Download PDF
Review  |  Open Access  |  5 May 2016

Murine double minute 2, a potential p53-independent regulator of liver cancer metastasis

Views: 4474 |  Downloads: 883 |  Cited:   5
Hepatoma Res 2016;2:114-21.
10.20517/2394-5079.2015.67 |  © 2016 Hepatoma Research
Author Information
Article Notes
Cite This Article

Abstract

Hepatocellular carcinoma (HCC) has emerged as one of the most commonly diagnosed forms of human cancer; yet, the mechanisms underlying HCC progression remain unclear. Unlike other cancers, systematic chemotherapy is not effective for HCC patients, while surgical resection and liver transplantation are the most viable treatment options. Thus, identifying factors or pathways that suppress HCC progression would be crucial for advancing treatment strategies for HCC. The murine double minute 2 (MDM2)-p53 pathway is impaired in most of the cancer types, including HCC, and MDM2 is overexpressed in approximately 30% of HCC. Overexpression of MDM2 is reported to be well correlated with metastasis, drug resistance, and poor prognosis of multiple cancer types, including HCC. Importantly, these correlations are observed even when p53 is mutated. Indeed, p53-independent functions of overexpressed MDM2 in cancer progression have been suitably demonstrated. In this review article, we summarize potential effectors of MDM2 that promote or suppress cancer metastasis and specifically discuss the p53-independent roles of MDM2 in liver cancer metastasis from clinical as well as biological perspectives.

Keywords

Murine double minute 2, metastasis, effectors, hepatocellular carcinoma, p53 independent

Introduction

Liver cancer is the 5th most frequently diagnosed cancer worldwide in males (9th in females) and is the 2nd leading cause of cancer-related death in males (6th in females).[1] Around 80% of hepatocellular carcinoma (HCC) cases occur in developing countries, mainly due to the incidence of hepatitis B and hepatitis C infections.[2] HCC is often diagnosed at late stages, and the 5-year survival rate for metastatic HCC is less than 10% (http://www.cancer.org/acs/groups/cid/documents/webcontent/003114-pdf.pdf).[3-5] Understanding the mechanisms involved in the regulation of HCC metastasis and discovering methods or compounds to suppress metastasis would be highly beneficial for HCC patients.[6]

Metastasis is a cellular process which involves multiple cascades including detachment of cancer cells from primary tumors, migration, intravasation, survival in the vasculature, extravasation, and colonization at a secondary site.[7] Multiple factors play a role in each metastatic step and the inhibition of any of these steps would be helpful in blocking the cancer spread. Although distant metastasis is not a common event in HCC, HCC often shows vascular invasion, intrahepatic colonization, and lymph node metastasis. This is most likely due to the dense hepatic vasculature which supports the intrahepatic metastasis of HCC.[8]

The murine double minute 2 (MDM2) was originally identified as a gene which was overexpressed in a spontaneously transformed mouse cell line (3T3-DM),[9] and the gene product was found to transform normal cells.[10] The primary function of MDM2 is to ubiquitinate the tumor suppressor p53 for inducing its degradation. Hence, MDM2 overexpression greatly contributes to tumor development through inhibition of p53 activity. MDM2 is also a transcriptional target of p53, hence forming autoregulatory negative feedback loop.[11]

Increasing evidence, however, indicates that MDM2 also has p53-independent functions toward malignant progression when overexpressed. Approximately 10% of human cancers have both MDM2 overexpression and mutant p53.[12] Mice carrying a MDM2 transgene develop a higher percentage of sarcomas regardless of p53 status, as compared with p53-null mice.[13] Ectopic expression of MDM2 in mammary epithelial cells of mice, as well as in mouse embryonic fibroblasts (MEFs), increases aneuploidy and chromosome/chromatid breaks regardless of p53 status.[14,15] MDM2 interacts with different proteins and alters their activities, leading to malignant progression independent of p53.[11] Specifically, MDM2 inhibits Nijmegen breakage syndrome 1, leading to inhibition of double-strand break repair.[16] MDM2 also promotes p21 degradation.[17,18] Additionally, MDM2 promotes cell cycle progression through activation of S-phase, via interaction with the retinoblastoma tumor suppressor protein and the transcriptional factor E2F.[19,20] MDM2 furthermore enhances doxorubicin resistance in acute lymphoblastic leukemia cells through its binding to the Sp1-binding site in the p65 promoter.[21] MDM2 is shown to bind to Sp1 and inhibit Sp1-dependent transcription.[22] Thus, numerous MDM2 binding partners and effectors contribute to its p53-independent functions.[23]

MDM2 overexpression is clinically correlated with metastasis of multiple cancer types including liver cancer,[24-27] but the underlying mechanisms remain unclear. In this review, we focus on p53-independent roles of MDM2 in cancer metastasis, specifically in liver cancer. We categorize effectors of MDM2 into metastasis promoters [Table 1] and suppressors [Table 2].

Table 1

Metastasis promoters interacting with MDM2

Gene Roles in liver cancer metastasisBinding to MDM2Functional association with MDM2References
HIF-1α Overexpression of HIF-1α is correlated with vascular invasion and poor survival in human HCC.Endogenous bindingMDM2 positively regulates HIF-1α expression in MEFs, colon cancer, and osteosarcoma cell lines independent of p53. Conversely, MDM2 is reported to destabilize HIF-1α by promoting its ubiquitination.[32-39]
Slug Overexpression of Slug is associated with invasion and metastasis of HCC by repressing E-cadherin.Endogenous bindingMDM2 stabilizes Slug mRNA in human non-small cell lung carcinoma and colon cancer cell lines.[41-44]
MMP-9 Overexpression of MMP-9 is well correlated with invasion, metastasis, and poor prognosis in liver cancer.UnknownMDM2 increases the MMP-9 promoter activity in breast cancer cell lines.[46-49,51,52]
HuR/ELAV1 HuR expression is positively correlated with advanced stages in HCC and poor outcomes in HCC patients.Endogenous bindingMDM2 neddylates HuR, protects it from degradation, and induces its nuclear localization in MEFs, mouse liver progenitor MLP29, colon cancer RKO, and HCC HepG2 cell lines.[58,60]
Table 2

Metastasis suppressors interacting with MDM2

Gene Roles in liver cancer metastasisBinding to MDM2Functional association with MDM2References
E-cadherin Reduced E-cadherin expression is associated with high tumor grade, vascular invasion, intrahepatic metastasis, disease progression, and poor outcomes.Endogenous bindingMDM2 promotes E-cadherin degradation in breast cancer cell lines.[68-72]
NME2 NME2 expression is increased in HCC.Endogenous bindingMDM2 suppresses the ability of NME2 to negatively regulate cell motility in renal cell carcinoma and lung cancer cell lines.[77-79]
TAp63 Role of TAp63 in HCC metastasis is not explored.Endogenous bindingMDM2 suppresses TAp63 activity by inhibiting its nuclear localization in MEFs and osteosarcoma cell lines. Conversely, MDM2 increases TAp63 levels and its transcriptional activity in osteosarcoma and monkey kidney fibroblast-like cell lines.[91,92,94]
FOXO family Direct association of FOXO proteins with HCC metastasis remains unknown.Endogenous bindingMDM2 degrades FOXO1, 3, and 4 in MEFs, breast cancer, and lung cancer cell lines.[110-112]
MTBP MTBP inhibits HCC migration and metastasis in ACTN4-dependent and -independent manners. Controversially, MTBP may increase HCC metastasis by stabilizing MDM2.ExogenousThe roles of MTBP in cancer metastasis, the underlying mechanisms, and functional association between MDM2 and MTBP remain to be further investigated.[114-117,122]

Metastasis promoters

Hypoxia-inducible factor-1-alpha

Hypoxia-inducible factor-1-alpha (HIF-1α) and HIF-1β are a class of transcription factors that play a key role in regulating cellular response against hypoxia.[28] While HIF-1β is constitutively expressed, expression of HIF-1α is dependent on oxygen tension. In normoxic conditions, it is rapidly degraded, whereas in hypoxic states, HIF-1α heterodimerizes with HIF-1β on hypoxia response elements in the promoter regions of numerous downstream target genes, thus promoting tumor invasion, angiogenesis, and metastasis.[29] For example, HIF-1α transactivates Snail1 and vascular endothelial growth factor (VEGF) that accelerate epithelial-mesenchymal transition (EMT), a crucial biologic process for epithelial tumors to gain metastatic potential, and angiogenesis, respectively, thereby enhancing invasion and metastasis.[30] HIF-1α is overexpressed in multiple types of human cancer including HCC.[31,32] Overexpression of HIF-1α is correlated with vascular invasion and poor survival in human HCC.[32-35]

MDM2 directly binds to HIF-1α, and overexpression of MDM2 results in accumulation of HIF-1α in hypoxic cells and increase in hypoxia-induced VEGF transcription.[36,37] Conversely, MDM2 is shown to degrade HIF-1α under hypoxic conditions, which is inhibited by phosphorylation of MDM2 at serine 166 by AKT.[38,39] Thus, the roles of MDM2 in regulating HIF-1α function need to be further investigated. Although both MDM2 and HIF-1α play roles in HCC progression, there is no existing study that directly shows MDM2 enhancing liver cancer metastasis through upregulation of HIF-1α.

Slug

Slug (also known as Snail family zinc finger 2: Snail2) is a member of the Snail family of transcription factors that induce EMT crucial for embryogenesis and cancer metastasis by repressing E-cadherin.[40] Slug is upregulated in many cancer types, including HCC, and its overexpression is associated with invasion and metastasis of HCC.[41-43]

MDM2 is shown to stabilize Slug mRNA in a p53-independent manner, while knockdown of Slug nullifies invasion of HCT116 p53-null colon cancer cells induced by MDM2 overexpression.[44] However, direct evidence demonstrating that MDM2’s involvement in promoting HCC metastasis via upregulation of Slug has not yet been demonstrated.

Matrix metalloproteinase-9

Matrix metalloproteinase 9 (MMP-9), is a type IV collagenase which is a group of zinc-containing endopeptidases to degrade structural proteins of extracellular matrix, thus playing a pivotal role in the metastatic process.[45] Overexpression of MMP-9 is well correlated with invasion, metastasis, and poor prognosis in liver cancer.[46-49] Correlation between the expression of MMP-9 and MDM2 is shown in benzopyrene-induced lung cancer in rats, where both protein expression is higher in stage III and IV lung cancer tissues as compared with stage I and II tissues.[50] Also, in human breast cancer, MDM2 expression is positively correlated with that of MMP-9, and is also negatively correlated with disease-free survival.[51] Moreover, knockdown of MDM2 in pancreatic carcinoma SW1990HM cells results in reduced MMP-9 protein expression,[52] and MDM2 promotes invasion of both MCF7 and MDA-MB-231 cell lines by increasing the MMP-9 promoter activity.[51] Although there is definite clinical and functional correlation between MMP-9 and MDM2, it remains unclear whether MDM2 induces invasion and metastasis in liver cancer through upregulation of MMP-9.

Hu antigen R

Hu antigen R (HuR, also known as ELAV-like protein 1) was first identified in drosophila as a member of the embryonic lethal abnormal vision (ELAV) family RNA-binding proteins.[53,54] HuR binds to AU-rich elements in the 3’ untranslated region of target mRNAs and stabilizes them, resulting in regulation of cell proliferation, survival, immune response, and differentiation.[55] Elevated expression of HuR is reported in many types of cancer.[56,57] Specifically, HuR is upregulated in HCC, and its expression is positively correlated with advanced stages of HCC, as well as poor outcomes in HCC patients.[58] HuR promotes proliferation and differentiation of hepatocytes, as well as HCC transformation.[59] Importantly, MDM2 neddylates HuR, protects it from degradation, and induces its nuclear localization in mouse liver progenitor, colon cancer, and HCC cell lines.[60] Although all the cell lines contain wild-type p53, neddylation of HuR by MDM2 is likely to be p53-independent, which needs to be clarified in the future. Importantly, it also remains unknown whether neddylated HuR by MDM2 enhances HCC metastasis.

Metastasis suppressors

E-cadherin

E-cadherin is a single transmembrane glycoprotein involved in Ca2+-mediated cell adhesion, mobility, and proliferation of epithelial cells and functions as a metastasis suppressor.[61,62] Reduced expression of E-cadherin is correlated with high potential of invasion and metastasis, as well as poor prognosis, in many cancer types including breast,[63] gastric,[64] lung,[65] colorectal,[66] and pancreatic cancer.[67] Also in HCC, reduced E-cadherin expression is associated with high tumor grade, vascular invasion, intrahepatic metastasis, disease progression, and poor outcomes.[68-71]

MDM2 is found to directly interact with E-cadherin and facilitate its degradation in a p53-independent manner.[72] Expression of MDM2 and E-cadherin is inversely correlated in breast cancer having lymph node metastasis.[72] However, it remains unclear whether or not MDM2 promotes HCC metastasis by degrading E-cadherin.

Non-metastatic cells 2

Non-metastatic cells 2 (NME2, also known as NDPK-B, NM23B, NM23-H2) belongs to the nonmetastatic family and functions as a metastasis suppressor.[73] Reduced NME2 expression is associated with increased metastatic potential of oral squamous cell carcinoma, lung, ovarian, colon, and breast cancer.[74-76] However, NME2 expression is found to be increased in HCC.[77,78]

MDM2 interacts with NME2 in H1299 lung cancer and HEK293 embryonic kidney cell lines and also suppresses the ability of NME2 to negatively regulate cell motility in renal cell carcinoma (UOK117 and its derivative 1.27) and H1299 cell lines.[79] However, the role of NME2 in metastasis suppression of HCC and its functional association with MDM2 in HCC remain to be investigated.

TAp63

TAp63, along with TAp73, are tumor suppressor proteins that belong to the p53 family with high homology in the DNA binding domain and recognize the same p53 responsive elements.[80] TAp63 suppresses migration and metastasis in many human cancer types including liver cancer, thus functioning as a metastasis suppressor.[81-86] On the other hand, isoforms of p63 lacking N-terminal domain show oncogenic function and are overexpressed in multiple cancer types.[85,87,88] Mice with deletion of the p63 gene spontaneously develop tumors, while compound knockout mice for p53 and p63 show high frequency of metastasis as compared with p53 or p63 knockout mice.[89,90]

TAp63 weakly binds to MDM2,[91] and MDM2 is shown to attenuate apoptotic function of TAp63 by inhibiting its nuclear localization.[92] However, it is unknown whether or not MDM2 inhibits metastasis suppressor function of TAp63. Conversely, it is also reported that MDM2 competes with TAp63 for binding to p53R175H mutant to restore p63 activity,[93] and overexpression of MDM2 increases the steady-state level of intracellular TAp63 and enhances its transcriptional activity.[94] Thus, the functional relationship of MDM2 with TAp63 is controversial.

Forkhead box O family

Forkhead box O (FOXO) proteins (FOXO1, 3, 4, and 6) are members of the forkhead family of transcription factors.[95] FOXO proteins have been implicated in suppression of tumor progression in multiple cancer types.[96-100] Expression of FOXO proteins is negatively correlated with migration, invasion, and metastasis of renal cell carcinoma,[101] lung cancer,[102] prostate cancer,[103] and urothelial cancer.[104] Importantly, FOXO3 inhibits EMT by suppressing activities of β-catenin in prostate cancer[103] and Twist1 in urothelial cancer,[104] while FOXO4 functions as a metastasis-suppressor through counteracting the PI3K/AKT signal pathway in prostate cancer[105] and inhibiting EMT in lung cancer.[106] Although reduced expression of FOXO proteins is correlated with hepatocarcinogenesis and poor survival of HCC patients, direct association of FOXO proteins with HCC metastasis remains unknown.[107-109] MDM2 functions as an E3 ubiquitin ligase for FOXO1, FOXO3, and FOXO4 to promote their degradation.[110-112] However, it remains unsolved whether degradation of FOXO proteins by MDM2 accelerates cancer metastasis.

MDM2 binding protein

MDM2 binding protein (MTBP) was originally identified as a protein that binds to MDM2.[113] Although these two proteins interact exogenously, their endogenous interactions have not yet been demonstrated. Overexpression of MTBP is shown to suppress cell migration and metastasis of osteosarcoma and HCC in alpha-actinin 4 (ACTN4)-dependent and -independent manners.[114-116] Also, in MTBP knockout mice, MTBP haploinsufficiency increases metastasis of tumors induced in the p53 heterozygous background.[117] Clinically, reduced MTBP expression is associated with reduced patient survival with head and neck carcinoma, as well as capsular/vascular invasion and lymph node metastasis in HCC.[116,118] On the other hand, increased MTBP expression is observed in B-cell lymphoma and triple negative breast cancer where it contributes to tumor progression through its interaction with Myc.[119-121] In another study on human HCC, increased MTBP expression is shown to be associated with increase in MDM2 levels and metastasis, as well as poor survival of HCC patients, which is contrary to previously published studies.[122] Thus, the roles of MTBP in cancer metastasis, the underlying mechanisms, and functional association between MDM2 and MTBP need to be further clarified in the future.

Conclusion

Approximately 30% of human cancers have MDM2 overexpression. Specifically, in well differentiated liposarcomas, MDM2 overexpression is detected in over 90% of the cases.[123] These observations indicate significance of MDM2 overexpression in cancer progression. The mechanisms of MDM2 overexpression or hyper-activation include MDM2 gene amplification,[124] single nucleotide polymorphisms in the MDM2 promoter,[125] silencing/inhibition of MDM2 negative regulators,[126] phosphorylation of MDM2,[127] enhanced translation,[128] or other mechanisms.[129] Although the best characterized function of MDM2 is to inhibit p53 activity, an increasing body of evidence suggests that MDM2 has a p53-independent function. Such function is found specifically when MDM2 is overexpressed. MDM2 mainly exerts its p53-independent function by interacting with its downstream effectors.[11] These effectors frequently play integral roles in cancer progression including cancer metastasis and drug resistance. Indeed, MDM2 overexpression is implicated in cancer metastasis through enhancing EMT, activation/upregulation of other oncoproteins, and suppression of tumor suppressors or metastasis suppressors. However, there is scarce evidence showing direct involvement of MDM2 in invasion and metastasis of HCC. It is thus imperative to have future studies that could appropriately demonstrate the direct role of overexpressed MDM2 in promoting HCC metastasis.

Acknowledgments

We thank Alejandro Parrales and Swathi V. Iyer for editing the manuscript and helpful discussion.

Financial support and sponsorship

This project is supported by NIH 1-R01-CA174735-01A1 (T.I.) and 5-P30-CA168524-02 (PI: Roy A. Jensen) grants.

Conflicts of interest

There are no conflicts of interest.

REFERENCES

1. Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin 2015;65:87-108.

2. Dhanasekaran R, Limaye A, Cabrera R. Hepatocellular carcinoma: current trends in worldwide epidemiology, risk factors, diagnosis, and therapeutics. Hepat Med 2012;4:19-37.

3. Bialecki ES, Di Bisceglie AM. Diagnosis of hepatocellular carcinoma. HPB (Oxford) 2005;7:26-34.

4. Zhang HY, Wu QJ, Xie CH, Zhang G. Long-term survival of patients with hepatocellular carcinoma with pulmonary and adrenal metastasis: A case report. Exp Ther Med 2012;3:699-702.

5. Davis GL, Dempster J, Meler JD, Orr DW, Walberg MW, Brown B, Berger BD, O'Connor JK, Goldstein RM. Hepatocellular carcinoma: management of an increasingly common problem. Proc (Bayl Univ Med Cent) 2008;21:266-80.

6. Meng X, Franklin DA, Dong J, Zhang Y. MDM2-p53 pathway in hepatocellular carcinoma. Cancer Res 2014;74:7161-7.

7. Valastyan S, Weinberg RA. Tumor metastasis: molecular insights and evolving paradigms. Cell 2011;147:275-92.

8. van Zijl F, Zulehner G, Petz M, Schneller D, Kornauth C, Hau M, Machat G, Grubinger M, Huber H, Mikulits W. Epithelial-mesenchymal transition in hepatocellular carcinoma. Future Oncol 2009;5:1169-79.

9. Cahilly-Snyder L, Yang-Feng T, Francke U, George DL. Molecular analysis and chromosomal mapping of amplified genes isolated from a transformed mouse 3T3 cell line. Somat Cell Mol Genet 1987;13:235-44.

10. Fakharzadeh SS, Trusko SP, George DL. Tumorigenic potential associated with enhanced expression of a gene that is amplified in a mouse tumor cell line. EMBO J 1991;10:1565-9.

11. Iwakuma T, Lozano G. MDM2, an introduction. Mol Cancer Res 2003;1:993-1000.

12. Onel K, Cordon-Cardo C. MDM2 and prognosis. Mol Cancer Res 2004;2:1-8.

13. Jones SN, Hancock AR, Vogel H, Donehower LA, Bradley A. Overexpression of Mdm2 in mice reveals a p53-independent role for Mdm2 in tumorigenesis. Proc Natl Acad Sci U S A 1998;95:15608-12.

14. Lundgren K, Montes de Oca Luna R, McNeill YB, Emerick EP, Spencer B, Barfield CR, Lozano G, Rosenberg MP, Finlay CA. Targeted expression of MDM2 uncouples S phase from mitosis and inhibits mammary gland development independent of p53. Genes Dev 1997;11:714-25.

15. Bouska A, Lushnikova T, Plaza S, Eischen CM. Mdm2 promotes genetic instability and transformation independent of p53. Mol Cell Biol 2008;28:4862-74.

16. Alt JR JR, Bouska A, Fernandez MR, Cerny RL, Xiao H, Eischen CM. Mdm2 binds to Nbs1 at sites of DNA damage and regulates double strand break repair. J Biol Chem 2005;280:18771-81.

17. Jin Y, Lee H, Zeng SX, Dai MS, Lu H. MDM2 promotes p21waf1/cip1 proteasomal turnover independently of ubiquitylation. Embo J 2003;22:6365-77.

18. Zhang Z, Wang H, Li M, Agrawal S, Chen X, Zhang R. MDM2 is a negative regulator of p21WAF1/CIP1, independent of p53. J Biol Chem 2004;279:16000-6.

19. Martin K, Trouche D, Hagemeier C, Sorensen TS, La Thangue NB, Kouzarides T. Stimulation of E2F1/DP1 transcriptional activity by MDM2 oncoprotein. Nature 1995;375:691-4.

20. Sdek P, Ying H, Zheng H, Margulis A, Tang X, Tian K, Xiao ZX. The central acidic domain of MDM2 is critical in inhibition of retinoblastoma-mediated suppression of E2F and cell growth. J Biol Chem 2004;279:53317-22.

21. Gu L, Findley HW, Zhou M. MDM2 induces NF-kappaB/p65 expression transcriptionally through Sp1-binding sites: a novel, p53-independent role of MDM2 in doxorubicin resistance in acute lymphoblastic leukemia. Blood 2002;99:3367-75.

22. Johnson-Pais T, Degnin C, Thayer MJ. pRB induces Sp1 activity by relieving inhibition mediated by MDM2. Proc Natl Acad Sci U S A 2001;98:2211-6.

23. Riley MF, Lozano G. The many faces of MDM2 binding partners. Genes Cancer 2012;3:226-39.

24. Zhu MH, Ni CR, Zhu Z, Li FM, Zhang SM. [Determination of expression of eight p53-related genes in hepatocellular carcinoma with tissue microarrays]. Ai Zheng 2003;22:680-5.

25. Qiu SJ, Ye SL, Wu ZQ, Tang ZY, Liu YK. The expression of the mdm2 gene may be related to the aberration of the p53 gene in human hepatocellular carcinoma. J Cancer Res Clin Oncol 1998;124:253-8.

26. Zhou XD. Recurrence and metastasis of hepatocellular carcinoma: progress and prospects. Hepatobiliary Pancreat Dis Int 2002;1:35-41.

27. Qiu S, Ye S, Wu Z. The expression of mdm2 gene related to the invasiveness of human hepatocellular carcinoma. Zhonghua Zhong Liu Za Zhi 1997;19:256-9.

28. Wang GL, Jiang BH, Rue EA, Semenza GL. Hypoxia-inducible factor 1 is a basic-helix-loop-helix-PAS heterodimer regulated by cellular O2 tension. Proc Natl Acad Sci U S A 1995;92:5510-4.

29. Luo D, Wang Z, Wu J, Jiang C, Wu J. The role of hypoxia inducible factor-1 in hepatocellular carcinoma. Biomed Res Int 2014;2014:409272.

30. Zhang L, Huang G, Li X, Zhang Y, Jiang Y, Shen J, Liu J, Wang Q, Zhu J, Feng X, Dong J, Qian C. Hypoxia induces epithelial-mesenchymal transition via activation of SNAI1 by hypoxia-inducible factor -1alpha in hepatocellular carcinoma. BMC Cancer 2013;13:108.

31. Zhong H, De Marzo AM, Laughner E, Lim M, Hilton DA, Zagzag D, Buechler P, Isaacs WB, Semenza GL, Simons JW. Overexpression of hypoxia-inducible factor 1alpha in common human cancers and their metastases. Cancer Res 1999;59:5830-5.

32. Tanaka H, Yamamoto M, Hashimoto N, Miyakoshi M, Tamakawa S, Yoshie M, Tokusashi Y, Yokoyama K, Yaginuma Y, Ogawa K. Hypoxia-independent overexpression of hypoxia-inducible factor 1alpha as an early change in mouse hepatocarcinogenesis. Cancer Res 2006;66:11263-70.

33. Zheng SS, Chen XH, Yin X, Zhang BH. Prognostic significance of HIF-1alpha expression in hepatocellular carcinoma: a meta-analysis. PLoS One 2013;8:e65753.

34. Dai CX, Gao Q, Qiu SJ, Ju MJ, Cai MY, Xu YF, Zhou J, Zhang BH, Fan J. Hypoxia-inducible factor-1 alpha, in association with inflammation, angiogenesis and MYC, is a critical prognostic factor in patients with HCC after surgery. BMC Cancer 2009;9:418.

35. Yang SL, Liu LP, Jiang JX, Xiong ZF, He QJ, Wu C. The correlation of expression levels of HIF-1alpha and HIF-2alpha in hepatocellular carcinoma with capsular invasion, portal vein tumor thrombi and patients' clinical outcome. Jpn J Clin Oncol 2014;44:159-67.

36. Chen D, Li M, Luo J, Gu W. Direct interactions between HIF-1 alpha and Mdm2 modulate p53 function. J Biol Chem 2003;278:13595-8.

37. Nieminen AL, Qanungo S, Schneider EA, Jiang BH, Agani FH. Mdm2 and HIF-1alpha interaction in tumor cells during hypoxia. J Cell Physiol 2005;204:364-9.

38. Joshi S, Singh AR, Durden DL. MDM2 regulates hypoxic hypoxia-inducible factor 1alpha stability in an E3 ligase, proteasome, and PTEN-phosphatidylinositol 3-kinase-AKT-dependent manner. J Biol Chem 2014;289:22785-97.

39. Ravi R, Mookerjee B, Bhujwalla ZM, Sutter CH, Artemov D, Zeng Q, Dillehay LE, Madan A, Semenza GL, Bedi A. Regulation of tumor angiogenesis by p53-induced degradation of hypoxia-inducible factor 1alpha. Genes Dev 2000;14:34-44.

40. Alves CC, Carneiro F, Hoefler H, Becker KF. Role of the epithelial-mesenchymal transition regulator Slug in primary human cancers. Front Biosci (Landmark Ed) 2009;14:3035-50.

41. Zhao X, Sun B, Sun D, Liu T, Che N, Gu Q, Dong X, Li R, Liu Y, Li J. Slug promotes hepatocellular cancer cell progression by increasing sox2 and nanog expression. Oncol Rep 2015;33:149-56.

42. Sun Y, Song GD, Sun N, Chen JQ, Yang SS. Slug overexpression induces stemness and promotes hepatocellular carcinoma cell invasion and metastasis. Oncol Lett 2014;7:1936-40.

43. Sun D, Sun B, Liu T, Zhao X, Che N, Gu Q, Dong X, Yao Z, Li R, Li J, Chi J, Sun R. Slug promoted vasculogenic mimicry in hepatocellular carcinoma. J Cell Mol Med 2013;17:1038-47.

44. Jung CH, Kim J, Park JK, Hwang SG, Moon SK, Kim WJ, Um HD. Mdm2 increases cellular invasiveness by binding to and stabilizing the Slug mRNA. Cancer Lett 2013;335:270-7.

45. Sternlicht MD, Werb Z. How matrix metalloproteinases regulate cell behavior. Annu Rev Cell Dev Biol 2001;17:463-516.

46. Arii S, Mise M, Harada T, Furutani M, Ishigami S, Niwano M, Mizumoto M, Fukumoto M, Imamura M. Overexpression of matrix metalloproteinase 9 gene in hepatocellular carcinoma with invasive potential. Hepatology 1996;24:316-22.

47. Sakamoto Y, Mafune K, Mori M, Shiraishi T, Imamura H, Mori M, Shiraishi T, Imamura H, Mori M, Takayama T, Makuuchi M. Overexpression of MMP-9 correlates with growth of small hepatocellular carcinoma. Int J Oncol 2000;17:237-43.

48. Zhao XL, Sun T, Che N, Sun D, Zhao N, Dong XY, Gu Q, Yao Z, Sun BC. Promotion of hepatocellular carcinoma metastasis through matrix metalloproteinase activation by epithelial-mesenchymal transition regulator Twist1. J Cell Mol Med 2011;15:691-700.

49. Yang P, Yuan W, He J, Wang J, Yu L, Jin X, Hu Y, Liao M, Chen Z, Zhang Y. Overexpression of EphA2, MMP-9, and MVD-CD34 in hepatocellular carcinoma: Implications for tumor progression and prognosis. Hepatol Res 2009;39:1169-77.

50. Zhang DH, Zhang LY, Liu DJ, Yang F, Zhao JZ. Expression and significance of MMP-9 and MDM2 in the oncogenesis of lung cancer in rats. Asian Pac J Trop Med 2014;7:585-8.

51. Chen X, Qiu J, Yang D, Lu J, Yan C, Zha X, Yin Y. MDM2 Promotes Invasion and Metastasis in Invasive Ductal Breast Carcinoma by Inducing Matrix Metalloproteinase-9. PLoS One 2013;8:e78794.

52. Shi W, Meng Z, Chen Z, Hua Y, Gao H, Wang P, Lin J, Zhou Z, Luo J, Liu L. RNA interference against MDM2 suppresses tumor growth and metastasis in pancreatic carcinoma SW1990HM cells. Mol Cell Biochem 2014;387:1-8.

53. Campos AR, Grossman D, White K. Mutant alleles at the locus elav in Drosophila melanogaster lead to nervous system defects. A developmental-genetic analysis. J Neurogenet 1985;2:197-218.

54. Ma WJ, Cheng S, Campbell C, Wright A, Furneaux H. Cloning and characterization of HuR, a ubiquitously expressed Elav-like protein. J Biol Chem 1996;271:8144-51.

55. Srikantan S, Gorospe M. HuR function in disease. Front Biosci (Landmark Ed) 2012;17:189-205.

56. Dixon DA, Tolley ND, King PH, Nabors LB, McIntyre TM, Zimmerman GA, Prescott SM. Altered expression of the mRNA stability factor HuR promotes cyclooxygenase-2 expression in colon cancer cells. J Clin Invest 2001;108:1657-65.

57. Lopez de Silanes I, Lal A, Gorospe M. HuR: post-transcriptional paths to malignancy. RNA Biol 2005;2:11-3.

58. Zhu H, Berkova Z, Mathur R, Sehgal L, Khashab T, Tao RH, Ao X, Feng L, Sabichi AL, Blechacz B, Rashid A, Samaniego F. HuR Suppresses Fas Expression and Correlates with Patient Outcome in Liver Cancer. Mol Cancer Res 2015;13:809-18.

59. Vazquez-Chantada M, Fernandez-Ramos D, Embade N, Martinez-Lopez N, Varela-Rey M, Woodhoo A, Luka Z, Wagner C, Anglim PP, Finnell RH, Caballería J, Laird-Offringa IA, Gorospe M, Lu SC, Mato JM, Martínez-Chantar ML. HuR/methyl-HuR and AUF1 regulate the MAT expressed during liver proliferation, differentiation, and carcinogenesis. Gastroenterology 2010;138:1943-53.

60. Embade N, Fernandez-Ramos D, Varela-Rey M, Beraza N, Sini M, Gutierrez de Juan V, Woodhoo A, Martínez-López N, Rodríguez-Iruretagoyena B, Bustamante FJ, de la Hoz AB, Carracedo A, Xirodimas DP, Rodríguez MS, Lu SC, Mato JM, Martínez-Chantar ML. Murine double minute 2 regulates Hu antigen R stability in human liver and colon cancer through NEDDylation. Hepatology 2012;55:1237-48.

61. Gumbiner BM. Regulation of cadherin-mediated adhesion in morphogenesis. Nat Rev Mol Cell Biol 2005;6:622-34.

62. Halbleib JM, Nelson WJ. Cadherins in development: cell adhesion, sorting, and tissue morphogenesis. Genes Dev 2006;20:3199-214.

63. Singhai R, Patil VW, Jaiswal SR, Patil SD, Tayade MB, Patil AV. E-Cadherin as a diagnostic biomarker in breast cancer. N Am J Med Sci 2011;3:227-33.

64. Oka H, Shiozaki H, Kobayashi K, Tahara H, Tamura S, Miyata M, Doki Y, Iihara K, Matsuyoshi N, Hirano S, Takeichi M, Mori T. Immunohistochemical evaluation of E-cadherin adhesion molecule expression in human gastric cancer. Virchows Arch A Pathol Anat Histopathol 1992;421:149-56.

65. Bremnes RM, Veve R, Hirsch FR, Franklin WA. The E-cadherin cell-cell adhesion complex and lung cancer invasion, metastasis, and prognosis. Lung Cancer 2002;36:115-24.

66. Dorudi S, Sheffield JP, Poulsom R, Northover JM, Hart IR. E-cadherin expression in colorectal cancer. An immunocytochemical and in situ hybridization study. Am J Pathol 1993;142:981-6.

67. von Burstin J, Eser S, Paul MC, Seidler B, Brandl M, Messer M, von Werder A, Schmidt A, Mages J, Pagel P, Schnieke A, Schmid RM, Schneider G, Saur D. E-cadherin regulates metastasis of pancreatic cancer in vivo and is suppressed by a SNAIL/HDAC1/HDAC2 repressor complex. Gastroenterology 2009;137:361-71, 71 e1-5.

68. Asayama Y, Taguchi Ki K, Aishima Si S, Nishi H, Masuda K, Tsuneyoshi M. The mode of tumour progression in combined hepatocellular carcinoma and cholangiocarcinoma: an immunohistochemical analysis of E-cadherin, alpha-catenin and beta-catenin. Liver 2002;22:43-50.

69. Chen J, Zhao J, Ma R, Lin H, Liang X, Cai X. Prognostic significance of e-cadherin expression in hepatocellular carcinoma: a meta-analysis. PLoS One 2014;9:e103952.

70. Gao ZH, Tretiakova MS, Liu WH, Gong C, Farris PD, Hart J. Association of E-cadherin, matrix metalloproteinases, and tissue inhibitors of metalloproteinases with the progression and metastasis of hepatocellular carcinoma. Mod Pathol 2006;19:533-40.

71. Schneider MR, Hiltwein F, Grill J, Blum H, Krebs S, Klanner A, Bauersachs S, Bruns C, Longerich T, Horst D, Brandl L, de Toni E, Herbst A, Kolligs FT. Evidence for a role of E-cadherin in suppressing liver carcinogenesis in mice and men. Carcinogenesis 2014;35:1855-62.

72. Yang JY, Zong CS, Xia W, Wei Y, Ali-Seyed M, Li Z, Broglio K, Berry DA, Hung MC. MDM2 promotes cell motility and invasiveness by regulating E-cadherin degradation. Mol Cell Biol 2006;26:7269-82.

73. Hartsough MT, Steeg PS. Nm23/nucleoside diphosphate kinase in human cancers. J Bioenerg Biomembr 2000;32:301-8.

74. Thakur RK, Yadav VK, Kumar A, Singh A, Pal K, Hoeppner L, Saha D, Purohit G, Basundra R, Kar A, Halder R, Kumar P, Baral A, Kumar MJ, Baldi A, Vincenzi B, Lorenzon L, Banerjee R, Kumar P, Shridhar V, Mukhopadhyay D, Chowdhury S. Non-metastatic 2 (NME2)-mediated suppression of lung cancer metastasis involves transcriptional regulation of key cell adhesion factor vinculin. Nucleic Acids Res 2014;42:11589-600.

75. Miyazaki H, Fukuda M, Ishijima Y, Takagi Y, Iimura T, Negishi A, Hirayama R, Ishikawa N, Amagasa T, Kimura N. Overexpression of nm23-H2/NDP kinase B in a human oral squamous cell carcinoma cell line results in reduced metastasis, differentiated phenotype in the metastatic site, and growth factor-independent proliferative activity in culture. Clin Cancer Res 1999;5:4301-7.

76. Thakur RK, Yadav VK, Kumar P, Chowdhury S. Mechanisms of non-metastatic 2 (NME2)-mediated control of metastasis across tumor types. Naunyn Schmiedebergs Arch Pharmacol 2011;384:397-406.

77. Lee MJ, Xu DY, Li H, Yu GR, Leem SH, Chu IS, Kim IH, Kim DG. Pro-oncogenic potential of NM23-H2 in hepatocellular carcinoma. Exp Mol Med 2012;44:214-24.

78. Iizuka N, Oka M, Noma T, Nakazawa A, Hirose K, Suzuki T. NM23-H1 and NM23-H2 messenger RNA abundance in human hepatocellular carcinoma. Cancer Res 1995;55:652-7.

79. Polanski R, Maguire M, Nield PC, Jenkins RE, Park BK, Krawczynska K, Devling T, Ray-Sinha A, Rubbi CP, Vlatkovic N, Boyd MT. MDM2 interacts with NME2 (non-metastatic cells 2, protein) and suppresses the ability of NME2 to negatively regulate cell motility. Carcinogenesis 2011;32:1133-42.

80. Yang A, Kaghad M, Wang Y, Gillett E, Fleming MD, Dötsch V, Andrews NC, Caput D, McKeon F. p63, a p53 homolog at 3q27-29, encodes multiple products with transactivating, death-inducing, and dominant-negative activities. Mol Cell 1998;2:305-16.

81. Adorno M, Cordenonsi M, Montagner M, Dupont S, Wong C, Hann B, Solari A, Bobisse S, Rondina MB, Guzzardo V, Parenti AR, Rosato A, Bicciato S, Balmain A, Piccolo S. A Mutant-p53/Smad complex opposes p63 to empower TGFbeta-induced metastasis. Cell 2009;137:87-98.

82. Gu X, Coates PJ, Boldrup L, Nylander K. p63 contributes to cell invasion and migration in squamous cell carcinoma of the head and neck. Cancer Lett 2008;263:26-34.

83. Melino G. p63 is a suppressor of tumorigenesis and metastasis interacting with mutant p53. Cell Death Differ 2011;18:1487-99.

84. Tucci P, Agostini M, Grespi F, Markert EK, Terrinoni A, Vousden KH, Muller PA, Dötsch V, Kehrloesser S, Sayan BS, Giaccone G, Lowe SW, Takahashi N, Vandenabeele P, Knight RA, Levine AJ, Melino G. Loss of p63 and its microRNA-205 target results in enhanced cell migration and metastasis in prostate cancer. Proc Natl Acad Sci U S A 2012;109:15312-7.

85. Park BJ, Lee SJ, Kim JI, Lee SJ, Lee CH, Chang SG, Park JH, Chi SG. Frequent alteration of p63 expression in human primary bladder carcinomas. Cancer Res 2000;60:3370-4.

86. Spiesbach K, Tannapfel A, Mossner J, Engeland K. TAp63gamma can substitute for p53 in inducing expression of the maspin tumor suppressor. Int J Cancer 2005;114:555-62.

87. Hara T, Kijima H, Yamamoto S, Kenmochi T, Kise Y, Tanaka H, Chino O, Shimada H, Takazawa K, Tanaka M, Inokuchi S, Makuuchi H. Ubiquitous p63 expression in human esophageal squamous cell carcinoma. Int J Mol Med 2004;14:169-73.

88. Inoue K, Fry EA. Alterations of p63 and p73 in human cancers. Subcell Biochem 2014;85:17-40.

89. Flores ER, Sengupta S, Miller JB, Newman JJ, Bronson R, Crowley D, Yang A, McKeon F, Jacks T. Tumor predisposition in mice mutant for p63 and p73: evidence for broader tumor suppressor functions for the p53 family. Cancer Cell 2005;7:363-73.

90. Iwakuma T, Lozano G, Flores ER. Li-Fraumeni syndrome: a p53 family affair. Cell Cycle 2005;4:865-7.

91. Zdzalik M, Pustelny K, Kedracka-Krok S, Huben K, Pecak A, Wladyka B, Jankowski S, Dubin A, Potempa J, Dubin G. Interaction of regulators Mdm2 and Mdmx with transcription factors p53, p63 and p73. Cell Cycle 2010;9:4584-91.

92. Kadakia M, Slader C, Berberich SJ. Regulation of p63 function by Mdm2 and MdmX. DNA Cell Biol 2001;20:321-30.

93. Stindt MH, Muller PA, Ludwig RL, Kehrloesser S, Dotsch V, Vousden KH. Functional interplay between MDM2, p63/p73 and mutant p53. Oncogene 2015;34:4300-10.

94. Calabro V, Mansueto G, Parisi T, Vivo M, Calogero RA, La Mantia G. The human MDM2 oncoprotein increases the transcriptional activity and the protein level of the p53 homolog p63. J Biol Chem 2002;277:2674-81.

95. Lam EW, Brosens JJ, Gomes AR, Koo CY. Forkhead box proteins: tuning forks for transcriptional harmony. Nat Rev Cancer 2013;13:482-95.

96. Xu D, He X, Chang Y, Xu C, Jiang X, Sun S, Lin J. Inhibition of miR-96 expression reduces cell proliferation and clonogenicity of HepG2 hepatoma cells. Oncol Rep 2013;29:653-61.

97. Arden KC. Multiple roles of FOXO transcription factors in mammalian cells point to multiple roles in cancer. Exp Gerontol 2006;41:709-17.

98. Yamaguchi F, Hirata Y, Akram H, Kamitori K, Dong Y, Sui L, Tokuda M. FOXO/TXNIP pathway is involved in the suppression of hepatocellular carcinoma growth by glutamate antagonist MK-801. BMC Cancer 2013;13:468.

99. Zou Y, Tsai WB, Cheng CJ, Hsu C, Chung YM, Li PC, Lin SH, Hu MC. Forkhead box transcription factor FOXO3a suppresses estrogen-dependent breast cancer cell proliferation and tumorigenesis. Breast Cancer Res 2008;10:R21.

100. Blake DC, Jr. , Mikse OR, Freeman WM, Herzog CR. FOXO3a elicits a pro-apoptotic transcription program and cellular response to human lung carcinogen nicotine-derived nitrosaminoketone (NNK). Lung Cancer 2010;67:37-47.

101. Ni D, Ma X, Li HZ, Gao Y, Li XT, Zhang Y, Ai Q, Zhang P, Song EL, Huang QB, Fan Y, Zhang X. Downregulation of FOXO3a promotes tumor metastasis and is associated with metastasis-free survival of patients with clear cell renal cell carcinoma. Clin Cancer Res 2014;20:1779-90.

102. Cheng CW, Chen PM, Hsieh YH, Weng CC, Chang CW, Yao CC, Hu LY, Wu PE, Shen CY. Foxo3a-mediated overexpression of microRNA-622 suppresses tumor metastasis by repressing hypoxia-inducible factor-1alpha in erk-responsive of lung cancer. Oncotarget 2015; doi: 10.18632/oncotarget.5826.

103. Liu H, Yin J, Wang H, Jiang G, Deng M, Zhang G, Bu X, Cai S, Du J, He Z. FOXO3a modulates WNT/beta-catenin signaling and suppresses epithelial-to-mesenchymal transition in prostate cancer cells. Cell Signal 2015;27:510-8.

104. Shiota M, Song Y, Yokomizo A, Kiyoshima K, Tada Y, Uchino H, Uchiumi T, Inokuchi J, Oda Y, Kuroiwa K, Tatsugami K, Naito S. Foxo3a suppression of urothelial cancer invasiveness through Twist1, Y-box-binding protein 1, and E-cadherin regulation. Clin Cancer Res 2010;16:5654-63.

105. Su B, Gao L, Baranowski C, Gillard B, Wang J, Ransom R, Ko HK, Gelman IH. A genome-wide RNAi screen identifies FOXO4 as a metastasis-suppressor through counteracting PI3K/AKT signal pathway in prostate cancer. PLoS One 2014;9:e101411.

106. Xu MM, Mao GX, Liu J, Li JC, Huang H, Liu YF, Liu JH. Low expression of the FoxO4 gene may contribute to the phenomenon of EMT in non-small cell lung cancer. Asian Pac J Cancer Prev 2014;15:4013-8.

107. Xie C, Song LB, Wu JH, Li J, Yun JP, Lai JM, Xie DY, Lin BL, Yuan YF, Li M, Gao ZL. Upregulator of cell proliferation predicts poor prognosis in hepatocellular carcinoma and contributes to hepatocarcinogenesis by downregulating FOXO3a. PLoS One 2012;7:e40607.

108. Lu M, Ma J, Xue W, Cheng C, Wang Y, Zhao Y, Ke Q, Liu H, Liu Y, Li P, Cui X, He S, Shen A. The expression and prognosis of FOXO3a and Skp2 in human hepatocellular carcinoma. Pathol Oncol Res 2009;15:679-87.

109. Carbajo-Pescador S, Mauriz JL, Garcia-Palomo A, Gonzalez-Gallego J. FoxO proteins: regulation and molecular targets in liver cancer. Curr Med Chem 2014;21:1231-46.

110. Yang JY, Zong CS, Xia W, Yamaguchi H, Ding Q, Xie X, Lang JY, Lai CC, Chang CJ, Huang WC, Huang H, Kuo HP, Lee DF, Li LY, Lien HC, Cheng X, Chang KJ, Hsiao CD, Tsai FJ, Tsai CH, Sahin AA, Muller WJ, Mills GB, Yu D, Hortobagyi GN, Hung MC. ERK promotes tumorigenesis by inhibiting FOXO3a via MDM2-mediated degradation. Nat Cell Biol 2008;10:138-48.

111. Fu W, Ma Q, Chen L, Li P, Zhang M, Ramamoorthy S, Nawaz Z, Shimojima T, Wang H, Yang Y, Shen Z, Zhang Y, Zhang X, Nicosia SV, Zhang Y, Pledger JW, Chen J, Bai W. MDM2 acts downstream of p53 as an E3 ligase to promote FOXO ubiquitination and degradation. J Biol Chem 2009;284:13987-4000.

112. Brenkman AB, de Keizer PL, van den Broek NJ, Jochemsen AG, Burgering BM. Mdm2 induces mono-ubiquitination of FOXO4. PLoS One 2008;3:e2819.

113. Boyd MT, Vlatkovic N, Haines DS. A novel cellular protein (MTBP) binds to MDM2 and induces a G1 arrest that is suppressed by MDM2. J Biol Chem 2000;275:31883-90.

114. Agarwal N, Tochigi Y, Adhikari AS, Cui S, Cui Y, Iwakuma T. MTBP plays a crucial role in mitotic progression and chromosome segregation. Cell Death Differ 2011;18:1208-19.

115. Agarwal N, Adhikari AS, Iyer SV, Hekmatdoost K, Welch DR, Iwakuma T. MTBP suppresses cell migration and filopodia formation by inhibiting ACTN4. Oncogene 2013;32:462-70.

116. Bi Q, Ranjan A, Fan R, Agarwal N, Welch DR, Weinman SA, Ding J, Iwakuma T. MTBP inhibits migration and metastasis of hepatocellular carcinoma. Clin Exp Metastasis 2015;32:301-11.

117. Iwakuma T, Tochigi Y, Van Pelt CS, Caldwell LC, Terzian T, Parant JM, Chau GP, Koch JG, Eischen CM, Lozano G. Mtbp haploinsufficiency in mice increases tumor metastasis. Oncogene 2008;27:1813-20.

118. Vlatkovic N, El-Fert A, Devling T, Ray-Sinha A, Gore DM, Rubbi CP, Dodson A, Jones AS, Helliwell TR, Jones TM, Boyd MT. Loss of MTBP expression is associated with reduced survival in a biomarker-defined subset of patients with squamous cell carcinoma of the head and neck. Cancer 2011;117:2939-50.

119. Odvody J, Vincent T, Arrate MP, Grieb B, Wang S, Garriga J, Lozano G, Iwakuma T, Haines DS, Eischen CM. A deficiency in Mdm2 binding protein inhibits Myc-induced B-cell proliferation and lymphomagenesis. Oncogene 2010;29:3287-96.

120. Grieb BC, Chen X, Eischen CM. MTBP is Over-expressed in Triple Negative Breast Cancer and Contributes to its Growth and Survival. Mol Cancer Res 2014;12:1216-24.

121. Grieb BC, Gramling MW, Arrate MP, Chen X, Beauparlant SL, Haines DS, Xiao H, Eischen CM. Oncogenic protein MTBP interacts with MYC to promote tumorigenesis. Cancer Res 2014;74:3591-602.

122. Lu S, Zhou W, Wei H, He L, Li L. MTBP Promotes the Invasion and Metastasis of Hepatocellular Carcinoma by Enhancing the MDM2-Mediated Degradation of E-Cadherin. Dig Dis Sci 2015;60:3681-90.

123. Aleixo PB, Hartmann AA, Menezes IC, Meurer RT, Oliveira AM. Can MDM2 and CDK4 make the diagnosis of well differentiated/dedifferentiated liposarcoma? An immunohistochemical study on 129 soft tissue tumours. J Clin Pathol 2009;62:1127-35.

124. Reifenberger G, Liu L, Ichimura K, Schmidt EE, Collins VP. Amplification and overexpression of the MDM2 gene in a subset of human malignant gliomas without p53 mutations. Cancer Res 1993;53:2736-9.

125. Bond GL, Hu W, Bond EE, Robins H, Lutzker SG, Arva NC, Bargonetti J, Bartel F, Taubert H, Wuerl P, Onel K, Yip L, Hwang SJ, Strong LC, Lozano G, Levine AJ. A single nucleotide polymorphism in the MDM2 promoter attenuates the p53 tumor suppressor pathway and accelerates tumor formation in humans. Cell 2004;119:591-602.

126. Weber JD, Taylor LJ, Roussel MF, Sherr CJ, Bar-Sagi D. Nucleolar Arf sequesters Mdm2 and activates p53. Nat Cell Biol 1999;1:20-6.

127. Malmlof M, Roudier E, Hogberg J, Stenius U. MEK-ERK-mediated phosphorylation of Mdm2 at Ser-166 in hepatocytes. Mdm2 is activated in response to inhibited Akt signaling. J Biol Chem 2007;282:2288-96.

128. Capoulade C, Bressac-de Paillerets B, Lefrere I, Ronsin M, Feunteun J, Tursz T, Wiels J. Overexpression of MDM2, due to enhanced translation, results in inactivation of wild-type p53 in Burkitt's lymphoma cells. Oncogene 1998;16:1603-10.

129. Zhao Y, Yu H, Hu W. The regulation of MDM2 oncogene and its impact on human cancers. Acta Biochim Biophys Sin (Shanghai) 2014;46:180-9.

Cite This Article

Export citation file: BibTeX | RIS

OAE Style

Ranjan A, Bera K, Iwakuma T. Murine double minute 2, a potential p53-independent regulator of liver cancer metastasis. Hepatoma Res 2016;2:114-21. http://dx.doi.org/10.20517/2394-5079.2015.67

AMA Style

Ranjan A, Bera K, Iwakuma T. Murine double minute 2, a potential p53-independent regulator of liver cancer metastasis. Hepatoma Research. 2016; 2: 114-21. http://dx.doi.org/10.20517/2394-5079.2015.67

Chicago/Turabian Style

Ranjan, Atul, Kaustav Bera, Tomoo Iwakuma. 2016. "Murine double minute 2, a potential p53-independent regulator of liver cancer metastasis" Hepatoma Research. 2: 114-21. http://dx.doi.org/10.20517/2394-5079.2015.67

ACS Style

Ranjan, A.; Bera K.; Iwakuma T. Murine double minute 2, a potential p53-independent regulator of liver cancer metastasis. Hepatoma. Res. 2016, 2, 114-21. http://dx.doi.org/10.20517/2394-5079.2015.67

About This Article

This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 License (http://creativecommons.org/licenses/by-nc-sa/3.0/), which allows others to remix, tweak and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.

Data & Comments

Data

Views
4474
Downloads
883
Citations
5
Comments
0
0

Comments

Comments must be written in English. Spam, offensive content, impersonation, and private information will not be permitted. If any comment is reported and identified as inappropriate content by OAE staff, the comment will be removed without notice. If you have any queries or need any help, please contact us at support@oaepublish.com.

0
Download PDF
Cite This Article 4 clicks
Like This Article 0 likes
Share This Article
Scan the QR code for reading!
See Updates
Contents
Figures
Related
Hepatoma Research
ISSN 2454-2520 (Online) 2394-5079 (Print)

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/