TMEFF2: Comparison
Please note this is a comparison between Version 2 by Vivi Li and Version 1 by Motasim Masood.

Transmembrane protein with an EGF-like and two follistatin-like domains 2 (TMEFF2) is a 374-residue long type-I transmembrane proteoglycan which is proteolytically shed from the cell surface. The protein is involved in a range of functions including metabolism, neuroprotection, apoptosis, embryonic development, onco-suppression and endocrine function. TMEFF2 is methylated in numerous cancers, and an inverse correlation with the stage, response to therapy and survival outcome has been observed. Moreover, TMEFF2 methylation increases with breast, colon and gastric cancer progression. TMEFF2 is methylated early during oncogenesis in breast and colorectal cancer, and the detection of methylated free-circulating TMEFF2 DNA has been suggested as a potential diagnostic tool. The TMEFF2 downregulation signature equals and sometimes outperforms the Gleason and pathological scores in prostate cancer. TMEFF2 is downregulated in glioma and cotricotropinomas, and it impairs the production of adrenocorticotropic hormone in glioma cells. Through binding the amyloid β protein, its precursor and derivatives, TMEFF2 provides neuroprotection in Alzheimer’s disease. Primary literature regarding TMEFF2 is incoherent and offers conflicting information, in particular, the oncogenic vs. onco-suppressive role of TMEFF2 in prostate cancer. 

  • HPP1
  • TPEF
  • TENB2
  • tomoregulin
  • follistatin domain
  • ectodomain shedding
  • interferon signalling
  • Alzheimer’s disease
  • epigenetic silencing
  • transmembrane proteoglycan
Please wait, diff process is still running!

References

  1. Tweedie, S.; Braschi, B.; Gray, K.; Jones, T.E.M.; Seal, R.L.; Yates, B.; Bruford, E.A. Genenames.org: The HGNC and VGNC resources in 2021. Nucleic Acids Res 2020.
  2. HGNC Database, HUGO Gene Nomenclature Committee (HGNC), European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus. Available online: https://www.genenames.org/ (accessed on 16 December 2020).
  3. Liang, G.; Robertson, K.D.; Talmadge, C.; Sumegi, J.; Jones, P.A. The Gene for a Novel Transmembrane Protein Containing Epidermal Growth Factor and Follistatin Domains Is Frequently Hypermethylated in Human Tumor Cells. Cancer Res. 2000, 60, 4907–4912.
  4. Horie, M.; Mitsumoto, Y.; Kyushiki, H.; Kanemoto, N.; Watanabe, A.; Taniguchi, Y.; Nishino, N.; Okamoto, T.; Kondo, M.; Mori, T.; et al. Identification and Characterization of TMEFF2, a Novel Survival Factor for Hippocampal and Mesencephalic Neurons. Genomics 2000, 67, 146–152.
  5. Ali, N.; Knaüper, V. Phorbol Ester-induced Shedding of the Prostate Cancer Marker Transmembrane Protein with Epidermal Growth Factor and Two Follistatin Motifs 2 Is Mediated by the Disintegrin and Metalloproteinase-17. J. Biol. Chem. 2007, 282, 37378–37388.
  6. Labeur, M.; Wolfel, B.; Stalla, J.; Stalla, G.K. TMEFF2 is an endogenous inhibitor of the CRH signal transduction pathway. J. Mol. Endocrinol. 2015, 54, 51–63.
  7. Hong, H.S.; Maezawa, I.; Petrlova, J.; Zhao, X.Y.; John, C.V.; Jin, L.W. Tomoregulin (TMEFF2) Binds Alzheimer’s Disease Amyloid-beta (Abeta) Oligomer and AbetaPP and Protects Neurons from Abeta-Induced Toxicity. J. Alzheimers Dis. 2015, 48, 731–743.
  8. Lin, K.; Taylor, J.R., Jr.; Wu, T.D.; Gutierrez, J.; Elliott, J.M.; Vernes, J.-M.; Koeppen, H.; Phillips, H.S.; de Sauvage, F.J.; Meng, Y.G. TMEFF2 is a PDGF-AA binding protein with methylation-associated gene silencing in multiple cancer types including glioma. PLoS ONE 2011, 6, e18608.
  9. Ren, J.; Wen, L.; Gao, X.; Jin, C.; Xue, Y.; Yao, X. DOG 1.0: Illustrator of protein domain structures. Cell Res. 2009, 19, 271–273.
  10. Young, J.; Biden, K.G.; Simms, L.A.; Huggard, P.; Karamatic, R.; Eyre, H.J.; Sutherland, G.R.; Herath, N.; Barker, M.; Anderson, G.J.; et al. HPP1: A transmembrane protein-encoding gene commonly methylated in colorectal polyps and cancers. Proc. Natl. Acad. Sci. USA 2001, 98, 265–270.
  11. Yonezawa, M.; Uchida, T.; Wada, K.; Akamatsu, T.; Mizoguchi, A.; Sakamoto, C.; Tsukui, T.; Sinoki, K.; Satou, J. A novel epidermal growth factor-like molecule containing two follistatin modules stimulates tyrosine phosphorylation of ERBB-4 in MKN28 gastric cancer cells. Gastroenterology 2000, 118, A557.
  12. Glynne-Jones, E.; Harper, M.E.; Seery, L.T.; James, R.; Anglin, I.; Morgan, H.E.; Taylor, K.M.; Gee, J.M.; Nicholson, R.I. TENB2, a proteoglycan identified in prostate cancer that is associated with disease progression and androgen independence. Int. J. Cancer 2001, 94, 178–184.
  13. Quayle, S.N.; Sadar, M.D. A truncated isoform of TMEFF2 encodes a secreted protein in prostate cancer cells. Genomics 2006, 87, 633–637.
  14. Uchida, T.; Wada, K.; Akamatsu, T.; Yonezawa, M.; Noguchi, H.; Mizoguchi, A.; Kasuga, M.; Sakamoto, C. A Novel Epidermal Growth Factor-like Molecule Containing Two Follistatin Modules Stimulates Tyrosine Phosphorylation of erbB-4 in MKN28 Gastric Cancer Cells. Biochem. Biophys. Res. Commun. 1999, 266, 593–602.
  15. Zhao, X.-Y.; Schneider, D.; Biroc, S.L.; Parry, R.; Alicke, B.; Toy, P.; Xuan, J.-A.; Sakamoto, C.; Wada, K.; Schulze, M.; et al. Targeting Tomoregulin for Radioimmunotherapy of Prostate Cancer. Cancer Res. 2005, 65, 2846–2853.
  16. Liang, G.; Salem, C.E.; Yu, M.C.; Nguyen, H.D.; Gonzales, F.A.; Nguyen, T.T.; Nichols, P.W.; Jones, P.A. DNA Methylation Differences Associated with Tumor Tissues Identified by Genome Scanning Analysis. Genomics 1998, 53, 260–268.
  17. Gawel-Beben, K.; Ali, N.; Ellis, V.; Velasco, G.; Poghosyan, Z.; Ager, A.; Knauper, V. TMEFF2 shedding is regulated by oxidative stress and mediated by ADAMs and transmembrane serine proteases implicated in prostate cancer. Cell Biol. Int. 2018, 42, 273–280.
  18. Mohler, J.L.; Morris, T.L.; Ford, O.H., 3rd; Alvey, R.F.; Sakamoto, C.; Gregory, C.W. Identification of differentially expressed genes associated with androgen-independent growth of prostate cancer. Prostate 2002, 51, 247–255.
  19. Chen, Q.; Watson, J.T.; Marengo, S.R.; Decker, K.S.; Coleman, I.; Nelson, P.S.; Sikes, R.A. Gene expression in the LNCaP human prostate cancer progression model: Progression associated expression in vitro corresponds to expression changes associated with prostate cancer progression in vivo. Cancer Lett. 2006, 244, 274–288.
  20. Green, T.; Chen, X.; Ryan, S.; Asch, A.S.; Ruiz-Echevarría, M.J. TMEFF2 and SARDH cooperate to modulate one-carbon metabolism and invasion of prostate cancer cells. Prostate 2013, 73, 1561–1575.
  21. Overcash, R.F.; Chappell, V.A.; Green, T.; Geyer, C.B.; Asch, A.S.; Ruiz-Echevarria, M.J. Androgen signaling promotes translation of TMEFF2 in prostate cancer cells via phosphorylation of the alpha subunit of the translation initiation factor 2. PLoS ONE 2013, 8, e55257.
  22. Chen, X.; Corbin, J.M.; Tipton, G.J.; Yang, L.V.; Asch, A.S.; Ruiz-Echevarría, M.J. The TMEFF2 tumor suppressor modulates integrin expression, RhoA activation and migration of prostate cancer cells. Biochim. Biophys. Acta 2014, 1843, 1216–1224.
  23. Torrecilla, D.; Lozano, M.V.; Lallana, E.; Neissa, J.I.; Novoa-Carballal, R.; Vidal, A.; Fernandez-Megia, E.; Torres, D.; Riguera, R.; Alonso, M.J.; et al. Anti-tumor efficacy of chitosan-g-poly(ethylene glycol) nanocapsules containing docetaxel: Anti-TMEFF-2 functionalized nanocapsules vs. non-functionalized nanocapsules. Eur. J. Pharm. Biopharm. 2013, 83, 330–337.
  24. Gery, S.; Sawyers, C.L.; Agus, D.B.; Said, J.W.; Koeffler, H.P. TMEFF2 is an androgen-regulated gene exhibiting antiproliferative effects in prostate cancer cells. Oncogene 2002, 21, 4739–4746.
  25. Afar, D.E.; Bhaskar, V.; Ibsen, E.; Breinberg, D.; Henshall, S.M.; Kench, J.G.; Drobnjak, M.; Powers, R.; Wong, M.; Evangelista, F.; et al. Preclinical validation of anti-TMEFF2-auristatin E-conjugated antibodies in the treatment of prostate cancer. Mol. Cancer Ther. 2004, 3, 921–932.
  26. Han, H.; Zhan, Z.; Xu, J.; Song, Z. TMEFF2 inhibits pancreatic cancer cells proliferation, migration, and invasion by suppressing phosphorylation of the MAPK signaling pathway. Onco Targets Ther. 2019, 12, 11371–11382.
  27. Corbin, J.M.; Overcash, R.F.; Wren, J.D.; Coburn, A.; Tipton, G.J.; Ezzell, J.A.; McNaughton, K.K.; Fung, K.-M.; Kosanke, S.D.; Ruiz-Echevarria, M.J. Analysis of TMEFF2 allografts and transgenic mouse models reveals roles in prostate regeneration and cancer. Prostate 2016, 76, 97–113.
  28. Kanemoto, N.; Horie, M.; Omori, K.; Nishino, N.; Kondo, M.; Noguchi, K.; Tanigami, A. Expression of TMEFF1 mRNA in the mouse central nervous system: Precise examination and comparative studies of TMEFF1 and TMEFF2. Brain Res. Mol. Brain Res. 2001, 86, 48–55.
  29. Chen, T.R.; Wang, P.; Carroll, L.K.; Zhang, Y.J.; Han, B.X.; Wang, F. Generation and characterization of TMEFF2 mutant mice. Biochem. Biophys. Res. Commun. 2012, 425, 189–194.
  30. Zhao, X.Y.; Liu, H.L.; Liu, B.; Willuda, J.; Siemeister, G.; Mahmoudi, M.; Dinter, H. Tomoregulin internalization confers selective cytotoxicity of immunotoxins on prostate cancer cells. Transl. Oncol. 2008, 1, 102–109.
  31. Chen, X.; Overcash, R.; Green, T.; Hoffman, D.; Asch, A.S.; Ruiz-Echevarria, M.J. The tumor suppressor activity of the transmembrane protein with epidermal growth factor and two follistatin motifs 2 (TMEFF2) correlates with its ability to modulate sarcosine levels. J. Biol. Chem. 2011, 286, 16091–16100.
  32. Huang, H.; Teng, P.; Mei, R.; Yang, A.; Zhang, Z.; Zhao, X.; Qiu, M. TMEFF2 is expressed in differentiating oligodendrocytes but dispensable for their differentiation in vivo. Sci. Rep. 2017, 7, 337.
  33. Elahi, A.; Zhang, L.; Yeatman, T.J.; Gery, S.; Sebti, S.; Shibata, D. HPP1-mediated tumor suppression requires activation of STAT1 pathways. Int. J. Cancer 2008, 122, 1567–1572.
  34. Hernandez, J.M.; Elahi, A.; Clark, W.; Humphries, L.A.; Wang, J.; Achille, A.; Seto, E.; Shibata, D. The Tumor Suppressive Effects of HPP1 Are Mediated Through JAK-STAT-Interferon Signaling Pathways. DNA Cell Biol. 2015, 34, 541–549.
  35. Sun, T.-T.; Tang, J.-Y.; Du, W.; Zhao, H.-J.; Zhao, G.; Yang, S.-L.; Chen, H.-Y.; Hong, J.; Fang, J.-Y. Bidirectional regulation between TMEFF2 and STAT3 may contribute to Helicobacter pylori-associated gastric carcinogenesis. Int. J. Cancer 2015, 136, 1053–1064.
  36. Sun, T.; Du, W.; Xiong, H.; Yu, Y.; Weng, Y.; Ren, L.; Zhao, H.; Wang, Y.; Chen, Y.; Xu, J.; et al. TMEFF2 deregulation contributes to gastric carcinogenesis and indicates poor survival outcome. Clin. Cancer Res. 2014, 20, 4689–4704.
  37. Patel, K. Follistatin. Int. J. Biochem. Cell Biol. 1998, 30, 1087–1093.
  38. Gao, L.; Nie, X.; Zheng, M.; Li, X.; Guo, Q.; Liu, J.; Liu, Q.; Hao, Y.; Lin, B. TMEFF2 is a novel prognosis signature and target for endometrial carcinoma. Life Sci. 2020, 243, 116910.
  39. Li, H.; Zhou, Y.; Cheng, H.; Tian, J.; Yang, S. Roles of a TMPO-AS1/microRNA-200c/TMEFF2 ceRNA network in the malignant behaviors and 5-FU resistance of ovarian cancer cells. Exp. Mol. Pathol. 2020, 115, 104481.
  40. Li, K.; Han, H.; Gu, W.; Cao, C.; Zheng, P. Long non-coding RNA LINC01963 inhibits progression of pancreatic carcinoma by targeting miR-641/TMEFF2. Biomed. Pharmacother. 2020, 129, 110346.
  41. Fan, J.M.; Zheng, Z.R.; Zeng, Y.M.; Chen, X.Y. MiR-323-3p Targeting Transmembrane Protein with EGF-Like and 2 Follistatin Domain (TMEFF2) Inhibits Human Lung Cancer A549 Cell Apoptosis by Regulation of AKT and ERK Signaling Pathways. Med. Sci. Monit. 2020, 26, e919454.
  42. Sato, F.; Shibata, D.; Harpaz, N.; Xu, Y.; Yin, J.; Mori, Y.; Wang, S.; Olaru, A.; Deacu, E.; Selaru, F.M.; et al. Aberrant methylation of the HPP1 gene in ulcerative colitis-associated colorectal carcinoma. Cancer Res. 2002, 62, 6820–6822.
  43. Lee, S.M.; Park, J.Y.; Kim, D.S. Methylation of TMEFF2 gene in tissue and serum DNA from patients with non-small cell lung cancer. Mol. Cells 2012, 34, 171–176.
  44. Young, J.; Barker, M.; Fraser, L.; Walsh, M.D.; Spring, K.; Biden, K.G.; Hopper, J.L.; Leggett, B.A.; Jass, J.R. Mutation searching in colorectal cancer studies: Experience with a denaturing high-pressure liquid chromatography system for exon-by-exon scanning of tumour suppressor genes. Pathology 2002, 34, 529–533.
  45. Fackler, M.J.; Umbricht, C.B.; Williams, D.; Argani, P.; Cruz, L.A.; Merino, V.F.; Teo, W.W.; Zhang, Z.; Huang, P.; Visvananthan, K.; et al. Genome-wide methylation analysis identifies genes specific to breast cancer hormone receptor status and risk of recurrence. Cancer Res. 2011, 71, 6195–6207.
  46. Fackler, M.J.; Lopez Bujanda, Z.; Umbricht, C.; Teo, W.W.; Cho, S.; Zhang, Z.; Visvanathan, K.; Jeter, S.; Argani, P.; Wang, C.; et al. Novel methylated biomarkers and a robust assay to detect circulating tumor DNA in metastatic breast cancer. Cancer Res. 2014, 74, 2160–2170.
  47. Park, S.Y.; Kwon, H.J.; Lee, H.E.; Ryu, H.S.; Kim, S.-W.; Kim, J.H.; Kim, I.A.; Jung, N.; Cho, N.-Y.; Kang, G.H. Promoter CpG island hypermethylation during breast cancer progression. Virchows Arch. 2011, 458, 73–84.
  48. de Groot, J.S.; Pan, X.; Meeldijk, J.; van der Wall, E.; van Diest, P.J.; Moelans, C.B. Validation of DNA promoter hypermethylation biomarkers in breast cancer—A short report. Cell. Oncol. 2014, 37, 297–303.
  49. Suzuki, M.; Shigematsu, H.; Shames, D.S.; Sunaga, N.; Takahashi, T.; Shivapurkar, N.; Iizasa, T.; Frenkel, E.P.; Minna, J.D.; Fujisawa, T.; et al. DNA methylation-associated inactivation of TGFβ-related genes DRM/Gremlin, RUNX3, and HPP1 in human cancers. Br. J. Cancer 2005, 93, 1029–1037.
  50. Zhang, C.; Zhao, H.; Li, J.; Liu, H.; Wang, F.; Wei, Y.; Su, J.; Zhang, D.; Liu, T.; Zhang, Y. The Identification of Specific Methylation Patterns across Different Cancers. PLoS ONE 2015, 10, e0120361.
  51. Belshaw, N.J.; Elliott, G.O.; Williams, E.A.; Bradburn, D.M.; Mills, S.J.; Mathers, J.C.; Johnson, I.T. Use of DNA from Human Stools to Detect Aberrant CpG Island Methylation of Genes Implicated in Colorectal Cancer. Cancer Epidemiol. Biomark. Prev. 2004, 13, 1495–1501.
  52. Wynter, C.V.; Walsh, M.D.; Higuchi, T.; Leggett, B.A.; Young, J.; Jass, J.R. Methylation patterns define two types of hyperplastic polyp associated with colorectal cancer. Gut 2004, 53, 573–580.
  53. Takahashi, T.; Shivapurkar, N.; Riquelme, E.; Shigematsu, H.; Reddy, J.; Suzuki, M.; Miyajima, K.; Zhou, X.; Bekele, B.N.; Gazdar, A.F.; et al. Aberrant promoter hypermethylation of multiple genes in gallbladder carcinoma and chronic cholecystitis. Clin. Cancer Res. 2004, 10, 6126–6133.
  54. Chen, E.; Zheng, F.; Yuan, X.; Ye, Y.; Li, X.; Dai, Y.; Chen, L. The effect of TMEFF2 methylation on the tumor stage and survival outcome of clear cell renal cell carcinoma. Cancer Biomark. 2017, 19, 207–212.
  55. Geddert, H.; Kiel, S.; Iskender, E.; Florl, A.R.; Krieg, T.; Vossen, S.; Gabbert, H.E.; Sarbia, M. Correlation of hMLH1 and HPP1 hypermethylation in gastric, but not in esophageal and cardiac adenocarcinoma. Int. J. Cancer 2004, 110, 208–211.
  56. Hadjinicolaou, A.V.; van Munster, S.N.; Achilleos, A.; Santiago Garcia, J.; Killcoyne, S.; Ragunath, K.; Bergman, J.J.G.H.M.; Fitzgerald, R.C.; di Pietro, M. Aneuploidy in targeted endoscopic biopsies outperforms other tissue biomarkers in the prediction of histologic progression of Barrett’s oesophagus: A multi-centre prospective cohort study. EBioMedicine 2020, 56, 102765.
  57. Shibata, D.M.; Sato, F.; Mori, Y.; Perry, K.; Yin, J.; Wang, S.; Xu, Y.; Olaru, A.; Selaru, F.; Spring, K.; et al. Hypermethylation of HPP1 is associated with hMLH1 hypermethylation in gastric adenocarcinomas. Cancer Res. 2002, 62, 5637–5640.
  58. Suzuki, M.; Toyooka, S.; Shivapurkar, N.; Shigematsu, H.; Miyajima, K.; Takahashi, T.; Stastny, V.; Zern, A.L.; Fujisawa, T.; Pass, H.I.; et al. Aberrant methylation profile of human malignant mesotheliomas and its relationship to SV40 infection. Oncogene 2005, 24, 1302–1308.
  59. Sabbioni, S.; Miotto, E.; Veronese, A.; Sattin, E.; Gramantieri, L.; Bolondi, L.; Calin, G.A.; Gafa, R.; Lanza, G.; Carli, G.; et al. Multigene methylation analysis of gastrointestinal tumors: TPEF emerges as a frequent tumor-specific aberrantly methylated marker that can be detected in peripheral blood. Mol. Diagn. 2003, 7, 201–207.
  60. Herbst, A.; Vdovin, N.; Gacesa, S.; Philipp, A.; Nagel, D.; Holdt, L.M.; op den Winkel, M.; Heinemann, V.; Stieber, P.; Graeven, U.; et al. Methylated free-circulating HPP1 DNA is an early response marker in patients with metastatic colorectal cancer. Int. J. Cancer 2017, 140, 2134–2144.
  61. Wang, J.; Elahi, A.; Ajidahun, A.; Clark, W.; Hernandez, J.; Achille, A.; Hao, J.H.; Seto, E.; Shibata, D. The interplay between histone deacetylases and c-Myc in the transcriptional suppression of HPP1 in colon cancer. Cancer Biol. Ther. 2014, 15, 1198–1207.
  62. Georgescu, C.; Corbin, J.M.; Thibivilliers, S.; Webb, Z.D.; Zhao, Y.D.; Koster, J.; Fung, K.-M.; Asch, A.S.; Wren, J.D.; Ruiz-Echevarría, M.J. A TMEFF2-regulated cell cycle derived gene signature is prognostic of recurrence risk in prostate cancer. BMC Cancer 2019, 19, 423.
  63. Pitale, P.M.; Gorbatyuk, O.; Gorbatyuk, M. Neurodegeneration: Keeping ATF4 on a Tight Leash. Front. Cell. Neurosci. 2017, 11.
  64. Harding, H.P.; Novoa, I.; Zhang, Y.; Zeng, H.; Wek, R.; Schapira, M.; Ron, D. Regulated Translation Initiation Controls Stress-Induced Gene Expression in Mammalian Cells. Mol. Cell 2000, 6, 1099–1108.
  65. Edwards, D.R.; Handsley, M.M.; Pennington, C.J. The ADAM metalloproteinases. Mol. Asp. Med. 2008, 29, 258–289.
  66. Lin, H.; Wada, K.; Yonezawa, M.; Shinoki, K.; Akamatsu, T.; Tsukui, T.; Sakamoto, C. Tomoregulin ectodomain shedding by proinflammatory cytokines. Life Sci. 2003, 73, 1617–1627.
  67. Blobel, C.P. ADAMs: Key components in EGFR signalling and development. Nat. Rev. Mol. Cell Biol. 2005, 6, 32–43.
  68. Elahi, A.; Ajidahun, A.; Hendrick, L.; Getun, I.; Humphries, L.A.; Hernandez, J.; Shibata, D. HPP1 Ectodomain Shedding is Mediated by ADAM17 and is Necessary for Tumor Suppression in Colon Cancer. J. Surg. Res. 2020, 254, 183–190.
  69. Hornbeck, P.V.; Zhang, B.; Murray, B.; Kornhauser, J.M.; Latham, V.; Skrzypek, E. PhosphoSitePlus, 2014: Mutations, PTMs and recalibrations. Nucleic Acids Res. 2014, 43, D512–D520.
More