Quantitative comparison of immunohistochemical and PCR analysis of midkine expression in breast cancer types and serum midkine level

Quantitative comparison of immunohistochemical and PCR analysis of midkine expression in breast cancer types and serum midkine level

Background/aim: Midkine (MK), a heparin-binding growth factor, has an important role in cancer progression. The aim of this study was to determine MK expression in breast tissue and the preoperative and postoperative serum levels of patients with breast cancer. Materials and methods: Sixty-one patients with breast cancer participated in our study. The MK serum levels were measured pre- and postoperatively for these patients. We also analyzed breast tissues of the 61 patients immunohistochemically. We examined serum midkine levels in 49 healthy volunteers. Results: MK expression was observed in 44 (72.1%) of 61 breast cancer patients. In breast cancer patients the serum MK levels (3.68 ± 2.13 ng/mL (mean ± SD)) were significantly higher than in the control group (1.77 ± 0.38 ng/mL) before tumor removal (P = 0.000). After tumor removal, serum MK levels (2.47 ± 1.00 ng/mL) were significantly (P = 0.000) decreased according to preoperative levels. Increased serum levels of MK were related with tumor stages when clinical parameters were analyzed. Conclusion: We found that increased serum MK levels and protein expressions were associated with the carcinogenesis of breast cancer. MK levels decreased after tumor removal. According to our findings, MK might be a useful tumor marker for patients with breast cancer.

___

  • 1. Hunter T. Signaling—2000 and beyond. Cell 2000; 100: 113– 127.
  • 2. Choudhuri R, Zhang HT, Donnini S, Ziche M, Bicknell R. An angiogenic role for the neurokines midkine and pleiotrophin in tumorigenesis. Cancer Res 1997; 57: 1814–1819.
  • 3. Qi M, Ikematsu S, Maeda N, Ichihara-Tanaka K, Sakuma S, Noda M, Muramatsu T, Kadomatsu K. Haptotactic migration induced by midkine. Involvement of protein-tyrosine phosphatase zeta. Mitogen-activated protein kinase, and phosphatidylinositol 3-kinase. J Biol Chem 2001; 276: 15868– 15875.
  • 4. Muramatsu T. Midkine and pleiotrophin: two related proteins involved in development, survival, inflammation and tumorigenesis. J Biochem 2002; 132: 359–371.
  • 5. Kadomatsu K, Muramatsu T. Midkine and pleiotrophin in neural development and cancer. Cancer Lett 2004; 204: 127– 143.
  • 6. Matsubara S, Tomomura M, Kadomatsu K, Muramatsu T. Structure of a retinoic acid-responsive gene, MK, which is transiently activated during the differentiation of embryonal carcinoma cells and the mid-gestation period of mouse embryogenesis. J Biol Chem 1990; 265: 9441–9443.
  • 7. Kaname T, Kuwano A, Murano I, Uehara K, Muramatsu T, Kajii T. Midkine gene (MDK), a gene for prenatal differentiation and neuroregulation, maps to band 11p11.2 by fluorescence in situ hybridization. Genomics 1993; 17: 514–515.
  • 8. Murasugi A, Tohma-Aiba Y. Production of native recombinant human midkine in the yeast, Pichia pastoris. Protein Expr Purif 2003; 27: 244–252.
  • 9. Kadomatsu K, Tomomura M, Muramatsu T. cDNA cloning and sequencing of a new gene intensely expressed in early differentiation stages of embryonal carcinoma cells and in midgestation period of mouse embryogenesis. Biochem Biophys Res Commun 1988; 151: 1312–1318.
  • 10. Kadomatsu K, Hagihara M, Akhter S, Fan QW, Muramatsu H, Muramatsu T. Midkine induces the transformation of NIH3T3 cells. Br J Cancer 1997; 75: 354–359.
  • 11. Ratovitski EA, Kotzbauer PT, Milbrandt J, Lowenstein CJ, Burrow CR. Midkine induces tumor cell proliferation and bind to a high affinity signaling receptor associated with JAK tyrosine kinase. J Biol Chem 1998; 273: 3654–3660.
  • 12. Muramatsu T. Midkine, a heparin-binding cytokine with multiple roles in development, repair and diseases. Proc Jpn Acad Ser B Phys Biol Sci 2010; 86: 410–425.
  • 13. Sakamoto K, Kadomatsu K. Midkine in the pathology of cancer, neural disease, and inflammation. Pathol Int 2012; 62: 445–455.
  • 14. Ikematsu S, Yano A, Aridome K, Kikuchi M, Kumai H, Nagano H, Okamoto K, Oda M, Sakuma S, Aikou T et al. Serum midkine levels are increased in patients with various types of carcinomas. Br J Cancer 2000; 83: 701–706.
  • 15. Edwards BK, Noone AM, Mariotto AB, Simard EP, Boscoe FP, Henley SJ, Jemal A, Cho H, Anderson RN, Kohler BA et al. Annual Report to the Nation on the status of cancer, 1975-2010, featuring prevalence of comorbidity and impact on survival among persons with lung, colorectal, breast, or prostate cancer. Cancer 2014; 120: 1290–1314.
  • 16. Perou CM, Sørlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, Pollack JR, Ross DT, Johnsen H, Akslen LA et al. Molecular portraits of human breast tumours. Nature 2000; 406: 747–752.
  • 17. Sørlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H, Hastie T, Eisen MB, van de Rijn M, Jeffrey SS et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. P Natl Acad Sci USA 2001; 98: 10869–10874.
  • 18. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2000; 100: 57–70.
  • 19. Cho H, Mariotto AB, Mann BS, Klabunde CN, Feuer EJ. Assessing non-cancer-related health status of US cancer patients: other-cause survival and comorbidity prevalence. Am J Epidemiol 2013; 178: 339–349.
  • 20. Kaifi JT, Fiegel HC, Rafnsdottir SL, Aridome K, Schurr PG, Reichelt U, Wachowiak R, Kleinhans H, Yekebas EF, Mann O et al. Midkine as a prognostic marker for gastrointestinal stromal tumors. J Cancer Res Clin Oncol 2007; 133: 431–435.
  • 21. Qin LL, Huang HL, Ping JL, Xu W, Li J, Dai LC. Expression of midkine and endoglin in breast carcinomas with different immunohistochemical profiles. APMIS 2010; 119: 103–110.
  • 22. Moon HS, Park W, Sung SH, Choi EA, Chung HW, Woo BH. Immunohistochemical and quantitative competitive PCR analyses of midkine and pleiotrophin expression in cervical cancer. Gynecol Oncol 2003; 88: 289–297.
  • 23. Kuroi K, Toi M. Circulating angiogenesis regulators in cancer patients. Int J Biol Markers 2001; 16: 5–26.
  • 24. Ibusuki M, Fujimori H, Yamamoto Y, Ota K, Ueda M, Shinriki S, Taketomi M, Sakuma S, Shinohara M, Iwase H et al. Midkine in plasma as a novel breast cancer marker. Cancer Sci 2009; 100: 1735–1739.
  • 25. Tsutsui J, Kadomatsu K, Matsubara S, Nakagawara A, Hamanoue M, Takao S, Shimazu H, Ohi Y, Muramatsu T. A new family of heparin-binding growth/differentiation factors: increased midkine expression in Wilms’ tumour and other human carcinomas. Cancer Res 1993; 53: 1281–1285.
  • 26. Garver RI Jr, Radford DM, Donis-Keller H, Wick MR, Milner PG. Midkine and pleiotrophin expression in normal and malignant breast tissue. Cancer 1994; 74: 1584–1590.
  • 27. Aridome K, Tsutsui J, Takao S, Kadomatsu K, Ozawa M, Aikou T, Muramatsu T. Increased midkine gene expression in human gastrointestinal cancers. Jpn J Cancer Res 1995; 86: 655–661.
  • 28. Nakagawara A, Milbrandt J, Muramatsu T, Deuel TF, Zhao H, Cnaan A, Brodeur GM. Differential expression of pleiotrophin and midkine in advanced neuroblastomas. Cancer Res 1995; 55: 1792–1797.
  • 29. O’Brien T, Cranston D, Fuggle S, Bicknell R, Harris AL. The angiogenic factor midkine is expressed in bladder cancer, and overexpression correlates with a poor outcome in patients with invasive cancers. Cancer Res 1996; 56: 2515–2518.
  • 30. Mishima K, Asai A, Kadomatsu K, Ino Y, Nomura K, Narita Y, Muramatsu T, Kurino T. Increased expression of midkine during the progression of human astrocytomas. Neurosci Lett 1997; 233: 29–32.
  • 31. Konishi N, Nakamura M, Nakaoka S, Hiasa Y, Cho M, Uemura H, Hirao Y, Muramatsu T, Kadomatsu K. Immunohistochemical analysis of midkine expression in human prostate carcinoma. Oncology 1999; 57: 253–257.
  • 32. Sueyoshi T, Jono H, Shinriki S, Ota K, Ota T, Tasaki M, Atsuyama E, Yakushiji T, Ueda M, Obayashi K et al. Therapeutic approaches targeting midkine suppress tumor growth and lung metastasis in osteosarcoma. Cancer Lett 2012; 316; 23–30.
  • 33. Kato M, Maeta H, Kato S, Shinozawa T, Terada T. Immunohistochemical and in situ hybridization analyses of midkine expression in thyroid papillary carcinoma. Mod Pathol 2000; 13: 1060–1065.
  • 34. Hu XF, Yao J, Gao SG, Yang YT, Peng XQ, Feng XS. Midkine and syndecan1 levels correlate with the progression of malignant gastric cardiac adenocarcinoma. Mol Med Rep 2014; 10: 1409– 1415.
  • 35. Miyashiro I, Kaname T, Shin E, Wakasugi E, Monden T, Takatsuka Y, Kikkawa N, Muramatsu T, Monden M, Akiyama T. Midkine expression in human breast cancers: expression of truncated form. Breast Cancer Res Treat 1997; 43: 1–6.
  • 36. Shimada H, Nabeya Y, Okazumi S, Matsubara H, Kadomatsu K, Muramatsu T, Ikematsu S, Sakuma S, Ochiai T. Increased serum midkine concentration as a possible tumor marker in patients with superficial esophageal cancer. Oncol Rep 2003; 10: 411–414.
  • 37. Shimada H, Nabeya Y, Tagawa M, Okazumi S, Matsubara H, Kadomatsu K, Muramatsu T, Ikematsu S, Sakuma S, Ochiai T. Preoperative serum midkine concentration is a prognostic marker for esophageal squamous cell carcinoma. Cancer Sci 2003; 94: 628–632.
  • 38. Krzystek-Korpacka M, Diakowska D, Neubauer K, Gamian A. Circulating midkine in malignant and non-malignant colorectal diseases. Cytokine 2013; 64: 158–164.
  • 39. Tanabe K, Matsumoto M, Ikematsu S, Nagase S, Hatakeyama A, Takano T, Niikura H, Ito K, Kadomatsu K, Hayashi S et al. Midkine and its clinical significance in endometrial carcinoma. Cancer Sci 2008; 99: 1125–1130.