Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 1175 word(s) 1175 2021-01-05 04:04:13 |
2 format correct -21 word(s) 1154 2021-01-12 06:53:47 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Lu, T.; Motolani, A.; Martin, M.; Sun, M. Phosphorylation of the NF-κB regulators. Encyclopedia. Available online: https://encyclopedia.pub/entry/6287 (accessed on 19 April 2024).
Lu T, Motolani A, Martin M, Sun M. Phosphorylation of the NF-κB regulators. Encyclopedia. Available at: https://encyclopedia.pub/entry/6287. Accessed April 19, 2024.
Lu, Tao, Aishat Motolani, Matthew Martin, Mengyao Sun. "Phosphorylation of the NF-κB regulators" Encyclopedia, https://encyclopedia.pub/entry/6287 (accessed April 19, 2024).
Lu, T., Motolani, A., Martin, M., & Sun, M. (2021, January 11). Phosphorylation of the NF-κB regulators. In Encyclopedia. https://encyclopedia.pub/entry/6287
Lu, Tao, et al. "Phosphorylation of the NF-κB regulators." Encyclopedia. Web. 11 January, 2021.
Phosphorylation of the NF-κB regulators
Edit

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

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 [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. 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][12][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 . 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][24][25][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].

References

  1. Hanahan, D.; Weinberg, R.A. Hallmarks of Cancer: The Next Generation. Cell 2011, 144, 646–674.
  2. Sever, R.; Brugge, J.S. Signal Transduction in Cancer. Cold Spring Harb. Perspect. Med. 2015, 5, a006098.
  3. Freudlsperger, C.; Bian, Y.; Contag, S.; Burnett, J.; Coupar, J.; Yang, X.; Chen, Z.; Van Waes, C. TGF-β and NF-κB signal pathway cross-talk is mediated through TAK1 and SMAD7 in a subset of head and neck cancers. Oncogene 2013, 32, 1549–1559.
  4. Xia, Y.; Shen, S.; Verma, I.M. NF-κB, an active player in human cancers. Cancer Immunol. Res. 2014, 2, 823–830.
  5. Samuels, Y.; Ericson, K. Oncogenic PI3K and its role in cancer. Curr. Opin. Oncol. 2006, 18, 77–82.
  6. Sen, R.; Baltimore, D. Multiple nuclear factors interact with the immunoglobulin enhancer sequences. Cell 1986, 46, 705–716.
  7. Sen, R.; Baltimore, D. Inducibility of κ immunoglobulin enhancer-binding protein NF-κB by a posttranslational mechanism. Cell 1986, 47, 921–928.
  8. Tripathi, P.; Aggarwal, A. NF-κB transcription factor: A key player in the generation of immune response. Curr. Sci. 2006, 90, 519.
  9. Zhang, Q.; Lenardo, M.J.; Baltimore, D. Leading Edge Review 30 Years of NF-κB: A Blossoming of Relevance to Human Pathobiology. Cell 2017, 168, 37–57.
  10. Giuliani, C.; Bucci, I.; Napolitano, G. The role of the transcription factor Nuclear Factor-kappa B in thyroid autoimmunity and cancer. Front. Endocrinol. 2018, 9, 471.
  11. Oeckinghaus, A.; Ghosh, S. The NF-kappaB family of transcription factors and its regulation. Cold Spring Harb. Perspect. Biol. 2009, 1, a000034.
  12. Lu, T.; Stark, G.R. Cytokine overexpression and constitutive NF-κB in cancer. Cell Cycle 2004, 3, 1114–1117.
  13. Wei, H.; Prabhu, L.; Hartley, A.-V.; Martin, M.; Sun, E.; Jiang, G.; Liu, Y.; Lu, T. Methylation of NF-κB and its Role in Gene Regulation. In Gene Expression and Regulation in Mammalian Cells—Transcription from General Aspects; IntechOpen: London, UK, 2018; 291.
  14. Sun, S.C. The noncanonical NF-κB pathway. Immunol. Rev. 2012, 246, 125–140.
  15. Dolcet, X.; Llobet, D.; Pallares, J.; Matias-Guiu, X. NF-κB in development and progression of human cancer. Virchows Arch. 2005, 446, 475–482.
  16. Chen, E.; Li, C.C.H. Association of Cdk2/cyclin E and NF-κB complexes at G1/S phase. Biochem. Biophys. Res. Commun. 1998, 249, 728–734.
  17. Hinz, M.; Krappmann, D.; Eichten, A.; Heder, A.; Scheidereit, C.; Strauss, M. NF-κB Function in Growth Control: Regulation of Cyclin D1 Expression and G0/G1-to-S-Phase Transition. Mol. Cell. Biol. 1999, 19, 2690–2698.
  18. Park, M.; Hong, J. Roles of NF-κB in Cancer and Inflammatory Diseases and Their Therapeutic Approaches. Cells 2016, 5, 15.
  19. Li, J.; Lau, G.K.-K.; Chen, L.; Dong, S.; Lan, H.-Y.; Huang, X.-R.; Li, Y.; Luk, J.M.; Yuan, Y.-F.; Guan, X. Interleukin 17A Promotes Hepatocellular Carcinoma Metastasis via NF-κB Induced Matrix Metalloproteinases 2 and 9 Expression. PLoS ONE 2011, 6, e21816.
  20. Huang, S.; Pettaway, C.A.; Uehara, H.; Bucana, C.D.; Fidler, I.J. Blockade of NF-κB activity in human prostate cancer cells is associated with suppression of angiogenesis, invasion, and metastasis. Oncogene 2001, 20, 4188–4197.
  21. Inoue, J.I.; Gohda, J.; Akiyama, T.; Semba, K. NF-κB activation in development and progression of cancer. Cancer Sci. 2007, 98, 268–274.
  22. Kim, Y.A.; Lee, W.H.; Choi, T.H.; Rhee, S.H.; Park, K.Y.; Choi, Y.H. Involvement of p21WAF1/CIP1, pRB, Bax and NF-kappaB in induction of growth arrest and apoptosis by resveratrol in human lung carcinoma A549 cells. Int. J. Oncol. 2003, 23, 1143–1149.
  23. Wang, C.Y.; Cusack, J.C.; Liu, R.; Baldwin, A.S. Control of inducible chemoresistance: Enhanced anti-tumor therapy through increased apoptosis by inhibition of NF-κB. Nat. Med. 1999, 5, 412–417.
  24. Luo, J.-L.; Kamata, H.; Karin, M. IKK/NF-k κB signaling: Balancing life and death-a new approach to cancer therapy. J. Clin. Investig. 2005, 115, 2625–2632.
  25. Sevilla, L.; Zaldumbide, A.; Pognonec, P.; Boulukos, K.E. Transcriptional regulation of the bcl-x gene encoding the an-ti-apoptotic Bcl-xL protein by Ets, Rel/NFkappaB, STAT and AP1 transcription factor families. Histol. Histopathol. 2001, 16, 595–601.
  26. Tergaonkar, V.; Pando, M.; Vafa, O.; Wahl, G.; Verma, I. p53 stabilization is decreased upon NFκB activation: A role for NFκB in acquisition of resistance to chemotherapy. Cancer Cell 2002, 1, 493–503.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , ,
View Times: 287
Revisions: 2 times (View History)
Update Date: 12 Jan 2021
1000/1000