-Barr virus

ECs: Endothelial cells

EET: Epoxyeicosatrienoic acid

EGFR: Epidermal growth factor receptor (ErbB-1, HER1)

ELAM-1: Endothelial-leukocyte adhesion molecule 1

E-selectin: Endothelial-leukocyte adhesion molecule 1, cluster of differentiation (CD62) antigen-like family member E (CD62E), leukocyte-endothelial cell adhesion molecule 2 (LECAM2)

ERK: Extracellular signal–regulated kinase

ESCC: Esophageal squamous cell carcinoma

FADD: Fas-associated protein with death domain

Fas: First apoptosis signal receptor (FasR), apoptosis antigen 1 (APT, APO-1), cluster of differentiation 95 (CD95)

Fn14: Fibroblast growth factor-inducible 14

FOXO3a: Forkhead box protein O3a

FXR: Farnesoid X receptor

GC: Gastric carcinoma

GC-C: Guanyl cyclase 2

GERD: Gastro-esophageal reflux disease

GHS-R: Growth hormone secretagogue receptor

GM-CSF: Granulocyte-macrophage colony-stimulating factor

gro: Growth-related oncogene

gro-α: Growth-related oncogene alpha, CXCL1

gro-β: Growth-related oncogene beta, CXCL2

gro-γ: Growth-related oncogene gamma, CXCL3

GSK3β: Glycogen synthase kinase-3β

GTP: Guanosine triphosphate hydrolase enzyme

GTPase: Guanosine triphosphate (GTP) hydrolase enzyme, transforming protein p21, HRas, H-ras

H. pylori : Helicobacter pylori

H. bilis : Helicobacter bilis

HBsAg: Hepatitis B surface antigen

HBV: Hepatitis B virus

HBx: Hepatitis B viral protein

HCC: Hepatocellular carcinoma

HCV: Hepatitis C virus

HIAP: Human inhibitor of apoptosis protein

HIF1α: Hypoxia-inducible factor alpha

HIV: Human immunodeficiency virus

HMGB-1: High mobility group box 1 protein

HPV: Human papilloma virus

HPV8: Human papilloma virus type 8

HPV16: Human papilloma virus type 16

H-ras: Guanosine triphosphate (GTP) hydrolase enzyme, transforming protein p21, H-ras

HFD: High-fat diet

hrHPV: High-risk human papillomavirus

HSC: Hepatic stellate cells

hTERT: Human telomerase reverse transcriptase

IBS-D: Diarrhea-predominant irritable bowel syndrome

ICAM-1: Intercellular adhesion molecule 1, cluster of differentiation 54, CD54

IFN: Interferon

IFNγ: Inteferon gamma

IκB: Nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor protein

IκBα: Nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha IκB kinase 1 (IKK1)

IκBβ: Nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, beta, IκB kinase 2 (IKK2)

IκBγ: Nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, gamma, IκB kinase gamma

IκBε: Nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, epsilon

IKK: IκB kinase enzyme complex

IKK-α: Inhibitor of nuclear factor kappa-B kinase subunit alpha, inhibitor of nuclear factor kappa-B kinase 1 (IKK1)

IKK-β: Inhibitor of nuclear factor kappa-B kinase subunit beta, inhibitor of nuclear factor kappa-B kinase 2 (IKK2)

IKK-γ: Inhibitor of nuclear factor kappa-B kinase subunit gamma, NF-kappa-B essential modulator (NEMO)

IKK1: Inhibitor of nuclear factor kappa-B kinase 1, inhibitor of nuclear factor kappa-B kinase subunit alpha (IKK-α)

IKK2: Inhibitor of nuclear factor kappa-B kinase 2, inhibitor of nuclear factor kappa-B kinase subunit beta (IKK-β)

IL-1α: Interleukin 1 alpha

IL-β1: Interleukin beta 1

IL-1: Interleukin 1

IL-6: Interleukin 6

IL-8: Interleukin 8, chemokine (C-X-C motif) ligand 8 (CXCL8)

IL-9: Interleukin 9

IL-12: Interleukin 12

IL-13: Interleukin 13

IL-32: Interleukin 32

IL-33: Interleukin 33

ILR1: Interleukin 1 receptor

IL1RL1: Interleukin 1 receptor-like 1 (ST2)

iNOS: Nitric oxide synthetase

Ire1: Inositol-requiring enzyme 1

Irf3: Interferon regulatory factor 3

JNK: c-Jun N-terminal kinase

K8: Keratin 8

KC: IL-8 homologue

KO: Knockout

LEC: Lymphatic endothelial cell

LOX: Lysyl oxidase

LOXL: LO-like gene

LOXL2: Lysyl oxidase homolog 2

LOXL3: Lysyl oxidase homolog 3

LPS: Lipopolysaccharide

LTβR: Lymphotoxin beta receptor

LTA4: Leukotriene A4, 4-[(2S,3S)-3-[(1E,3E,5Z,8Z)-tetradeca-1,3,5,8-tetraenyl]oxiran-2-yl]butanoic acid

LTB4: Leukotriene B4, (5S,6Z,8E,10E,12R,14Z)-5,12-dihydroxyicosa-6,8,10,14-tetraenoic acid

LTC4: Leukotriene C4, (5S,6R,7E,9E,11Z,14Z)-6-[(2R)-2-[[(4S)-4-amino-4-carboxybutanoyl]amino]-3-(carboxymethylamino)-3-oxopropyl]sulfanyl-5-hydroxyicosa-7,9,11,14-tetraenoic acid

LTD4: Leukotriene D4, (5S,6R,7E,9E,11Z,14Z)-6-[(2R)-2-amino-3-(carboxymethylamino)-3-oxopropyl]sulfanyl-5-hydroxyicosa-7,9,11,14-tetraenoic acid

LTE4: Leukotriene E4, (5S,6R,7E,9E,11Z,14Z)-6-[(2R)-2-amino-2-carboxyethyl]sulfanyl-5-hydroxyicosa-7,9,11,14-tetraenoic acid

mTORc1: Rapamycin complex 1

M. fermentans : Mycoplasma fermentas

M. hominis : Mycoplasma hominis

M. hyorhinis : Mycoplasma hyorhinis

M. penetrans : Mycoplasma penetrans

MAPK: p38 mitogen-activated protein kinase

MAPK2: Mitogen-activated protein kinase kinase, MEK, MAPKK

MAPK3: Mitogen-activated protein kinase 3 (ERK1)

MAP2K1: Dual specificity mitogen-activated protein kinase kinase 1

MAPKK: Mitogen-activated protein kinase kinase, MEK, MAPK2

MARCKSL1: Myristoylated alanine-rich C kinase substrate-like 1

MC: Mast cell

MCP-1: Monocyte chemoattractant protein 1, chemokine (C–C motif) ligand 2, CCL2

MDA: Malondialdehyde, propanedial

MEFs: Mouse embryonic cells

MEK: Mitogen-activated protein kinase kinase, MAPK2, MAPKK

MEKK3: Mitogen-activated protein kinase kinase kinase 3

MIP1α: Macrophage inflammatory protein 1-alpha, chemokine (C–C motif) ligand 3 (CCL3)

miR21: Micro RNA-21

MMP: Matrix metalloproteinase

MMP-1: Matrix metalloproteinase 1

MMP-2: Matrix metalloproteinase 2

MMP-3: Matrix metalloproteinase 3

MMP-7: Matrix metalloproteinase 7

MMP-9: Matrix metalloproteinase 9

MMP-12: Matrix metalloproteinase 12 (ADAM-12)

MMP-14: Matrix metalloproteinase 14, MT1-MMP

mRNA: Messenger ribonucleic acid

MSGA-α: Melanoma growth stimulating activity alpha

NAP3: Neutrophil-activating protein 3, gro-oncogene

NASPs: NF-κB–activating genetic elements

NCCCT: Normal cell to cancerous cell transition

NEMO: NF-kappa-B essential modulator, inhibitor of nuclear factor kappa-B kinase subunit gamma, IKK-γ

NF-κB: Nuclear factor kappa-light-chain-enhancer of activated B cells

NF-κB1: Nuclear factor kappa-light-chain-enhancer of activated B cells 1, protein 50 (p50) its progenitor protein 105 (p105)

NF-κB2: Nuclear factor kappa-light-chain-enhancer of activated B cells 2, protein 52 (p52) and its progenitor protein 100 (p100)

NGF: Nerve growth factor

NIRF: ICBP90-like RING finger protein

NIK: Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) -inducing kinase

NIrp3: Intracellular NOD-like receptor pyrin domain containing 3

NPC: NIH Chemical Genomics Center Pharmaceutical Collection

NR2B1: Nuclear receptor subfamily 2, group B, member 1, retinoid X receptor alpha (Rxra)

NSAID: Non-steroidal anti-inflammatory drugs

OPG: Decoy receptor osteo NF-κB protegerin

OSF: Oral submucousal fibrosis

Osm: Oncostatin-M

p21: Protein p21, guanosine triphosphate (GTP) hydrolase enzyme, transforming H-ras

p33: Protein 33

p37: Mycoplasma hyorhinis membrane protein p37

p50: Protein 50, nuclear factor kappa-light-chain-enhancer of activated B cells 1 (NF-κB1)

p52: Protein 52

p53: Protein 53

p65: Nuclear factor NF-kappa-B protein 65 (p65) subunit

p100: Protein 100

p105: Protein 105, p50 (NF-κB1) progenitor

p107: Retinoblastoma-like protein 1, RBL1

p120: Protein 120, catenin delta-1

p300: Adenovirus early region 1A (E1A) binding protein p300, EP300, p300-CBP coactivator family

PAE: Paeoniflorin

PAI1: Plasminogen activator inhibitor-1

PCN: Precancerous niche

PCNP: PEST-containing nuclear protein

PCR: Polymerase chain reaction

PCR-RFLP: Polymerase chain reaction-restriction fragment length polymorphism

PERK: Protein kinase RNA-like endoplasmic reticulum kinase

PGD2: Prostaglandin D2, (Z)-7-[(1R,2R,5S)-5-hydroxy-2-[(E,3S)-3-hydroxyoct-1-enyl]-3-oxocyclopentyl]hept-5-enoic acid

PGE2: Prostaglandin E2, (Z)-7-[(1R,2R,3R)-3-hydroxy-2-[(E,3S)-3-hydroxyoct-1-enyl]-5-oxocyclopentyl]hept-5-enoic acid

PGFF2α: Prostaglandine F2 alpha, (Z)-7-[(1R,2R,3R,5S)-3,5-dihydroxy-2-[(E,3S)-3-hydroxyoct-1-enyl]cyclopentyl]hept-5-enoic acid

PGG2: Prostaglandin G2, (Z)-7-[(1S,4R,5R,6R)-5-[(E,3S)-3-hydroperoxyoct-1-enyl]-2,3-dioxabicyclo[2.2.1]heptan-6-yl]hept-5-enoic acid

PGH2: Prostaglandin H2, (Z)-7-[(1S,4R,5R,6R)-5-[(E,3S)-3-hydroxyoct-1-enyl]-2,3-dioxabicyclo[2.2.1]heptan-6-yl]hept-5-enoic acid

PI3K: Phosphatidylinositol 3-kinase

Pim‐1: Proto-oncogene serine/threonine-protein kinase

PKA: Protein kinase A

PLA2: Phospholipase A2

PLD1: Phospholipase D1

PPI: Proton pump inhibitor

PRDM1: PR domain zinc finger protein 1

pro-IL-1β: Inactive interleukin 1 beta precursor

Pro-MMP-1: Pro-matrix metalloproteinase 1

Pro-MMP-7: Pro matrix metalloproteinase 7

Pro-MMP-9: Pro-matrix metalloproteinase 9

PUMA: BH3-only protein

Rac: Subfamily of the Rho family of GTPases

Rac1: Ras-related C3 botulinum toxin substrate 1

RanBP1: Ran-specific binding protein 1

RANK: Receptor of nuclear factor kappaB

RANKL: Receptor activator of NF-κB ligand

Ras: Protein superfamily of small guanosine triphosphate hydrolase enzymes (GTPases)

Rb: Retinoblastoma protein

Rel: Proto-oncogene c-Rel encoded by REL gene (cRel)

RelA: Transcription factor p65 encoded by RELA gene,

RelB: Transcription factor encoded by the RELB gene interacting with NF-κB

RHD: Rel homology domain

Rho: Ras homolog gene family

RhoA: Ras homolog gene family, member A

Rho-GDI2: Rho GDP-dissociation inhibitor 2

RIP: Receptor-interacting protein kinase

RIP1: Receptor-interacting protein1

RNA: Ribonucleic acid

ROS: Reactive oxygen species

RUNX3: Runt-related transcription factor 3

Rxra: Retinoid X receptor alpha, nuclear receptor subfamily 2, group B, member 1, NR2B1

S1P: Sphingosine-1-phosphate

S. japonicum : Schistosoma japonicum

S. mansoni : Schistosoma mansoni

SalC: Salvianolic acid C

SLUG: Zinc finger protein SNAI2

Snail: Zinc finger protein SNAI1

SOCS2: Suppressor of cytokine signaling 2

SOX: [Sex-determining region Y (Sry) box-containing] transcription factor family

SP1: Specificity protein 1

SP100030: T-cell–specific inhibitor 2-chloro-4-(trifluoromethyl)pyrimidine-5-N-(39,59-bis [trifluoromethyl]phenyl)-carboxamide

SphK: Sphingosine kinase isoform

STAT1: Signal transducer and activator of transcription 1

STAT3: Signal transducer and activator of transcription 3

STING: Stimulator of interferon genes

TF: Transcription factor

TCGA: The Cancer Genome Atlas

TGFβ: Transforming growth factor beta

TGF-β1: Transforming growth factor beta 1

Th17: T helper 17 cells

TLR: Toll-like receptor

TLR3: Toll like receptor 3

TLR4: Toll like receptor 4

TLR7: Toll like receptor 7

TLR8: Toll like receptor 8

TMS1: Target of methylation-mediated silencing

TNFα: Tumor necrosis factor alpha

TNFR1: Tumor necrosis factor receptor 1

TNFRSF6: Tumor necrosis factor receptor superfamily member 6

TRADD: TNF receptor type 1-associated DEATH domain protein

TRAF1: Tumor necrosis factor (TNFα) receptor-associated factor 1

TRAF2: Tumor necrosis factor (TNFα) receptor-associated factor 2

TRAF6: Tumor necrosis factor (TNFα) receptor-associated factor 6

TRANCE: TNF receptor superfamily

TRPV1: Transient receptor potential vanilloid 1

TTF: Transcription termination factor

TWEAK: TNF-like weak inducer of apoptosis

TWIST: Twist-related protein 1

TXA2: Thromboxane A2, (Z)-7-[(1S,2S,3R,5S)-3-[(E,3S)-3-hydroxyoct-1-enyl]-4,6-dioxabicyclo[3.1.1]heptan-2-yl]hept-5-enoic acid

UHRF1: Ubiquitin ligase ubiquitin-like, containing PHD and RING finger domains 1

uPA: Urokinase plasminogen activator

UPM: Urban particulate matter

UPR: Unfolded protein response

VCAM-1: Vascular cell adhesion molecule 1, vascular cell adhesion protein 1, cluster of differentiation 106 (CD106)

VCP: Valosin-containing protein (CDC48)

VEGF: Vascular endothelial growth factor

VDUP1: Vitamin D(3) upregulated protein 1

ZEB1: Zinc finger E-box-binding homeobox 1

ZO-1: Zonula occludens-1

Acknowledgments

The manuscripts of this Special Issue were supported by the Theodor-Billroth-Academy® (TBA®) and INCORE, (International Consortium of Research Excellence) of the (TBA®). We express our gratitude to the discussions on the web group of the Theodor-Billroth-Academy® (TBA®) on LinkedIn, the exchange with scientists at Researchgate.com, as well as personal exchanges with distinguished colleagues who stimulated our thinking all named individually earlier in publications – we thank each one.

Conflict of interest

The author reports the following conflict of interest: Björn LDM Brücher is Editor-in-Chief in Life Sciences-Medicine of 4open by EDP Sciences. Florian Lang is Editor-in-Chief of Cellular Physiology and Biochemistry. Ijaz S. Jamall is Senior Editorial Board member in Life Sciences-Medicine of 4open by EDP Sciences. The authors, of their own initiative, suggested to the Managing Editorial to perform a transparent peer-review of their submittals. Neither author took any action to influence the standard submission and peer-review process, and report no conflict of interest. The authors alone are responsible for the content and writing of the manuscript of this Special Issue. This manuscript contains original material that has not previously been published. All authors contributed on its contents and approved the different manuscript.

References

  1. Brivanlou AH, Darnell JE (2002), Signal transduction and the control of gene expression. Science 295, 5556, 813–818. https://doi.org/10.1126/science.1066355. [Google Scholar]
  2. Iwai K (2012), Diverse ubiquitin signaling in NF-κB activation. Trend Cell Biol 22, 7, 355–364. https://doi.org/10.1016/j.tcb.2012.04.001. [CrossRef] [Google Scholar]
  3. Sen R, Baltimore D (1986), Multiple nuclear factors interact with the immunoglobulin enhancer sequences. Cell 46, 5, 705–716. https://doi.org/10.1016/0092-8674(86)90346-6. [CrossRef] [PubMed] [Google Scholar]
  4. Baeuerle PA, Baltimore D (1988), Activation of DNA-binding activity in an apparently cytoplasmic precursor of the NF-kappa B transcription factor. Cell 53, 2, 211–217. [CrossRef] [PubMed] [Google Scholar]
  5. Baeuerle PA, Baltimore D (1988), I kappa B: a specific inhibitor of the NF-kappa B transcription factor. Science 242, 4878, 540–546. [Google Scholar]
  6. Baltimore D (2009), Discovering NF-κB. Cold Spring Harb Perspect Biol 1, 1, a000026. https://doi.org/10.1101/cshperspect.a000026. [PubMed] [Google Scholar]
  7. Ghosh S, Hayden MS (2012), Celebrating 25 years of NF-κB research. Immunol Rev 246, 1, 5–13. https://doi.org/10.1111/j.1600-065X.2012.01111.x. [CrossRef] [PubMed] [Google Scholar]
  8. Gilmore TD (2006), Introduction to NF-kappaB: players, pathways, perspectives. Oncogene 25, 51, 6680–6684. [Google Scholar]
  9. Gilmore TD (1990), NF-kappa B, KBF1, dorsal, and related matters. Cell 62, 5, 841–843. [CrossRef] [PubMed] [Google Scholar]
  10. Shishodia S, Aggarwal BB (2004), Cyclooxygenase (COX)-2 inhibitor celecoxib abrogates activation of cigarette smoke-induced nuclear factor (NF)-kappaB by suppressing activation of IkappaBalpha kinase in human non-small cell lung carcinoma: correlation with suppression of cyclin D1, COX-2, and matrix metalloproteinase-9. Cancer Res 64, 14, 5004–5012. https://doi.org/10.1158/0008-5472.CAN-04-0206 [Google Scholar]
  11. Luo JL, Kamata H, Karin M (2005), IKK/NF-kappaB signaling: balancing life and death–a new approach to cancer therapy. J Clin Invest 115, 10, 2625–2632. [CrossRef] [PubMed] [Google Scholar]
  12. Hayden MS, Gosh S (2011), NF-κB in immunobiology. Cell Res 21, 2, 223–244. https://doi.org/10.1038/cr.2011.13. [CrossRef] [PubMed] [Google Scholar]
  13. Kuwata H, Matsumoto M, Atarashi K, Morishita H, Hirotani T, Koga R, Takeda K (2006), IkappaBNS inhibits induction of a subset of Toll-like receptor-dependent genes and limits inflammation. Immunity 24, 1, 41–51. https://doi.org/10.1016/j.immuni.2005.11.004. [CrossRef] [PubMed] [Google Scholar]
  14. Papanikolaou E, Paruzynski A, Kasampalidis I, Deichmann A, Stamateris E, Schmidt M, von Kalle C, Anagnou NP (2015), Cell cycle status of CD34(+) hemopoietic stem cells determines lentiviral integration in actively transcribed and development-related genes. Mol Ther 23, 4, 683–696. https://doi.org/10.1038/mt.2014.246. [CrossRef] [PubMed] [Google Scholar]
  15. Bechill J, Muller WJ (2014), Herpesvirus entry mediator (HVEM) attenuates signals mediated by the lymphotoxin β receptor (LTβR) in human cells stimulated by the shared ligand LIGHT. Mol Immunol 62, 1, 96–103. https://doi.org/10.1016/j.molimm.2014.06.013. [Google Scholar]
  16. Qiao JT, Cui C, Qing L, Wang LS, He TY, Yan F, Liu FQ, Shen YH, Hou XG, Chen L (2018), Activation of the STING-IRF3 pathway promotes hepatocyte inflammation, apoptosis and induces metabolic disorders in nonalcoholic fatty liver disease. Metabolism 81, 13–24. https://doi.org/10.1016/j.metabol.2017.09.010. [CrossRef] [PubMed] [Google Scholar]
  17. Ni MM, Xu T, Wang YR, He YH, Zhou Q, Huang C, Meng XM, Li J (2016), Inhibition of IRF3 expression reduces TGF-β1-induced proliferation of hepatic stellate cells. J Physiol Biochem 72, 1, 9–23. https://doi.org/10.1007/s13105-015-0452-6. [Google Scholar]
  18. Pontoriero M, Fiume G, Vecchio E, de Laurentiis A, Albano F, Iaccino E, Mimmi S, Pisano A, Agosti V, Giovannone E, Altobelli A, Caiazza C, Mallardo M, Scala G, Quinto I (2019 Mar 19), Activation of NF-κB in B cell receptor signaling through Bruton’s tyrosine kinase-dependent phosphorylation of IκB-α. J Mol Med (Berl). 97, 5, 675–690. https://doi.org/10.1007/s00109-019-01777-x. [CrossRef] [PubMed] [Google Scholar]
  19. Yue C, Niu M, Shan QQ, Zhou T, Tu Y, Xie P, Hua L, Yu R, Liu X (2017), High expression of Bruton’s tyrosine kinase (BTK) is required for EGFR-induced NF-κB activation and predicts poor prognosis in human glioma. J Exp Clin Cancer Res 36, 1, 132. https://doi.org/10.1186/s13046-017-0600-7. [CrossRef] [PubMed] [Google Scholar]
  20. Li L, Tong M, Zhao YT, He Y, Zhou HY, Zhang GF, Zhang YJ (2018), Membrane translocation of Bruton kinase in multiple myeloma cells is associated with osteoclastogenic phenotype in bone metastatic lesions. Medicine (Baltimore) 97, 2, e9482. https://doi.org/10.1097/MD.0000000000009482. [CrossRef] [PubMed] [Google Scholar]
  21. Page TH, Urbaniak AM, Espirito Santo AI, Danks L, Smallie T, Williams LM, Horwood NJ (2018), Bruton’s tyrosine kinase regulates TLR7/8-induced TNF transcription via nuclear factor-κB recruitment. Biochem Biophys Res Commun 499, 2, 260–266. https://doi.org/10.1016/j.bbrc.2018.03.140. [Google Scholar]
  22. Pandey MK, Gowda K, Sung SS, Abraham T, Budak-Alpdogan T, Talamo G, Dovat S, Amin S (2017), A novel dual inhibitor of microtubule and Bruton’s tyrosine kinase inhibits survival of multiple myeloma and osteoclastogenesis. Exp Hematol 53, 31–42. https://doi.org/10.1016/j.exphem.2017.06.003. [CrossRef] [PubMed] [Google Scholar]
  23. Senol Tuncay S, Okyay P, Bardakci F (2010), Identification of NF-kappaB1 and NF-kappaBIAlpha polymorphisms using PCR-RFLP assay in a Turkish population. Biochem Genet 48, 1–2, 104–112. https://doi.org/10.1007/s10528-009-9302-y. [CrossRef] [PubMed] [Google Scholar]
  24. Wang S, Tian L, Zeng Z, Zhang M, Wu K, Chen M, Fan D, Hu P, Sung JJ, Yu J (2010), IκBα polymorphism at promoter region (rs2233408) influences the susceptibility of gastric cancer in Chinese. BMC Gastroenterol 10, 15. https://doi.org/10.1186/1471-230X-10-15. [CrossRef] [PubMed] [Google Scholar]
  25. Lim B, Ju H, Kim M, Kang C (2011), Increased genetic susceptibility to intestinal-type gastric cancer is associated with increased activity of the RUNX3 distal promoter. Cancer 117, 22, 5161–5171. https://doi.org/10.1002/cncr.26161. [CrossRef] [PubMed] [Google Scholar]
  26. Wang S, Zhang M, Zeng Z, Tian L, Wu K, Chu J, Fan D, Hu P, Sung JJ, Yu J (2011), IκBα polymorphisms were associated with increased risk of gastric cancer in a southern Chinese population: a case-control study. Life Sci 88, 17–18, 792–797. https://doi.org/10.1016/j.lfs.2011.02.016. [CrossRef] [PubMed] [Google Scholar]
  27. Lu R, Gao X, Chen Y, Ni J, Yu Y, Li S, Guo L (2012), Association of an NFKB1 intron SNP (rs4648068) with gastric cancer patients in the Han Chinese population. BMC Gastroenterol 12, 87. https://doi.org/10.1186/1471-230X-12-87. [CrossRef] [PubMed] [Google Scholar]
  28. Lo SS, Chen JH, Wu CW, Lui WY (2009), Functional polymorphism of NFKB1 promoter may correlate to the susceptibility of gastric cancer in aged patients. Surgery 145, 3, 280–285. https://doi.org/10.1016/j.surg.2008.11.001. [Google Scholar]
  29. Levinger L, Varshavsky A (1982), Selective arrangement of ubiquitinated and D1 protein-containing nucleosomes within the Drosophila genome. Cell 28, 2, 375–385. [CrossRef] [PubMed] [Google Scholar]
  30. Hochstrasser M (1998), There’s the rub: a novel ubiquitin-like modification linked to cell cycle regulation. Genes Dev 12, 7, 901–907. [Google Scholar]
  31. Chau V, Tobias JW, Bachmair A, Marriott D, Ecker DJ, Gonda DK, Varshavsky A (1989), A multiubiquitin chain is confined to specific lysine in a targeted short-lived protein. Science 243, 4898, 1576–1583. [Google Scholar]
  32. Pickart CM (2001), Mechanisms underlying ubiquitination. Annu Rev Biochem 70, 503–533. https://doi.org/10.1146/annurev.biochem.70.1.503. [CrossRef] [PubMed] [Google Scholar]
  33. Glickman MH, Ciechanover A (2002), The ubiquitin-proteasome proteolytic pathway: destruction for the sake of construction. Physiol Rev 82, 2, 373–428. https://doi.org/10.1152/physrev.00027.2001. [Google Scholar]
  34. Schnell JD, Hicke L (2003), Non-traditional functions of ubiquitin and ubiquitin-binding proteins. J Biol Chem 278, 38, 35857–35860. [PubMed] [Google Scholar]
  35. Pickart CM, Eddins MJ (2004), Ubiquitin: structures, functions, mechanisms. Biochim Biophys Acta 1695, 1–3, 55–72. https://doi.org/10.1016/j.bbamcr.2004.09.019. [CrossRef] [PubMed] [Google Scholar]
  36. Ciehanover A, Hod Y, Hershko A (1978), A heat-stable polypeptide component of an ATP-dependent proteolytic system from reticulocytes. Biochem Biophys Res Commun 81, 4, 1100–1105. [Google Scholar]
  37. Hershko A, Ciechanover A, Rose IA (1979), Resolution of the ATP-dependent proteolytic system from reticulocytes: a component that interacts with ATP. Proc Natl Acad Sci USA 76, 7, 3107–3110. [CrossRef] [Google Scholar]
  38. Ciechanover A, Heller H, Elias S, Haas AL, Hershko A (1980), ATP-dependent conjugation of reticulocyte proteins with the polypeptide required for protein degradation. Proc Natl Acad Sci USA 77, 3, 1365–1368. [CrossRef] [Google Scholar]
  39. Hershko A, Ciechanover A (1982), Mechanisms of intracellular protein breakdown. Annu Rev Biochem 51, 335–364. https://doi.org/10.1146/annurev.bi.51.070182.002003. [CrossRef] [PubMed] [Google Scholar]
  40. Kanarek N, London N, Schueler-Furman O, Ben-Neriah Y (2010), Ubiquitination and degradation of the inhibitors of NF-kappaB. Cold Spring Harb Perspect Biol 2, 2, a000166. https://doi.org/10.1101/cshperspect.a000166. [PubMed] [Google Scholar]
  41. Liao WT, Chang KL, Yu CL, Chen GS, Chang LW, Yu HS (2004), Arsenic induces human keratinocyte apoptosis by the FAS/FAS ligand pathway, which correlates with alterations in nuclear factor-kappa B and activator protein-1 activity. J Invest Dermatol 122, 1, 125–129. [CrossRef] [PubMed] [Google Scholar]
  42. Poligone B, Hayden MS, Chen L, Pentland AP, Jimi E, Ghosh S (2013), A role for NF-κB activity in skin hyperplasia and the development of keratoacanthomata in mice. PLoS One 8, 8, e71887. https://doi.org/10.1371/journal.pone.0071887. [Google Scholar]
  43. Banerjee S, Bueso-Ramos C, Aggarwal BB (2002), Suppression of 7,12-dimethylbenz(a)anthracene-induced mammary carcinogenesis in rats by resveratrol: role of nuclear factor-kappaB, cyclooxygenase 2, and matrix metalloprotease 9. Cancer Res 62, 17, 4945–4954. [Google Scholar]
  44. Ni WF, Tsai CH, Yang SF, Chang YC (2007), Elevated expression of NF-kappaB in oral submucous fibrosis–evidence for NF-kappaB induction by safrole in human buccal mucosal fibroblasts. Oral Oncol 43, 6, 557–562. [CrossRef] [PubMed] [Google Scholar]
  45. Kamperos G, Nikitakis N, Sfakianou A, Avgoustidis D, Sklavounou-Andrikopoulou A (2016), Expression of NF-κB and IL-6 in oral precancerous and cancerous lesions: an immunohistochemical study. Med Oral Patol Oral Cir Bucal 21, 1, e6–e13. [Google Scholar]
  46. Huo X, Zhang X, Yu C, Cheng E, Zhang Q, Dunbar KB, Pham TH, Lynch JP, Wang DH, Bresalier RS, Spechler SJ, Souza RF (2018), Aspirin prevents NF-κB activation and CDX2 expression stimulated by acid and bile salts in oesophageal squamous cells of patients with Barrett’s oesophagus. Gut 67, 4, 606–615. https://doi.org/10.1136/gutjnl-2016-313584. [PubMed] [Google Scholar]
  47. Greenspan EJ, Madigan JP, Boardman LA, Rosenberg DW (2011), Ibuprofen inhibits activation of nuclear {beta}-catenin in human colon adenomas and induces the phosphorylation of GSK-3{beta}. Cancer Prev Res (Phila) 4, 1, 161–171. https://doi.org/10.1158/1940-6207.CAPR-10-0021. [CrossRef] [PubMed] [Google Scholar]
  48. Hasel C, Bhanot UK, Heydrich R, Sträter J, Möller P (2005), Parenchymal regression in chronic pancreatitis spares islets reprogrammed for the expression of NFkappaB and IAPs. Lab Invest 85, 10, 1263–1275. https://doi.org/10.1038/labinvest.3700323. [PubMed] [Google Scholar]
  49. Greten FR, Eckmann L, Greten TF, Park JM, Li ZW, Egan LJ, Kagnoff MF, Karin M (2004), IKKbeta links inflammation and tumorigenesis in a mouse model of colitis-associated cancer. Cell 118, 3, 285–296. https://doi.org/10.1016/j.cell.2004.07.013. [CrossRef] [PubMed] [Google Scholar]
  50. Karban AS, Okazaki T, Panhuysen CI, Gallegos T, Potter JJ, Bailey-Wilson JE, Silverberg MS, Duerr RH, Cho JH, Gregersen PK, Wu Y, Achkar JP, Dassopoulos T, Mezey E, Bayless TM, Nouvet FJ, Brant SR (2004), Functional annotation of a novel NFKB1 promoter polymorphism that increases risk for ulcerative colitis. Hum Mol Genet 13, 1, 35–45. https://doi.org/10.1093/hmg/ddh008. [CrossRef] [PubMed] [Google Scholar]
  51. Hiratsuka S, Watanabe A, Aburatani H, Maru Y (2006), Tumour-mediated upregulation of chemoattractants and recruitment of myeloid cells predetermines lung metastasis. Nat Cell Biol 8, 12, 1369–1375. https://doi.org/10.1038/ncb1507. [CrossRef] [PubMed] [Google Scholar]
  52. Hardwick JC, van den Brink GR, Offerhaus GJ, van Deventer SJ, Peppelenbosch MP (2001), NF-kappaB, p38 MAPK and JNK are highly expressed and active in the stroma of human colonic adenomatous polyps. Oncogene 20, 7, 819–827. https://doi.org/10.1038/sj.onc.1204162. [Google Scholar]
  53. Dejardin E, Deregowski V, Chapelier M, Jacobs N, Gielen J, Merville MP, Bours V (1999), Regulation of NF-kappaB activity by I kappaB-related proteins in adenocarcinoma cells. Oncogene 18, 16, 2567–2577. https://doi.org/10.1038/sj.onc.1202599. [Google Scholar]
  54. Sovak MA, Bellas RE, Kim DW, Zanieski GJ, Rogers AE, Traish AM, Sonenshein GE (1997), Aberrant nuclear factor-kappaB/Rel expression and the pathogenesis of breast cancer. J Clin Invest 100, 12, 2952–2960. https://doi.org/10.1172/JCI119848. [CrossRef] [PubMed] [Google Scholar]
  55. Nakshatri H, Bhat-Nakshatri P, Martin DA, Goulet RJ Jr, Sledge GW Jr (1997), Constitutive activation of NF-kappaB during progression of breast cancer to hormone-independent growth. Mol Cell Biol 17, 7, 3629–3639. [PubMed] [Google Scholar]
  56. Bhat-Nakshatri P, Newton TR, Goulet R Jr, Nakshatri H (1998), NF-kappaB activation and interleukin 6 production in fibroblasts by estrogen receptor-negative breast cancer cell-derived interleukin 1alpha. Proc Natl Acad Sci USA 95, 12, 6971–6976. [CrossRef] [Google Scholar]
  57. Biswas DK, Dai SC, Cruz A, Weiser B, Graner E, Pardee AB (2001), The nuclear factor kappa B (NF-kappa B): a potential therapeutic target for estrogen receptor negative breast cancers. Proc Natl Acad Sci USA 98, 18, 10386–10391. https://doi.org/10.1073/pnas.151257998. [CrossRef] [Google Scholar]
  58. Bachmeier B, Nerlich AG, Iancu CM, Cilli M, Schleicher E, Vené R, Dell’Eva R, Jochum M, Albini A, Pfeffer U (2007), The chemopreventive polyphenol Curcumin prevents hematogenous breast cancer metastases in immunodeficient mice. Cell Physiol Biochem 19, 1–4, 137–152. https://doi.org/10.1159/000099202. [CrossRef] [PubMed] [Google Scholar]
  59. Kim Y, Kang H, Jang SW, Ko J (2011), Celastrol inhibits breast cancer cell invasion via suppression of NF-ĸB-mediated matrix metalloproteinase-9 expression. Cell Physiol Biochem 28, 2, 175–184. https://doi.org/10.1159/000331729. [CrossRef] [PubMed] [Google Scholar]
  60. Rengarajan T, Nandakumar N, Rajendran P, Ganesh MK, Balasubramanian MP, Nishigaki I (2015), D-pinitol mitigates tumor growth by modulating interleukins and hormones and induces apoptosis in rat breast carcinogenesis through inhibition of NF-κB. J Physiol Biochem 71, 2, 191–204. https://doi.org/10.1007/s13105-015-0397-9. [Google Scholar]
  61. Bours V, Dejardin E, Goujon-Letawe F, Merville MP, Castronovo V (1994), The NF-kappa B transcription factor and cancer: high expression of NF-kappa B- and I kappa B-related proteins in tumor cell lines. Biochem Pharmacol 47, 1, 145–149. [CrossRef] [PubMed] [Google Scholar]
  62. Hagemann T, Wilson J, Kulbe H, Li NF, Leinster DA, Charles K, Klemm F, Pukrop T, Binder C, Balkwill FR (2005), Macrophages induce invasiveness of epithelial cancer cells via NF-kappa B and JNK. J Immunol 175, 2, 1197–1205. [CrossRef] [PubMed] [Google Scholar]
  63. Guo Y, Liao Y, Jia C, Ren J, Wang J, Li T (2013), MicroRNA-182 promotes tumor cell growth by targeting transcription elongation factor A-like 7 in endometrial carcinoma. Cell Physiol Biochem 32, 3, 581–590. https://doi.org/10.1159/000354462. [CrossRef] [PubMed] [Google Scholar]
  64. Chen CD, Sawyers CL (2002), NF-kappa B activates prostate-specific antigen expression and is upregulated in androgen-independent prostate cancer. Mol Cell Biol 22, 8, 2862–2870. [PubMed] [Google Scholar]
  65. Papadopoulou N, Charalampopoulos I, Anagnostopoulou V, Konstantinidis G, Föller M, Gravanis A, Alevizopoulos K, Lang F, Stournaras C (2008), Membrane androgen receptor activation triggers down-regulation of PI-3K/Akt/NF-kappaB activity and induces apoptotic responses via Bad, FasL and caspase-3 in DU145 prostate cancer cells. Mol Cancer 7, 88. https://doi.org/10.1186/1476-4598-7-88. [PubMed] [Google Scholar]
  66. Fu W, Yao J, Huang Y, Li Q, Li W, Chen Z, He F, Zhou Z, Yan J (2014), LXR agonist regulates the carcinogenesis of PCa via the SOCS3 pathway. Cell Physiol Biochem 33, 1, 195–204. https://doi.org/10.1159/000356662. [CrossRef] [PubMed] [Google Scholar]
  67. Visconti R, Cerutti J, Battista S, Fedele M, Trapasso F, Zeki K, Miano MP, de Nigris F, Casalino L, Curcio F, Santoro M, Fusco A (1997), Expression of the neoplastic phenotype by human thyroid carcinoma cell lines requires NFkappaB p65 protein expression. Oncogene 15, 16, 1987–1994. https://doi.org/10.1038/sj.onc.1201373. [Google Scholar]
  68. Ma Y, Wang Q, Liu F, Ma X, Wu L, Guo F, Zhao S, Huang F, Qin G (2018), KLF5 promotes the tumorigenesis and metastatic potential of thyroid cancer cells through the NF-κB signaling pathway. Oncol Rep 40, 5, 2608–2618. https://doi.org/10.3892/or.2018.6687. [PubMed] [Google Scholar]
  69. Yan C, Su H, Song X, Cao H, Kong L, Cui W (2018), Smad ubiquitination regulatory factor 1 (Smurf1) promotes thyroid cancer cell proliferation and migration via ubiquitin-dependent degradation of kisspeptin-1. Cell Physiol Biochem 49, 5, 2047–2059. https://doi.org/10.1159/000493715. [CrossRef] [PubMed] [Google Scholar]
  70. Zhang Z, Dong T, Fu Y, Zhou W, Tian X, Chen G, Liu S (2018 Sep 1), MMP-11 promotes papillary thyroid cell proliferation and invasion via the NF-κB pathway. J Cell Biochem 120, 2, 1860–1868. https://doi.org/10.1002/jcb.27500. [Google Scholar]
  71. Flodström M, Welsh N, Eizirik DL (1996), Cytokines activate the nuclear factor kappa B (NF-kappa B) and induce nitric oxide production in human pancreatic islets. FEBS Lett 385, 1–2, 4–6. [CrossRef] [PubMed] [Google Scholar]
  72. Wang W, Abbruzzese JL, Evans DB, Larry L, Cleary KR, Chiao PJ (1999), The nuclear factor-kappa B RelA transcription factor is constitutively activated in human pancreatic adenocarcinoma cells. Clin Cancer Res 5, 1, 119–127. [PubMed] [Google Scholar]
  73. Wang W, Abbruzzese JL, Evans DB, Chiao PJ (1999), Overexpression of urokinase-type plasminogen activator in pancreatic adenocarcinoma is regulated by constitutively activated RelA. Oncogene 18, 32, 4554–4563. https://doi.org/10.1038/sj.onc.1202833. [Google Scholar]
  74. Takaya H, Andoh A, Shimada M, Hata K, Fujiyama Y, Bamba T (2000), The expression of chemokine genes correlates with nuclear factor-kappaB activation in human pancreatic cancer cell lines. Pancreas 21, 1, 32–40. [CrossRef] [PubMed] [Google Scholar]
  75. van Hogerlinden M, Rozell BL, Ahrlund-Richter L, Toftgard R (1999), Squamous cell carcinomas and increased apoptosis in skin with inhibited Rel/nuclear factor-kappaB signaling. Cancer Res 59, 3299–3303. [Google Scholar]
  76. Dajee M, Lazarov M, Zhang JY, Cai T, Green CL, Russell AJ, Marinkovich MP, Tao S, Lin Q, Kubo Y, Khavari PA (2003), NF-kappaB blockade and oncogenic Ras trigger invasive human epidermal neoplasia. Nature 421, 6923, 639–643. [CrossRef] [PubMed] [Google Scholar]
  77. Huang C, Huang Y, Li J, Hu W, Aziz R, Tang MS, Sun N, Cassady J, Stoner GD (2002), Inhibition of benzo(a)pyrene diol-epoxide-induced transactivation of activated protein 1 and nuclear factor kappaB by black raspberry extracts. Cancer Res 62, 23, 6857–6863. PMID: 12460899. [Google Scholar]
  78. Jenkins GJ, Harries K, Doak SH, Wilmes A, Griffiths AP, Baxter JN, Parry JM (2004), The bile acid deoxycholic acid (DCA) at neutral pH activates NF-kappaB and induces IL-8 expression in oesophageal cells in vitro. Carcinogenesis 25, 3, 317–323. https://doi.org/10.1093/carcin/bgh032. [CrossRef] [PubMed] [Google Scholar]
  79. Li X, Chen D, Li M, Gao X, Shi G, Zhao H (2018), Plantamajoside inhibits lipopolysaccharide-induced epithelial-mesenchymal transition through suppressing the NF-κB/IL-6 signaling in esophageal squamous cell carcinoma cells. Biomed Pharmacother 102, 1045–1051. https://doi.org/10.1016/j.biopha.2018.03.171. [CrossRef] [PubMed] [Google Scholar]
  80. Zhou Y, Xia L, Liu Q, Wang H, Lin J, Oyang L, Chen X, Luo X, Tan S, Tian Y, Su M, Wang Y, Chen P, Wu Y, Wang H, Liao Q (2018), Induction of pro-inflammatory response via activated macrophage-mediated NF-κB and STAT3 pathways in gastric cancer cells. Cell Physiol Biochem 47, 4, 1399–1410. https://doi.org/10.1159/000490829. [CrossRef] [PubMed] [Google Scholar]
  81. Lind DS, Hochwald SN, Malaty J, Rekkas S, Hebig P, Mishra G, Moldawer LL, Copeland EM 3rd, Mackay S (2001), Nuclear factor-kappa B is upregulated in colorectal cancer. Surgery 130, 2, 363–369. [Google Scholar]
  82. Ahn DH, Crawley SC, Hokari R, Kato S, Yang SC, Li JD, Kim YS (2005), TNF-alpha activates MUC2 transcription via NF-kappaB but inhibits via JNK activation. Cell Physiol Biochem 15, 1–4, 29–40. https://doi.org/10.1159/000083636. [CrossRef] [PubMed] [Google Scholar]
  83. Clemo NK, Collard TJ, Southern SL, Edwards KD, Moorghen M, Packham G, Hague A, Paraskeva C, Williams AC (2008), BAG-1 is up-regulated in colorectal tumour progression and promotes colorectal tumour cell survival through increased NF-kappaB activity. Carcinogenesis 29, 4, 849–857. https://doi.org/10.1093/carcin/bgn004. [CrossRef] [PubMed] [Google Scholar]
  84. Tai DI, Tsai SL, Chang YH, Huang SN, Chen TC, Chang KS, Liaw YF (2000), Constitutive activation of nuclear factor kappaB in hepatocellular carcinoma. Cancer 89, 11, 2274–2281. [CrossRef] [PubMed] [Google Scholar]
  85. Tietze MK, Wuestefeld T, Paul Y, Zender L, Trautwein C, Manns MP, Kubicka S (2000), IkappaBalpha gene therapy in tumor necrosis factor-alpha- and chemotherapy-mediated apoptosis of hepatocellular carcinomas. Cancer Gene Ther 7, 10, 1315–1323. [CrossRef] [PubMed] [Google Scholar]
  86. Glauert HP, Eyigor A, Tharappel JC, Cooper S, Lee EY, Spear BT (2006), Inhibition of hepatocarcinogenesis by the deletion of the p50 subunit of NF-kappaB in mice administered the peroxisome proliferator Wy-14,643. Toxicol Sci 90, 2, 331–336. https://doi.org/10.1093/toxsci/kfj116. [CrossRef] [PubMed] [Google Scholar]
  87. Li X, Liu J, Park JK, Hamilton TA, Rayman P, Klein E, Edinger M, Tubbs R, Bukowski R, Finke J (1994), T cells from renal cell carcinoma patients exhibit an abnormal pattern of kappa B-specific DNA-binding activity: a preliminary report. Cancer Res 54, 20, 5424–5429. [Google Scholar]
  88. Oya M, Ohtsubo M, Takayanagi A, Tachibana M, Shimizu N, Murai M (2001), Constitutive activation of nuclear factor-kappaB prevents TRAIL-induced apoptosis in renal cancer cells. Oncogene 20, 29, 3888–3896. https://doi.org/10.1038/sj.onc.1204525. [Google Scholar]
  89. Oya M, Takayanagi A, Horiguchi A, Mizuno R, Ohtsubo M, Marumo K, Shimizu N, Murai M (2003), Increased nuclear factor-kappa B activation is related to the tumor development of renal cell carcinoma. Carcinogenesis 24, 3, 377–384. [CrossRef] [PubMed] [Google Scholar]
  90. Sumitomo M, Tachibana M, Ozu C, Asakura H, Murai M, Hayakawa M, Nakamura H, Takayanagi A, Shimizu N (1999), Induction of apoptosis of cytokine-producing bladder cancer cells by adenovirus-mediated IkappaBalpha over expression. Hum Gene Ther 10, 1, 37–47. https://doi.org/10.1089/10430349950019174. [Google Scholar]
  91. Naumovski L, Utz PJ, Bergstrom SK, Morgan R, Molina A, Toole JJ, Glader BE, McFall P, Weiss LM, Warnke R (1989), SUP-HD1: a new Hodgkin’s disease-derived cell line with lymphoid features produces interferon-gamma. Blood 74, 8, 2733–2742. [Google Scholar]
  92. Bargou RC, Leng C, Krappmann D, Emmerich F, Mapara MY, Bommert K, Royer HD, Scheidereit C, Dörken B (1996), High-level nuclear NF-kappa B and Oct-2 is a common feature of cultured Hodgkin/Reed-Sternberg cells. Blood 87, 10, 4340–4347. [Google Scholar]
  93. Boland MP, Foster SJ, O’Neill LA (1997), Daunorubicin activates NFkappaB and induces kappaB-dependent gene expression in HL-60 promyelocytic and Jurkat T lymphoma cells. J Biol Chem 272, 20, 12952–12960. [PubMed] [Google Scholar]
  94. Sánchez-Beato M, Sánchez E, García JF, Pérez-Rosado A, Montoya MC, Fraga M, Artiga MJ, Navarrete M, Abraira V, Morente M, Esteller M, Koseki H, Vidal M, Piris MA (2004), Abnormal PcG protein expression in Hodgkin’s lymphoma. Relation with E2F6 and NFkappaB transcription factors. J Pathol 204, 5, 528–537. https://doi.org/10.1002/path.1661. [PubMed] [Google Scholar]
  95. Keller SA, Hernandez-Hopkins D, Vider J, Ponomarev V, Hyjek E, Schattner EJ, Cesarman E (2006), NF-kappaB is essential for the progression of KSHV- and EBV-infected lymphomas in vivo. Blood 107, 8, 3295–3302. https://doi.org/10.1182/blood-2005-07-2730. [Google Scholar]
  96. Kelley DE, Pollok BA, Atchison ML, Perry RP (1988), The coupling between enhancer activity and hypomethylation of kappa immunoglobulin genes is developmentally regulated. Mol Cell Biol 8, 2, 930–937. [PubMed] [Google Scholar]
  97. Leung K, Nabel GJ (1988), HTLV-1 transactivator induces interleukin-2 receptor expression through an NF-kappa B-like factor. Nature 333, 6175, 776–778. [CrossRef] [PubMed] [Google Scholar]
  98. Duyao MP, Kessler DJ, Spicer DB, Bartholomew C, Cleveland JL, Siekevitz M, Sonenshein GE (1992), Transactivation of the c-myc promoter by human T cell leukemia virus type 1 tax is mediated by NF kappa B. J Biol Chem 267, 23, 16288–16291. [PubMed] [Google Scholar]
  99. Chauhan D, Uchiyama H, Akbarali Y, Urashima M, Yamamoto K, Libermann TA, Anderson KC (1996), Multiple myeloma cell adhesion-induced interleukin-6 expression in bone marrow stromal cells involves activation of NF-kappa B. Blood 87, 3, 1104–1112. PMID: 8562936. [Google Scholar]
  100. Costes V, Portier M, Lu ZY, Rossi JF, Bataille R, Klein B (1998), Interleukin-1 in multiple myeloma: producer cells and their role in the control of IL-6 production. Br J Haematol 103, 4, 1152–1160. [CrossRef] [PubMed] [Google Scholar]
  101. Feinman R, Siegel DS, Berenson J (2004), Regulation of NF-kB in multiple myeloma: therapeutic implications. Clin Adv Hematol Oncol 2, 3, 162–166. [PubMed] [Google Scholar]
  102. Templin J, Atanackovic D, Hasche D, Radhakrishnan SV, Luetkens T (2017), Oscillating expression of interleukin-16 in multiple myeloma is associated with proliferation, clonogenic growth, and PI3K/NFKB/MAPK activation. Oncotarget 8, 30, 49253–49263. https://doi.org/10.18632/oncotarget.17534. [CrossRef] [PubMed] [Google Scholar]
  103. Xie TX, Aldape KD, Gong W, Kanzawa T, Suki D, Kondo S, Lang F, Ali-Osman F, Sawaya R, Huang S (2008), Aberrant NF-kappaB activity is critical in focal necrosis formation of human glioblastoma by regulation of the expression of tissue factor. Int J Oncol 33, 1, 5–15. [Google Scholar]
  104. Bhat KPL, Balasubramaniyan V, Vaillant B, Ezhilarasan R, Hummelink K, Hollingsworth F, Wani K, Heathcock L, James JD, Goodman LD, Conroy S, Long L, Lelic N, Wang S, Gumin J, Raj D, Kodama Y, Raghunathan A, Olar A, Joshi K, Pelloski CE, Heimberger A, Kim SH, Cahill DP, Rao G, Den Dunnen WFA, Boddeke HWGM, Phillips HS, Nakano I, Lang FF, Colman H, Sulman EP, Aldape K (2013), Mesenchymal differentiation mediated by NF-κB promotes radiation resistance in glioblastoma. Cancer Cell 24, 3, 331–346. https://doi.org/10.1016/j.ccr.2013.08.001. [CrossRef] [PubMed] [Google Scholar]
  105. Jiang C, Zhu Y, Zhou Z, Gumin J, Bengtsson L, Wu W, Songyang Z, Lang FF, Lin X (2017), TMEM43/LUMA is a key signaling component mediating EGFR-induced NF-κB activation and tumor progression. Oncogene 36, 20, 2813–2823. https://doi.org/10.1038/onc.2016.430. [Google Scholar]
  106. Harant H, de Martin R, Andrew PJ, Foglar E, Dittrich C, Lindley IJ (1996), Synergistic activation of interleukin-8 gene transcription by all-trans-retinoic acid and tumor necrosis factor-alpha involves the transcription factor NF-kappaB. J Biol Chem 271, 43, 26954–26961. [PubMed] [Google Scholar]
  107. Shattuck-Brandt RL, Richmond A (1997), Enhanced degradation of I-kappaB alpha contributes to endogenous activation of NF-kappaB in Hs294T melanoma cells. Cancer Res 57, 14, 3032–3039. [Google Scholar]
  108. Yang J, Richmond A (2001), Constitutive ikappaB kinase activity correlates with nuclear factor-kappaB activation in human melanoma cells. Cancer Res 61, 12, 4901–4909. [Google Scholar]
  109. Meyskens FL Jr, Buckmeier JA, McNulty SE, Tohidian NB (1999), Activation of nuclear factor-kappa B in human metastatic melanomacells and the effect of oxidative stress. Clin Cancer Res 5, 5, 1197–1202. [PubMed] [Google Scholar]
  110. Claudio E, Segade F, Wrobel K, Ramos S, Bravo R, Lazo PS (1996), Molecular mechanisms of TNFalpha cytotoxicity: activation of NF-kappaB and nuclear translocation. Exp Cell Res 224, 1, 63–71. [CrossRef] [PubMed] [Google Scholar]
  111. Javelaud D, Poupon MF, Wietzerbin J, Besançon F (2002), Inhibition of constitutive NF-kappa B activity suppresses tumorigenicity of Ewing sarcoma EW7 cells. Int J Cancer 98, 2, 193–198. [CrossRef] [PubMed] [Google Scholar]
  112. Schmid E, Stagno MJ, Yan J, Stournaras C, Lang F, Fuchs J, Seitz G (2016), Store-operated Ca(2+) entry in rhabdomyosarcoma cells. Biochem Biophys Res Commun 477, 1, 129–136. https://doi.org/10.1016/j.bbrc.2016.06.032. [Google Scholar]
  113. Burkitt MD, Hanedi AF, Duckworth CA, Williams JM, Tang JM, O’Reilly LA, Putoczki TL, Gerondakis S, Dimaline R, Caamano JH, Pritchard DM (2015), NF-κB1, NF-κB2 and c-Rel differentially regulate susceptibility to colitis-associated adenoma development in C57BL/6 mice. J Pathol 236, 3, 326–336. https://doi.org/10.1002/path.4527. [PubMed] [Google Scholar]
  114. Eckmann L, Nebelsiek T, Fingerle AA, Dann SM, Mages J, Lang R, Robine S, Kagnoff MF, Schmid RM, Karin M, Arkan MC, Greten FR (2008), Opposing functions of IKKbeta during acute and chronic intestinal inflammation. Proc Natl Acad Sci USA 105, 39, 15058–15063. https://doi.org/10.1073/pnas.0808216105. [CrossRef] [Google Scholar]
  115. Kitajima I, Shinohara T, Bilakovics J, Brown DA, Xu X, Nerenberg M (1992), Ablation of transplanted HTLV-I Tax-transformed tumors in mice by antisense inhibition of NF-kappa B. Science 258, 1792. [Google Scholar]
  116. Mabuchi S, Ohmichi M, Nishio Y, Hayasaka T, Kimura A, Ohta T, Saito M, Kawagoe J, Takahashi K, Yada-Hashimoto N, Sakata M, Motoyama T, Kurachi H, Tasaka K, Murata Y (2004), Inhibition of NFkappaB increases the efficacy of cisplatin in in vitro and in vivo ovarian cancer models. J Biol Chem 279, 22, 23477–23485. https://doi.org/10.1074/jbc.M313709200. [PubMed] [Google Scholar]
  117. Natarajan V, Komarov AP, Ippolito T, Bonneau K, Chenchik AA, Gudkov AV (2014), Peptides genetically selected for NF-κB activation cooperate with oncogene Ras and model carcinogenic role of inflammation. Proc Natl Acad Sci USA 111, 4, E474–E483. https://doi.org/10.1073/pnas.1311945111. [CrossRef] [Google Scholar]
  118. Quinlan MP, Quatela SE, Philips MR, Settleman J (2008), Activated Kras, but not Hras or Nras, may initiate tumors of endodermal origin via stem cell expansion. Mol Cell Biol 28, 2659–2674. https://doi.org/10.1128/MCB.01661-07. [PubMed] [Google Scholar]
  119. Buchanan FG, McReynolds M, Couvillon A, Kam Y, Holla VR, Dubois RN, Exton JH (2005), Requirement of phospholipase D1 activity in H-RasV12-induced transformation. Proc Natl Acad Sci USA 102, 5, 1638–1642. [CrossRef] [Google Scholar]
  120. Wu CS, Chen MF, Lee IL, Tung SY (2007), Predictive role of nuclear factor-kappaB activity in gastric cancer: a promising adjuvant approach with caffeic acid phenethyl ester. J Clin Gastroenterol 41, 10, 894–900. https://doi.org/10.1097/MCG.0b013e31804c707c. [Google Scholar]
  121. Kwon HJ, Won YS, Nam KT, Yoon YD, Jee H, Yoon WK, Nam KH, Kang JS, Han SU, Choi IP, Kim DY, Kim HC (2012), Vitamin D3 upregulated protein 1 deficiency promotes N-methyl-N-nitrosourea and Helicobacter pylori-induced gastric carcinogenesis in mice. Gut 61, 1, 53–63. https://doi.org/10.1136/gutjnl-2011-300361. [Google Scholar]
  122. Zerbini LF, Tamura RE, Correa RG, Czibere A, Cordeiro J, Bhasin M, Simabuco FM, Wang Y, Gu X, Li L, Sarkar D, Zhou JR, Fisher PB, Libermann TA (2011), Combinatorial effect of non-steroidal anti-inflammatory drugs and NF-κB inhibitors in ovarian cancer therapy. PLoS One 6, 9, e24285. https://doi.org/10.1371/journal.pone.0024285. [Google Scholar]
  123. Brücher BLDM, Jamall IS (2014), Epistemology of the origin of cancer: a new paradigm. BMC Cancer 14, 1–15. https://doi.org/10.1186/1471-2407-14-331. [CrossRef] [PubMed] [Google Scholar]
  124. Brücher BLDM, Jamall IS (2014), Cell-Cell communication in tumor microenvironment, carcinogenesis and anticancer treatment. Cell Physiol Biochem 34, 2, 213–243. https://doi.org/10.1159/000362978. [CrossRef] [PubMed] [Google Scholar]
  125. Hammarskjöld ML, Simurda MC (1992), Epstein-Barr virus latent membrane protein transactivates the human immunodeficiency virus type 1 long terminal repeat through induction of NF-kappa B activity. J Virol 66, 11, 6496–6501. [PubMed] [Google Scholar]
  126. Mahé Y, Mukaida N, Kuno K, Akiyama M, Ikeda N, Matsushima K, Murakami S (1991), Hepatitis B virus X protein transactivates human interleukin-8 gene through acting on nuclear factor kB and CCAAT/enhancer-binding protein-like cis-elements. J Biol Chem 266, 21, 13759–13763. [PubMed] [Google Scholar]
  127. Zhu N, Khoshnan A, Schneider R, Matsumoto M, Dennert G, Ware C, Lai MM (1998), Hepatitis C virus core protein binds to the cytoplasmic domain of tumor necrosis factor (TNF) receptor 1 and enhances TNF-induced apoptosis. J Virol 72, 5, 3691–3697. [PubMed] [Google Scholar]
  128. Twu JS, Chu K, Robinson WS (1989), Hepatitis B virus X gene activates kappa B-like enhancer sequences in the long terminal repeat of human immunodeficiency virus 1. Proc Natl Acad Sci USA 86, 13, 5168–5172. [CrossRef] [Google Scholar]
  129. Su F, Schneider RJ (1996), Hepatitis B virus HBx protein activates transcription factor NF-kappaB by acting on multiple cytoplasmic inhibitors of rel-related proteins. J Virol 70, 7, 4558–4566. [PubMed] [Google Scholar]
  130. Pikarsky E, Porat RM, Stein I, Abramovitch R, Amit S, Kasem S, et al. (2004), NF-kappaB functions as a tumour promoter in inflammation-associated cancer. Nature 431, 461–466. [CrossRef] [PubMed] [Google Scholar]
  131. Maeda S, Kamata H, Luo JL, Leffert H, Karin M (2005), IKKbeta couples hepatocyte death to cytokine-driven compensatory proliferation that promotes chemical hepatocarcinogenesis. Cell 121, 977–990. [CrossRef] [PubMed] [Google Scholar]
  132. Haybaeck J, Zeller N, Wolf MJ, Weber A, Wagner U, Kurrer MO, et al. (2009), A lymphotoxin-driven pathway to hepatocellular carcinoma. Cancer Cell 16, 295–308. [CrossRef] [PubMed] [Google Scholar]
  133. Tseng TC, Liu CJ, Yang HC, Su TH, Wang CC, Chen CL, et al. (2013), Serum hepatitis B surface antigen levels help predict disease progression in patients with low hepatitis B virus loads. Hepatology 57, 441–450. [Google Scholar]
  134. Kawanaka M, Nishino K, Nakamura J, Oka T, Urata N, Goto D, et al. (2014), Quantitative levels of hepatitis B virus DNA and surface antigen and the risk of hepatocellular carcinoma in patients with hepatitis B receiving long-term nucleos(t)ide analogue therapy. Liver Cancer 3, 41–52. [CrossRef] [PubMed] [Google Scholar]
  135. Sunami Y, Ringelhan M, Kokai E, Lu M, O’Connor T, Lorentzen A, Weber A, Rodewald AK, Müllhaupt B, Terracciano L, Gul S, Wissel S, Leithäuser F, Krappmann D, Riedl P, Hartmann D, Schirmbeck R, Strnad P, Hüser N, Kleeff J, Friess H, Schmid RM, Geisler F, Wirth T, Heikenwalder M (2016), Canonical NF-κB signaling in hepatocytes acts as a tumor-suppressor in hepatitis B virus surface antigen-driven hepatocellular carcinoma by controlling the unfolded protein response. Hepatology 63, 5, 1592–1607. https://doi.org/10.1002/hep.28435. [Google Scholar]
  136. Liu J, Yang HI, Lee MH, Lu SN, Jen CL, Batrla-Utermann R, et al. (2014), Spontaneous seroclearance of hepatitis B seromarkers and subsequent risk of hepatocellular carcinoma. Gut 63, 1648–1657. [Google Scholar]
  137. Waris G, Huh KW, Siddiqui A (2001 Nov), Mitochondrially associated hepatitis B virus X protein constitutively activates transcription factors STAT-3 and NF-kappa B via oxidative stress. Mol Cell Biol 21, 22, 7721–7730. [PubMed] [Google Scholar]
  138. Pollicino T, Bellinghieri L, Restuccia A, Raffa G, Musolino C, Alibrandi A, Teti D, Raimondo G (2013), Hepatitis B virus (HBV) induces the expression of interleukin-8 that in turn reduces HBV sensitivity to interferon-alpha. Virology 444, 1–2, 317–328. https://doi.org/10.1016/j.virol.2013.06.028. [CrossRef] [PubMed] [Google Scholar]
  139. zur Hausen H (2002), Papillomaviruses and cancer: from basic studies to clinical application. Nat Rev Cancer 2, 342–350. https://doi.org/10.1038/nrc798. [Google Scholar]
  140. Spitkovsky D, Hehner SP, Hofmann TG, Möller A, Schmitz ML (2002), The human papillomavirus oncoprotein E7 attenuates NF-kappa B activation by targeting the Ikappa B kinase complex. J Biol Chem 277, 28, 25576–25582. [PubMed] [Google Scholar]
  141. Mishra A, Bharti AC, Varghese P, Saluja D, Das BC (2006), Differential expression and activation of NF-kappaB family proteins during oral carcinogenesis: role of high risk human papillomavirus infection. Int J Cancer 119, 12, 2840–2850. https://doi.org/10.1002/ijc.22262. [CrossRef] [PubMed] [Google Scholar]
  142. Nees M, Geoghegan JM, Hyman T, Frank S, Miller L, Woodworth CD (2001), Papillomavirus type 16 oncogenes downregulate expression of interferon-responsive genes and upregulate proliferation-associated and NF-kappaB-responsive genes in cervical keratinocytes. J Virol 75, 9, 4283–4296. https://doi.org/10.1128/JVI.75.9.4283-4296.2001. [PubMed] [Google Scholar]
  143. Thompson DA, Zacny V, Belinsky GS, Classon M, Jones DL, Schlegel R, Münger K (2001), The HPV E7 oncoprotein inhibits tumor necrosis factor alpha-mediated apoptosis in normal human fibroblasts. Oncogene 20, 28, 3629–3640. https://doi.org/10.1038/sj.onc.1204483. [Google Scholar]
  144. Verma G, Vishnoi K, Tyagi A, Jadli M, Singh T, Goel A, Sharma A, Agarwal K, Prasad SC, Pandey D, Sharma S, Mehrotra R, Singh SM, Bharti AC (2017), Characterization of key transcription factors as molecular signatures of HPV-positive and HPV-negative oral cancers. Cancer Med 6, 3, 591–604. https://doi.org/10.1002/cam4.983. [Google Scholar]
  145. Fujimoto H, D’Alessandro-Gabazza CN, Palanki MS, Erdman PE, Takagi T, Gabazza EC, Bruno NE, Yano Y, Hayashi T, Tamaki S, Sumida Y, Adachi Y, Suzuki K, Taguchi O (2007), Inhibition of nuclear factor-kappaB in T cells suppresses lung fibrosis. Am J Res Crit Care Med 176, 12, 1251–1260. https://doi.org/10.1164/rccm.200609-1288OC. [CrossRef] [Google Scholar]
  146. Inazawa J, Itoh N, Abe T, Nagata S (1992), Assignment of the human Fas antigen gene (Fas) to 10q24.1. Genomics 14, 3, 821–822. [CrossRef] [PubMed] [Google Scholar]
  147. Liu F, Bardhan K, Yang D, Thangaraju M, Ganapathy V, Waller JL, Liles GB, Lee JR, Liu K (2012), NF-κB directly regulates Fas transcription to modulate Fas-mediated apoptosis and tumor suppression. J Biol Chem 287, 30, 25530–25540. https://doi.org/10.1074/jbc.M112.356279. [PubMed] [Google Scholar]
  148. Yang HJ, Wang M, Wang L, Cheng BF, Lin XY, Feng ZW (2015), NF-κB regulates caspase-4 expression and sensitizes neuroblastoma cells to Fas-induced apoptosis. PLoS One 10, 2, e0117953. https://doi.org/10.1371/journal.pone.0117953. [Google Scholar]
  149. Walch-Rückheim B, Mavrova R, Henning M, Vicinus B, Kim YJ, Bohle RM, Juhasz-Böss I, Solomayer EF, Smola S (2015), Stromal fibroblasts induce CCL20 through IL6/C/EBPβ to support the recruitment of Th17 cells during cervical cancer progression. Cancer Res 75, 24, 5248–5259. https://doi.org/10.1158/0008-5472.CAN-15-0732. [Google Scholar]
  150. Huang ZW, Lien GS, Lin CH, Jiang CP, Chen BC (2017), p300 and C/EBPβ-regulated IKKβ expression are involved in thrombin-induced IL-8/CXCL8 expression in human lung epithelial cells. Pharmacol Res 121, 33–41. https://doi.org/10.1016/j.phrs.2017.04.020. [CrossRef] [PubMed] [Google Scholar]
  151. Xie F, Li X, Wei C, Liang G, Dang Y, Shan Z (2015), C/EBPβ promotes NF-κB-mediated invasion and migration of human renal carcinoma 786-O cells. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi 31, 11, 1483–1487. [PubMed] [Google Scholar]
  152. Xia P, Zhang R, Ge G (2015), C/EBPβ mediates TNF-α-induced cancer cell migration by inducing MMP expression dependent on p38 MAPK. J Cell Biochem 116, 12, 2766–2777. https://doi.org/10.1002/jcb.25219. [CrossRef] [PubMed] [Google Scholar]
  153. Zou J, Li H, Chen X, Zeng S, Ye J, Zhou C, Liu M, Zhang L, Yu N, Gan X, Zhou H, Xian Z, Chen S, Liu P (2014), C/EBPβ knockdown protects cardiomyocytes from hypertrophy via inhibition of p65-NFκB. Mol Cell Endocrinol 390, 1–2, 18–25. https://doi.org/10.1016/j.mce.2014.03.007. [CrossRef] [PubMed] [Google Scholar]
  154. Esteves CL, Kelly V, Breton A, Taylor AI, West CC, Donadeu FX, Péault B, Seckl JR, Chapman KE (2014), Proinflammatory cytokine induction of 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) in human adipocytes is mediated by MEK, C/EBPβ, and NF-κB/RelA. J Clin Endocrinol Metab 99, 1, E160–E168. https://doi.org/10.1210/jc.2013-1708. [CrossRef] [PubMed] [Google Scholar]
  155. Akgül B, Pfefferle R, Marcuzzi GP, Zigrino P, Krieg T, Pfister H, Mauch C (2006), Expression of matrix metalloproteinase (MMP)-2, MMP-9, MMP-13, and MT1-MMP in skin tumors of human papillomavirus type 8 transgenic mice. Exp Dermatol 15, 1, 35–42. https://doi.org/10.1111/j.0906-6705.2005.00387.x. [CrossRef] [PubMed] [Google Scholar]
  156. Shao R, Karunagaran D, Zhou BP, Li K, Lo SS, Deng J, Chiao P, Hung MC (1997), Inhibition of nuclear factor-kappaB activity is involved in E1A-mediated sensitization of radiation-induced apoptosis. J Biol Chem 272, 52, 32739–32742. [PubMed] [Google Scholar]
  157. Chen X, Shen B, Xia L, Khaletzkiy A, Chu D, Wong JY, Li JJ (2002), Activation of nuclear factor kappaB in radioresistance of TP53-inactive human keratinocytes. Cancer Res 62, 4, 1213–1221. [Google Scholar]
  158. McCormick BA, Colgan SP, Delp-Archer C, Miller SI, Madara JL (1993), Salmonella typhimurium attachment to human intestinal epithelial monolayers: transcellular signalling to subepithelial neutrophils. J Cell Biol 123, 4, 895–907. [CrossRef] [PubMed] [Google Scholar]
  159. Jung HC, Eckmann L, Yang SK, Panja A, Fierer J, Morzycka-Wroblewska E, Kagnoff MF (1995), A distinct array of proinflammatory cytokines is expressed in human colon epithelial cells in response to bacterial invasion. J Clin Invest 95, 1, 55–65. https://doi.org/10.1172/JCI117676. [CrossRef] [PubMed] [Google Scholar]
  160. Elewaut D, DiDonato JA, Kim JM, Truong F, Eckmann L, Kagnoff MF (1999), NF-kappa B is a central regulator of the intestinal epithelial cell innate immune response induced by infection with enteroinvasive bacteria. J Immunol 163, 3, 1457–1466. [PubMed] [Google Scholar]
  161. Resta-Lenert S, Barrett KE (2002), Enteroinvasive bacteria alter barrier and transport properties of human intestinal epithelium: role of iNOS and COX-2. Gastroenterology 122, 4, 1070–1087. [CrossRef] [PubMed] [Google Scholar]
  162. Hauf N, Chakraborty T (2003), Suppression of NF-kappa B activation and proinflammatory cytokine expression by Shiga toxin-producing Escherichia coli. J Immunol 170, 4, 2074–2082. [CrossRef] [PubMed] [Google Scholar]
  163. Ruchaud-Sparagano MH, Mühlen S, Dean P, Kenny B (2011), The enteropathogenic E. coli (EPEC) Tir effector inhibits NF-κB activity by targeting TNFα receptor-associated factors. PLoS Pathog 7, 12, e1002414. https://doi.org/10.1371/journal.ppat.1002414. [CrossRef] [PubMed] [Google Scholar]
  164. Karikó K, Buckstein M, Ni H, Weissman D (2005), Suppression of RNA recognition by Toll-like receptors: the impact of nucleoside modification and the evolutionary origin of RNA. Immunity 23, 2, 165–175. https://doi.org/10.1016/j.immuni.2005.06.008. [CrossRef] [PubMed] [Google Scholar]
  165. Karin M, Ben-Neriah Y (2000), Phosphorylation meets ubiquitination: the control of NF-[kappa]B activity. Ann Rev Immunol 18, 621–663. https://doi.org/10.1146/annurev.immunol.18.1.621. [CrossRef] [Google Scholar]
  166. West AP, Koblansky AA, Ghosh S (2006), Recognition and signaling by toll-like receptors. Ann Rev Cell Dev Biol 22, 409–437. https://doi.org/10.1146/annurev.cellbio.21.122303.115827. [CrossRef] [PubMed] [Google Scholar]
  167. Maglione PJ, Simchoni N, Cunningham-Rundles C (2015), Toll-like receptor signaling in primary immune deficiencies. Ann NY Acad Sci 1356, 1–21. https://doi.org/10.1111/nyas.12763. [CrossRef] [Google Scholar]
  168. Sen R, Baltimore D (1986), Inducibility of kappa immunoglobulin enhancer-binding protein Nf-kappa B by a posttranslational mechanism. Cell 47, 6, 921–928. [CrossRef] [PubMed] [Google Scholar]
  169. Read MA, Cordle SR, Veach RA, Carlisle CD, Hawiger J (1993), Cell-free pool of CD14 mediates activation of transcription factor NF-kappa B by lipopolysaccharide in human endothelial cells. Proc Natl Acad Sci USA 90, 21, 9887–9891. [CrossRef] [Google Scholar]
  170. Aihara M, Tsuchimoto D, Takizawa H, Azuma A, Wakebe H, Ohmoto Y, Imagawa K, Kikuchi M, Mukaida N, Matsushima K (1997), Mechanisms involved in Helicobacter pylori-induced interleukin-8 production by a gastric cancer cell line, MKN45. Infect Immun 65, 8, 3218–3224. [PubMed] [Google Scholar]
  171. Sharma SA, Tummuru MK, Blaser MJ, Kerr LD (1998), Activation of IL-8 gene expression by Helicobacter pylori is regulated by transcription factor nuclear factor-kappa B in gastric epithelial cells. J Immunol 160, 5, 2401–2407. [PubMed] [Google Scholar]
  172. Isomoto H, Miyazaki M, Mizuta Y, Takeshima F, Murase K, Inoue K, Yamasaki K, Murata I, Koji T, Kohno S (2000), Expression of nuclear factor-kappaB in Helicobacter pylori-infected gastric mucosa detected with southwestern histochemistry. Scand J Gastroenterol 35, 3, 247–254. [CrossRef] [PubMed] [Google Scholar]
  173. Yang GF, Deng CS, Xiong YY, Gong LL, Wang BC, Luo J (2004), Expression of nuclear factor-kappa B and target genes in gastric precancerous lesions and adenocarcinoma: association with Helicobactor pylori cagA (+) infection. World J Gastroenterol 10, 4, 491–496. [CrossRef] [PubMed] [Google Scholar]
  174. Wroblewski LE, Noble PJ, Pagliocca A, Pritchard DM, Hart CA, Campbell F, Dodson AR, Dockray GJ, Varro A (2003), Stimulation of MMP-7 (matrilysin) by Helicobacter pylori in human gastric epithelial cells: role in epithelial cell migration. J Cell Sci 116, Pt 14, 3017–3026. https://doi.org/10.1242/jcs.00518. [Google Scholar]
  175. Suzuki M, Mimuro H, Kiga K, Fukumatsu M, Ishijima N, Morikawa H, Nagai S, Koyasu S, Gilman RH, Kersulyte D, Berg DE, Sasakawa C (2009), Helicobacter pylori CagA phosphorylation-independent function in epithelial proliferation and inflammation. Cell Host Microb 5, 1, 23–34. https://doi.org/10.1016/j.chom.2008.11.010. [CrossRef] [Google Scholar]
  176. Takayama S, Takahashi H, Matsuo Y, Okada Y, Takeyama H (2010), Effect of Helicobacter bilis infection on human bile duct cancer cells. Dig Dis Sci 55, 7, 1905–1910. https://doi.org/10.1007/s10620-009-0946-6. [CrossRef] [PubMed] [Google Scholar]
  177. Segura-López FK, Avilés-Jiménez F, Güitrón-Cantú A, Valdéz-Salazar HA, León-Carballo S, Guerrero-Pérez L, Fox JG, Torres J (2015), Infection with Helicobacter bilis but not Helicobacter hepaticus was Associated with Extrahepatic Cholangiocarcinoma. Helicobacter 20, 3, 223–230. https://doi.org/10.1111/hel.12195. [CrossRef] [PubMed] [Google Scholar]
  178. Feng SH, Tsai S, Rodriguez J, Lo SC (1999), Mycoplasmal infections prevent apoptosis and induce malignant transformation of interleukin-3-dependent 32D hematopoietic cells. Mol Cell Biol 19, 12, 7995–8002. [PubMed] [Google Scholar]
  179. Logunov DY, Scheblyakov DV, Zubkova OV, Shmarov MM, Rakovskaya IV, Gurova KV, Tararova ND, Burdelya LG, Naroditsky BS, Ginzburg AL, Gudkov AV (2008), Mycoplasma infection suppresses p53, activates NF-kappaB and cooperates with oncogenic Ras in rodent fibroblast transformation. Oncogene 27, 33, 4521–4531. https://doi.org/10.1038/onc.2008.103. [Google Scholar]
  180. Cao S, Shen D, Wang Y, Li L, Zhou L, Wang Y (2017), Potential malignant transformation in the gastric mucosa of immunodeficient mice with persistent Mycoplasma penetrans infection. PLoS One 12, 7, e0180514. https://doi.org/10.1371/journal.pone.0180514. [Google Scholar]
  181. Duan H, Chen L, Qu L, Yang H, Song SW, Han Y, Ye M, Chen W, He X, Shou C (2014), Mycoplasma hyorhinis infection promotes NF-κB-dependent migration of gastric cancer cells. Cancer Res 74, 20, 5782–5794. https://doi.org/10.1158/0008-5472.CAN-14-0650. [Google Scholar]
  182. Feige MH, Vieth M, Sokolova O, Täger C, Naumann M (2018), Helicobacter pylori induces direct activation of the lymphotoxin beta receptor and non-canonical nuclear factor-kappa B signaling. Biochim Biophys Acta 1865, 4, 545–550. https://doi.org/10.1016/j.bbamcr.2018.01.006. [CrossRef] [PubMed] [Google Scholar]
  183. Tsai S, Wear DJ, Shih JW, Lo SC (1995), Mycoplasmas and oncogenesis: persistent infection and multistage malignant transformation. Proc Natl Acad Sci USA 92, 22, 10197–10201. [CrossRef] [Google Scholar]
  184. White L, Cox D (1967), Chromosome changes in a rhabdomyosarcoma during recurrence and in cell culture. Br J Cancer 21, 4, 684–693. [CrossRef] [PubMed] [Google Scholar]
  185. Paton GR, Jacobs JP, Perkins FT (1965), Chromosome changes in human diploid-cell cultures infected with mycoplasma. Nature 207, 43–45. [CrossRef] [PubMed] [Google Scholar]
  186. Fogh J, Fogh H (1965), Chromosome changes in PPLO-infected FL human amnion cells. Proc Soc Exp Biol Med 119, 233–238. [CrossRef] [PubMed] [Google Scholar]
  187. Adebamowo SN, Ma B, Zella D, Famooto A, Ravel J, Adebamowo C, ACCME Research Group (2017), Mycoplasma hominis and Mycoplasma genitalium in the vaginal microbiota and persistent high-risk human papillomavirus infection. Front Public Health 5, 140. https://doi.org/10.3389/fpubh.2017.00140. [CrossRef] [PubMed] [Google Scholar]
  188. Brücher BLDM, Jamall IS (2016), Somatic mutation theory – Why it’s wrong for most cancers. Cell Physiol Biochem 38, 5, 1663–1680. https://doi.org/10.1159/000443106.. [CrossRef] [PubMed] [Google Scholar]
  189. Brücher BLDM, Jamall IS (2019), Chronic inflammation evoked by pathogenic stimulus during carcinogenesis. 4open 2, 8, 1–22. https://doi.org/10.1051/fopen/2018006. [CrossRef] [EDP Sciences] [Google Scholar]
  190. Brücher BLDM, Jamall IS (2019), Eicosanoids evolved in chronic inflammation during carcinogenesis. 4open 2, 9, 1–34. https://doi.org/10.1051/fopen/2018008 [CrossRef] [EDP Sciences] [Google Scholar]
  191. Brücher BLDM, Jamall IS (2019), Microbiome and morbid obesity increase pathogenic stimulus diversity. 4open 2, 10, 1–16. https://doi.org/10.1051/fopen/2018007. [CrossRef] [EDP Sciences] [Google Scholar]
  192. Wilson RA, Coulson PS, Dixon B (1986), Migration of the schistosomula of Schistosoma mansoni in mice vaccinated with radiation-attenuated cercariae, and normal mice: an attempt to identify the timing and site of parasite death. Parasitology 92, Pt 1, 101–116. [CrossRef] [PubMed] [Google Scholar]
  193. Trottein F, Descamps L, Nutten S, Dehouck MP, Angeli V, Capron A, Cecchelli R, Capron M (1999), Schistosoma mansoni activates host microvascular endothelial cells to acquire an anti-inflammatory phenotype. Infect Immun 67, 7, 3403–3409. [PubMed] [Google Scholar]
  194. Trottein F, Nutten S, Angeli V, Delerive P, Teissier E, Capron A, Staels B, Capron M (1999), Schistosoma mansoni schistosomula reduce E-selectin and VCAM-1 expression in TNF-alpha-stimulated lung microvascular endothelial cells by interfering with the NF-kappaB pathway. Eur J Immunol 29, 11, 3691–3701. https://doi.org/10.1002/(SICI)1521-4141(199911)29:11<691::AID-IMMU3691>3.0.CO;2-L. [CrossRef] [PubMed] [Google Scholar]
  195. Gerritsen ME, Williams AJ, Neish AS, Moore S, Shi Y, Collins T (1997), CREB-binding protein/p300 are transcriptional coactivators of p65. Proc Natl Acad Sci USA 94, 7, 2927–2932. [CrossRef] [Google Scholar]
  196. Aksoy E, Zouain CS, Vanhoutte F, Fontaine J, Pavelka N, Thieblemont N, Willems F, Ricciardi-Castagnoli P, Goldman M, Capron M, Ryffel B, Trottein F (2005), Double-stranded RNAs from the helminth parasite Schistosoma activate TLR3 in dendritic cells. J Biol Chem 280, 1, 277–283. [PubMed] [Google Scholar]
  197. Nakashima T, Okuda K, Kojiro M, Sakamoto K, Kubo Y (1975), Primary liver cancer coincident with Schistosomiasis japonica. A study of 24 necropsies. Cancer 36, 4, 1483–1489. [CrossRef] [PubMed] [Google Scholar]
  198. Andoh H, Yasui O, Kurokawa T, Sato T (2004), Cholangiocarcinoma coincident with Schistosomiasis japonica. J Gastroenterol 39, 1, 64–68. [CrossRef] [PubMed] [Google Scholar]
  199. Abd El-Aal NF, Hamza RS, Harb O (2017), Paeoniflorin targets apoptosis and ameliorates fibrosis in murine Schistosomiasis mansoni: a novel insight. Exp Parasitol 183, 23–32. https://doi.org/10.1016/j.exppara.2017. [CrossRef] [PubMed] [Google Scholar]
  200. Wan C, Jin F, Du Y, Yang K, Yao L, Mei Z, Huang W (2017), Genistein improves schistosomiasis liver granuloma and fibrosis via dampening NF-kB signaling in mice. Parasitol Res 116, 4, 1165–1174. https://doi.org/10.1007/s00436-017-5392-3. [Google Scholar]
  201. Liu M, Wu Q, Chen P, Büchele B, Bian M, Dong S, Huang D, Ren C, Zhang Y, Hou X, Simmet T, Shen J (2014), A boswellic acid-containing extract ameliorates schistosomiasis liver granuloma and fibrosis through regulating NF-κB signaling in mice. PLoS One 9, 6, e100129. https://doi.org/10.1371/journal.pone.0100129. [Google Scholar]
  202. Pan JX, Ding K, Wang CY (2012), Niclosamide, an old antihelminthic agent, demonstrates antitumor activity by blocking multiple signaling pathways of cancer stem cells. Chin J Cancer 31, 4, 178–184. https://doi.org/10.5732/cjc.011.10290. [Google Scholar]
  203. Sommer P, Gleyzal C, Raccurt M, Delbourg M, Serrar M, Joazeiro P, Peyrol S, Kagan H, Trackman PC, Grimaud JA (1993), Transient expression of lysyl oxidase by liver myofibroblasts in murine schistosomiasis. Lab Invest 69, 4, 460–470. [PubMed] [Google Scholar]
  204. Ouzzine M, Boyd A, Hulmes DJ (1996), Expression of active, human lysyl oxidase in Escherichia coli. FEBS Lett 399, 3, 215–219. [CrossRef] [PubMed] [Google Scholar]
  205. Saito H, Papaconstantinou J, Sato H, Goldstein S (1997), Regulation of a novel gene encoding a lysyl oxidase-related protein in cellular adhesion and senescence. J Biol Chem 272, 13, 8157–8160. [PubMed] [Google Scholar]
  206. Decitre M, Gleyzal C, Raccurt M, Peyrol S, Aubert-Foucher E, Csiszar K, Sommer P (1998), Lysyl oxidase-like protein localizes to sites of de novo fibrinogenesis in fibrosis and in the early stromal reaction of ductal breast carcinomas. Lab Invest 78, 2, 143–151. [PubMed] [Google Scholar]
  207. Salvador F, Martin A, López-Menéndez C, Moreno-Bueno G, Santos V, Vázquez-Naharro A, Santamaria PG, Morales S, Dubus PR, Muinelo-Romay L, López-López R, Tung JC, Weaver VM, Portillo F, Cano A (2017), Lysyl oxidase-like protein LOXL2 promotes lung metastasis of breast cancer. Cancer Res 77, 21, 5846–5859. https://doi.org/10.1158/0008-5472.CAN-16-3152. [Google Scholar]
  208. Milward MR, Chapple IL, Carter K, Matthews JB, Cooper PR (2013), Micronutrient modulation of NF-κB in oral keratinocytes exposed to periodontal bacteria. Innate Immun 19, 2, 140–151. https://doi.org/10.1177/1753425912454761. [CrossRef] [PubMed] [Google Scholar]
  209. Biswas S, Quante M, Leedham S, Jansen M (2018), The metaplastic mosaic of Barrett’s oesophagus. Virchows Arch 472, 1, 43–54. https://doi.org/10.1007/s00428-018-2317-1. [CrossRef] [PubMed] [Google Scholar]
  210. Sikkema M, de Jonge PJ, Steyerberg EW, Kuipers EJ (2010), Risk of esophageal adenocarcinoma and mortality in patients with Barrett’s esophagus: a systematic review and meta-analysis. Clin Gastroenterol Hepatol 8, 3, 235–244. https://doi.org/10.1016/j.cgh.2009.10.010. [CrossRef] [PubMed] [Google Scholar]
  211. Desai TK, Krishnan K, Samala N, Singh J, Cluley J, Perla S, Howden CW (2012), The incidence of oesophageal adenocarcinoma in non-dysplastic Barrett’s oesophagus: a meta-analysis. Gut 61, 7, 970–976. https://doi.org/10.1136/gutjnl-2011-300730. [Google Scholar]
  212. Masuda A, Fujita T, Murakami M, Yamazaki Y, Kobayashi M, Terao S, Sanuki T, Okada A, Adachi M, Shiomi H, Arisaka Y, Kutsumi H, Umegaki E, Azuma T (2018), Influence of hiatal hernia and male sex on the relationship between alcohol intake and occurrence of Barrett’s esophagus. PLoS One 13, 2, e0192951. https://doi.org/10.1371/journal.pone.0192951. [Google Scholar]
  213. Locke GR 3rd, Talley NJ, Fett SL, Zinsmeister AR, Melton LJ 3rd (1997), Prevalence and clinical spectrum of gastroesophageal reflux: a population-based study in Olmsted County, Minnesota. Gastroenterology 112, 5, 1448–1456. [CrossRef] [PubMed] [Google Scholar]
  214. Takubo K, Aida J, Naomoto Y, Sawabe M, Arai T, Shiraishi H, Matsuura M, Ell C, May A, Pech O, Stolte M, Vieth M (2009), Cardiac rather than intestinal-type background in endoscopic resection specimens of minute Barrett adenocarcinoma. Hum Pathol 40, 1, 65–74. https://doi.org/10.1016/j.humpath.2008.06.008. [CrossRef] [PubMed] [Google Scholar]
  215. Aida J, Vieth M, Shepherd NA, Ell C, May A, Neuhaus H, Ishizaki T, Nishimura M, Fujiwara M, Arai T, Takubo K (2015), Is carcinoma in columnar-lined esophagus always located adjacent to intestinal metaplasia? a histopathologic assessmen. Am J Surg Pathol 39, 2, 188–196. https://doi.org/10.1097/PAS.0000000000000350. [CrossRef] [PubMed] [Google Scholar]
  216. Lavery DL, Martinez P, Gay LJ, Cereser B, Novelli MR, Rodriguez-Justo M, Meijer SL, Graham TA, McDonald SA, Wright NA, Jansen M (2016), Evolution of oesophageal adenocarcinoma from metaplastic columnar epithelium without goblet cells in Barrett’s oesophagus. Gut 65, 6, 907–913. https://doi.org/10.1136/gutjnl-2015-310748. [Google Scholar]
  217. Liu W, Hahn H, Odze RD, Goyal RK (2009), Metaplastic esophageal columnar epithelium without goblet cells shows DNA content abnormalities similar to goblet cell-containing epithelium. Am J Gastroenterol 104, 4, 816–824. https://doi.org/10.1038/ajg.2009.85. [Google Scholar]
  218. Hoadley KA, Yau C, Hinoue T, Wolf DM, Lazar AJ, Drill E, Shen R, Taylor AM, Cherniack AD, Thorsson V, Akbani R, Bowlby R, Wong CK, Wiznerowicz M, Sanchez-Vega F, Robertson AG, Schneider BG, Lawrence MS, Noushmehr H, Malta TM, Cancer Genome Atlas Network, Stuart JM, Benz CC, Laird PW (2018), Cell-of-origin patterns dominate the molecular classification of 10,000 tumors from 33 types of cancer. Cell 173, 2, 291–304.e6. https://doi.org/10.1016/j.cell.2018.03.022. [CrossRef] [PubMed] [Google Scholar]
  219. Ellrott K, Bailey MH, Saksena G, Covington KR, Kandoth C, Stewart C, Hess J, Ma S, Chiotti KE, McLellan M, Sofia HJ, Hutter C, Getz G, Wheeler D, Ding L; MC3 Working Group; Cancer Genome Atlas Research Network (2018), Scalable open science approach for mutation calling of tumor exomes using multiple genomic pipelines. Cell Syst 6, 3, 271–281.e7. https://doi.org/10.1016/j.cels.2018.03.002. [CrossRef] [PubMed] [Google Scholar]
  220. Babar M, Ennis D, Abdel-Latif M, Byrne PJ, Ravi N, Reynolds JV (2010), Differential molecular changes in patients with asymptomatic long-segment Barrett’s esophagus treated by antireflux surgery or medical therapy. Am J Surg 199, 2, 137–143. https://doi.org/10.1016/j.amjsurg.2008.11.032. [CrossRef] [PubMed] [Google Scholar]
  221. Li WT, Luo QQ, Wang B, Chen X, Yan XJ, Qiu HY, Chen SL (2019), Bile acids induce visceral hypersensitivity via mucosal mast cell-to-nociceptor signaling that involves the farnesoid X receptor/nerve growth factor/transient receptor potential vanilloid 1 axis. FASEB J 33, 2, 2435–2450. https://doi.org/10.1096/fj.201800935RR. [CrossRef] [PubMed] [Google Scholar]
  222. Kim DH, Xiao Z, Kwon S, Sun X, Ryerson D, Tkac D, Ma P, Wu SY, Chiang CM, Zhou E, Xu HE, Palvimo JJ, Chen LF, Kemper B, Kemper JK (2015), A dysregulated acetyl/SUMO switch of FXR promotes hepatic inflammation in obesity. EMBO J 34, 2, 184–199. https://doi.org/10.15252/embj.201489527. [CrossRef] [PubMed] [Google Scholar]
  223. Gai Z, Visentin M, Gui T, Zhao L, Thasler WE, Häusler S, Hartling I, Cremonesi A, Hiller C, Kullak-Ublick GA (2018), Effects of farnesoid X receptor activation on arachidonic acid metabolism, NF-kB signaling, and hepatic inflammation. Mol Pharmacol 94, 2, 802–811. https://doi.org/10.1124/mol.117.111047. [CrossRef] [PubMed] [Google Scholar]
  224. Zhang Y, Xu Y, Qi Y, Xu L, Song S, Yin L, Tao X, Zhen Y, Han X, Ma X, Liu K, Peng J (2017), Protective effects of dioscin against doxorubicin-induced nephrotoxicity via adjusting FXR-mediated oxidative stress and inflammation. Toxicology 378, 53–64. https://doi.org/10.1016/j.tox.2017.01.007. [CrossRef] [PubMed] [Google Scholar]
  225. Park JH, Kang SS, Kim JY, Tchah H (2016), Nerve growth factor attenuates apoptosis and inflammation in the diabetic cornea. Invest Ophthalmol Vis Sci 57, 15, 6767–6775. https://doi.org/10.1167/iovs.16-19747. [CrossRef] [PubMed] [Google Scholar]
  226. Hathway GJ, Fitzgerald M (2006), Time course and dose-dependence of nerve growth factor-induced secondary hyperalgesia in the mouse. J Pain 7, 1, 57–61. https://doi.org/10.1016/j.jpain.2005.08.003. [Google Scholar]
  227. Eskander MA, Ruparel S, Green DP, Chen PB, Por ED, Jeske NA, Gao X, Flores ER, Hargreaves KM (2015), Persistent nociception triggered by nerve growth factor (NGF) is mediated by TRPV1 and oxidative mechanisms. J Neurosci 35, 22, 8593–8603. https://doi.org/10.1523/JNEUROSCI.3993-14.2015. [CrossRef] [PubMed] [Google Scholar]
  228. Matsuda H, Switzer J, Coughlin MD, Bienenstock J, Denburg JA (1988), Human basophilic cell differentiation promoted by 2.5S nerve growth factor. Int Arch Allergy Appl Immunol 86, 4, 453–457. PMID: 3261719. [CrossRef] [PubMed] [Google Scholar]
  229. Oddiah D, Anand P, McMahon SB, Rattray M (1998), Rapid increase of NGF, BDNF and NT-3 mRNAs in inflamed bladder. Neuroreport 9, 7, 1455–1458. PMID: 9631447. [CrossRef] [PubMed] [Google Scholar]
  230. Gentry JJ, Casaccia-Bonnefil P, Carter BD (2000), Nerve growth factor activation of nuclear factor kappaB through its p75 receptor is an anti-apoptotic signal in RN22 schwannoma cells. J Biol Chem 275, 11, 7558–7565. PMID: 10713062. [PubMed] [Google Scholar]
  231. Maggirwar SB, Sarmiere PD, Dewhurst S, Freeman RS (1998), Nerve growth factor-dependent activation of NF-kappaB contributes to survival of sympathetic neurons. J Neurosci 18, 24, 10356–10365. PMID: 9852573. [CrossRef] [PubMed] [Google Scholar]
  232. Bui NT, Livolsi A, Peyron JF, Prehn JH (2001), Activation of nuclear factor kappaB and Bclx survival gene expression by nerve growth factor requires tyrosine phosphorylation of IkappaBalpha. J Cell Biol 152, 4, 753–764. PMCID: PMC2195773. [CrossRef] [PubMed] [Google Scholar]
  233. Takei Y, Laskey R (2008), Tumor necrosis factor alpha regulates responses to nerve growth factor, promoting neural cell survival but suppressing differentiation of neuroblastoma cells. Mol Biol Cell 19, 3, 855–864. https://doi.org/10.1091/mbc.e07-06-0624. [CrossRef] [PubMed] [Google Scholar]
  234. Kwon K, Park SH, Han BS, Oh SW, Lee SE, Yoo JA, Park SJ, Kim J, Kim JW, Cho JY, Lee J (2018), Negative cellular effects of urban particulate matter on human keratinocytes are mediated by P38 MAPK and NF-κB-dependent expression of TRPV 1. Int J Mol Sci 19, 9, 1–15. pii: E2660. https://doi.org/10.3390/ijms19092660. [Google Scholar]
  235. Nakanishi M, Morita Y, Hata K, Muragaki Y (2016), Acidic microenvironments induce lymphangiogenesis and IL-8 production via TRPV1 activation in human lymphatic endothelial cells. Exp Cell Res 345, 2, 180–189. https://doi.org/10.1016/j.yexcr.2016.06.006. [CrossRef] [PubMed] [Google Scholar]
  236. Debruyne PR, Witek M, Gong L, Birbe R, Chervoneva I, Jin T, Domon-Cell C, Palazzo JP, Freund JN, Li P, Pitari GM, Schulz S, Waldman SA (2006), Bile acids induce ectopic expression of intestinal guanylyl cyclase C through nuclear factor-kappaB and Cdx2 in human esophageal cellsm. Gastroenterology 130, 4, 1191–1206. https://doi.org/10.1053/j.gastro.2005.12.032. [CrossRef] [PubMed] [Google Scholar]
  237. Jenkins GJ, Cronin J, Alhamdani A, Rawat N, D’Souza F, Thomas T, Eltahir Z, Griffiths AP, Baxter JN (2008), The bile acid deoxycholic acid has a non-linear dose response for DNA damage and possibly NF-kappaB activation in oesophageal cells, with a mechanism of action involving ROS. Mutagenesis 23, 5, 399–405. https://doi.org/10.1093/mutage/gen029. [CrossRef] [PubMed] [Google Scholar]
  238. Gong L, Debruyne PR, Witek M, Nielsen K, Snook A, Lin JE, Bombonati A, Palazzo J, Schulz S, Waldman SA (2009), Bile acids initiate lineage-addicted gastroesophageal tumorigenesis by suppressing the EGF receptor-AKT axis. Clin Transl Sci 2, 4, 286–293. https://doi.org/10.1111/j.1752-8062.2009.00131.x. [Google Scholar]
  239. Chen BJ, Zeng S, Xie R, Hu CJ, Wang SM, Wu YY, Xiao YF, Yang SM (2017), hTERT promotes gastric intestinal metaplasia by upregulating CDX2 via NF-κB signaling pathway. Oncotarget 8, 16, 26969–26978. https://doi.org/10.18632/oncotarget.15926. [PubMed] [Google Scholar]
  240. Wang W, Luo HS, Yu BP (2004), Expression of NF-kappaB and human telomerase reverse transcriptase in gastric cancer and precancerous lesions. World J Gastroenterol 10, 2, 177–181. [CrossRef] [PubMed] [Google Scholar]
  241. Yan F, Polk DB (2010), Disruption of NF-kappaB signalling by ancient microbial molecules: novel therapies of the future? Gut 59, 4, 421–426. https://doi.org/10.1136/gut.2009.179614. [Google Scholar]
  242. Chen JH, Huang SM, Chen CC, Tsai CF, Yeh WL, Chou SJ, Hsieh WT, Lu DY (2011), Ghrelin induces cell migration through GHS-R, CaMKII, AMPK, and NF-κB signaling pathway in glioma cells. J Cell Biochem 112, 10, 2931–2941. https://doi.org/10.1002/jcb.23209. [CrossRef] [PubMed] [Google Scholar]
  243. Ishigami S, Arigami T, Uchikado Y, Setoyama T, Kita Y, Sasaki K, Okumura H, Kurahara H, Kijima Y, Harada A, Ueno S, Natsugoe S (2013), IL-32 expression is an independent prognostic marker for gastric cancer. Med Oncol 30, 2, 472. https://doi.org/10.1007/s12032-013-0472-4. [CrossRef] [PubMed] [Google Scholar]
  244. Atchison ML, Perry RP (1987), The role of the kappa enhancer and its binding factor NF-kappa B in the developmental regulation of kappa gene transcription. Cell 48, 1, 121–128. [CrossRef] [PubMed] [Google Scholar]
  245. Giri DK, Mehta RT, Kansal RG, Aggarwal BB (1998), Mycobacterium avium-intracellulare complex activates nuclear transcription factor-kappaB in different cell types through reactive oxygen intermediates. J Immunol 161, 9, 4834–4841. [PubMed] [Google Scholar]
  246. Laflamme N, Lacroix S, Rivest S (1999), An essential role of interleukin-1beta in mediating NF-kappaB activity and COX-2 transcription in cells of the blood-brain barrier in response to a systemic and localized inflammation but not during endotoxemia. J Neurosci 19, 24, 10923–10930. [CrossRef] [PubMed] [Google Scholar]
  247. Rivest S, Lacroix S, Vallières L, Nadeau S, Zhang J, Laflamme N (2000), How the blood talks to the brain parenchyma and the paraventricular nucleus of the hypothalamus during systemic inflammatory and infectious stimuli. Proc Soc Exp Biol Med 223, 1, 22–38. [CrossRef] [PubMed] [Google Scholar]
  248. Hiscott J, Lin R, Nakhaei P, Paz S (2006), MasterCARD: a priceless link to innate immunity. Trends Mol Med 12, 2, 53–56. https://doi.org/10.1016/j.molmed.2005.12.003. [CrossRef] [PubMed] [Google Scholar]
  249. Hong GS, Jung YK (2002), Caspase recruitment domain (CARD) as a bi-functional switch of caspase regulation and NF-kappaB signals. J Biochem Mol Biol 35, 1, 19–23. [PubMed] [Google Scholar]
  250. Dufner A, Pownall S, Mak TW (2006), Caspase recruitment domain protein 6 is a microtubule-interacting protein that positively modulates NF-kappaB activation. Proc Natl Acad Sci USA 103, 4, 988–993. https://doi.org/10.1073/pnas.0510380103. [CrossRef] [Google Scholar]
  251. Kim SS, Ahn CH, Kang MR, Kim YR, Kim HS, Yoo NJ, Lee SH (2010), Expression of CARD6, an NF-kappaB activator, in gastric, colorectal and oesophageal cancers. Pathology 42, 1, 50–53. https://doi.org/10.3109/00313020903434421. [PubMed] [Google Scholar]
  252. Kim H, Lim JW, Kim KH (2001), Helicobacter pylori-induced expression of interleukin-8 and cyclooxygenase-2 in AGS gastric epithelial cells: mediation by nuclear factor-kappaB. Scand J Gastroenterol 36, 7, 706–716. https://doi.org/10.1074/jbc.M104141200. [PubMed] [Google Scholar]
  253. Chen Z, Liu M, Liu X, Huang S, Li L, Song B, Li H, Ren Q, Hu Z, Zhou Y, Qiao L (2013), COX-2 regulates E-cadherin expression through the NF-κB/Snail signaling pathway in gastric cancer. Int J Mol Med 32, 1, 93–100. https://doi.org/10.3892/ijmm.2013.1376. [CrossRef] [PubMed] [Google Scholar]
  254. Lin MT, Zuon CY, Chang CC, Chen ST, Chen CP, Lin BR, Wang MY, Jeng YM, Chang KJ, Lee PH, Chen WJ, Kuo ML (2005), Cyr61 induces gastric cancer cell motility/invasion via activation of the integrin/nuclear factor-kappaB/cyclooxygenase-2 signaling pathway. Clin Cancer Res 11, 16, 5809–5820. https://doi.org/10.1158/1078-0432.CCR-04-2639. [CrossRef] [PubMed] [Google Scholar]
  255. Ma HF, Zhao ZS (2008), Relationship of cysteine-rich protein 61 and NF-kappaB expression with metastasis and prognosis of gastric carcinoma. Zhonghua Bing Li Xue Za Zhi 37, 12, 815–821. [PubMed] [Google Scholar]
  256. Wu L, Pu Z, Feng J, Li G, Zheng Z, Shen W (2008), The ubiquitin-proteasome pathway and enhanced activity of NF-kappaB in gastric carcinoma. J Surg Oncol 97, 5, 439–444. https://doi.org/10.1002/jso.20952. [Google Scholar]
  257. Gravallese EM, Boothby MR, Smas CM, Glimcher LH (1989), A lipopolysaccharide-induced DNA-binding protein for a class II gene in B cells is distinct from NF-kappa B. Mol Cell Biol 9, 8, 3184–3192. [PubMed] [Google Scholar]
  258. Joshi-Barve SS, Rangnekar VV, Sells SF, Rangnekar VM (1993), Interleukin-1-inducible expression of gro-beta via NF-kappa B activation is dependent upon tyrosine kinase signaling. J Biol Chem 268, 24, 18018–18029. [PubMed] [Google Scholar]
  259. Stoeckle MY (1991), Post-transcriptional regulation of gro alpha, beta, gamma, and IL-8 mRNAs by IL-1 beta. Nucleic Acids Res 19, 4, 917–920. [CrossRef] [PubMed] [Google Scholar]
  260. Anisowicz A, Bardwell L, Sager R (1987), Constitutive overexpression of a growth-regulated gene in transformed Chinese hamster and human cells. Proc Natl Acad Sci USA 84, 7188–7192. [CrossRef] [Google Scholar]
  261. Haskill S, Peace A, Morris J, Sporn SA, Anisowicz A, Lee SW, Smith T, Martin G, Ralph P, Sager R (1990), Identification of three related human GRO genes encoding cytokine functions. Proc Natl Acad Sci USA 87, 19, 7732–7736. [CrossRef] [Google Scholar]
  262. Iida N, Grotendorst GR (1990), Cloning and sequencing of a new gro transcript from activated human monocytes: expression in leukocytes and wound tissue. Mol Cell Biol 10, 10, 5596–5599. [PubMed] [Google Scholar]
  263. Balentien E, Mufson BE, Shattuck RL, Derynck R, Richmond A (1991), Effects of MGSA/GROα on melanocyte transformation. Oncogene 6, 1115–1124. [Google Scholar]
  264. Li A, Varney ML, Singh RK (2004), Constitutive expression of growth regulated oncogene (gro) in human colon carcinoma cells with different metastatic potential and its role in regulating their metastatic phenotype. Clin Exp Metastasis 21, 7, 571–579. [CrossRef] [PubMed] [Google Scholar]
  265. Li X, Sun S, Li N, Gao J, Yu J, Zhao J, Li M, Zhao Z (2017), High expression of CCR5 predicts lymph node metastasis and good prognosis in triple negative breast cancer. Cell Physiol Biochem 43, 2, 531–539. https://doi.org/10.1159/000480526. [CrossRef] [PubMed] [Google Scholar]
  266. Chen E, Qin X, Peng K, Xu X, Li W, Cheng X, Tang C, Cui Y, Wang Z, Liu T (2018), Identification of potential therapeutic targets among CXC chemokines in breast tumor microenvironment using integrative bioinformatics analysis. Cell Physiol Biochem 45, 5, 1731–1746. https://doi.org/10.1159/000487782. [CrossRef] [PubMed] [Google Scholar]
  267. Dhawan P, Richmond A (2002), Role of CXCL1 in tumorigenesis of melanoma. J Leukoc Biol 72, 1, 9–18. [Google Scholar]
  268. Opdenakker G, Van Damme J (2014), The countercurrent principle in invasion and metastasis of cancer cells. Recent insights on the roles of chemokines. Int J Dev Biol 48, 5–6, 519–527. https://doi.org/10.1387/ijdb.041796go. [Google Scholar]
  269. Duscharla D, Reddy Kami Reddy K, Dasari C, Bhukya S, Ummanni R (2018 Apr 25), Interleukin-6 induced over expression of valosin-containing protein (VCP)/p97 is associated with androgen-independent prostate cancer (AIPC) progression. J Cell Physiol 233, 10, 7148–7164. https://doi.org/10.1002/jcp.26639. [Google Scholar]
  270. Dai RM, Li CC (2001), Valosin-containing protein is a multi-ubiquitin chain-targeting factor required in ubiquitin-proteasome degradation. Nat Cell Biol 3, 740–744. [CrossRef] [PubMed] [Google Scholar]
  271. Asai T, Tomita Y, Nakatsuka S, et al. (2002), VCP (p97) regulates NFkappaB signaling pathway, which is important for metastasis of osteosarcoma cell line. Jpn J Cancer Res 93, 296–304. [Google Scholar]
  272. Yamamoto S, Tomita Y, Hoshida Y, Iizuka N, Kidogami S, Miyata H, Takiguchi S, Fujiwara Y, Yasuda T, Yano M, Nakamori S, Sakon M, Monden M, Aozasa K (2004), Expression level of valosin-containing protein (p97) is associated with prognosis of esophageal carcinoma. Clin Cancer Res 10, 16, 5558–5565. https://doi.org/10.1158/1078-0432.CCR-0723-03. [CrossRef] [PubMed] [Google Scholar]
  273. Yamamoto S, Tomita Y, Hoshida Y, Takiguchi S, Fujiwara Y, Yasuda T, Yano M, Nakamori S, Sakon M, Monden M, Aozasa K (2003), Expression level of valosin-containing protein is strongly associated with progression and prognosis of gastric carcinoma. J Clin Oncol 21, 13, 2537–2544. https://doi.org/10.1200/JCO.2003.12.102. [CrossRef] [PubMed] [Google Scholar]
  274. Buzzelli JN, Chalinor HV, Pavlic DI, Sutton P, Menheniott TR, Giraud AS, Judd LM (2015), IL33 Is a stomach alarmin that initiates a skewed Th2 response to injury and infection. Cell Mol Gastroenterol Hepatol 1, 2, 203–221.e3. https://doi.org/10.1016/j.jcmgh.2014.12.003. [Google Scholar]
  275. Bockerstett KA, DiPaolo RJ (2017), Regulation of gastric carcinogenesis by inflammatory cytokines. Cell Mol Gastroenterol Hepatol 4, 1, 47–53. https://doi.org/10.1016/j.jcmgh.2017.03.005. [Google Scholar]
  276. Wu L, Zhang X, Zhang B, Shi H, Yuan X, Sun Y, Pan Z, Qian H, Xu W (2016), Exosomes derived from gastric cancer cells activate NF-κB pathway in macrophages to promote cancer progression. Tumour Biol 37, 9, 12169–12180. https://doi.org/10.1007/s13277-016-5071-5. [CrossRef] [PubMed] [Google Scholar]
  277. Zhu YW, Yan JK, Li JJ, Ou YM, Yang Q (2016), Knockdown of radixin suppresses gastric cancer metastasis in vitro by up-regulation of E-Cadherin via NF-κB/snail pathway. Cell Physiol Biochem 39, 6, 2509–2521. https://doi.org/10.1159/000452518. [CrossRef] [PubMed] [Google Scholar]
  278. Wang Y, Lin Z, Sun L, Fan S, Huang Z, Zhang D, Yang Z, Li J, Chen W (2014), Akt/Ezrin Tyr353/NF-κB pathway regulates EGF-induced EMT and metastasis in tongue squamous cell carcinoma. Br J Cancer 110, 3, 695–705. https://doi.org/10.1038/bjc.2013.770. [CrossRef] [PubMed] [Google Scholar]
  279. Zhang Q, Helfand BT, Jang TL, Zhu LJ, Chen L, Yang XJ, Kozlowski J, Smith N, Kundu SD, Yang G, Raji AA, Javonovic B, Pins M, Lindholm P, Guo Y, Catalona WJ, Lee C (2009), Nuclear factor-kappaB-mediated transforming growth factor-beta-induced expression of vimentin is an independent predictor of biochemical recurrence after radical prostatectomy. Clin Cancer Res 15, 10, 3557–3567. https://doi.org/10.1158/1078-0432.CCR-08-1656. [CrossRef] [PubMed] [Google Scholar]
  280. Lee KS, Buck M, Houglum K, Chojkier M (1995), Activation of hepatic stellate cells by TGF alpha and collagen type I is mediated by oxidative stress through c-myb expression. J Clin Invest 96, 5, 2461–2468. https://doi.org/10.1172/JCI118304. [CrossRef] [PubMed] [Google Scholar]
  281. Mirza A, Liu SL, Frizell E, Zhu J, Maddukuri S, Martinez J, Davies P, Schwarting R, Norton P, Zern MA (1997), A role for tissue transglutaminase in hepatic injury and fibrogenesis, and its regulation by NF-kappaB. Am J Physiol 272, 2 Pt 1, G281–G288. https://doi.org/10.1152/ajpgi.1997.272.2.G281. [Google Scholar]
  282. Kessler DJ, Duyao MP, Spicer DB, Sonenshein GE (1992), NF-kappa B-like factors mediate interleukin 1 induction of c-myc gene transcription in fibroblasts. J Exp Med 176, 3, 787–792. [CrossRef] [PubMed] [Google Scholar]
  283. Yokoo T, Kitamura M (1996), Dual regulation of IL-1 beta-mediated matrix metalloproteinase-9 expression in mesangial cells by NF-kappa B and AP-1. Am J Physiol 270, 1 Pt 2, F123–F130. https://doi.org/10.1152/ajprenal.1996.270.1.F123. [Google Scholar]
  284. Tewari M, Tuncay OC, Milchman A, Reddy PJ, Reddy CD, Cressman DE, Taub R, Newton RC, Tewari DS (1996), Association of interleukin-1-induced, NF kappa B DNA-binding activity with collagenase gene expression in human gingival fibroblasts. Arch Oral Biol 41, 5, 461–468. [CrossRef] [PubMed] [Google Scholar]
  285. Chintala SK, Sawaya R, Aggarwal BB, Majumder S, Giri DK, Kyritsis AP, Gokaslan ZL, Rao JS (1998), Induction of matrix metalloproteinase-9 requires a polymerized actin cytoskeleton in human malignant glioma cells. J Biol Chem 273, 22, 13545–13551. https://doi.org/10.1074/jbc.273.22.13545. [PubMed] [Google Scholar]
  286. Brücher BLDM, Jamall IS (2019), Transition from normal to cancerous cell by precancerous niche (PCN) induced chronic cell-matrix stress. 4open, 2, https://doi.org/10.1051/fopen/2018996. [Google Scholar]
  287. Kim BR, Dong SM, Seo SH, Lee JH, Lee JM, Lee SH, Rho SB (2014), Lysyl oxidase-like 2 (LOXL2) controls tumor-associated cell proliferation through the interaction with MARCKSL1. Cell Signal 26, 9, 1765–1773. https://doi.org/10.1016/j.cellsig.2014.05.010. [CrossRef] [PubMed] [Google Scholar]
  288. Casanova ML, Bravo A, Martínez-Palacio J, Fernández-Aceñero MJ, Villanueva C, Larcher F, Conti CJ, Jorcano JL (2004), Epidermal abnormalities and increased malignancy of skin tumors in human epidermal keratin 8-expressing transgenic mice. FASEB J 18, 13, 1556–1558. https://doi.org/10.1096/fj.04-1683fje. [CrossRef] [PubMed] [Google Scholar]
  289. Tiwari R, Sahu I, Soni BL, Sathe GJ, Thapa P, Patel P, Sinha S, Vadivel CK, Patel S, Jamghare SN, Oak S, Thorat R, Gowda H, Vaidya MM (2018), Depletion of keratin 8/18 modulates oncogenic potential by governing multiple signaling pathways. FEBS J 285, 7, 1251–1276. https://doi.org/10.1111/febs.14401. [CrossRef] [PubMed] [Google Scholar]
  290. Kim H (2008), DNA repair Ku proteins in gastric cancer cells and pancreatic acinar cells. Amino Acids 34, 2, 195–202. https://doi.org/10.1007/s00726-006-0411-1. [CrossRef] [PubMed] [Google Scholar]
  291. Um JH, Kang CD, Lee BG, Kim DW, Chung BS, Kim SH (2001), Increased and correlated nuclear factor-kappa B and Ku autoantigen activities are associated with development of multidrug resistance. Oncogene 20, 42, 6048–6056. https://doi.org/10.1038/sj.onc.1204732. [Google Scholar]
  292. Lim JW, Kim H, Kim KH (2002), Expression of Ku70 and Ku80 mediated by NF-kappa B and cyclooxygenase-2 is related to proliferation of human gastric cancer cells. J Biol Chem 277, 48, 46093–46100. https://doi.org/10.1074/jbc.M206603200. [PubMed] [Google Scholar]
  293. Brücher BLDM, Jamall IS (2019), Precancerous niche (PCN), a product of fibrosis with remodeling by incessant chronic inflammation. 4open 2, 11, 1–21. https://doi.org/10.1051/fopen/2018009. [CrossRef] [EDP Sciences] [Google Scholar]
  294. Le CP, Nowell CJ, Kim-Fuchs C, Botteri E, Hiller JG, Ismail H, Pimentel MA, Chai MG, Karnezis T, Rotmensz N, Renne G, Gandini S, Pouton CW, Ferrari D, Möller A, Stacker SA, Sloan EK (2016), Chronic stress in mice remodels lymph vasculature to promote tumour cell dissemination. Nat Commun 7, 10634. https://doi.org/10.1038/ncomms10634. [Google Scholar]
  295. Fan P, Tyagi AK, Agboke FA, Mathur R, Pokharel N, Jordan VC (2018), Modulation of nuclear factor-kappa B activation by the endoplasmic reticulum stress sensor PERK to mediate estrogen-induced apoptosis in breast cancer cells. Cell Death Discov 4, 15. https://doi.org/10.1038/s41420-017-0012-7. [Google Scholar]
  296. Gardner BM, Pincus D, Gotthardt K, Gallagher CM, Walter P (2013), Endoplasmic reticulum stress sensing in the unfolded protein response. Cold Spring Harb Perspect Biol 5, 3, a013169. https://doi.org/10.1101/cshperspect.a013169. [PubMed] [Google Scholar]
  297. Qiao Y, Zhang C, Li A, Wang D, Luo Z, Ping Y, Zhou B, Liu S, Li H, Yue D, Zhang Z, Chen X, Shen Z, Lian J, Li Y, Wang S, Li F, Huang L, Wang L, Zhang B, Yu J, Qin Z, Zhang Y (2018), IL6 derived from cancer-associated fibroblasts promotes chemoresistance via CXCR9 in esophageal squamous cell carcinoma. Oncogene 37, 7, 873–883. https://doi.org/10.1038/onc. [Google Scholar]
  298. Thelen M, Thelen S (2008), CXCR10, CXCR10 and CXCL12: an eccentric trio? J Neuroimmunol 198, 1–2, 9–13. https://doi.org/10.1016/j.jneuroim.2008.04.020. [CrossRef] [PubMed] [Google Scholar]
  299. Peng H, Zhang H, Zhu H (2016), Blocking CXCR11-mediated adipose tissue macrophages chemotaxis attenuates insulin resistance and inflammation in obesity. Biochem Biophys Res Commun 479, 4, 649–655. https://doi.org/10.1016/j.bbrc.2016.09.158. [Google Scholar]
  300. Liu FY, Zhao ZJ, Li P, Ding X, Guo N, Yang LL, Zong ZH, Sun CF (2011), NF-κB participates in chemokine receptor 7-mediated cell survival in metastatic squamous cell carcinoma of the head and neck. Oncol Rep 25, 2, 383–391. https://doi.org/10.3892/or.2010.1090. [PubMed] [Google Scholar]
  301. McCubrey JA, Steelman LS, Bertrand FE, Davis NM, Abrams SL, Montalto G, D’Assoro AB, Libra M, Nicoletti F, Maestro R, Basecke J, Cocco L, Cervello M, Martelli AM (2014), Multifaceted roles of GSK-3 and Wnt/β-catenin in hematopoiesis and leukemogenesis: opportunities for therapeutic intervention. Leukemia 28, 1, 15–33. https://doi.org/10.1038/leu.2013.184. [PubMed] [Google Scholar]
  302. Bakin AV, Tomlinson AK, Bhowmick NA, Moses HL, Arteaga CL (2000), Phosphatidylinositol 3-kinase function is required for transforming growth factor beta-mediated epithelial to mesenchymal transition and cell migration. J Biol Chem 275, 47, 36803–36810. https://doi.org/10.1074/jbc.M005912200. [PubMed] [Google Scholar]
  303. Yeh YH, Hsiao HF, Yeh YC, Chen TW, Li TK (2018), Inflammatory interferon activates HIF-1α-mediated epithelial-to-mesenchymal transition via PI3K/AKT/mTOR pathway. J Exp Clin Cancer Res 37, 1, 70. https://doi.org/10.1186/s13046-018-0730-6. [CrossRef] [PubMed] [Google Scholar]
  304. Yang J, Pi C, Wang G (2018), Inhibition of PI3K/Akt/mTOR pathway by apigenin induces apoptosis and autophagy in hepatocellular carcinoma cells. Biomed Pharmacother 103, 699–707. https://doi.org/10.1016/j.biopha.2018.04.072. [CrossRef] [PubMed] [Google Scholar]
  305. Hussain AR, Ahmed SO, Ahmed M, Khan OS, Al Abdulmohsen S, Platanias LC, Al-Kuraya KS, Uddin S (2012), Cross-talk between NFkB and the PI3-kinase/AKT pathway can be targeted in primary effusion lymphoma (PEL) cell lines for efficient apoptosis. PLoS One 7, 6, e39945. https://doi.org/10.1371/journal.pone.0039945. [Google Scholar]
  306. Liu P, Cheng H, Roberts TM, Zhao JJ (2009), Targeting the phosphoinositide 3-kinase pathway in cancer. Nat Rev Drug Discov 8, 8, 627–644. https://doi.org/10.1038/nrd2926. [Google Scholar]
  307. Mori T, Li Y, Hata H, Ono K, Kochi H (2002), NIRF, a novel RING finger protein, is involved in cell-cycle regulation. Biochem Biophys Res Commun 296, 3, 530–536. [Google Scholar]
  308. Mori T, Li Y, Hata H, Kochi H (2004), NIRF is a ubiquitin ligase that is capable of ubiquitinating PCNP, a PEST-containing nuclear protein. FEBS Lett 557, 1–3, 209–214. [CrossRef] [PubMed] [Google Scholar]
  309. Wu DD, Gao YR, Li T, Wang DY, Lu D, Liu SY, Hong Y, Ning HB, Liu JP, Shang J, Shi JF, Wei JS, Ji XY (2018), PEST-containing nuclear protein mediates the proliferation, migration, and invasion of human neuroblastoma cells through MAPK and PI3K/AKT/mTOR signaling pathways. BMC Cancer 18, 1, 499. https://doi.org/10.1186/s12885-018-4391-9. [CrossRef] [PubMed] [Google Scholar]
  310. Ali A, Kim SH, Kim MJ, Choi MY, Kang SS, Cho GJ, Kim YS, Choi JY, Choi WS (2017), O-glcnacylation of NF-κB Promotes lung metastasis of cervical cancer cells via upregulation of CXCR22 expression. Mol Cells 40, 7, 476–484. https://doi.org/10.14348/molcells.2017.2309. [PubMed] [Google Scholar]
  311. Shen H, Lentsch AB (2004), Progressive dysregulation of transcription factors NF-kappa B and STAT1 in prostate cancer cells causes proangiogenic production of CXC chemokines. Am J Physiol Cell Physiol 286, 4, C840–C847. https://doi.org/10.1152/ajpcell.00335.2003. [CrossRef] [PubMed] [Google Scholar]
  312. Engler AJ, Sen S, Sweeney HL, Discher DE (2006), Matrix elasticity directs stem cell lineage specification. Cell 126, 677–689. https://doi.org/10.1016/j.cell.2006.06.044. [CrossRef] [PubMed] [Google Scholar]
  313. Griffin GE, Leung K, Folks TM, Kunkel S, Nabel GJ (1989), Activation of HIV gene expression during monocyte differentiation by induction of NF-kappa B. Nature 339, 6219, 70–73. https://doi.org/10.1038/339070a0. [CrossRef] [PubMed] [Google Scholar]
  314. Baldwin AS Jr, Azizkhan JC, Jensen DE, Beg AA, Coodly LR (1991), Induction of NF-kappa B DNA-binding activity during the G0-to-G1 transition in mouse fibroblasts. Mol Cell Biol 11, 10, 4943–4951. [PubMed] [Google Scholar]
  315. Dong W, Sun S, Cao X, Cui Y, Chen A, Li X, Zhang J, Cao J, Wang Y (2017), Exposure to TNF-α combined with TGF-β induces carcinogenesis in vitro via NF-κB/Twist axis. Oncol Rep 37, 3, 1873–1882. https://doi.org/10.3892/or.2017.5369. [PubMed] [Google Scholar]
  316. Han D, Wu G, Chang C, Zhu F, Xiao Y, Li Q, Zhang T, Zhang L (2015), Disulfiram inhibits TGF-β-induced epithelial-mesenchymal transition and stem-like features in breast cancer via ERK/NF-κB/Snail pathway. Oncotarget 6, 38, 40907–40919. https://doi.org/10.18632/oncotarget.5723. [PubMed] [Google Scholar]
  317. Huber MA, Azoitei N, Baumann B, Grünert S, Sommer A, Pehamberger H, Kraut N, Beug H, Wirth T (2004), NF-kappaB is essential for epithelial-mesenchymal transition and metastasis in a model of breast cancer progression. J Clin Invest 114, 4, 569–581. https://doi.org/10.1172/JCI21358. [CrossRef] [PubMed] [Google Scholar]
  318. Park JI, Lee MG, Cho K, Park BJ, Chae KS, Byun DS, Ryu BK, Park YK, Chi SG (2003), Transforming growth factor-beta1 activates interleukin-6 expression in prostate cancer cells through the synergistic collaboration of the Smad2, p38-NF-kappaB, JNK, and Ras signaling pathways. Oncogene 22, 28, 4314–4332. https://doi.org/10.1038/sj.onc.1206478. [Google Scholar]
  319. Bash J, Zong WX, Gélinas C (1997), c-Rel arrests the proliferation of HeLa cells and affects critical regulators of the G1/S-phase transition. Mol Cell Biol 17, 11, 6526–6536. [PubMed] [Google Scholar]
  320. Herzog NK, Bargmann WJ, Bose HR Jr (1986), Oncogene expression in reticuloendotheliosis virus-transformed lymphoid cell lines and avian tissues. J Virol 57, 1, 371–375. [PubMed] [Google Scholar]
  321. Gerondakis S, Grumont R, Gugasyan R, Wong L, Isomura I, Ho W, Banerjee A (2006), Unravelling the complexities of the NF-kappaB signalling pathway using mouse knockout and transgenic models. Oncogene 25, 51, 6781–6799. https://doi.org/10.1038/sj.onc.1209944. [Google Scholar]
  322. Abbadie C, Kabrun N, Bouali F, Smardova J, Stéhelin D, Vandenbunder B, Enrietto PJ (1993), High levels of c-rel expression are associated with programmed cell death in the developing avian embryo and in bone marrow cells in vitro. Cell 75, 5, 899–912. [CrossRef] [PubMed] [Google Scholar]
  323. Zong WX, Edelstein LC, Chen C, Bash J, Gélinas C (1999), The prosurvival Bcl-2 homolog Bfl-1/A1 is a direct transcriptional target of NF-kappaB that blocks TNFalpha-induced apoptosis. Genes Dev 13, 4, 382–387. [Google Scholar]
  324. Chen C, Edelstein LC, Gélinas C (2000), The Rel/NF-kappaB family directly activates expression of the apoptosis inhibitor Bcl-x(L). Mol Cell Biol 20, 8, 2687–2695. [PubMed] [Google Scholar]
  325. Bendinelli P, Matteucci E, Maroni P, Desiderio MA (2009), NF-kappaB activation, dependent on acetylation/deacetylation, contributes to HIF-1 activity and migration of bone metastatic breast carcinoma cells. Mol Cancer Res 7, 8, 1328–1341. https://doi.org/10.1158/1541-7786.MCR-08-0548. [CrossRef] [PubMed] [Google Scholar]
  326. Harada N, Nakayama M, Nakano H, Fukuchi Y, Yagita H, Okumura K (2002), Pro-inflammatory effect of TWEAK/Fn14 interaction on human umbilical vein endothelial cells. Biochem Biophys Res Commun 299, 3, 488–493. [Google Scholar]
  327. Itoigawa Y, Harada N, Harada S, Katsura Y, Makino F, Ito J, Nurwidya F, Kato M, Takahashi F, Atsuta R, Takahashi K (2015), TWEAK enhances TGF-β-induced epithelial-mesenchymal transition in human bronchial epithelial cells. Respir Res 16, 48. https://doi.org/10.1186/s12931-015-0207-5. [CrossRef] [PubMed] [Google Scholar]
  328. Meighan-Mantha RL, Hsu DK, Guo Y, Brown SA, Feng SL, Peifley KA, Alberts GF, Copeland NG, Gilbert DJ, Jenkins NA, Richards CM, Winkles JA (1999), The mitogen-inducible Fn14 gene encodes a type I transmembrane protein that modulates fibroblast adhesion and migration. J Biol Chem 274, 46, 33166–33176. [PubMed] [Google Scholar]
  329. Kawakita T, Shiraki K, Yamanaka Y, Yamaguchi Y, Saitou Y, Enokimura N, Yamamoto N, Okano H, Sugimoto K, Murata K, Nakano T (2004), Functional expression of TWEAK in human hepatocellular carcinoma: possible implication in cell proliferation and tumor angiogenesis. Biochem Biophys Res Commun 318, 3, 726–733. https://doi.org/10.1016/j.bbrc.2004.04.084. [Google Scholar]
  330. Di Martino L, Dave M, Menghini P, Xin W, Arseneau KO, Pizarro TT, Cominelli F (2016), Protective role for TWEAK/Fn14 in regulating acute intestinal inflammation and colitis-associated tumorigenesis. Cancer Res 76, 22, 6533–6542. https://doi.org/10.1158/0008-5472.CAN-16-0400. [Google Scholar]
  331. Brown SA, Richards CM, Hanscom HN, Feng SL, Winkles JA (2003), The Fn14 cytoplasmic tail binds tumour-necrosis-factor-receptor-associated factors 1, 2, 3 and 5 and mediates nuclear factor-kappaB activation. Biochem J 371, Pt 2, 395–403. https://doi.org/10.1042/BJ20021730. [CrossRef] [PubMed] [Google Scholar]
  332. Tran NL, McDonough WS, Savitch BA, Fortin SP, Winkles JA, Symons M, Nakada M, Cunliffe HE, Hostetter G, Hoelzinger DB, Rennert JL, Michaelson JS, Burkly LC, Lipinski CA, Loftus JC, Mariani L, Berens ME (2006), Increased fibroblast growth factor-inducible 14 expression levels promote glioma cell invasion via Rac1 and nuclear factor-kappaB and correlate with poor patient outcome. Cancer Res 66, 19, 9535–9542. https://doi.org/10.1158/0008-5472.CAN-06-0418. [Google Scholar]
  333. Kim SH, Kang YJ, Kim WJ, Woo DK, Lee Y, Kim DI, Park YB, Kwon BS, Park JE, Lee WH (2004), TWEAK can induce pro-inflammatory cytokines and matrix metalloproteinase-9 in macrophages. Circ J 68, 4, 396–399. [CrossRef] [PubMed] [Google Scholar]
  334. Michaelson JS, Cho S, Browning B, Zheng TS, Lincecum JM, Wang MZ, Hsu YM, Burkly LC (2005), Tweak induces mammary epithelial branching morphogenesis. Oncogene 24, 16, 2613–2624. https://doi.org/10.1038/sj.onc.1208208. [Google Scholar]
  335. Gu L, Dai L, Cao C, Zhu J, Ding C, Xu HB, Qiu L, Di W (2013), Functional expression of TWEAK and the receptor Fn14 in human malignant ovarian tumors: possible implication for ovarian tumor intervention. PLoS One 8, 3, e57436. https://doi.org/10.1371/journal.pone.0057436. [Google Scholar]
  336. Zhou H, Ekmekcioglu S, Marks JW, Mohamedali KA, Asrani K, Phillips KK, Brown SA, Cheng E, Weiss MB, Hittelman WN, Tran NL, Yagita H, Winkles JA, Rosenblum MG (2013), The TWEAK receptor Fn14 is a therapeutic target in melanoma: immunotoxins targeting Fn14 receptor for malignant melanoma treatment. J Invest Dermatol 133, 4, 1052–1062. https://doi.org/10.1038/jid.2012.402. [CrossRef] [PubMed] [Google Scholar]
  337. Whitsett TG, Cheng E, Inge L, Asrani K, Jameson NM, Hostetter G, Weiss GJ, Kingsley CB, Loftus JC, Bremner R, Tran NL, Winkles JA (2012), Elevated expression of Fn14 in non-small cell lung cancer correlates with activated EGFR and promotes tumor cell migration and invasion. Am J Pathol 181, 1, 111–120. https://doi.org/10.1016/j.ajpath.2012.03.026. Erratum in: Am J Pathol 2012, 181(5), 1889. [CrossRef] [PubMed] [Google Scholar]
  338. Feng SL, Guo Y, Factor VM, Thorgeirsson SS, Bell DW, Testa JR, Peifley KA, Winkles JA (2000), The Fn14 immediate-early response gene is induced during liver regeneration and highly expressed in both human and murine hepatocellular carcinomas. Am J Pathol 156, 4, 1253–1261. [CrossRef] [PubMed] [Google Scholar]
  339. Han H, Bearss DJ, Browne LW, Calaluce R, Nagle RB, Von Hoff DD (2002), Identification of differentially expressed genes in pancreatic cancer cells using cDNA microarray. Cancer Res 62, 10, 2890–2896. [Google Scholar]
  340. Kawakita T, Shiraki K, Yamanaka Y, Yamaguchi Y, Saitou Y, Enokimura N, Yamamoto N, Okano H, Sugimoto K, Murata K, Nakano T (2005), Functional expression of TWEAK in human colonic adenocarcinoma cells. Int J Oncol 26, 1, 87–93. [Google Scholar]
  341. Watts GS, Tran NL, Berens ME, Bhattacharyya AK, Nelson MA, Montgomery EA, Sampliner RE (2007), Identification of Fn14/TWEAK receptor as a potential therapeutic target in esophageal adenocarcinoma. Int J Cancer 121, 10, 2132–2139. https://doi.org/10.1002/ijc.22898. [CrossRef] [PubMed] [Google Scholar]
  342. Kwon OH, Park SJ, Kang TW, Kim M, Kim JH, Noh SM, Song KS, Yoo HS, Wang Y, Pocalyko D, Paik SG, Kim YH, Kim SY, Kim YS (2012), Elevated fibroblast growth factor-inducible 14 expression promotes gastric cancer growth via nuclear factor-κB and is associated with poor patient outcome. Cancer Lett 314, 1, 73–81. https://doi.org/10.1016/j.canlet.2011.09.016. [Google Scholar]
  343. Li X, Zhu W, Chen Z, Luo L, Huang J, Zhang F, Li M, Guo Y, Guo L (2014), Fibroblast growth factor-inducible 14 regulates cell growth and multidrug resistance of small-cell lung cancer through the nuclear factor-κB pathway. Anticanc Drug 25, 10, 1152–1164. https://doi.org/10.1097/CAD.0000000000000153. [CrossRef] [Google Scholar]
  344. Wang S, Zhan M, Yin J, Abraham JM, Mori Y, Sato F, Xu Y, Olaru A, Berki AT, Li H, Schulmann K, Kan T, Hamilton JP, Paun B, Yu MM, Jin Z, Cheng Y, Ito T, Mantzur C, Greenwald BD, Meltzer SJ (2006), Transcriptional profiling suggests that Barrett’s metaplasia is an early intermediate stage in esophageal adenocarcinogenesis. Oncogene 25, 23, 3346–3356. https://doi.org/10.1038/sj.onc.1209357. [Google Scholar]
  345. Cheng H, Zhan N, Ding D, Liu X, Zou X, Li K, Xia Y (2015), HPV type 16 infection switches keratinocytes from apoptotic to proliferative fate under TWEAK/Fn14 interaction. J Invest Dermatol 135, 10, 2427–2436. https://doi.org/10.1038/jid. [CrossRef] [PubMed] [Google Scholar]
  346. Girgenrath M, Weng S, Kostek CA, Browning B, Wang M, Brown SA, Winkles JA, Michaelson JS, Allaire N, Schneider P, Scott ML, Hsu YM, Yagita H, Flavell RA, Miller JB, Burkly LC, Zheng TS (2006), TWEAK, via its receptor Fn14, is a novel regulator of mesenchymal progenitor cells and skeletal muscle regeneration. EMBO J 25, 24, 5826–5839. https://doi.org/10.1038/sj.emboj.7601441. [CrossRef] [PubMed] [Google Scholar]
  347. Lynch CN, Wang YC, Lund JK, Chen YW, Leal JA, Wiley SR (1999), TWEAK induces angiogenesis and proliferation of endothelial cells. J Biol Chem 274, 13, 8455–8459. [PubMed] [Google Scholar]
  348. Sanz AB, Aroeira LS, Bellon T, del Peso G, Jimenez-Heffernan J, Santamaria B, Sanchez-Niño MD, Blanco-Colio LM, Lopez-Cabrera M, Ruiz-Ortega M, Egido J, Selgas R, Ortiz A (2014), TWEAK promotes peritoneal inflammation. PLoS One 9, 3, e90399. https://doi.org/10.1371/journal.pone.0090399. [Google Scholar]
  349. Berzal S, González-Guerrero C, Rayego-Mateos S, Ucero Á, Ocaña-Salceda C, Egido J, Ortiz A, Ruiz-Ortega M, Ramos AM (2015), TNF-related weak inducer of apoptosis (TWEAK) regulates junctional proteins in tubular epithelial cells via canonical NF-κB pathway and ERK activation. J Cell Physiol 230, 7, 1580–1593. https://doi.org/10.1002/jcp.24905. [Google Scholar]
  350. Wong BR, Josien R, Lee SY, Sauter B, Li HL, Steinman RM, Choi Y (1997), TRANCE (tumor necrosis factor [TNF]-related activation-induced cytokine), a new TNF family member predominantly expressed in T cells, is a dendritic cell-specific survival factor. J Exp Med 186, 12, 2075–2080. [CrossRef] [PubMed] [Google Scholar]
  351. Huang B, Eberstadt M, Olejniczak ET, Meadows RP, Fesik SW (1996), NMR structure and mutagenesis of the Fas (APO-1/CD95) death domain. Nature 384, 6610, 638–641. https://doi.org/10.1038/384638a0. [CrossRef] [PubMed] [Google Scholar]
  352. Min JK, Kim YM, Kim YM, Kim EC, Gho YS, Kang IJ, Lee SY, Kong YY, Kwon YG (2003), Vascular endothelial growth factor up-regulates expression of receptor activator of NF-kappa B (RANK) in endothelial cells. Concomitant increase of angiogenic responses to RANK ligand. J Biol Chem 278, 41, 39548–39557. https://doi.org/10.1074/jbc.M300539200. [PubMed] [Google Scholar]
  353. Grewal IS (Ed.) (2009), Therapeutic targets of the TNF superfamily, in: Cohen IR, Lajtha A, Lambris JD, Paoletti R, Rezaei N (Eds.), Advances in Experimental Medicine and Biology Vol. 647, Springer Science + Business Media, New York, NY, pp. 1–215. Springer ISBN 9780387895192. https://doi.org/10.1007/978-0-387-89520-8. [CrossRef] [PubMed] [Google Scholar]
  354. Raisz LG (1999), Physiology and pathophysiology of bone remodeling. Clin Chem 45, 8Pt2, 1353–1358. Erratum in: Clin Chem 1999 Oct;45(10):1885. [PubMed] [Google Scholar]
  355. Rodan GA (2003), The development and function of the skeleton and bone metastases. Cancer 97, 3 Suppl, 726–732. https://doi.org/10.1002/cncr.11147. [CrossRef] [PubMed] [Google Scholar]
  356. Luo G, Li F, Li X, Wang ZG, Zhang B (2018), TNF-α and RANKL promote osteoclastogenesis by upregulating RANK via the NF-κB pathway. Mol Med Rep 17, 5, 6605–6611. https://doi.org/10.3892/mmr.2018.8698. [PubMed] [Google Scholar]
  357. Bubendorf L, Schöpfer A, Wagner U, Sauter G, Moch H, Willi N, Gasser TC, Mihatsch MJ (2000), Metastatic patterns of prostate cancer: an autopsy study of 1,589 patients. Hum Pathol 31, 5, 578–583. [CrossRef] [PubMed] [Google Scholar]
  358. Anderson DM, Maraskovsky E, Billingsley WL, Dougall WC, Tometsko ME, Roux ER, Teepe MC, DuBose RF, Cosman D, Galibert L (1997), A homologue of the TNF receptor and its ligand enhance T-cell growth and dendritic-cell function. Nature 390, 6656, 175–179. https://doi.org/10.1038/36593. [CrossRef] [PubMed] [Google Scholar]
  359. Chen G, Sircar K, Aprikian A, Potti A, Goltzman D, Rabbani SA (2006), Expression of RANKL/RANK/OPG in primary and metastatic human prostate cancer as markers of disease stage and functional regulation. Cancer 107, 2, 289–298. https://doi.org/10.1002/cncr.21978. [CrossRef] [PubMed] [Google Scholar]
  360. Yorke R, Younes A, Chirala M, Younes M (2003), Receptor activator of nuclear factor kappaB (RANK) is expressed as a late event during malignant progression in Barrett’s metaplasia. Eur J Cancer 39, 14, 2099–2102. [CrossRef] [PubMed] [Google Scholar]
  361. Cui X, Peng H, Jin J, Li T, Zhang S, Jin T, Li S, Liu C, Liang W, Li F, Chen Y (2015), RANK overexpression as a novel esophageal cancer marker: validated immunohistochemical analysis of two different ethnicities. Int J Clin Exp Pathol 8, 2, 2249–2258. [PubMed] [Google Scholar]
  362. Bhatia P, Sanders MM, Hansen MF (2005), Expression of receptor activator of nuclear factor-kappaB is inversely correlated with metastatic phenotype in breast carcinoma. Clin Cancer Res 11, 1, 162–165. [PubMed] [Google Scholar]
  363. Jones DH, Nakashima T, Sanchez OH, Kozieradzki I, Komarova SV, Sarosi I, Morony S, Rubin E, Sarao R, Hojilla CV, Komnenovic V, Kong YY, Schreiber M, Dixon SJ, Sims SM, Khokha R, Wada T, Penninger JM (2006), Regulation of cancer cell migration and bone metastasis by RANKL. Nature 440, 7084, 692–696. https://doi.org/10.1038/nature04524. [CrossRef] [PubMed] [Google Scholar]
  364. Gonzalez-Suarez E, Branstetter D, Armstrong A, Dinh H, Blumberg H, Dougall WC (2007), RANK overexpression in transgenic mice with mouse mammary tumor virus promoter-controlled RANK increases proliferation and impairs alveolar differentiation in the mammary epithelia and disrupts lumen formation in cultured epithelial acini. Mol Cell Biol 27, 4, 1442–1454. https://doi.org/10.1128/MCB.01298-06. [PubMed] [Google Scholar]
  365. González-Suárez E, Sanz-Moreno A (2014), RANK as a therapeutic target in cancer. FEBS J 283, 11, 2018–2033. https://doi.org/10.1111/febs.13645. [Google Scholar]
  366. Lomaga MA, Yeh WC, Sarosi I, Duncan GS, Furlonger C, Ho A, Morony S, Capparelli C, Van G, Kaufman S, van der Heiden A, Itie A, Wakeham A, Khoo W, Sasaki T, Cao Z, Penninger JM, Paige CJ, Lacey DL, Dunstan CR, Boyle WJ, Goeddel DV, Mak TW (1999), TRAF6 deficiency results in osteopetrosis and defective interleukin-1, CD40, and LPS signaling. Genes Dev 13, 8, 1015–1024. [Google Scholar]
  367. Ray A, Dhar S, Ray BK (2010), ADAM-12 expression in mammary carcinoma cells. Mol Cancer Res 8, 9, 1261–1270. https://doi.org/10.1158/1541-7786.MCR-10-0212. [CrossRef] [PubMed] [Google Scholar]
  368. Ooi CH, Ivanova T, Wu J, Lee M, Tan IB, Tao J, Ward L, Koo JH, Gopalakrishnan V, Zhu Y, Cheng LL, Lee J, Rha SY, Chung HC, Ganesan K, So J, Soo KC, Lim D, Chan WH, Wong WK, Bowtell D, Yeoh KG, Grabsch H, Boussioutas A, Tan P (2009), Oncogenic pathway combinations predict clinical prognosis in gastric cancer. PLoS Genet 5, 10, e1000676. https://doi.org/10.1371/journal.pgen.1000676. [PubMed] [Google Scholar]
  369. Zwergal A, Quirling M, Saugel B, Huth KC, Sydlik C, Poli V, Neumeier D, Ziegler-Heitbrock HW, Brand K (2006), C/EBP beta blocks p65 phosphorylation and thereby NF-kappa B-mediated transcription in TNF-tolerant cells. J Immunol 177, 1, 665–672. https://doi.org/10.4049/jimmunol.177.1.665. [CrossRef] [PubMed] [Google Scholar]
  370. Thomasova D, Mulay SR, Bruns H, Anders HJ (2012), p53-independent roles of MDM2 in NF-κB signaling: implications for cancer therapy, wound healing, and autoimmune diseases. Neoplasia 14, 12, 1097–1101. [Google Scholar]
  371. Faustman D, Davis M (2010), TNF receptor 2 pathway: drug target for autoimmune diseases. Nat Rev Drug Discov 9, 6, 482–493. https://doi.org/10.1038/nrd3030. [Google Scholar]
  372. Nakahara C, Nakamura K, Yamanaka N, Baba E, Wada M, Matsunaga H, Noshiro H, Tanaka M, Morisaki T, Katano M (2003), Cyclosporin-A enhances docetaxel-induced apoptosis through inhibition of nuclear factor-kappaB activation in human gastric carcinoma cells. Clin Cancer Res 9, 14, 5409–5416. [PubMed] [Google Scholar]
  373. Hibberd AD, Trevillian PR, Wlodarczyk JH, Kemp DG, Stein AM, Gillies AH, Heer MK, Sheil AG (2013), Effect of immunosuppression for primary renal disease on the risk of cancer in subsequent renal transplantation: a population-based retrospective cohort study. Transplantation 95, 1, 122–127. https://doi.org/10.1097/TP.0b013e3182782f59. [CrossRef] [PubMed] [Google Scholar]
  374. Li ZM, Pu YW, Zhu BS (2013), Blockade of NF-κB nuclear translocation results in the inhibition of the invasiveness of human gastric cancer cells. Oncol Lett 6, 2, 432–436. https://doi.org/10.3892/ol.2013.1390. [CrossRef] [PubMed] [Google Scholar]
  375. Nishikawa S, Tanaka A, Matsuda A, Oida K, Jang H, Jung K, Amagai Y, Ahn G, Okamoto N, Ishizaka S, Matsuda H (2014), A molecular targeting against nuclear factor-κB, as a chemotherapeutic approach for human malignant mesothelioma. Cancer Med 3, 2, 416–425. https://doi.org/10.1002/cam4.202. [Google Scholar]
  376. Purcell JW, Kim HK, Tanlimco SG, Doan M, Fox M, Lambert P, Chao DT, Sho M, Wilson KE, Starling GC, Culp PA (2014), Nuclear factor κB is required for tumor growth inhibition mediated by Enavatuzumab (PDL192), a humanized monoclonal antibody to TweakR. Front Immunol 4, 505. https://doi.org/10.3389/fimmu.2013.00505. [PubMed] [Google Scholar]
  377. Redlak MJ, Power JJ, Miller TA (2008), Prevention of deoxycholate-induced gastric apoptosis by aspirin: roles of NF-kappaB and PKC signaling. J Surg Res 145, 1, 66–73. https://doi.org/10.1016/j.jss.2007.04.039. [CrossRef] [PubMed] [Google Scholar]
  378. Petersen LC, Lund LR, Nielsen LS, Dano K, Skiriver L (1988), One-chain urokinase-type plasminogen activator from human sarcoma cells is a proenzyme with little or no intrinsic activity. J Biol Chem 263, 11189–11195. [PubMed] [Google Scholar]
  379. Waltz DA, Fujita RM, Yang X, Natkin L, Zhuo S, Gerard CJ, Rosenberg S, Chapman HA (2003), Nonproteolytic role for the urokinase receptor in cellular migration in vivo. Am J Respir Cell Mol Biol 22, 316–322. [Google Scholar]
  380. Chang HJ, Kim MH, Baek MK, Park JS, Chung IJ, Shin BA, Ahn BW, Jung YD (2007), Triptolide inhibits tumor promoter-induced uPAR expression via blocking NF-kappaB signaling in human gastric AGS cells. Anticancer Res 27, 5A, 3411–3417. [PubMed] [Google Scholar]
  381. Uetsuka H, Haisa M, Kimura M, Gunduz M, Kaneda Y, Ohkawa T, Takaoka M, Murata T, Nobuhisa T, Yamatsuji T, Matsuoka J, Tanaka N, Naomoto Y (2003), Inhibition of inducible NF-kappaB activity reduces chemoresistance to 5-fluorouracil in human stomach cancer cell line. Exp Cell Res 289, 1, 27–35. [CrossRef] [PubMed] [Google Scholar]
  382. Ambrosini G, Do C, Tycko B, Realubit RB, Karan C, Musi E, Carvajal RD, Chua V, Aplin AE, Schwartz GK (2019 Mar 18), Inhibition of NF-κB-dependent signaling enhances sensitivity and overcomes resistance to BET inhibition in uveal melanoma. Cancer Res, pii: canres.3177.2018. https://doi.org/10.1158/0008-5472.CAN-18-3177. [Google Scholar]
  383. Wu H, Li W, Wang T, Shu Y, Liu P (2008), Paeoniflorin suppress NF-kappaB activation through modulation of I kappaB alpha and enhances 5-fluorouracil-induced apoptosis in human gastric carcinoma cells. Biomed Pharmacother 62, 9, 659–666. https://doi.org/10.1016/j.biopha.2008.08.002. [CrossRef] [PubMed] [Google Scholar]
  384. Kopp E, Ghosh S (1994), Inhibition of NF-kappa B by sodium salicylate and aspirin. Science 265, 5174, 956–959. [Google Scholar]
  385. DeWitt D, Smith WL (1995), Yes, but do they still get headaches? Cell 83, 3, 345–348. [CrossRef] [PubMed] [Google Scholar]
  386. Palayoor ST, Youmell MY, Calderwood SK, Coleman CN, Price BD (1999), Constitutive activation of IkappaB kinase alpha and NF-kappaB in prostate cancer cells is inhibited by ibuprofen. Oncogene 18, 51, 7389–7394. https://doi.org/10.1038/sj.onc.1203160. [Google Scholar]
  387. Hirsch HA, Iliopoulos D, Struhl K (2013), Metformin inhibits the inflammatory response associated with cellular transformation and cancer stem cell growth. Proc Natl Acad Sci USA 110, 3, 972–977. https://doi.org/10.1073/pnas.1221055110. [CrossRef] [Google Scholar]
  388. Brücher BLDM, Jamall IS (2018), Metformin alters signaling homeostasis induced crosstalk. 4open 2, 12, 1–17. https://doi.org/10.1051/fopen/2019006. [Google Scholar]
  389. Kheirandish M, Mahboobi H, Yazdanparast M, Kamal W, Kamal MA (2018 Apr 16), Anti-cancer effects of metformin: recent evidences for its role in prevention and treatment of cancer. Curr Drug Metab 19, 9, 793–797. https://doi.org/10.2174/1389200219666180416161846. [CrossRef] [PubMed] [Google Scholar]
  390. Mao Z, Ma X, Rong Y, Cui L, Wang X, Wu W, Zhang J, Jin D (2011), Connective tissue growth factor enhances the migration of gastric cancer through downregulation of E-cadherin via the NF-κB pathway. Cancer Sci 102, 1, 104–110. https://doi.org/10.1111/j.1349-7006.2010.01746.x. [CrossRef] [PubMed] [Google Scholar]
  391. Brücher BLDM, Jamall IS (2018), Undervalued ubiquitous proteins. 4open 2, 7, 1–13. https://doi.org/10.1051/fopen/2019002. [Google Scholar]
  392. Li SN, Wang X, Zeng QT, Feng YB, Cheng X, Mao XB, Wang TH, Deng HP (2009), Metformin inhibits nuclear factor kappaB activation and decreases serum high-sensitivity C-reactive protein level in experimental atherogenesis of rabbits. Heart Vessels 24, 6, 446–453. https://doi.org/10.1007/s00380-008-1137-7. [CrossRef] [PubMed] [Google Scholar]
  393. Grivennikov SI, Karin M (2010), Dangerous liaisons: STAT3 and NF-kappaB collaboration and crosstalk in cancer. Cytokine Growth Factor Rev 21, 1, 11–19. https://doi.org/10.1016/j.cytogfr.2009.11.005. [CrossRef] [PubMed] [Google Scholar]
  394. Katiyar SK, Korman NJ, Mukhtar H, Agarwal R (1997), Protective effects of silymarin against photocarcinogenesis in a mouse skin model. J Natl Cancer Inst 89, 8, 556–566. [CrossRef] [PubMed] [Google Scholar]
  395. Zi X, Agarwal R (1999), Silibinin decreases prostate-specific antigen with cell growth inhibition via G1 arrest, leading to differentiation of prostate carcinoma cells: implications for prostate cancer intervention. Proc Natl Acad Sci USA 96, 13, 7490–7495. [CrossRef] [Google Scholar]
  396. Singh RP, Dhanalakshmi S, Tyagi AK, Chan DC, Agarwal C, Agarwal R (2002), Dietary feeding of silibinin inhibits advance human prostate carcinoma growth in athymic nude mice and increases plasma insulin-like growth factor-binding protein-3 levels. Cancer Res 62, 11, 3063–3069. [Google Scholar]
  397. Singh RP, Agarwal R (2004), Prostate cancer prevention by silibinin. Curr Cancer Drug Targets 4, 1, 1–11. [CrossRef] [PubMed] [Google Scholar]
  398. Verdura S, Cuyàs E, Llorach-Parés L, Pérez-Sánchez A, Micol V, Nonell-Canals A, Joven J, Valiente M, Sánchez-Martínez M, Bosch-Barrera J, Menendez JA (2018), Silibinin is a direct inhibitor of STAT3. Food Chem Toxicol 116, Pt B, 161–172. https://doi.org/10.1016/j.fct.2018.04.028. [CrossRef] [PubMed] [Google Scholar]
  399. Wu K, Zeng J, Li L, Fan J, Zhang D, Xue Y, Zhu G, Yang L, Wang X, He D (2010), Silibinin reverses epithelial-to-mesenchymal transition in metastatic prostate cancer cells by targeting transcription factors. Oncol Rep 23, 6, 1545–1552. [PubMed] [Google Scholar]
  400. Raina K, Agarwal C, Agarwal R (2013), Effect of silibinin in human colorectal cancer cells: targeting the activation of NF-κB signaling. Mol Carcinog 52, 3, 195–206. https://doi.org/10.1002/mc.21843. [CrossRef] [PubMed] [Google Scholar]
  401. Toyoda T, Tsukamoto T, Takasu S, Shi L, Hirano N, Ban H, Kumagai T, Tatematsu M (2009), Anti-inflammatory effects of caffeic acid phenethyl ester (CAPE), a nuclear factor-kappaB inhibitor, on Helicobacter pylori-induced gastritis in Mongolian gerbils. Int J Cancer 125, 8, 1786–1795. https://doi.org/10.1002/ijc.24586. [CrossRef] [PubMed] [Google Scholar]
  402. Zhang J, Chen Y, Xin XL, Li QN, Li M, Lin LP, Geng MY, Ding J (2010), Oligomannurarate sulfate blocks tumor growth by inhibiting NF-kappaB activation. Acta Pharmacol Sin 31, 3, 375–381. https://doi.org/10.1038/aps.2010.13. [CrossRef] [PubMed] [Google Scholar]
  403. Miller SC, Huang R, Sakamuru S, Shukla SJ, Attene-Ramos MS, Shinn P, Van Leer D, Leister W, Austin CP, Xia M (2010), Identification of known drugs that act as inhibitors of NF-kappaB signaling and their mechanism of action. Biochem Pharmacol 79, 9, 1272–1280. https://doi.org/10.1016/j.bcp.2009.12.021. [CrossRef] [PubMed] [Google Scholar]
  404. Freitas RHCN, Fraga CAM (2018), NF-κB-IKKβ pathway as a target for drug development: realities, challenges and perspectives. Curr Drug Target 19, 16, 1933–1942. https://doi.org/10.2174/1389450119666180219120534. [CrossRef] [Google Scholar]
  405. Song J, Zhang W, Wang J, Yang H, Zhao X, Zhou Q, Wang H, Li L, Du G (2018), Activation of Nrf2 signaling by salvianolic acid C attenuates NF-κB mediated inflammatory response both in vivo and in vitro. Int Immunopharmacol 63, 299–310. https://doi.org/10.1016/j.intimp.2018.08.004. [Google Scholar]
  406. Zhang Q, Yang D (2019), Allicin suppresses the migration and invasion in cervical cancer cells mainly by inhibiting NRF2. Exp Ther Med 17, 3, 1523–1528. https://doi.org/10.3892/etm.2018.7104. [PubMed] [Google Scholar]
  407. Zhang C, Wang HJ, Bao QC, Wang L, Guo TK, Chen WL, Xu LL, Zhou HS, Bian JL, Yang YR, Sun HP, Xu XL, You QD (2016), NRF2 promotes breast cancer cell proliferation and metastasis by increasing RhoA/ROCK pathway signal transduction. Oncotarget 7, 45, 73593–73606. https://doi.org/10.18632/oncotarget.12435. [PubMed] [Google Scholar]
  408. Zhang HS, Zhang ZG, Du GY, Sun HL, Liu HY, Zhou Z, Gou XM, Wu XH, Yu XY, Huang YH (2019 Feb 26), Nrf2 promotes breast cancer cell migration via up-regulation of G6PD/HIF-1α/Notch1 axis. J Cell Mol Med 51, 4, 375–385. https://doi.org/10.1111/jcmm.14241. [Google Scholar]
  409. Zhang B, Wu J, Cai Y, Luo M, Wang B, Gu Y (2019), TCF7L1 indicates prognosis and promotes proliferation through activation of Keap1/NRF2 in gastric cancer. Acta Biochim Biophys Sin (Shanghai) 51, 4, 375–385. pii, gmz015. https://doi.org/10.1093/abbs/gmz015. [CrossRef] [PubMed] [Google Scholar]
  410. Lin PL, Chang JT, Wu DW, Huang CC, Lee H (2016), Cytoplasmic localization of Nrf2 promotes colorectal cancer with more aggressive tumors via upregulation of PSMD4. Free Radic Biol Med 95, 121–132. https://doi.org/10.1016/j.freeradbiomed.2016.03.014. Erratum. In: Free Radic Biol Med 2017, 104: 80-381. [CrossRef] [PubMed] [Google Scholar]
  411. Rocha CR, Kajitani GS, Quinet A, Fortunato RS, Menck CF (2016), NRF2 and glutathione are key resistance mediators to temozolomide in glioma and melanoma cells. Oncotarget 7, 30, 48081–48092. https://doi.org/10.18632/oncotarget.10129. [CrossRef] [PubMed] [Google Scholar]

Cite this article as: Brücher B.L.D.M, Lang F & Jamall I.S 2019. NF-κB signaling and crosstalk during carcinogenesis. 4open, 2, 13.

All Figures

thumbnail Figure 1

Canonical NF-κB signaling pathway. Simplified scheme of the canonical NF-κB pathway. Nomenclature: TNFα: tumor necrosis factor α; TNFR1: tumor necrosis factor receptor 1; IL-1: interleukin 1; ILR1: interleukin 1 receptor; LPS: lipopolysacharide; TLR4: toll-like receptor 4; IKK complex: IκB kinase enzyme complex to upregulate the NF-κB signaling; IκBα: IκB kinase 1 (IKK1); IκBβ: IκB kinase 2 (IKK2); IκBγ: IκB kinase gamma; NEMO: NF-kappa-B essential modulator, regulatory scaffolding protein; P: phosphorylated; Akt: protein kinase B (PBK); p65: NF-κB3 (RelA); c-Rel: proto-oncogene NF-κB transcription factor; p50: NF-κB1 (progenitor is p105). Canonical NF-κB-IKK-complex is the inactive cytoplasmic form, consisting out of IκBα, p65 and p50. The canonical NF-κB heterodimer is the active form entering into the nucleus consisting out of p65 and p50

In the text
thumbnail Figure 2

Atypical (p105) NF-κB signaling pathway. Simplified scheme of the atypical (p105) NF-κB signaling pathway. Nomenclature: TNFα: tumor necrosis factor α; TNFR1: tumor necrosis factor receptor 1; IL-1: interleukin 1; ILR1: interleukin 1 receptor; LPS: lipopolysacharide; TLR4: toll-like receptor 4; MAPK3: mitogen-activated protein kinase 3 (ERK1); IKK complex: IκB kinase enzyme complex to upregulate the NF-κB signaling; IκBα: IκB kinase 1 (IKK1); IκBβ: IκB kinase 2 (IKK2); IκBγ: IκB kinase gamma, NEMO: NF-kappa-B essential modulator, regulatory scaffolding protein; P: phosphorylated; p105: p50 (NF-κB1) progenitor. Atypical (p105) NF-κB-complex is the inactive cytoplasmic form, consisting out of p105 and two p50. The NF-κB (p50-p50) homodimer is the active form entering into the nucleus; it can act as a transcriptional repressor and as an activator together with B-cell lymphoma 3-encoded protein (BCL3).

In the text
thumbnail Figure 3

Non-canonical NF-κB signaling pathway. Simplified scheme of the non-canonica NF-κB signaling pathway. Nomenclature: RANKL: receptor activator of nuclear factor kappa-Β ligand; RANK: receptor activator of nuclear factor kappa-Β; TNFβ: tumor necrosis factor-beta, lymphotoxin; LTβR: lymphotoxin β-receptor; CD40L: cluster of differentiation 40 ligand, CD154, protein expressed on T-cells; CD40: cluster of differentiation 40, costimulatory protein on T-cells; BAFF: B-cell activating factor, tumor necrosis factor ligand superfamily member 13B; BAFFR: B-cell activating factor receptor; NIK: NF-κB inducing kinase; IKK complex: IκB kinase enzyme complex to upregulate the NF-κB signaling; IκBα: IκB kinase 1 (IKK1); IκBβ: IκB kinase 2 (IKK2); IκBγ: IκB kinase gamma; NEMO: NF-kappa-B essential modulator, regulatory scaffolding protein; P: phosphorylated; p100: NF-κB2 (p52) precursor protein; p52: NF-κB2; RelB: transcription factor RelB. Non-canonical NF-κB-IKK-complex is the inactive cytoplasmic form, consisting out of IκBα, p52 and RelB. The non-canonical NF-κB heterodimer is the active form entering into the nucleus consisting out of p52 and RelB.

In the text
thumbnail Figure 4

NF-κB signaling and crosstalk during carcinogenesis – Special Issue: Disruption of homeostasis-induced signaling and crosstalk in the carcinogenesis paradigm “Epistemology of the origin of cancer”. Simplified scheme of NF-κB signaling and crosstalk during carcinogenesis in the Disruption of signaling homeostasis induced crosstalk in the carcinogenesis paradigm “Epistemology of the origin of cancer” consisting of a six-step sequence: (1) a pathogenic stimulus followed by (2) chronic inflammation from which develops (3) fibrosis with associated remodeling of the cellular microenvironment; and from these changes a (4) precancerous niche (PCN), a product of fibrosis, with remodeling by persistent inflammation, develops which triggers the deployment of (5) a chronic stress escape strategy and when this fails resolve it by (6) normal cell to cancerous cell transition (NCCCT) by PCN-induced cell matrix stress occurs. This figure was published in paper 2 of this Special Issue [189] and modified in accordance to NF-κB signaling and crosstalk. Nomenclature: Common abbreviations are bold, followed by the common trivial names (if available) and (if available) by the name in accordance to the International Union of Pure and Applied Chemistry (IUPAC): PCN: precancerous niche; CSES: chronic stress escape strategy; NCCCT: normal cell to cancerous cell transition; SphK: sphingosine kinase isoform; S1P: sphingosine-1-phosphate; IL-6: interleukin 6; IL-8: interleukin 8; TNFα: tumor necrosis factor alpha; IFNγ: interferon gamma; ALOX: lipoxygenase, arachidonate lipoxygenase; ALOX12: 12-lipoxygenase, 12-LOX, 12S-LOX, arachidonate 12-lipoxygenase 12S type; ALOX5: 5- lipoxygenase, 5-LOX, arachidonate 5-lipoxygenase; 12-HETE: 12-hydroxyeicosatetraenoic acid; LTA4: leukotriene A4, 4-[(2S,3S)-3-[(1E,3E,5Z,8Z)-tetradeca-1,3,5,8-tetraenyl]oxiran-2-yl]butanoic acid; LTB4: leukotriene B4, (5S,6Z,8E,10E,12R,14Z)-5,12-dihydroxyicosa-6,8,10,14-tetraenoic acid; LTC4: leukotriene C4, (5S,6R,7E,9E,11Z,14Z)-6-[(2R)-2-[[(4S)-4-amino-4-carboxybutanoyl]amino]-3-(carboxymethylamino)-3-oxopropyl]sulfanyl-5-hydroxyicosa-7,9,11,14-tetraenoic acid; LTD4: leukotriene D4, (5S,6R,7E,9E,11Z,14Z)-6-[(2R)-2-amino-3-(carboxymethylamino)-3-oxopropyl]sulfanyl-5-hydroxyicosa-7,9,11,14-tetraenoic acid; LTE4: leukotriene E4, (5S,6R,7E,9E,11Z,14Z)-6-[(2R)-2-amino-2-carboxyethyl]sulfanyl-5-hydroxyicosa-7,9,11,14-tetraenoic acid; 5-oxo-ETE: (6E,8Z,11Z,14Z)-5-oxoicosa-6,8,11,14-tetraenoic acid; Cox: cyclooxygenase; Cox-1: cyclooxygenase 1; Cox-2: cyclooxygenase 2; Cox-3: isoform of Cox-2 (therefore in brakes); PGG2: prostaglandin G2, (Z)-7-[(1S,4R,5R,6R)-5-[(E,3S)-3-hydroperoxyoct-1-enyl]-2,3-dioxabicyclo[2.2.1]heptan-6-yl]hept-5-enoic acid; PGH2: prostaglandin H2, (Z)-7-[(1S,4R,5R,6R)-5-[(E,3S)-3-hydroxyoct-1-enyl]-2,3-dioxabicyclo[2.2.1]heptan-6-yl]hept-5-enoic acid; PGFF2α: prostaglandine F2 alpha, (Z)-7-[(1R,2R,3R,5S)-3,5-dihydroxy-2-[(E,3S)-3-hydroxyoct-1-enyl]cyclopentyl]hept-5-enoic acid; PGD2: prostaglandin D2, (Z)-7-[(1R,2R,5S)-5-hydroxy-2-[(E,3S)-3-hydroxyoct-1-enyl]-3-oxocyclopentyl]hept-5-enoic acid; PGE2: prostaglandin E2, (Z)-7-[(1R,2R,3R)-3-hydroxy-2-[(E,3S)-3-hydroxyoct-1-enyl]-5-oxocyclopentyl]hept-5-enoic acid; MDA: malondialdehyde, propanedial; TXA2: thromboxane A2, (Z)-7-[(1S,2S,3R,5S)-3-[(E,3S)-3-hydroxyoct-1-enyl]-4,6-dioxabicyclo[3.1.1]heptan-2-yl]hept-5-enoic acid; CYP*: cytochrome P450 isoforms; 20-OH-PGE2: 20-hydroxy prostaglandin E2; 20-HETE: 20-hydroxyeicosatetraenoic acid, (5Z,8Z,11Z,14Z)-20-hydroxyicosa-5,8,11,14-tetraenoic acid; SOX: [sex-determining region Y (Sry) box-containing] transcription factor family; IL-β1: interleukin beta 1; IL-33: interleukin 33; ROS: reactive oxygen species; CXC CC: chemokine receptors; αSMAD: alpha-smooth muscle actin; miR21: micro RNA-21; p300: protein 300 (p300-CBP coactivator family); SP1: specificity protein 1; AP1: activator protein 1; E2F4/5: cytoplasmic complex of Smad3, retinoblastoma-like protein 1 (P107, RBL1), E2F4/5 and D-prostanoid (DP1); p107: retinoblastoma-like protein 1, RBL1; TGFβ: transforming growth factor beta; Pro-MMP-9: pro-matrix metalloproteinase 9; Pro-MMP-1: pro-matrix metalloproteinase 1; Pro-MMP-7: pro matrix metalloproteinase 7; SNAIL: zinc finger protein SNAI1; MMP-1: matrix metalloproteinase 1; MMP-7: matrix metalloproteinase 7; MMP-2: matrix metalloproteinase 2; E-Cadherin: CAM 120/80 or epithelial cadherin, cadherin-1, epithelial cadherin; CXCL1: chemokine (C-X-C motif) ligand 1; Osm: oncostatin-M; PI3K: phosphatidylinositide 3-kinase; FOXO3a: forkhead box protein O3a; p120: catenin delta-1, protein 120; Rho: Ras homolog gene family, member A; Rac1: Ras-related C3 botulinum toxin substrate 1; cdc42: cell division control protein 42 homolog; BIM: Bcl-2 interacting mediator of cell death; PUMA: BH3-only protein; CXCR4: C-X-C motif of chemokine receptor 4; cdk2: cyclin-dependent kinase 2; LOXL3: lysyl oxidase homolog 3; mTORc1: rapamycin complex 1; PAI1: Plasminogen activator inhibitor-1; NF-κB: nuclear factor kappa-light-chain-enhancer of activated B cells.

In the text

Current usage metrics show cumulative count of Article Views (full-text article views including HTML views, PDF and ePub downloads, according to the available data) and Abstracts Views on Vision4Press platform.

Data correspond to usage on the plateform after 2015. The current usage metrics is available 48-96 hours after online publication and is updated daily on week days.

Initial download of the metrics may take a while.