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 + 1826 word(s) 1826 2022-03-18 05:02:35 |
2 corrected the format -35 word(s) 1791 2022-03-25 06:56:03 |

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.
Li, Q. EZH2 and Endometrial Cancer Development. Encyclopedia. Available online: https://encyclopedia.pub/entry/21004 (accessed on 27 July 2024).
Li Q. EZH2 and Endometrial Cancer Development. Encyclopedia. Available at: https://encyclopedia.pub/entry/21004. Accessed July 27, 2024.
Li, Qinglei. "EZH2 and Endometrial Cancer Development" Encyclopedia, https://encyclopedia.pub/entry/21004 (accessed July 27, 2024).
Li, Q. (2022, March 24). EZH2 and Endometrial Cancer Development. In Encyclopedia. https://encyclopedia.pub/entry/21004
Li, Qinglei. "EZH2 and Endometrial Cancer Development." Encyclopedia. Web. 24 March, 2022.
EZH2 and Endometrial Cancer Development
Edit

Enhancer of zeste homolog 2 (EZH2), a core component of polycomb repressive complex 2, plays an important role in cancer development. As both oncogenic and tumor suppressive functions of EZH2 have been documented in the literature, the objective of this study is to determine the impact of Ezh2 deletion on the development and progression of endometrial cancer induced by inactivation of phosphatase and tensin homolog (PTEN), a tumor suppressor gene frequently dysregulated in endometrial cancer patients.

Ezh2 endometrial cancer Pten

1. Introduction

Endometrial cancer is the most common cancer in the genital tract in women, with approximately 65,570 new cases and 12,940 deaths each year in the United States [1]. Endometrial cancer is classified into two distinct types [2]. The type I cancer represents the major type (~90%) and is often companied by endometrial hyperplasia [2][3]. The type II cancer accounts for ~10% of the total cases and is more aggressive than the type I cancer [2][3][4][5]. Histologically, the type I cancer is endometrioid carcinoma while the type II cancer consists of several subtypes, including serous carcinoma and clear-cell carcinoma [6]. Notably, the type I, but not the type II, endometrial cancer is related to estrogen stimulation [7]. Using molecular sequencing technologies, endometrial cancer has been classified into the following types by The Cancer Genome Atlas (TCGA) Research Network: DNA polymerase epsilon catalytic subunit (POLE) (ultramutated), microsatellite-instability (MSI) (hypermutated), copy-number low, as well as copy-number high [8]. To facilitate the classification in clinical practice, the Proactive Molecular Risk Classifier for Endometrial Cancer (ProMisE) has been developed and validated, with the inclusion of immunohistochemical analysis of DNA mismatch repair (MMR) protein and tumor protein p53 (TP53) [9][10][11]. Interestingly, a report shows that a combination of tumor-infiltrating lymphocytes pattern and MMR may be used as a surrogate for the POLE mutation group [12]. ProMisE has been used in molecular diagnosis of human endometrial cancer [13].
Significant challenges remain for endometrial cancer treatment. Determining the histological subtype of endometrial cancer is an effective strategy that guides cancer treatment, with an emerging need to incorporate more molecular details into clinical interventions [14]. While surgery remains to be the most common option to treat this gynecological malignancy, new therapeutic strategies targeting actionable mutations and/or molecular pathways are potentially valuable [15][16]. Of particular importance, knowledge gaps need to be filled in areas of early cancer diagnostics, cancer risk stratification, and molecular identity-based treatment options [14].
Phosphatase and tensin homolog (PTEN), a tumor suppressor gene, is frequently dysregulated in the type I endometrial cancer patients [2]. Loss of heterozygosity of chromosome 10q where PTEN is located (chromosome 10q23.3) or intragenic mutation of PTEN has been identified in endometrial cancer [2][17][18][19]. Conditional deletion of Pten in the mouse uterus promotes endometrial cancer development, lending credence to the role of PTEN in the pathogenesis of endometrial cancer [20]. Dysregulation of the phosphatidylinositol 3-kinase (PI3K) pathway, mitogen-activated protein kinase (MAPK) pathway, catenin beta 1 (CTNNB1), or AT-rich interaction domain 1A (ARID1A or BAF250) appears common in endometrial cancer patients [21]. Meanwhile, mutations in phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), phosphoinositide-3-kinase regulatory subunit 1 (PIK3R1), KRAS proto-oncogene, GTPase (KRAS), fibroblast growth factor receptor 2 (FGFR2), protein phosphatase 2 scaffold subunit Aalpha (PPP2R1A), and tumor protein p53 (TP53) have also been identified in endometrioid carcinoma and serous endometrial cancer [21].
Enhancer of zeste homolog 2 (EZH2) is a core component of polycomb repressive complex 2 (PRC2) [22]. EZH2 is a well-established histone methyltransferase that regulates gene expression via inducing the tri-methylation of lysine 27 on histone H3 (H3K27) [23]. EZH2 is overexpressed in both human endometrial cancer cell lines and endometrial cancer tissues [24]. Moreover, gain-of-function [25][26] or loss-of-function [27][28] mutations of EZH2 frequently occur in cancers [23][29]. Of note, both tumor-promoting and tumor-suppressive effects of EZH2 have been documented in cancer development [29]. However, the role of EZH2 in endometrial cancer remains poorly defined.

2. EZH2 in Endometrial Cancer

Both PTEN and EZH2 play important roles in endometrial cancer. The mutation of PTEN gene has been identified in ~20% of human endometrial hyperplasia, suggesting its importance in early cancer development [30]. The frequency of PTEN mutation appears to be associated with the histotypes of endometrial cancer, as PTEN mutation occurs in ~40% of endometrioid cancers but only 5% of serous or clear cell endometrial cancers [19]. EZH2 is overexpressed in endometrial cancer, and its downregulation in endometrial cancer cells inhibits cell proliferation [24][31]. A study has identified a correlation between overexpression of EZH2 in endometrial cancer patients and disease-free and overall survival [32]. This report has further demonstrated that silencing EZH2 in endometrial cancer cells impairs the expression of growth-related genes such as peroxiredoxin 6 (PRDX6) [32]. The mechanisms underlying EZH2 action in endometrial cancer progression remain incompletely understood. However, it appears that microRNA-361/Twist axis plays an important role in mediating the role of EZH2 in driving endometrial cancer development [33]. The evidence points to the therapeutic potential of targeting EZH2. However, EZH2 may also function as a tumor suppressor in myeloma and pancreatic tumor [34][35]. It has been shown that loss of EZH2 in the mouse uterus enhances epithelial cell proliferation [36][37][38] and induces epithelial stratification [39]. Herein, it is found that conditional deletion of both Ezh2 and Pten reduced cell proliferation and uterine growth during early carcinogenesis but exacerbated intraluminal neutrophil accumulation and chronic inflammation during tumor progression, leading to an unfavorable disease outcome. Current results revealed dual roles of EZH2 in the development of endometrial cancer lacking Pten, a gene frequently mutated in endometrioid carcinomas.
The uterine weights of Ptend/d; Ezh2d/d mice were lower than those of Ptend/d mice at three weeks of age, accompanied by reduced cell proliferation revealed by Ki67-staining. As EZH2 inhibits uterine epithelial cell proliferation and uterine growth [39][36][37][38], the results suggest that EZH2 plays distinct roles in normal uterine epithelial cells versus malignant epithelial cells. Supporting the assumption that the role of EZH2 in PTEN-depleted epithelial cells differs from that in PTEN-expressing epithelial cells, it was reported that loss of PTEN or activation of AKT switches the tumor suppressive role of EZH2 to an oncogenic function 
Neutrophils are the first-line defenders that actively participate in host defense, tissue damage, and inflammatory disease [42]. Tumor-associated neutrophils play important roles in tumor microenvironment, where N1 neutrophils are anti-tumorigenic and N2 neutrophils are pro-tumorigenic [43][44]. The pro-tumorigenic action of neutrophils is generally associated with their effects on cancer cell invasion, extracellular matrix remodeling, and angiogenesis [44]. Although the oncogenic role of EZH2 has been documented, some in vivo experiments suggest a tumor-suppressive function of EZH2. One study showed that loss of EZH2 promotes KRasG12D-driven oncogenesis in pancreatic cancer [35]. In another report, deletion of Ezh2 accelerates Kras-driven lung adenocarcinoma in a mouse model [45]. In both cases, EZH2 appears to play a role in controlling inflammatory microenvironment [35][45]. It is found that tumor burden was reduced in Ptend/d; Ezh2d/d mice during early tumor development, revealing an oncogenic role of EZH2 in endometrial cancer development. However, unfavorable cancer outcomes were observed in these mice compared with Ptend/d mice. The latter effect is likely non-cell autonomous, as dysregulation of EZH2 in cancer cells is known to alter immune response [46]. Indeed, massive accumulation of intraluminal neutrophils is a hallmark of the endometrial cancer in Ptend/d; Ezh2d/d mice at nine weeks of age. The finding is also consistent with a previous report that increased levels of intratumoral neutrophils correlate with a poor cancer outcome [47].
The underlying mechanisms that promote the heightened inflammation in Ptend/d; Ezh2d/d mice remain unclear. However, several important contributing factors were identified by the present study. First, it is found reduced hypoxia in the uteri of Ptend/d; Ezh2d/d mice at 1 month of age. Elegant studies have demonstrated that hypoxia increases neutrophil recruitment in endometrial cancer induced by PTEN depletion, which serves to restrain the development of endometrial cancer by debridement of the malignant cells [48][49]. Interestingly, reduction of hypoxia causes attenuated neutrophil infiltration. However, these neutrophils gain more efficient capability of attacking cancer cells [49]. Loss of EZH2 limited the extent of hypoxia in Ptend/d; Ezh2d/d mice, likely enhancing the tumoricidal effect of neutrophils [49]. Intraluminal accumulation of cancer cells/debris would in turn stimulate neutrophil influx and cause heightened immune reactions, forming a vicious cycle and resulting in chronic inflammation and/or eliciting secondary infectious event. The exact reasons of how EZH2 ablation led to reduced hypoxia is unclear. However, increased vascularization in Ptend/d; Ezh2d/d uteri (Fang X and Li Q, unpublished observation) may be one of the reasons. Second, conditional deletion of Ezh2 potentiated epithelial stratification in Ptend/d mice. The uterus contains simple columnar epithelial cells expressing KRT8 but not KRT14 and p63 [50]. Current results showed that stratified epithelial markers KRT14 and ΔNp63 were expressed earlier in Ptend/d; Ezh2d/d uteri than Ptend/d uteri, consistent with the previous finding that loss of EZH2 in the uterus promotes the development of basal cells and stratified epithelia [39]. The intensified epithelial stratification in Ptend/d; Ezh2d/d uteri likely reflected the additive effect of loss of EZH2 and PTEN. Uterine epithelial stratification is a pathological event that alters the polarity and function of epithelial cells [51][52]. It is possible that epithelial stratification adversely impacts the progression of endometrial cancer due to altered epithelial cell properties. The role of epithelial stratification in endometrial cancer development in the model requires further investigation. Finally, it was found that epithelia adjacent to the uterine lumen had reduced expression of PGR in Ptend/d; Ezh2d/d mice at one month of age, when epithelial stratification intensified and marked accumulation of intraluminal neutrophils occurred. PGR loss has been associated with increased cell proliferation and metastasis [53][54]. PGR signaling antagonizes estrogen signaling during tumor development [55]. Estrogen is known to promote neutrophil recruitment during mammary involution or breast cancer development [56][57]. Thus, it is tempting to speculate that the reduction of PGR expression is associated with estrogen-directed neutrophil infiltration and heightened inflammation, which merits further investigation.
Endometrial cancer in Ptend/d mice is not metastatic to other organs even at 25–36 weeks of age [20][58][59]. However, dysregulation of several key regulators/signaling pathways may trigger metastasis. It have shown that conditional deletion of transforming growth factor β type 1 receptor (Tgfbr1) in Ptend/d mice promotes pulmonary metastases [58]. Lung metastasis was also reported in a mouse model where PTEN-ablated and K-ras expressed endometrial cancer cells were grafted [60]. In addition, conditional deletion of both Pten and dicer 1, ribonuclease type III (Dicer1) in the mouse uterus triggers adnexal metastasis [61]. EZH2 expression has been linked to endometrial cancer cell invasion and metastasis [31]. As mice conditionally overexpressing EZH2 are available [62], future investigations are needed to determine whether conditional overexpression of EZH2 in PTEN-depleted uteri impacts metastasis.
From a systems biology perspective, the functions of cells are achieved and coordinated by numerous genes/pathways within a highly interactive network [63]. Cancer may develop when perturbations of protein-protein interactions occur due to gene mutations [64]. Studies on protein-protein interaction networks in cancer may benefit cancer treatment by gaining a holistic view of mechanisms governing tumor development and discovering novel cancer drivers as well as therapeutic targets [65][66]. Studies have begun to explore protein-protein interaction networks in female reproductive cancers including endometrial cancer using an integrative computational approach [67]. Defining the interactome of endometrial cancer remains to be one of the key goals in the future.

References

  1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics, 2021. CA Cancer J. Clin. 2021, 71, 7–33.
  2. Di Cristofano, A.; Ellenson, L.H. Endometrial carcinoma. Annu. Rev. Pathol. 2007, 2, 57–85.
  3. Malik, T.Y.; Chishti, U.; Aziz, A.B.; Sheikh, I. Comparison of risk factors and survival of type 1 and type ii endometrial cancers. Pak. J. Med. Sci. 2016, 32, 886–890.
  4. Moore, K.N.; Fader, A.N. Uterine papillary serous carcinoma. Clin. Obstet. Gynecol. 2011, 54, 278–291.
  5. Lobo, F.D.; Thomas, E. Type ii endometrial cancers: A case series. J. Midlife Health 2016, 7, 69–72.
  6. Horn, L.C.; Meinel, A.; Handzel, R.; Einenkel, J. Histopathology of endometrial hyperplasia and endometrial carcinoma: An update. Ann. Diagn. Pathol. 2007, 11, 297–311.
  7. Sherman, M.E.; Sturgeon, S.; Brinton, L.A.; Potischman, N.; Kurman, R.J.; Berman, M.L.; Mortel, R.; Twiggs, L.B.; Barrett, R.J.; Wilbanks, G.D. Risk factors and hormone levels in patients with serous and endometrioid uterine carcinomas. Mod. Pathol. 1997, 10, 963–968.
  8. Levine, D.A.; The Cancer Genome Atlas Research Network. Integrated genomic characterization of endometrial carcinoma. Nature 2013, 497, 67–73.
  9. Talhouk, A.; McConechy, M.K.; Leung, S.; Li-Chang, H.H.; Kwon, J.S.; Melnyk, N.; Yang, W.; Senz, J.; Boyd, N.; Karnezis, A.N.; et al. A clinically applicable molecular-based classification for endometrial cancers. Br. J. Cancer 2015, 113, 299–310.
  10. Talhouk, A.; McConechy, M.K.; Leung, S.; Yang, W.; Lum, A.; Senz, J.; Boyd, N.; Pike, J.; Anglesio, M.; Kwon, J.S.; et al. Confirmation of promise: A simple, genomics-based clinical classifier for endometrial cancer. Cancer 2017, 123, 802–813.
  11. Kommoss, S.; McConechy, M.K.; Kommoss, F.; Leung, S.; Bunz, A.; Magrill, J.; Britton, H.; Kommoss, F.; Grevenkamp, F.; Karnezis, A.; et al. Final validation of the promise molecular classifier for endometrial carcinoma in a large population-based case series. Ann. Oncol. 2018, 29, 1180–1188.
  12. Raffone, A.; Travaglino, A.; Raimondo, D.; Boccellino, M.P.; Maletta, M.; Borghese, G.; Casadio, P.; Insabato, L.; Mollo, A.; Zullo, F.; et al. Tumor-infiltrating lymphocytes and pole mutation in endometrial carcinoma. Gynecol. Oncol. 2021, 161, 621–628.
  13. Huvila, J.; Orte, K.; Vainio, P.; Mettala, T.; Joutsiniemi, T.; Hietanen, S. Molecular subtype diagnosis of endometrial carcinoma: Comparison of the next-generation sequencing panel and proactive molecular risk classifier for endometrial cancer classifier. Hum. Pathol. 2021, 111, 98–109.
  14. Urick, M.E.; Bell, D.W. Clinical actionability of molecular targets in endometrial cancer. Nat. Rev. Cancer 2019, 19, 510–521.
  15. Soumerai, T.E.; Donoghue, M.T.A.; Bandlamudi, C.; Srinivasan, P.; Chang, M.T.; Zamarin, D.; Cadoo, K.A.; Grisham, R.N.; O’Cearbhaill, R.E.; Tew, W.P.; et al. Clinical utility of prospective molecular characterization in advanced endometrial cancer. Clin. Cancer Res. 2018, 24, 5939–5947.
  16. Tsimberidou, A.M.; Iskander, N.G.; Hong, D.S.; Wheler, J.J.; Falchook, G.S.; Fu, S.; Piha-Paul, S.; Naing, A.; Janku, F.; Luthra, R.; et al. Personalized medicine in a phase i clinical trials program: The md anderson cancer center initiative. Clin. Cancer Res. 2012, 18, 6373–6383.
  17. Tashiro, H.; Blazes, M.S.; Wu, R.; Cho, K.R.; Bose, S.; Wang, S.I.; Li, J.; Parsons, R.; Ellenson, L.H. Mutations in pten are frequent in endometrial carcinoma but rare in other common gynecological malignancies. Cancer Res. 1997, 57, 3935–3940.
  18. Peiffer, S.L.; Herzog, T.J.; Tribune, D.J.; Mutch, D.G.; Gersell, D.J.; Goodfellow, P.J. Allelic loss of sequences from the long arm of chromosome 10 and replication errors in endometrial cancers. Cancer Res. 1995, 55, 1922–1926.
  19. Risinger, J.I.; Hayes, K.; Maxwell, G.L.; Carney, M.E.; Dodge, R.K.; Barrett, J.C.; Berchuck, A. Pten mutation in endometrial cancers is associated with favorable clinical and pathologic characteristics. Clin. Cancer Res. 1998, 4, 3005–3010.
  20. Daikoku, T.; Hirota, Y.; Tranguch, S.; Joshi, A.R.; DeMayo, F.J.; Lydon, J.P.; Ellenson, L.H.; Dey, S.K. Conditional loss of uterine pten unfailingly and rapidly induces endometrial cancer in mice. Cancer Res. 2008, 68, 5619–5627.
  21. O’Hara, A.J.; Bell, D.W. The genomics and genetics of endometrial cancer. Adv. Genom. Genet. 2012, 2012, 33–47.
  22. Jani, K.S.; Jain, S.U.; Ge, E.J.; Diehl, K.L.; Lundgren, S.M.; Muller, M.M.; Lewis, P.W.; Muir, T.W. Histone h3 tail binds a unique sensing pocket in ezh2 to activate the prc2 methyltransferase. Proc. Natl. Acad. Sci. USA 2019, 116, 8295–8300.
  23. Kim, K.H.; Roberts, C.W. Targeting ezh2 in cancer. Nat. Med. 2016, 22, 128–134.
  24. Oki, S.; Sone, K.; Oda, K.; Hamamoto, R.; Ikemura, M.; Maeda, D.; Takeuchi, M.; Tanikawa, M.; Mori-Uchino, M.; Nagasaka, K.; et al. Oncogenic histone methyltransferase ezh2: A novel prognostic marker with therapeutic potential in endometrial cancer. Oncotarget 2017, 8, 40402–40411.
  25. Bachmann, I.M.; Halvorsen, O.J.; Collett, K.; Stefansson, I.M.; Straume, O.; Haukaas, S.A.; Salvesen, H.B.; Otte, A.P.; Akslen, L.A. Ezh2 expression is associated with high proliferation rate and aggressive tumor subgroups in cutaneous melanoma and cancers of the endometrium, prostate, and breast. J. Clin. Oncol. 2006, 24, 268–273.
  26. Varambally, S.; Dhanasekaran, S.M.; Zhou, M.; Barrette, T.R.; Kumar-Sinha, C.; Sanda, M.G.; Ghosh, D.; Pienta, K.J.; Sewalt, R.G.; Otte, A.P.; et al. The polycomb group protein ezh2 is involved in progression of prostate cancer. Nature 2002, 419, 624–629.
  27. Sashida, G.; Harada, H.; Matsui, H.; Oshima, M.; Yui, M.; Harada, Y.; Tanaka, S.; Mochizuki-Kashio, M.; Wang, C.; Saraya, A.; et al. Ezh2 loss promotes development of myelodysplastic syndrome but attenuates its predisposition to leukaemic transformation. Nat. Commun. 2014, 5, 4177.
  28. Ernst, T.; Chase, A.J.; Score, J.; Hidalgo-Curtis, C.E.; Bryant, C.; Jones, A.V.; Waghorn, K.; Zoi, K.; Ross, F.M.; Reiter, A.; et al. Inactivating mutations of the histone methyltransferase gene ezh2 in myeloid disorders. Nat. Genet. 2010, 42, 722–726.
  29. Yan, K.S.; Lin, C.Y.; Liao, T.W.; Peng, C.M.; Lee, S.C.; Liu, Y.J.; Chan, W.P.; Chou, R.H. Ezh2 in cancer progression and potential application in cancer therapy: A friend or foe? Int. J. Mol. Sci. 2017, 18, 1172.
  30. Maxwell, G.L.; Risinger, J.I.; Gumbs, C.; Shaw, H.; Bentley, R.C.; Barrett, J.C.; Berchuck, A.; Futreal, P.A. Mutation of the pten tumor suppressor gene in endometrial hyperplasias. Cancer Res. 1998, 58, 2500–2503.
  31. Gu, Y.; Zhang, J.; Guan, H. Expression of ezh2 in endometrial carcinoma and its effects on proliferation and invasion of endometrial carcinoma cells. Oncol. Lett. 2017, 14, 7191–7196.
  32. Roh, J.W.; Choi, J.E.; Han, H.D.; Hu, W.; Matsuo, K.; Nishimura, M.; Lee, J.S.; Kwon, S.Y.; Cho, C.H.; Kim, J.; et al. Clinical and biological significance of ezh2 expression in endometrial cancer. Cancer Biol. Ther. 2020, 21, 147–156.
  33. Ihira, K.; Dong, P.; Xiong, Y.; Watari, H.; Konno, Y.; Hanley, S.J.; Noguchi, M.; Hirata, N.; Suizu, F.; Yamada, T.; et al. Ezh2 inhibition suppresses endometrial cancer progression via mir-361/twist axis. Oncotarget 2017, 8, 13509–13520.
  34. Kikuchi, J.; Koyama, D.; Wada, T.; Izumi, T.; Hofgaard, P.O.; Bogen, B.; Furukawa, Y. Phosphorylation-mediated ezh2 inactivation promotes drug resistance in multiple myeloma. J. Clin. Investig. 2015, 125, 4375–4390.
  35. Mallen-St Clair, J.; Soydaner-Azeloglu, R.; Lee, K.E.; Taylor, L.; Livanos, A.; Pylayeva-Gupta, Y.; Miller, G.; Margueron, R.; Reinberg, D.; Bar-Sagi, D. Ezh2 couples pancreatic regeneration to neoplastic progression. Genes Dev. 2012, 26, 439–444.
  36. Nanjappa, M.K.; Mesa, A.M.; Medrano, T.I.; Jefferson, W.N.; DeMayo, F.J.; Williams, C.J.; Lydon, J.P.; Levin, E.R.; Cooke, P.S. The histone methyltransferase ezh2 is required for normal uterine development and function in mice. Biol. Reprod. 2019, 101, 306–317.
  37. Mesa, A.M.; Mao, J.D.; Nanjappa, M.K.; Medrano, T.I.; Tevosian, S.; Yu, F.H.; Kinkade, J.; Lyu, Z.; Liu, Y.; Joshi, T.; et al. Mice lacking uterine enhancer of zeste homolog 2 have transcriptomic changes associated with uterine epithelial proliferation. Physiol. Genom. 2020, 52, 81–95.
  38. Mesa, A.M.; Mao, J.; Medrano, T.I.; Bivens, N.J.; Jurkevich, A.; Tuteja, G.; Cooke, P.S.; Rosenfeld, C.S. Spatial transcriptomics analysis of uterine gene expression in enhancer of zeste homolog 2 conditional knockout micedagger. Biol. Reprod. 2021, 105, 1126–1139.
  39. Fang, X.; Ni, N.; Lydon, J.P.; Ivanov, I.; Bayless, K.J.; Rijnkels, M.; Li, Q. Enhancer of zeste 2 polycomb repressive complex 2 subunit is required for uterine epithelial integrity. Am. J. Pathol. 2019, 189, 1212–1225.
  40. Xie, Y.; Naizabekov, S.; Chen, Z.; Tokay, T. Power of pten/akt: Molecular switch between tumor suppressors and oncogenes. Oncol. Lett. 2016, 12, 375–378.
  41. Sirohi, V.K.; Medrano, T.I.; Mesa, A.M.; Kannan, A.; Bagchi, I.C.; Cooke, P.S. Regulation of akt signaling in mouse uterus. Endocrinology 2022, 163, bqab233.
  42. Kruger, P.; Saffarzadeh, M.; Weber, A.N.; Rieber, N.; Radsak, M.; von Bernuth, H.; Benarafa, C.; Roos, D.; Skokowa, J.; Hartl, D. Neutrophils: Between host defence, immune modulation, and tissue injury. PLoS Pathog. 2015, 11, e1004651.
  43. Wu, L.; Saxena, S.; Awaji, M.; Singh, R.K. Tumor-associated neutrophils in cancer: Going pro. Cancers 2019, 11, 564.
  44. Masucci, M.T.; Minopoli, M.; Carriero, M.V. Tumor associated neutrophils. Their role in tumorigenesis, metastasis, prognosis and therapy. Front. Oncol. 2019, 9, 1146.
  45. Wang, Y.; Hou, N.; Cheng, X.; Zhang, J.; Tan, X.; Zhang, C.; Tang, Y.; Teng, Y.; Yang, X. Ezh2 acts as a tumor suppressor in kras-driven lung adenocarcinoma. Int. J. Biol. Sci. 2017, 13, 652–659.
  46. Gan, L.; Yang, Y.; Li, Q.; Feng, Y.; Liu, T.; Guo, W. Epigenetic regulation of cancer progression by ezh2: From biological insights to therapeutic potential. Biomark Res. 2018, 6, 10.
  47. Shen, M.; Hu, P.; Donskov, F.; Wang, G.; Liu, Q.; Du, J. Tumor-associated neutrophils as a new prognostic factor in cancer: A systematic review and meta-analysis. PLoS ONE 2014, 9, e98259.
  48. Blaisdell, A.; Crequer, A.; Columbus, D.; Daikoku, T.; Mittal, K.; Dey, S.K.; Erlebacher, A. Neutrophils oppose uterine epithelial carcinogenesis via debridement of hypoxic tumor cells. Cancer Cell 2015, 28, 785–799.
  49. Mahiddine, K.; Blaisdell, A.; Ma, S.; Crequer-Grandhomme, A.; Lowell, C.A.; Erlebacher, A. Relief of tumor hypoxia unleashes the tumoricidal potential of neutrophils. J. Clin. Investig. 2020, 130, 389–403.
  50. Cunha, G.R.; Kurita, T.; Cao, M.; Shen, J.; Robboy, S.; Baskin, L. Molecular mechanisms of development of the human fetal female reproductive tract. Differentiation 2017, 97, 54–72.
  51. McCaffrey, L.M.; Macara, I.G. Epithelial organization, cell polarity and tumorigenesis. Trends Cell Biol. 2011, 21, 727–735.
  52. Filant, J.; DeMayo, F.J.; Pru, J.K.; Lydon, J.P.; Spencer, T.E. Fibroblast growth factor receptor two (fgfr2) regulates uterine epithelial integrity and fertility in mice. Biol. Reprod. 2014, 90, 7.
  53. Tangen, I.L.; Werner, H.M.; Berg, A.; Halle, M.K.; Kusonmano, K.; Trovik, J.; Hoivik, E.A.; Mills, G.B.; Krakstad, C.; Salvesen, H.B. Loss of progesterone receptor links to high proliferation and increases from primary to metastatic endometrial cancer lesions. Eur. J. Cancer 2014, 50, 3003–3010.
  54. Kim, J.J.; Chapman-Davis, E. Role of progesterone in endometrial cancer. Semin. Reprod. Med. 2010, 28, 81–90.
  55. Rodriguez, A.C.; Blanchard, Z.; Maurer, K.A.; Gertz, J. Estrogen signaling in endometrial cancer: A key oncogenic pathway with several open questions. Horm. Cancer 2019, 10, 51–63.
  56. Chung, H.H.; Or, Y.Z.; Shrestha, S.; Loh, J.T.; Lim, C.L.; Ong, Z.; Woo, A.R.E.; Su, I.H.; Lin, V.C.L. Estrogen reprograms the activity of neutrophils to foster protumoral microenvironment during mammary involution. Sci. Rep. 2017, 7, 46485.
  57. Vazquez Rodriguez, G.; Abrahamsson, A.; Jensen, L.D.; Dabrosin, C. Estradiol promotes breast cancer cell migration via recruitment and activation of neutrophils. Cancer Immunol. Res. 2017, 5, 234–247.
  58. Gao, Y.; Lin, P.; Lydon, J.P.; Li, Q. Conditional abrogation of transforming growth factor-beta receptor 1 in pten-inactivated endometrium promotes endometrial cancer progression in mice. J. Pathol. 2017, 243, 89–99.
  59. Lindberg, M.E.; Stodden, G.R.; King, M.L.; MacLean, J.A., 2nd; Mann, J.L.; DeMayo, F.J.; Lydon, J.P.; Hayashi, K. Loss of cdh1 and pten accelerates cellular invasiveness and angiogenesis in the mouse uterus. Biol. Reprod. 2013, 89, 8.
  60. Fedorko, A.M.; Kim, T.H.; Broaddus, R.; Schmandt, R.; Chandramouli, G.V.R.; Kim, H.I.; Jeong, J.W.; Risinger, J.I. An immune competent orthotopic model of endometrial cancer with metastasis. Heliyon 2020, 6, e04075.
  61. Wang, X.; Wendel, J.R.H.; Emerson, R.E.; Broaddus, R.R.; Creighton, C.J.; Rusch, D.B.; Buechlein, A.; DeMayo, F.J.; Lydon, J.P.; Hawkins, S.M. Pten and dicer1 loss in the mouse uterus causes poorly differentiated endometrial adenocarcinoma. Oncogene 2020, 39, 6286–6299.
  62. Koppens, M.A.; Tanger, E.; Nacerddine, K.; Westerman, B.; Song, J.Y.; van Lohuizen, M. A new transgenic mouse model for conditional overexpression of the polycomb group protein ezh2. Transgenic Res. 2017, 26, 187–196.
  63. Ideker, T.; Galitski, T.; Hood, L. A new approach to decoding life: Systems biology. Annu. Rev. Genom. Hum. Genet. 2001, 2, 343–372.
  64. Qiu, J.; Chen, K.; Zhong, C.; Zhu, S.; Ma, X. Network-based protein-protein interaction prediction method maps perturbations of cancer interactome. PLoS Genet. 2021, 17, e1009869.
  65. Kim, M.; Park, J.; Bouhaddou, M.; Kim, K.; Rojc, A.; Modak, M.; Soucheray, M.; McGregor, M.J.; O’Leary, P.; Wolf, D.; et al. A protein interaction landscape of breast cancer. Science 2021, 374, eabf3066.
  66. Pilot-Storck, F.; Chopin, E.; Rual, J.F.; Baudot, A.; Dobrokhotov, P.; Robinson-Rechavi, M.; Brun, C.; Cusick, M.E.; Hill, D.E.; Schaeffer, L.; et al. Interactome mapping of the phosphatidylinositol 3-kinase-mammalian target of rapamycin pathway identifies deformed epidermal autoregulatory factor-1 as a new glycogen synthase kinase-3 interactor. Mol. Cell Proteom. 2010, 9, 1578–1593.
  67. Pane, K.; Affinito, O.; Zanfardino, M.; Castaldo, R.; Incoronato, M.; Salvatore, M.; Franzese, M. An integrative computational approach based on expression similarity signatures to identify protein-protein interaction networks in female-specific cancers. Front. Genet. 2020, 11, 612521.
More
Information
Subjects: Oncology
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 351
Revisions: 2 times (View History)
Update Date: 29 Mar 2022
1000/1000
Video Production Service