Skip to main content

Advertisement

Log in

Clinicopathological significance of p38β, p38γ, and p38δ and its biological roles in esophageal squamous cell carcinoma

  • Original Article
  • Published:
Tumor Biology

Abstract

P38β, p38γ, and p38δ have been sporadically and scarcely reported to be involved in the carcinogenesis of cancers, compared with p38α isoform. However, little has been known regarding their clinicopathological significance and biological roles in esophageal squamous cell carcinoma (ESCC). Expression status of p38β, p38γ, and p38δ was assayed using immunohistochemistry with ESCC tissue microarray; ensuing clinicopathological significance was statistically analyzed. To define its biological roles on proliferation, migration and invasion of ESCC cell line Eca109 in vitro, MTT, wound healing, and Transwell assays were employed, respectively. As confirmation, athymic nude mice were taken to verify the effect over proliferation in vivo. It was found that both p38β and p38δ expression, other than p38γ, were significantly higher in ESCC tissues compared with paired normal controls. In terms of prognosis, only p38β expression was observed to be significantly associated with overall prognosis. Clinicopathologically, there was significant association between p38γ expression and clinical stage, lymph nodes metastases, and tumor volume. No significant association was found for p38β and p38δ between its expression and other clinicopathological parameters other than significant difference of expression between ESCC versus normal control. In Eca109, it was observed that p38β, p38γ, and p38δ can promote the cell growth and motility. As verification, over-expression of p38δ can promote, whereas knockdown of p38γ can prevent, the tumorigenesis in nude mice model xenografted with Eca109 cells whose basal level of p38δ was stably over-expressed and p38γ was stably knocked down. Together, our results demonstrate that p38β, p38γ, and p38δ played oncogenic roles in ESCC.

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
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

References

  1. Cuenda A, Rousseau S. p38 MAP-kinases pathway regulation, function and role in human diseases. Biochim Biophys Acta. 2007;1773(8):1358–75.

    Article  CAS  PubMed  Google Scholar 

  2. Risco A, Cuenda A. New insights into the p38gamma and p38delta MAPK Pathways. J Signal Transduct. 2012;2012:520289.

    Article  CAS  PubMed  Google Scholar 

  3. Zheng ST, Liu T, Liu Q, Lu M, Gao XP, Sheyhidin I, et al. Investigation on role of p38alpha mitogen-activated protein kinases in human esophageal squamous cell carcinoma cell line Eca109. Zhonghua yu fang yi xue za zhi Chin J Prev Med. 2013;47(8):757–61.

    CAS  Google Scholar 

  4. O’Callaghan C, Fanning LJ, Barry OP. p38delta MAPK phenotype: an indicator of chemotherapeutic response in oesophageal squamous cell carcinoma. Anti-Cancer Drugs. 2015;26(1):46–55.

    Article  CAS  PubMed  Google Scholar 

  5. O’Callaghan C, Fanning LJ, Houston A, Barry OP. Loss of p38delta mitogen-activated protein kinase expression promotes oesophageal squamous cell carcinoma proliferation, migration and anchorage-independent growth. Int J Oncol. 2013;43(2):405–15.

    PubMed  PubMed Central  Google Scholar 

  6. Del Reino P, Alsina-Beauchamp D, Escos A, Cerezo-Guisado MI, Risco A, Aparicio N, et al. Pro-oncogenic role of alternative p38 mitogen-activated protein kinases p38gamma and p38delta, linking inflammation and cancer in colitis-associated colon cancer. Cancer Res. 2014;74(21):6150–60.

    Article  CAS  PubMed  Google Scholar 

  7. Meng F, Zhang H, Liu G, Kreike B, Chen W, Sethi S, et al. p38gamma mitogen-activated protein kinase contributes to oncogenic properties maintenance and resistance to poly (ADP-ribose)-polymerase-1 inhibition in breast cancer. Neoplasia. 2011;13(5):472–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Rosenthal DT, Iyer H, Escudero S, Bao L, Wu Z, Ventura AC, et al. p38gamma promotes breast cancer cell motility and metastasis through regulation of RhoC GTPase, cytoskeletal architecture, and a novel leading edge behavior. Cancer Res. 2011;71(20):6338–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Yin N, Qi X, Tsai S, Lu Y, Basir Z, Oshima K, Thomas JP, Myers CR, Stoner G, Chen G. p38gamma MAPK is required for inflammation-associated colon tumorigenesis. Oncogene 2015.

  10. Singh AK, Pandey R, Gill K, Singh R, Saraya A, Chauhan SS, et al. p38beta MAP kinase as a therapeutic target for pancreatic cancer. Chem Biol Drug Des. 2012;80(2):266–73.

    Article  CAS  PubMed  Google Scholar 

  11. He Z, He J, Liu Z, Xu J, Yi SF, Liu H, et al. MAPK11 in breast cancer cells enhances osteoclastogenesis and bone resorption. Biochimie. 2014;106:24–32.

    Article  CAS  PubMed  Google Scholar 

  12. Kukkonen-Macchi A, Sicora O, Kaczynska K, Oetken-Lindholm C, Pouwels J, Laine L, et al. Loss of p38gamma MAPK induces pleiotropic mitotic defects and massive cell death. J Cell Sci. 2011;124(Pt 2):216–27.

    Article  CAS  PubMed  Google Scholar 

  13. Hewitt SM, Baskin DG, Frevert CW, Stahl WL, Rosa-Molinar E. Controls for immunohistochemistry: the Histochemical Society’s standards of practice for validation of immunohistochemical assays. J Histochem Cytochem Off J Histochem Soc. 2014;62(10):693–7.

    Article  CAS  Google Scholar 

  14. Burry RW. Controls for immunocytochemistry: an update. J Histochem Cytochem Off J Histochem Soc. 2011;59(1):6–12.

    Article  CAS  Google Scholar 

  15. Liu H, He J, Yang J. Tumor cell p38 MAPK: a trigger of cancer bone osteolysis. Cancer Cell Microenviron. 2015;2(1).

  16. Gonzalez-Villasana V, Fuentes-Mattei E, Ivan C, Dalton HJ, Rodriguez-Aguayo C, Fernandez-de Thomas RJ, et al. Rac1/Pak1/p38/MMP-2 axis regulates angiogenesis in ovarian cancer. Clin Cancer Res Off J Am Assoc Cancer Res. 2015;21(9):2127–37.

    Article  CAS  Google Scholar 

  17. Rennefahrt U, Janakiraman M, Ollinger R, Troppmair J. Stress kinase signaling in cancer: fact or fiction? Cancer Lett. 2005;217(1):1–9.

    Article  CAS  PubMed  Google Scholar 

  18. Loesch M, Chen G. The p38 MAPK stress pathway as a tumor suppressor or more? Front Biosci J Virtual Libr. 2008;13:3581–93.

    Article  CAS  Google Scholar 

  19. Chen L, Mayer JA, Krisko TI, Speers CW, Wang T, Hilsenbeck SG, et al. Inhibition of the p38 kinase suppresses the proliferation of human ER-negative breast cancer cells. Cancer Res. 2009;69(23):8853–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Radziwon-Balicka A, Santos-Martinez MJ, Corbalan JJ, O’Sullivan S, Treumann A, Gilmer JF, et al. Mechanisms of platelet-stimulated colon cancer invasion: role of clusterin and thrombospondin 1 in regulation of the P38MAPK-MMP-9 pathway. Carcinogenesis. 2014;35(2):324–32.

    Article  CAS  PubMed  Google Scholar 

  21. Grossi V, Peserico A, Tezil T, Simone C. p38alpha MAPK pathway: a key factor in colorectal cancer therapy and chemoresistance. World J Gastroenterol WJG. 2014;20(29):9744–58.

    Article  CAS  PubMed  Google Scholar 

  22. Tai TW, Su FC, Chen CY, Jou IM, Lin CF. Activation of p38 MAPK-regulated Bcl-xL signaling increases survival against zoledronic acid-induced apoptosis in osteoclast precursors. Bone. 2014;67:166–74.

    Article  CAS  PubMed  Google Scholar 

  23. Germani A, Matrone A, Grossi V, Peserico A, Sanese P, Liuzzi M, et al. Targeted therapy against chemoresistant colorectal cancers: inhibition of p38alpha modulates the effect of cisplatin in vitro and in vivo through the tumor suppressor FoxO3A. Cancer Lett. 2014;344(1):110–8.

    Article  CAS  PubMed  Google Scholar 

  24. Gupta J, del Barco BI, Igea A, Sakellariou S, Pateras IS, Gorgoulis VG, et al. Dual function of p38alpha MAPK in colon cancer: suppression of colitis-associated tumor initiation but requirement for cancer cell survival. Cancer Cell. 2014;25(4):484–500.

    Article  CAS  PubMed  Google Scholar 

  25. Yu L, Yuan X, Wang D, Barakat B, Williams ED, Hannigan GE. Selective regulation of p38beta protein and signaling by integrin-linked kinase mediates bladder cancer cell migration. Oncogene. 2014;33(6):690–701.

    Article  CAS  PubMed  Google Scholar 

  26. O’Callaghan C, Fanning LJ, Barry OP. p38delta MAPK: emerging roles of a neglected isoform. Int J Cell Biol. 2014;2014:272689.

    PubMed  PubMed Central  Google Scholar 

  27. Meng F, Wu G. Is p38gamma MAPK a metastasis-promoting gene or an oncogenic property-maintaining gene? Cell Cycle. 2013;12(14):2329–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Yang K, Liu Y, Liu Z, Liu J, Liu X, Chen X, et al. p38gamma overexpression in gliomas and its role in proliferation and apoptosis. Sci Rep. 2013;3:2089.

    PubMed  PubMed Central  Google Scholar 

  29. Junttila MR, Ala-Aho R, Jokilehto T, Peltonen J, Kallajoki M, Grenman R, et al. p38alpha and p38delta mitogen-activated protein kinase isoforms regulate invasion and growth of head and neck squamous carcinoma cells. Oncogene. 2007;26(36):5267–79.

    Article  CAS  PubMed  Google Scholar 

  30. Tan FL, Ooi A, Huang D, Wong JC, Qian CN, Chao C, et al. p38delta/MAPK13 as a diagnostic marker for cholangiocarcinoma and its involvement in cell motility and invasion. Int J Cancer J Int Cancer. 2010;126(10):2353–61.

    CAS  Google Scholar 

  31. Choi YK, Woo SM, Cho SG, Moon HE, Yun YJ, Kim JW, et al. Brain-metastatic triple-negative breast cancer cells regain growth ability by altering gene expression patterns. Cancer Genomics Proteomics. 2013;10(6):265–75.

    PubMed  Google Scholar 

  32. Kaplan RM, Chambers DA, Glasgow RE. Big data and large sample size: a cautionary note on the potential for bias. Clin Transl Sci. 2014;7(4):342–6.

    Article  PubMed  Google Scholar 

  33. Meier DT, Entrup L, Templin AT, Hogan MF, Samarasekera T, Zraika S, et al. Determination of optimal sample size for quantification of beta-cell area, amyloid area and beta-cell apoptosis in isolated islets. J Histochem Cytochem Off J Histochem Soc. 2015;63(8):663–73.

    Article  CAS  Google Scholar 

  34. Baker M. Reproducibility crisis: blame it on the antibodies. Nature. 2015;521(7552):274–6.

    Article  CAS  PubMed  Google Scholar 

  35. Helsby MA, Fenn JR, Chalmers AD. Reporting research antibody use: how to increase experimental reproducibility. F1000Res. 2013;2:153.

    PubMed  PubMed Central  Google Scholar 

  36. Helsby MA, Leung MY, Chalmers AD. The F1000 research antibody validation article collection. F1000Res. 2014;3:241.

    PubMed  PubMed Central  Google Scholar 

  37. Linderoth J, Ehinger M, Akerman M, Cavallin-Stahl E, Enblad G, Erlanson M, et al. Tissue microarray is inappropriate for analysis of BCL6 expression in diffuse large B-cell lymphoma. Eur J Haematol. 2007;79(2):146–9.

    Article  PubMed  Google Scholar 

Download references

Acknowledgments

The study was supported by National Science Foundation of China (no. 81360357, 81160303, 81260359, 81201891, U1303321), from Major Science and Technology Projects of the Xinjiang Uygur Autonomous Region (no. 201430123-1) and Opening Project of Xinjiang Medical Animal Model Research Key Laboratory (XJDX1103-2012-05). We are especially appreciative of Professor Ana Cuenda in the Department of Immunology and Oncology, Centro National de Biotecnología/CSIC, Madrid, Spain, for kindly proof reading and giving constructive comments in the manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Xiaomei Lu.

Ethics declarations

Conflicts of interest

None

Electronic supplementary material

Below is the link to the electronic supplementary material.

ESM 1

(DOCX 1925 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zheng, S., Yang, C., Liu, T. et al. Clinicopathological significance of p38β, p38γ, and p38δ and its biological roles in esophageal squamous cell carcinoma. Tumor Biol. 37, 7255–7266 (2016). https://doi.org/10.1007/s13277-015-4610-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13277-015-4610-9

Keywords

Navigation