Phosphorylation of the NF-κB regulators: History
Please note this is an old version of this entry, which may differ significantly from the current revision.

The nuclear factor kappa B (NF-κB) is a ubiquitous transcription factor central to inflammation and various malignant diseases in humans. The regulation of NF-κB can be influenced by a myriad of post-translational modifications (PTMs), including phosphorylation, one of the most popular PTM formats in NF-κB signaling. The regulation by phosphorylation modification is not limited to NF-κB subunits, but it also encompasses the diverse regulators of NF-κB signaling. The differential site-specific phosphorylation of NF-κB itself or some NF-κB regulators can result in dysregulated NF-κB signaling, often culminating in events that induce cancer progression and other hyper NF-κB related diseases, such as inflammation, cardiovascular diseases, diabetes, as well as neuro-degenerative diseases, etc.

  • cancer signaling
  • NF-κB
  • phosphorylation
  • PRMT5
  • YBX1

Note: The following contents are extract from your paper. The entry will be online only after author check and submit it.

1. Brief Overview of Cancer and Key Signaling Pathways

Cancer is a diverse and multifactorial genetic disease that arises through a multistep accumulation of genetic alterations, which causes genomic instability in a cell. This genomic instability results in aberrant cellular functions, such as uncontrolled growth, cell death resistance, increased cell migration and invasion, evasion of immune surveillance, metabolic reprograming, etc. [1]. The progression of cancer is further driven by the complex interaction of malignant cells with neighboring cells in their microenvironment [2].

Genetic mutations in multiple signaling pathways have been linked to cancer progression. Several typical examples include the receptor tyrosine kinase/Ki-ras2 Kirsten rat sarcoma viral oncogene homolog (RTK/KRAS) pathway, tumor protein P53 (p53) pathway, transforming growth factor β (TGFβ) pathway, and phosphoinositide 3-kinase/Akt (PI-3-kinase/Akt) pathway, etc. Interestingly, ample evidences suggest that these signaling pathways frequently promote cancer progression through the nuclear factor κB (NF-κB) activation [3,4]. For example, KRAS oncogenic mutation and p53 loss of function mutation, which is present in approximately 25% and 50% of human tumors, respectively, leads to the constitutive activation of NF-κB, thereby promoting cell survival in multiple cancers. [4]. Similarly, mutations in the PI-3-Kinase/Akt pathway, which exist in over 30% of solid tumors, promotes the activation of components in NF-κB pathway [2,5]. These examples, among many others, demonstrate the complex signaling interactions in cancer and the pivotal role that NF-κB plays in enabling cancer progression. Hence, it is of great clinical importance to fully understand the different facets of NF-κB regulation in cancer. In this review, we will further elaborate on the complexity of NF-κB regulation in cancer, with the goal of providing deep insight and aiding the development strategies of novel NF-κB targeted cancer therapeutics.

2. Overview of NF-κB Signaling

Gene transcription plays a fundamental role in mediating several biological processes. Thus, strict regulation of transcription factors is necessary to maintain cellular homeostasis. One such transcription factor is NF-κB. The omnipresent NF-κB is a group of homo- and hetero-dimeric proteins, which was discovered about three decades ago in B lymphocytes. NF-κB was found to bind to a B motif at the enhancer element of the κ light-chain gene to regulate its expression [6,7]. The κB motif, as it is currently termed, consists of the following sequence: 5′‐GGGRNNYYCC‐3′; wherein Y = pyrimidine, R = purine, and N, any nucleotide. After years of continuous studies on NF-κB, additional mechanistic insights into the roles of NF-κB and its signaling cascade—beyond B-cells—have been elucidated [8]. Mammalian NF-κB is composed of five-member subunits that dimerize at gene promoters to control differential gene expression. The members of the NF-κB family include p65/RelA, RelB, c-Rel, p50/p105 (NF-κB1), and p52/p100 (NF-κB2) [9]. These subunits are characterized by the Rel-homology domains in their N-terminal, which contains a DNA-binding domain, a nuclear localization sequence, and a dimerization domain [8]. Additionally, p65, RelB, and c-Rel contain a transactivation domain in their C-terminal, enabling their transcriptional activity. In contrast, the C-terminal region of p105 and p100, the precursors of p50 and p52 respectively, lacks a transactivation domain but contains several ankyrin repeat sequences that function to inhibit NF-κB [10]. As shown in Figure 1, the mechanism of NF-κB induction is grouped into two pathways: canonical and non-canonical pathways. In the canonical pathway, external stimuli such as growth factors and cytokines bind to NF-κB cell-surface receptors to activate the phosphorylation of Inhibitor of κB (IκB) by Inhibitor of κB kinase (IKK). This phosphorylation results in IκBα degradation, causing the translocation of p65/p50 dimer to the nucleus and enabling its binding to the respective κB elements on the genes [11–13] (Figure 1). Comparably, the non-canonical pathway involves signaling via Cluster of differentiation 40 (CD40) receptor, Lymphotoxin-β receptor (LTβR), and BLyS receptor 3 (BR3) receptors, triggering NF-κB -inducing kinase (NIK) phosphorylation of IKKα dimers, and subsequent phosphorylation of p100 by IKKα [9]. This phosphorylation cascade triggers the translocation of RelB/p52 dimers into the nucleus to modulate gene transcription [14] (Figure1).

3. Implication of NF-κB Signaling in Cancer

Considering the unique mechanism of gene regulation, NF-κB has been implicated in a diverse range of cellular processes such as inflammation, cell survival, and cell differentiation. Notably, there has been a growing amount of evidence indicating the pivotal role of NF-κB in cancer initiation and progression [15]. NF-κB is highly involved in cell proliferation via the regulation of cell cycle proteins. For instance, NF-κB was reported to trigger cyclin D1 expression in breast carcinoma cells and was found to interact with cyclin-dependent kinase 2 (CDK2) and cyclin E complex in lymphocytes [16,17]. Additionally, NF-κB has been shown to contribute to most cancer hallmarks including promoting metastasis, enabling angiogenesis, altering the tumor microenvironment, evading apoptosis, among others, in different tumor types [18]. Cytokines such as interleukin-17A (IL-17A) was shown to cause metastasis in hepatocellular carcinoma (HCC) by upregulating the levels of metalloproteinases (MMP) 2 and 9 through NF-κB induction [19]. Additionally, the constitutive activity of NF-κB in human prostate tumors was reported to be associated with the expression of key angiogenesis promoters such as vascular endothelial growth factor (VEGF), MMP 9, and interleukin-8 (IL-8) [20]. Unsurprisingly, the tumor microenvironment, which consists of various immune cells, is invariably transformed into a pro-tumorigenic microenvironment through NF-κB signaling. NF-κB activates the expression of distinct pro-inflammatory cytokines that engage in a feedback-loop to promote NF-κB dependent transcription of oncogenes [21]. Alongside enabling tumor growth, NF-κB also plays a vital role in preventing apoptosis in many cancers. For instance, inhibition of NF-κB activity triggered apoptosis in both lung cancer and colorectal cancer cell lines [22,23]. Cancer cells can evade apoptosis by upregulating a number of NF-κB-dependent anti-apoptotic genes such as B-cell lymphoma-extra-large (Bcl-xL), FLICE-inhibitory protein (FLIP), cellular inhibitor of apoptosis protein (c-IAP), and mouse double minute 2 homolog (Mdm2), a negative regulator of p53. [23–26]. p53 plays a key role in preserving the genomic integrity of a cell in response to cellular stress by activating cell cycle arrest or inducing apoptosis. Thus, NF-κB signaling has been speculated to hinder p53-induced apoptosis in response to chemotherapeutic agents used for cancer treatment [26].

This entry is adapted from the peer-reviewed paper 10.3390/biom11010015

This entry is offline, you can click here to edit this entry!