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 -- 1215 2023-01-30 20:40:48 |
2 update references and layout Meta information modification 1215 2023-01-31 03:34:04 |

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.
Bartolomeo, L.D.;  Vaccaro, F.;  Irrera, N.;  Borgia, F.;  Pomi, F.L.;  Squadrito, F.;  Vaccaro, M. Wnt Signaling Pathways, Inflammation and Carcinogenesis. Encyclopedia. Available online: https://encyclopedia.pub/entry/40620 (accessed on 29 April 2024).
Bartolomeo LD,  Vaccaro F,  Irrera N,  Borgia F,  Pomi FL,  Squadrito F, et al. Wnt Signaling Pathways, Inflammation and Carcinogenesis. Encyclopedia. Available at: https://encyclopedia.pub/entry/40620. Accessed April 29, 2024.
Bartolomeo, Luca Di, Federico Vaccaro, Natasha Irrera, Francesco Borgia, Federica Li Pomi, Francesco Squadrito, Mario Vaccaro. "Wnt Signaling Pathways, Inflammation and Carcinogenesis" Encyclopedia, https://encyclopedia.pub/entry/40620 (accessed April 29, 2024).
Bartolomeo, L.D.,  Vaccaro, F.,  Irrera, N.,  Borgia, F.,  Pomi, F.L.,  Squadrito, F., & Vaccaro, M. (2023, January 30). Wnt Signaling Pathways, Inflammation and Carcinogenesis. In Encyclopedia. https://encyclopedia.pub/entry/40620
Bartolomeo, Luca Di, et al. "Wnt Signaling Pathways, Inflammation and Carcinogenesis." Encyclopedia. Web. 30 January, 2023.
Wnt Signaling Pathways, Inflammation and Carcinogenesis
Edit

Wnt signaling is responsible for the regulation of different intracellular signal transduction pathways, which are essential for embryogenic development, cellular migration, polarization and differentiation as well as stem cell biology control and growth. Wnt signaling activation is related to the binding of Wnt ligands to a specific cell surface receptor which belongs to the Frizzled (Fzd) family, thus inducing the canonical (β-catenin-dependent) or non-canonical (β-catenin-independent) pathway.

Wnt signaling basal cell carcinoma squamous cell carcinoma

1. Wnt Signaling

The Fzd receptor interacts with other coreceptors, such as the receptor-like tyrosine kinase (Ryk), the receptor tyrosine kinase-like orphan receptor 2 (Ror2) or the low-density lipoprotein receptor-related protein (Lrp)-5/6, with the consequent activation of Disheveled (Dvl) and the downstream pathways [1]. Generally, cytoplasmatic β-catenin is phosphorylated by a complex formed by glycogen synthase kinase 3β (Gsk3β), adenomatous polyposis coli (Apc), casein kinase 1α (Ck1α) and Axin; β-catenin phosphorylation leads to its degradation by proteasome [2][3]. β-catenin is stabilized when the degradation complex is inhibited by Dvl following the canonical Wnt pathway activation [4]. β-catenin translocation into the nucleus allows the interaction with transcription factors, such as lymphoid-enhancing factor/T-cell factor (Lef/Tcf) transcription proteins, thus promoting Wnt target genes transcription [5]. Canonical Wnt signaling role starts from stem cell differentiation until cell proliferation, both during embryogenesis and adult tissue homeostasis [6].
The non-canonical Wnt signaling pathways are divided into Wnt/Calcium (Ca2+) and Wnt/Planar cell polarity (PCP) pathways [1]. In particular, in the Wnt/Calcium (Ca2+) pathway, the activated Fzd-co-receptor-Dvl complex induces phospholipase C γ, which converts phosphatidylinositol 4,5-bisphosphate (PIP2) into diacylglycerol (DAG) and inositol 1,4,5-triphosphate (IP3), resulting in the increased release of intracellular Ca2+. The release of Ca2+ leads to the activation of calcium-dependent kinases, such as Ca2+-dependent phosphatase calcineurin (CaN), Ca2+-calmodulin dependent kinase II (CAMKII) or protein kinase C (PKC) [7]. The activation of CaMKII may induce the phosphorylation of TGFβ-activated kinase 1 (TAK1), resulting in Nemo-like kinase (NLK) activation. TAK1-NLK pathway stimulation antagonizes the canonical Wnt/β-catenin pathway [8]; on the other hand, CaN induces the translocation of nuclear factor of activated T-cells (NFAT) family proteins into the nucleus, thus inducing the transcription of their target genes. In the Wnt/PCP pathway, activated Fzd-co-receptor-Dvl complex induces the activation of Rho family small GTPases, including RhoA, Rac and Cdc42 [9]. Cdc42 and Rac promote the c-Jun N-terminal kinase (JNK) signaling, resulting into the activating protein-1 (AP-1) complex activation [10]. On the other hand, RhoA induces the activation of Rho-associated kinase (ROCK) [11]. These pathways result in the regulation of cell motility and polarity [12][13].

2. Wnt Pathway, Inflammation and Carcinogenesis

Chronic inflammation is a widely recognized risk factor for several skin cancers, especially cutaneous squamous cell carcinoma (cSCC) [14]. The role of Wnt signaling in inflammation is complex, because it includes both anti- and proinflammatory functions [15]. Particularly, Wnt signaling would exert its immunomodulation acting on important inflammatory cytokines, such as the nuclear factor kappa B (NF-kB) and its target genes IL6, IL8 and TNFA, encoding tumor necrosis factor-α (TNF-α) [15][16][17][18]. Wnt signaling alterations are involved in the pathogenesis of cutaneous chronic inflammatory diseases, as well as those affecting the skin, such as psoriasis, and autoimmune diseases [16][19][20]. In psoriasis, non-canonical Wnt pathway increases keratinocyte proliferation and secretion of pro-inflammatory cytokines, such as TNF-α, interleukin-12 (IL-12) and (IL-23) [21]. Moreover, it is important to mention the role of canonical and non-canonical Wnt pathways in regulation of immune cells, particularly T cells and dendritic cells. The non-canonical Wnt pathway promotes migration of T cells via the CXC chemokine ligand-12 (CXCL12)–CXC chemokine receptor-4 (CXCR4) signaling [22]. Nevertheless, Wnt5a, via non-canonical Ca(2+)/CaMKII /NF-κB signaling, may also induce an abnormal phenotype of dendritic cells, which show an altered response to Toll-like receptor (TLR) ligands [23]. This phenotype is tolerogenic and characterized by increased production of IL-10 [23]. On the other hand, the canonical Wnt pathway represses the T regulatory cells function, promoting autoimmune response [24]. The imbalance of immune responses mediated by Wnt pathways may result in psoriatic inflammation but also in autoimmune disease, such as systemic lupus erythematosus (SLE) or systemic sclerosis (SSc) [21]. Canonical Wnt pathway is hyperactivated in SLE and plays a role in renal fibrosis of lupus nephritis, promoting the differentiation of T cell in Th17 clones [21]. An increased activation of Wnt signaling is present also in skin fibroblasts of patients with SSc, leading to skin fibrosis [21]. The deficiency of Wnt inhibitory factor-1 (WIF1) in fibroblasts of SSc patients is correlated with an hyperactivation of Wnt/β-catenin and, thus, with an increased production of collagen [25]. It is well known that reactive oxygen species (ROS) play an important role in fibrotic processes in SSc [26]. Preventing the accumulation of ROS in cultured SSc patient cells restored WIF-1 expression, thus avoiding collagen accumulation [25]. In conclusion, ROS promote Wnt activation, which contributes to fibrosis [25].
As in cutaneous inflammatory and autoimmune diseases, the Wnt pathway may modulate inflammatory responses in cutaneous cancers and influence tumor microenvironment (TME), thus promoting tumor development [27]. Wnt pathway and cancer are in relation to the complement system, which is involved not only in anti-tumor but also in pro-tumorigenic immune responses [14]. Complement component 3 (C3) and its active form, complement anaphylatoxin (C3a), may promote the epithelial-mesenchymal transition (EMT), thus giving invasive properties by decreasing E-cadherin expression [28]. C3a effects are also mediated by the transcription factor Twist [28], which induces the EMT through β-catenin [29]. In vitro experiments showed that exposure to C3 induces cyclin D1 and metalloproteases up-regulation which promote proliferation and migration of cSCC cells [30]. Moreover, C3a exposure stimulates β-catenin and Sox-2, a transcription factor which regulates cell stemness and induces pluripotent stem cells; in fact, C3a receptor silencing led to the down-regulation of β-catenin and Sox-2 with the consequent decrease of tumor volume [30]. The most important contribution of Wnt signaling to pro-tumorigenic inflammation is not limited to the complement system but also concerns T cells function: the thymic stromal lymphopoietin (TSLP) is a pro-inflammatory cytokine which antagonizes skin carcinogenesis by regulating the functions of CD8 and CD4 T cells [31]. TSLP-deficient mice show pro-tumorigenic inflammation and a predisposition to develop tumor growth through Wnt/β-catenin signaling involvement [31]. In fact, Wnt signaling may influence cancer immune-surveillance and facilitate a tumor immune escape, by reducing the recruitment of dendritic cells and by impairing the activity of T regulatory cells and cytotoxic T lymphocytes [32][33]. Tumors responding better to immunotherapy are characterized by a T cell-inflamed tumor microenvironment (TME), namely infiltrating antigen-specific T cells [33]. The TME may influence the growth and progression of tumors [33]. The activation of Wnt/β-catenin pathway prevent T-cell infiltration and activity in TME [33]. This results in progression of tumors and resistance to immunotherapy [33]. There are three main mechanisms underlying tumor immune escape by the Wnt/β-catenin pathway. First, the canonical Wnt pathway induces the activating transcription factor 3 (ATF3), thus inhibiting the transcription of C-C motif chemokine ligand 4 (CCL4) in a mouse model. This results in defective infiltration and activation of dendritic cells and T-cells. Another mechanism involved in immune escape concerns the crosstalk between tumor cells and tumor-associated macrophages (TAMs). Tumor cells may stimulate IL-1β production in TAMs via Snail, a transcription factor of canonical Wnt pathway. In turn, IL-β may increase the availability of β-catenin in tumor cells. Finally, canonical Wnt pathway may influence the activity of T regulatory cells, enhancing their survival [33]. A combination therapy based on the use of immune check point inhibitors plus Wnt signaling inhibitors may improve antitumor immunity [34].

References

  1. Veltri, A.; Lang, C.; Lien, W.H. Concise Review: Wnt Signaling Pathways in Skin Development and Epidermal Stem Cells. Stem Cells 2018, 36, 22–35.
  2. Liu, C.; Li, Y.; Semenov, M.; Han, C.; Baeg, G.H.; Tan, Y.; Zhang, Z.; Lin, X.; He, X. Control of beta-catenin phosphorylation/degradation by a dual-kinase mechanism. Cell 2002, 108, 837–847.
  3. Lee, E.; Salic, A.; Krüger, R.; Heinrich, R.; Kirschner, M.W. The roles of APC and Axin derived from experimental and theoretical analysis of the Wnt pathway. PLoS Biol. 2003, 1, E10.
  4. Axelrod, J.D.; Miller, J.R.; Shulman, J.M.; Moon, R.T.; Perrimon, N. Differential recruitment of Dishevelled provides signaling specificity in the planar cell polarity and Wingless signaling pathways. Genes Dev. 1998, 12, 2610–2622.
  5. Logan, C.Y.; Nusse, R. The Wnt signaling pathway in development and disease. Annu. Rev. Cell. Dev. Biol. 2004, 20, 781–810.
  6. Steinhart, Z.; Angers, S. Wnt signaling in development and tissue homeostasis. Development 2018, 145, dev146589.
  7. Sheldahl, L.C.; Slusarski, D.C.; Pandur, P.; Miller, J.R.; Kühl, M.; Moon, R.T. Dishevelled activates Ca2+ flux, PKC, and CamKII in vertebrate embryos. J. Cell Biol. 2003, 161, 769–777.
  8. Ishitani, T.; Kishida, S.; Hyodo-Miura, J.; Ueno, N.; Yasuda, J.; Waterman, M.; Shibuya, H.; Moon, R.T.; Ninomiya-Tsuji, J.; Matsumoto, K. The TAK1-NLK mitogen-activated protein kinase cascade functions in the Wnt-5a/Ca(2+) pathway to antagonize Wnt/beta-catenin signaling. Mol. Cell Biol. 2003, 23, 131–139.
  9. Schlessinger, K.; Hall, A.; Tolwinski, N. Wnt signaling pathways meet Rho GTPases. Genes Dev. 2009, 23, 265–277.
  10. Yamanaka, H.; Moriguchi, T.; Masuyama, N.; Kusakabe, M.; Hanafusa, H.; Takada, R.; Takada, S.; Nishida, E. JNK functions in the non-canonical Wnt pathway to regulate convergent extension movements in vertebrates. EMBO Rep. 2002, 3, 69–75.
  11. Habas, R.; Kato, Y.; He, X. Wnt/Frizzled activation of Rho regulates vertebrate gastrulation and requires a novel Formin homology protein Daam1. Cell 2001, 107, 843–854.
  12. Endo, Y.; Wolf, V.; Muraiso, K.; Kamijo, K.; Soon, L.; Uren, A.; Barshishat-Küpper, M.; Rubin, J.S. Wnt-3a-dependent cell motility involves RhoA activation and is specifically regulated by dishevelled-2. J. Biol. Chem. 2005, 280, 777–786.
  13. Schlessinger, K.; McManus, E.J.; Hall, A. Cdc42 and noncanonical Wnt signal transduction pathways cooperate to promote cell polarity. J. Cell Biol. 2007, 178, 355–361.
  14. Riihilä, P.; Nissinen, L.; Knuutila, J.; Rahmati Nezhad, P.; Viiklepp, K.; Kähäri, V.M. Complement System in Cutaneous Squamous Cell Carcinoma. Int. J. Mol. Sci. 2019, 20, 3550.
  15. Ma, B.; Hottiger, M.O. Crosstalk between Wnt/β-Catenin and NF-κB Signaling Pathway during Inflammation. Front. Immunol. 2016, 7, 378.
  16. Jridi, I.; Canté-Barrett, K.; Pike-Overzet, K.; Staal, F.J.T. Inflammation and Wnt Signaling: Target for Immunomodulatory Therapy? Front. Cell. Dev. Biol. 2021, 8, 615131.
  17. Picciolo, G.; Pallio, G.; Altavilla, D.; Vaccaro, M.; Oteri, G.; Irrera, N.; Squadrito, F. β-Caryophyllene Reduces the Inflammatory Phenotype of Periodontal Cells by Targeting CB2 Receptors. Biomedicines 2020, 8, 164.
  18. Custurone, P.; Di Bartolomeo, L.; Irrera, N.; Borgia, F.; Altavilla, D.; Bitto, A.; Pallio, G.; Squadrito, F.; Vaccaro, M. Role of Cytokines in Vitiligo: Pathogenesis and Possible Targets for Old and New Treatments. Int. J. Mol. Sci. 2021, 22, 11429.
  19. Irrera, N.; Bitto, A.; Vaccaro, M.; Mannino, F.; Squadrito, V.; Pallio, G.; Arcoraci, V.; Minutoli, L.; Ieni, A.; Lentini, M.; et al. PDRN, a Bioactive Natural Compound, Ameliorates Imiquimod-Induced Psoriasis through NF-κB Pathway Inhibition and Wnt/β-Catenin Signaling Modulation. Int. J. Mol. Sci. 2020, 21, 1215.
  20. Picciolo, G.; Mannino, F.; Irrera, N.; Altavilla, D.; Minutoli, L.; Vaccaro, M.; Arcoraci, V.; Squadrito, V.; Picciolo, G.; Squadrito, F.; et al. PDRN, a natural bioactive compound, blunts inflammation and positively reprograms healing genes in an "in vitro" model of oral mucositis. Biomed. Pharmacother. 2021, 138, 111538.
  21. Tian, F.; Mauro, T.M.; Li, Z. The pathological role of Wnt5a in psoriasis and psoriatic arthritis. J. Cell. Mol. Med. 2019, 23, 5876–5883.
  22. Ghosh, M.C.; Collins, G.D.; Vandanmagsar, B.; Patel, K.; Brill, M.; Carter, A.; Lustig, A.; Becker, K.G.; Wood, W.W., 3rd; Emeche, C.D.; et al. Activation of Wnt5A signaling is required for CXC chemokine ligand 12-mediated T-cell migration. Blood 2009, 114, 1366–1373.
  23. Valencia, J.; Hernández‐López, C.; Martínez, V.G.; Hidalgo, L.; Zapata, A.G.; Vicente, Á.; Varas, A.; Sacedón, R. Wnt5a skews dendritic cell differentiation to an unconventional phenotype with tolerogenic features. J. Immunol. 2011, 187, 4129–4139.
  24. van Loosdregt, J.; Fleskens, V.; Tiemessen, M.M.; Mokry, M.; van Boxtel, R.; Meerding, J.; Pals, C.E.; Kurek, D.; Baert, M.R.; Delemarre, E.M.; et al. Canonical Wnt signaling negatively modulates regulatory T cell function. Immunity 2013, 39, 298–310.
  25. Svegliati, S.; Marrone, G.; Pezone, A.; Spadoni, T.; Grieco, A.; Moroncini, G.; Grieco, D.; Vinciguerra, M.; Agnese, S.; Jüngel, A.; et al. Oxidative DNA damage induces the ATM-mediated transcriptional suppression of the Wnt inhibitor WIF-1 in systemic sclerosis and fibrosis. Sci. Signal 2014, 7, ra84.
  26. Bagnato, G.L.; Irrera, N.; Pizzino, G.; Santoro, D.; Roberts, W.N.; Bagnato, G.; Pallio, G.; Vaccaro, M.; Squadrito, F.; Saitta, A.; et al. Dual αvβ3 and αvβ5 blockade attenuates fibrotic and vascular alterations in a murine model of systemic sclerosis. Clin. Sci. 2018, 132, 231–242.
  27. Mantovani, A.; Allavena, P.; Sica, A.; Balkwill, F. Cancer-related inflammation. Nature 2008, 454, 436–444.
  28. Cho, M.S.; Rupaimoole, R.; Choi, H.J.; Noh, K.; Chen, J.; Hu, Q.; Sood, A.K.; Afshar-Kharghan, V. Complement Component 3 Is Regulated by TWIST1 and Mediates Epithelial-Mesenchymal Transition. J. Immunol. 2016, 196, 1412–1418.
  29. Li, J.; Zhou, B.P. Activation of β-catenin and Akt pathways by Twist are critical for the maintenance of EMT associated cancer stem cell-like characters. BMC Cancer 2011, 11, 49.
  30. Fan, Z.; Qin, J.; Wang, D.; Geng, S. Complement C3a promotes proliferation, migration and stemness in cutaneous squamous cell carcinoma. J. Cell. Mol. Med. 2019, 23, 3097–3107.
  31. Di Piazza, M.; Nowell, C.S.; Koch, U.; Durham, A.D.; Radtke, F. Loss of cutaneous TSLP-dependent immune responses skews the balance of inflammation from tumor protective to tumor promoting. Cancer Cell. 2012, 22, 479–493.
  32. Spranger, S.; Gajewski, T.F. A new paradigm for tumor immune escape: β-catenin-driven immune exclusion. J. Immunother. Cancer 2015, 3, 43.
  33. Li, X.; Xiang, Y.; Li, F.; Yin, C.; Li, B.; Ke, X. WNT/β-Catenin Signaling Pathway Regulating T Cell-Inflammation in the Tumor Microenvironment. Front. Immunol. 2019, 10, 2293.
  34. Takeuchi, Y.; Tanegashima, T.; Sato, E.; Irie, T.; Sai, A.; Itahashi, K.; Kumagai, S.; Tada, Y.; Togashi, Y.; Koyama, S.; et al. Highly immunogenic cancer cells require activation of the WNT pathway for immunological escape. Sci. Immunol. 2021, 6, eabc6424.
More
Information
Subjects: Dermatology
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: 418
Revisions: 2 times (View History)
Update Date: 31 Jan 2023
1000/1000