Skip to main content

Advertisement

Log in

Emerging role of nuclear protein 1 (NUPR1) in cancer biology

  • NON-THEMATIC REVIEW
  • Published:
Cancer and Metastasis Reviews Aims and scope Submit manuscript

Abstract

NUPR1, or p8 or com1, was first identified from rat pancreas during acute pancreatitis and later as a gene whose expression was upregulated in metastatic breast cancer cells. NUPR1 is a molecule whose expression is upregulated in response to stress and is hence influenced by the host microenvironment. While NUPR1 has been implicated in several diseases, there is no singular biochemical pathway that can be attributed to its role in cancer. NUPR1 has been found to aid the establishment of metastasis and to play a key role in the progression of several malignancies including those of breast, thyroid, brain and pancreas. NUPR1 has been implicated in inducing chemoresistance in pancreatic and breast cancer cells, protecting them from apoptosis and making tumor cells genetically unstable. In prostate cancer, however, NUPR1 appears to have tumor suppressive activity. Understanding the mechanism of action of the multifaceted functions of NUPR1 may open up new dimensions towards creating novel therapies against cancer as well as other pathologies. This review draws on several published studies on NUPR1, mainly in cancer biology, and assesses NUPR1 from the perspective of its functional role in making cancer cells resistant to the action of conventional chemotherapeutic drugs.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

Abbreviations

NUPR1:

Nuclear protein 1

NLS:

Nuclear localization signal

NES:

Nuclear export signal

MEF:

Mouse embryonic fibroblasts

References

  1. Mallo, G. V., Fiedler, F., Calvo, E. L., Ortiz, E. M., Vasseur, S., Keim, V., et al. (1997). Cloning and expression of the rat p8 cDNA, a new gene activated in pancreas during the acute phase of pancreatitis, pancreatic development, and regeneration, and which promotes cellular growth. Journal of biological chemistry, 272, 32360–32369.

    Article  PubMed  CAS  Google Scholar 

  2. Ree, A. H., Tvermyr, M., Engebraaten, O., Rooman, M., Rosok, O., Hovig, E., et al. (1999). Expression of a novel factor in human breast cancer cells with metastatic potential. Cancer research, 59, 4675–4680.

    PubMed  CAS  Google Scholar 

  3. Giroux, V., Malicet, C., Barthet, M., Gironella, M., Archange, C., Dagorn, J. C., et al. (2006). p8 is a new target of gemcitabine in pancreatic cancer cells. Clinical cancer research, 12, 235–241.

    Article  PubMed  CAS  Google Scholar 

  4. Clark, D. W., Mitra, A., Fillmore, R. A., Jiang, W. G., Samant, R. S., Fodstad, O., et al. (2008). NUPR1 interacts with p53, transcriptionally regulates p21 and rescues breast epithelial cells from doxorubicin-induced genotoxic stress. Current cancer drug targets, 8(5), 421–430.

    Article  PubMed  CAS  Google Scholar 

  5. Vasseur, S., Vidal Mallo, G., Fiedler, F., Bodeker, H., Canepa, E., Moreno, S., et al. (1999). Cloning and expression of the human p8, a nuclear protein with mitogenic activity. European journal of biochemistry, 259, 670–675.

    Article  PubMed  CAS  Google Scholar 

  6. Vasseur, S., Hoffmeister, A., Garcia-Montero, A., Barthet, M., Saint-Michel, L., Berthezene, P., et al. (2003). Mice with targeted disruption of p8 gene show increased sensitivity to lipopolysaccharide and DNA microarray analysis of livers reveals an aberrant gene expression response. BMC Gastroenterol, 3, 25.

    Article  PubMed  Google Scholar 

  7. Taieb, D., Malicet, C., Garcia, S., Rocchi, P., Arnaud, C., Dagorn, J. C., et al. (2005). Inactivation of stress protein p8 increases murine carbon tetrachloride hepatotoxicity via preserved CYP2E1 activity. Hepatology, 42, 176–182.

    Article  PubMed  CAS  Google Scholar 

  8. Vasseur, S., Hoffmeister, A., Garcia-Montero, A., Mallo, G. V., Feil, R., Kuhbandner, S., et al. (2002). p8-deficient fibroblasts grow more rapidly and are more resistant to adriamycin-induced apoptosis. Oncogene, 21, 1685–1694.

    Article  PubMed  CAS  Google Scholar 

  9. Courjal, F., & Theillet, C. (1997). Comparative genomic hybridization analysis of breast tumors with predetermined profiles of DNA amplification. Cancer research, 57, 4368–4377.

    PubMed  CAS  Google Scholar 

  10. Valacco, M. P., Varone, C., Malicet, C., Canepa, E., Iovanna, J. L., Moreno, S. (2006). Cell growth-dependent subcellular localization of p8. Journal of cellular biochemistry, 97, 1066–1079.

    Article  PubMed  CAS  Google Scholar 

  11. Malicet, C., Lesavre, N., Vasseur, S., & Iovanna, J. L. (2003). p8 inhibits the growth of human pancreatic cancer cells and its expression is induced through pathways involved in growth inhibition and repressed by factors promoting cell growth. Molecular cancer, 2, 37.

    Article  PubMed  Google Scholar 

  12. Dingwall, C., & Laskey, R. A. (1991). Nuclear targeting sequences—a consensus? Trends in biochemical sciences, 16, 478–481.

    Article  PubMed  CAS  Google Scholar 

  13. Goruppi, S., & Kyriakis, J. M. (2004). The pro-hypertrophic basic helix-loop-helix protein p8 is degraded by the ubiquitin/proteasome system in a protein kinase B/Akt— and glycogen synthase kinase-3-dependent manner, whereas endothelin induction of p8 mRNA and renal mesangial cell hypertrophy require NFAT4. Journal of biological chemistry, 279, 20950–20958.

    Article  PubMed  CAS  Google Scholar 

  14. Path, G., Opel, A., Knoll, A., & Seufert, J. (2004). Nuclear protein p8 is associated with glucose-induced pancreatic beta-cell growth. Diabetes, 53(Suppl 1), S82–85.

    Article  PubMed  Google Scholar 

  15. Vasseur, S., Folch-Puy, E., Hlouschek, V., Garcia, S., Fiedler, F., Lerch, M. M., et al. (2004). p8 improves pancreatic response to acute pancreatitis by enhancing the expression of the anti-inflammatory protein pancreatitis-associated protein I. Journal of biological chemistry, 279, 7199–7207.

    Article  PubMed  CAS  Google Scholar 

  16. Carracedo, A., Egia, A., Guzman, M., & Velasco, G. (2006). p8 Upregulation sensitizes astrocytes to oxidative stress. FEBS letters, 580, 1571–1575.

    Article  PubMed  CAS  Google Scholar 

  17. Goruppi, S., Patten, R. D., Force, T., & Kyriakis, J. M. (2007). Helix-loop-helix protein p8, a transcriptional regulator required for cardiomyocyte hypertrophy and cardiac fibroblast matrix metalloprotease induction. Molecular and cellular biology, 27, 993–1006.

    Article  PubMed  CAS  Google Scholar 

  18. Malicet, C., Giroux, V., Vasseur, S., Dagorn, J. C., Neira, J. L., & Iovanna, J. L. (2006). Regulation of apoptosis by the p8/prothymosin alpha complex. Proceedings of the National Academy of Sciences of the United States of America, 103, 2671–2676.

    Article  PubMed  CAS  Google Scholar 

  19. Ree, A. H., Pacheco, M. M., Tvermyr, M., Fodstad, O., & Brentani, M. M. (2000). Expression of a novel factor, com1, in early tumor progression of breast cancer. Clinical cancer research, 6, 1778–1783.

    PubMed  CAS  Google Scholar 

  20. Su, S. B., Motoo, Y., Iovanna, J. L., Berthezene, P., Xie, M. J., Mouri, H., et al. (2001). Overexpression of p8 is inversely correlated with apoptosis in pancreatic cancer. Clinical cancer research, 7, 1320–1324.

    PubMed  CAS  Google Scholar 

  21. Su, S. B., Motoo, Y., Iovanna, J. L., Xie, M. J., Mouri, H., Ohtsubo, K., et al. (2001). Expression of p8 in human pancreatic cancer. Clinical cancer research, 7, 309–313.

    PubMed  CAS  Google Scholar 

  22. Quirk, C. C., Seachrist, D. D., & Nilson, J. H. (2003). Embryonic expression of the luteinizing hormone beta gene appears to be coupled to the transient appearance of p8, a high mobility group-related transcription factor. Journal of biological chemistry, 278, 1680–1685.

    Article  PubMed  CAS  Google Scholar 

  23. Mohammad, H. P., Seachrist, D. D., Quirk, C. C., & Nilson, J. H. (2004). Reexpression of p8 contributes to tumorigenic properties of pituitary cells and appears in a subset of prolactinomas in transgenic mice that hypersecrete luteinizing hormone. Molecular endocrinology, 18, 2583–2593.

    Article  PubMed  CAS  Google Scholar 

  24. Brannon, K. M., Million Passe, C. M., White, C. R., Bade, N. A., King, M. W., & Quirk, C. C. (2007). Expression of the high mobility group A family member p8 is essential to maintaining tumorigenic potential by promoting cell cycle dysregulation in LbetaT2 cells. Cancer letter, 254, 146–155.

    Article  CAS  Google Scholar 

  25. Ito, Y., Yoshida, H., Motoo, Y., Miyoshi, E., Iovanna, J. L., Tomoda, C., et al. (2003). Expression and cellular localization of p8 protein in thyroid neoplasms. Cancer letter, 201, 237–244.

    Article  CAS  Google Scholar 

  26. Ree, A. H., Bratland, A., Kroes, R. A., Aasheim, H. C., Florenes, V. A., Moskal, J. R., et al. (2002). Clinical and cell line specific expression profiles of a human gene identified in experimental central nervous system metastases. Anticancer research, 22, 1949–1957.

    PubMed  Google Scholar 

  27. Bratland, A., Risberg, K., Maelandsmo, G. M., Gutzkow, K. B., Olsen, O. E., Moghaddam, A., et al. (2000). Expression of a novel factor, com1, is regulated by 1,25-dihydroxyvitamin D3 in breast cancer cells. Cancer research, 60, 5578–5583.

    PubMed  CAS  Google Scholar 

  28. Vasseur, S., Hoffmeister, A., Garcia, S., Bagnis, C., Dagorn, J. C., & Iovanna, J. L. (2002). p8 is critical for tumour development induced by rasV12 mutated protein and E1A oncogene. EMBO reports, 3, 165–170.

    Article  PubMed  CAS  Google Scholar 

  29. Blazquez, C., Salazar, M., Carracedo, A., Lorente, M., Egia, A., Gonzalez-Feria, L., et al. (2008). Cannabinoids inhibit glioma cell invasion by down-regulating matrix metalloproteinase-2 expression. Cancer research, 68, 1945–1952.

    Article  PubMed  CAS  Google Scholar 

  30. Blazquez, C., Carracedo, A., Salazar, M., Lorente, M., Egia, A., Gonzalez-Feria, L., et al. (2008). Down-regulation of tissue inhibitor of metalloproteinases-1 in gliomas: a new marker of cannabinoid antitumoral activity? Neuropharmacology, 54, 235–243.

    Article  PubMed  CAS  Google Scholar 

  31. Carracedo, A., Gironella, M., Lorente, M., Garcia, S., Guzman, M., Velasco, G., et al. (2006). Cannabinoids induce apoptosis of pancreatic tumor cells via endoplasmic reticulum stress-related genes. Cancer research, 66, 6748–6755.

    Article  PubMed  CAS  Google Scholar 

  32. Jiang, W. G., Watkins, G., Douglas-Jones, A., Mokbel, K., Mansel, R. E., & Fodstad, O. (2005). Expression of Com-1/P8 in human breast cancer and its relevance to clinical outcome and ER status. International journal of cancer, 117, 730–737.

    Article  CAS  Google Scholar 

  33. Jiang, W. G., Davies, G., & Fodstad, O. (2005). Com-1/P8 in oestrogen regulated growth of breast cancer cells, the ER-beta connection. Biochemical and biophysical research communications, 330, 253–262.

    Article  PubMed  CAS  Google Scholar 

  34. Jiang, W. G., Davies, G., Martin, T. A., Kynaston, H., Mason, M. D., & Fodstad, O. (2006). Com-1/p8 acts as a putative tumour suppressor in prostate cancer. International Journal of Molecular Medicine, 18, 981–986.

    PubMed  CAS  Google Scholar 

  35. Ishida, M., Miyamoto, M., Naitoh, S., Tatsuda, D., Hasegawa, T., Nemoto, T., et al. (2007). The SYT-SSX fusion protein down-regulates the cell proliferation regulator COM1 in t(x;18) synovial sarcoma. Molecular and cellular biology, 27, 1348–1355.

    Article  PubMed  CAS  Google Scholar 

  36. Dulic, V., Stein, G. H., Far, D. F., & Reed, S. I. (1998). Nuclear accumulation of p21Cip1 at the onset of mitosis: a role at the G2/M-phase transition. Molecular and cellular biology, 18, 546–557.

    PubMed  CAS  Google Scholar 

  37. Medema, R. H., Klompmaker, R., Smits, V. A., & Rijksen, G. (1998). p21waf1 can block cells at two points in the cell cycle, but does not interfere with processive DNA-replication or stress-activated kinases. Oncogene, 16, 431–441.

    Article  PubMed  CAS  Google Scholar 

  38. Niculescu 3rd, A. B., Chen, X., Smeets, M., Hengst, L., Prives, C., & Reed, S. I. (1998). Effects of p21(Cip1/Waf1) at both the G1/S and the G2/M cell cycle transitions: pRb is a critical determinant in blocking DNA replication and in preventing endoreduplication. Molecular and cellular biology, 18, 629–643.

    PubMed  CAS  Google Scholar 

  39. Smits, V. A., & Medema, R. H. (2001). Checking out the G(2)/M transition. Biochimica et biophysica acta, 1519, 1–12.

    PubMed  CAS  Google Scholar 

  40. Staversky, R. J., Vitiello, P. F., Gehen, S. C., Helt, C. E., Rahman, A., Keng, P. C., et al. (2006). p21(Cip1/Waf1/Sdi1) protects against hyperoxia by maintaining expression of Bcl-X(L). Free radical biology & medicine, 41, 601–609.

    Article  CAS  Google Scholar 

  41. Tian, H., Wittmack, E. K., & Jorgensen, T. J. (2000). p21WAF1/CIP1 antisense therapy radiosensitizes human colon cancer by converting growth arrest to apoptosis. Cancer research, 60, 679–684.

    PubMed  CAS  Google Scholar 

  42. Zhang, Y., Fujita, N., & Tsuruo, T. (1999). Caspase-mediated cleavage of p21Waf1/Cip1 converts cancer cells from growth arrest to undergoing apoptosis. Oncogene, 18, 1131–1138.

    Article  PubMed  CAS  Google Scholar 

  43. Vitiello, P. F., Staversky, R. J., Gehen, S. C., Johnston, C. J., Finkelstein, J. N., Wright, T. W., et al. (2006). p21Cip1 protection against hyperoxia requires Bcl-XL and is uncoupled from its ability to suppress growth. American journal of pathology, 168, 1838–1847.

    Article  PubMed  CAS  Google Scholar 

  44. Wang, Z., Goulet 3rd, R., Stanton, K. J., Sadaria, M., & Nakshatri, H. (2005). Differential effect of anti-apoptotic genes Bcl-xL and c-FLIP on sensitivity of MCF-7 breast cancer cells to paclitaxel and docetaxel. Anticancer research, 25, 2367–2379.

    PubMed  CAS  Google Scholar 

  45. Shoemaker, A. R., Oleksijew, A., Bauch, J., Belli, B. A., Borre, T., Bruncko, M., et al. (2006). A small-molecule inhibitor of Bcl-XL potentiates the activity of cytotoxic drugs in vitro and in vivo. Cancer research, 66, 8731–8739.

    Article  PubMed  CAS  Google Scholar 

  46. Trump, D. L., Muindi, J., Fakih, M., Yu, W. D., & Johnson, C. S. (2006). Vitamin D compounds: clinical development as cancer therapy and prevention agents. Anticancer research, 26, 2551–2556.

    PubMed  CAS  Google Scholar 

  47. Garcia-Montero, A. C., Vasseur, S., Giono, L. E., Canepa, E., Moreno, S., Dagorn, J. C., et al. (2001). Transforming growth factor beta-1 enhances Smad transcriptional activity through activation of p8 gene expression. Biochemical journal, 357, 249–253.

    Article  PubMed  CAS  Google Scholar 

  48. Massague, J. (1998). TGF-beta signal transduction. Annual Reviews of Biochemical, 67, 753–791.

    Article  CAS  Google Scholar 

  49. Chen, Y. G., Hata, A., Lo, R. S., Wotton, D., Shi, Y., Pavletich, N., et al. (1998). Determinants of specificity in TGF-beta signal transduction. Genes & development, 12, 2144–2152.

    Article  CAS  Google Scholar 

  50. Kretzschmar, M., & Massague, J. (1998). SMADs: mediators and regulators of TGF-beta signaling. Current opinion in genetics & development, 8, 103–111.

    Article  CAS  Google Scholar 

  51. Hoffmeister, A., Ropolo, A., Vasseur, S., Mallo, G. V., Bodeker, H., Ritz-Laser, B., et al. (2002). The HMG-I/Y-related protein p8 binds to p300 and Pax2 trans-activation domain-interacting protein to regulate the trans-activation activity of the Pax2A and Pax2B transcription factors on the glucagon gene promoter. Journal of biological chemistry, 277, 22314–22319.

    Article  PubMed  CAS  Google Scholar 

  52. Carracedo, A., Lorente, M., Egia, A., Blazquez, C., Garcia, S., Giroux, V., et al. (2006). The stress-regulated protein p8 mediates cannabinoid-induced apoptosis of tumor cells. Cancer Cell, 9, 301–312.

    Article  PubMed  CAS  Google Scholar 

  53. Gort, E. H., van Haaften, G., Verlaan, I., Groot, A. J., Plasterk, R. H., Shvarts, A., et al. (2008). The TWIST1 oncogene is a direct target of hypoxia-inducible factor-2alpha. Oncogene, 27, 1501–1510.

    Article  PubMed  CAS  Google Scholar 

  54. Gort, E. H., Groot, A. J., van der Wall, E., van Diest, P. J., & Vooijs, M. A. (2008). Hypoxic regulation of metastasis via hypoxia-inducible factors. Current molecular medicine, 8, 60–67.

    Article  PubMed  CAS  Google Scholar 

  55. Bristow, R. G., & Hill, R. P. (2008). Hypoxia and metabolism. Hypoxia, DNA repair and genetic instability. Nature reviews. Cancer, 8, 180–192.

    Article  PubMed  CAS  Google Scholar 

  56. Bindra, R. S., Schaffer, P. J., Meng, A., Woo, J., Maseide, K., Roth, M. E., et al. (2005). Alterations in DNA repair gene expression under hypoxia: elucidating the mechanisms of hypoxia-induced genetic instability. Annals of the New York Academy of Sciences, 1059, 184–195.

    Article  PubMed  CAS  Google Scholar 

  57. Rankin, E. B., & Giaccia, A. J. (2008). The role of hypoxia-inducible factors in tumorigenesis. Cell death and differentiation, 15, 678–685.

    Article  PubMed  CAS  Google Scholar 

  58. Mehlen, P., & Puisieux, A. (2006). Metastasis: a question of life or death. Nature reviews. Cancer, 6, 449–458.

    Article  PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Lalita A. Shevde.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Chowdhury, U.R., Samant, R.S., Fodstad, O. et al. Emerging role of nuclear protein 1 (NUPR1) in cancer biology. Cancer Metastasis Rev 28, 225–232 (2009). https://doi.org/10.1007/s10555-009-9183-x

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10555-009-9183-x

Keywords

Navigation