Vol 10, No 1 (2019)
Review paper
Published online: 2019-06-19

open access

Page views 1281
Article views/downloads 2220
Get Citation

Connect on Social Media

Connect on Social Media

The importance of neprilysin (CD10) in hematooncology

Kamil Sokół1, Monika Prochorec-Sobieszek1, Anna Szumera-Ciećkiewicz1
DOI: 10.5603/Hem.2019.0013
Hematologia 2019;10(1):19-28.

Abstract

CD10 is an extracellular protein with enzymatic activity. Its substrates are various substances with the properties of signal particles. CD10 also has in itself the propriety of transmitting signals via the membrane, and affecting various intracellular signal transduction pathways, through interaction with numerous protein complexes. CD10 expression is observed in many normal tissues as well as in selected tumors. It is therefore particularly important in histopathological differential diagnosis. The following article presents the structure, location and functions of the CD10. The effect of CD10 expression on the clinical course and prognosis in hematological and non-haematological malignancies is also described. 

Article available in PDF format

View PDF (Polish) Download PDF file

References

  1. Erdös EG, Skidgel RA. Neutral endopeptidase 24.11 (enkephalinase) and related regulators of peptide hormones. FASEB J. 1989; 3(2): 145–151.
  2. Loke SL, Leung CY, Chiu KY, et al. Localisation of CD10 to biliary canaliculi by immunoelectron microscopical examination. J Clin Pathol. 1990; 43(8): 654–656.
  3. Sato Y, Itoh F, Hinoda Y, et al. Expression of CD10/neutral endopeptidase in normal and malignant tissues of the human stomach and colon. J Gastroenterol. 1996; 31(1): 12–17.
  4. Song J, Aumüller G, Xiao F, et al. Cell specific expression of CD10/neutral endopeptidase 24.11 gene in human prostatic tissue and cells. Prostate. 2004; 58(4): 394–405.
  5. Clark EA, Lane PJ. Regulation of human B-cell activation and adhesion. Annu Rev Immunol. 1991; 9: 97–127.
  6. Janossy G, Bofill M, Schuurman HJ. Human B-lymphoid differentiation: normal versus malignant. Neth J Med. 1991; 39(3-4): 232–243.
  7. Barker PE, Shipp MA, D'Adamio L, et al. The common acute lymphoblastic leukemia antigen gene maps to chromosomal region 3 (q21-q27). J Immunol. 1989; 142(1): 283–287.
  8. D'Adamio L, Shipp MA, Masteller EL, et al. Organization of the gene encoding common acute lymphoblastic leukemia antigen (neutral endopeptidase 24.11): multiple miniexons and separate 5' untranslated regions. Proc Natl Acad Sci U S A. 1989; 86(18): 7103–7107.
  9. Malfroy B, Kuang WJ, Seeburg PH, et al. Molecular cloning and amino acid sequence of human enkephalinase (neutral endopeptidase). FEBS Lett. 1988; 229(1): 206–210.
  10. Iwase A, Shen R, Navarro D, et al. Direct binding of neutral endopeptidase 24.11 to ezrin/radixin/moesin (ERM) proteins competes with the interaction of CD44 with ERM proteins. J Biol Chem. 2004; 279(12): 11898–11905.
  11. Sumitomo M, Shen R, Nanus DM. Involvement of neutral endopeptidase in neoplastic progression. Biochim Biophys Acta. 2005; 1751(1): 52–59.
  12. Zheng R, Horiguchi A, Iida K, et al. Neutral endopeptidase is a myristoylated protein. Mol Cell Biochem. 2010; 335(1-2): 173–180.
  13. Dale GE, D'Arcy B, Yuvaniyama C, et al. Purification and crystallization of the extracellular domain of human neutral endopeptidase (neprilysin) expressed in Pichia pastoris. Acta Crystallogr D Biol Crystallogr. 2000; 56(Pt 7): 894–897.
  14. Oefner C, D'Arcy A, Hennig M, et al. Structure of human neutral endopeptidase (Neprilysin) complexed with phosphoramidon. J Mol Biol. 2000; 296(2): 341–349.
  15. Marie-Claire C, Tiraboschi G, Ruffet E, et al. Exploration of the S(')(1) subsite of neprilysin: a joined molecular modeling and site-directed mutagenesis study. Proteins. 2000; 39(4): 365–371.
  16. David C, Bischoff L, Meudal H, et al. Investigation of subsite preferences in aminopeptidase A (EC 3.4.11.7) led to the design of the first highly potent and selective inhibitors of this enzyme. J Med Chem. 1999; 42(25): 5197–5211.
  17. Iijima-Ando K, Hearn SA, Granger L, et al. Overexpression of neprilysin reduces alzheimer amyloid-beta42 (Abeta42)-induced neuron loss and intraneuronal Abeta42 deposits but causes a reduction in cAMP-responsive element-binding protein-mediated transcription, age-dependent axon pathology, and premature death in Drosophila. J Biol Chem. 2008; 283(27): 19066–19076.
  18. Helisalmi S, Hiltunen M, Vepsäläinen S, et al. Polymorphisms in neprilysin gene affect the risk of Alzheimer's disease in Finnish patients. J Neurol Neurosurg Psychiatry. 2004; 75(12): 1746–1748.
  19. Shipp MA, Stefano GB, Switzer SN, et al. CD10 (CALLA)/neutral endopeptidase 24.11 modulates inflammatory peptide-induced changes in neutrophil morphology, migration, and adhesion proteins and is itself regulated by neutrophil activation. Blood. 1991; 78(7): 1834–1841.
  20. Lu B, Gerard NP, Kolakowski LF, et al. Neutral endopeptidase modulation of septic shock. J Exp Med. 1995; 181(6): 2271–2275.
  21. Sapino A, Macrì L, Tonda L, et al. Oxytocin enhances myoepithelial cell differentiation and proliferation in the mouse mammary gland. Endocrinology. 1993; 133(2): 838–842.
  22. Zheng R, Iwase A, Shen R, et al. Neuropeptide-stimulated cell migration in prostate cancer cells is mediated by RhoA kinase signaling and inhibited by neutral endopeptidase. Oncogene. 2006; 25(44): 5942–5952.
  23. Goodman OB, Febbraio M, Simantov R, et al. Neprilysin inhibits angiogenesis via proteolysis of fibroblast growth factor-2. J Biol Chem. 2006; 281(44): 33597–33605.
  24. Sumitomo M, Iwase A, Zheng R, et al. Synergy in tumor suppression by direct interaction of neutral endopeptidase with PTEN. Cancer Cell. 2004; 5(1): 67–78.
  25. Horejsí V, Drbal K, Cebecauer M, et al. GPI-microdomains: a role in signalling via immunoreceptors. Immunol Today. 1999; 20(8): 356–361.
  26. Béné MC. Immunophenotyping of acute leukaemias. Immunol Lett. 2005; 98(1): 9–21.
  27. Ganju RK, Shpektor RG, Brenner DG, et al. CD10/neutral endopeptidase 24.11 is phosphorylated by casein kinase II and coassociates with other phosphoproteins including the lyn src-related kinase. Blood. 1996; 88(11): 4159–4165.
  28. Osman I, Yee H, Taneja SS, et al. Neutral endopeptidase protein expression and prognosis in localized prostate cancer. Clin Cancer Res. 2004; 10(12 Pt 1): 4096–4100.
  29. Pesando JM, Ritz J, Lazarus H, et al. Leukemia-associated antigens in ALL. Blood. 1979; 54(6): 1240–1248.
  30. Swerdlow SH, Campo E, Pileri SA, et al. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood. 2016; 127(20): 2375–2390.
  31. Wood BL, Arroz M, Barnett D, et al. 2006 Bethesda International Consensus recommendations on the immunophenotypic analysis of hematolymphoid neoplasia by flow cytometry: optimal reagents and reporting for the flow cytometric diagnosis of hematopoietic neoplasia. Cytometry B Clin Cytom. 2007; 72 Suppl 1: S14–S22.
  32. Borowitz MJ, Devidas M, Hunger SP, et al. Children's Oncology Group. Clinical significance of minimal residual disease in childhood acute lymphoblastic leukemia and its relationship to other prognostic factors: a Children's Oncology Group study. Blood. 2008; 111(12): 5477–5485.
  33. Li HF, Meng WT, Jia YQ, et al. Development-associated immunophenotypes reveal the heterogeneous and individualized early responses of adult B-acute lymphoblastic leukemia. Medicine (Baltimore). 2016; 95(34): e4128.
  34. Martin-Kleiner I, Svoboda-Beusan I, Gabrilovac J. PMA and doxorubicin decrease viability, MTT activity and expression of CD10 marker on NALM-1 leukemic cells. Immunopharmacol Immunotoxicol. 2006; 28(3): 411–420.
  35. Oliveira E, Bacelar TS, Ciudad J, et al. Altered neutrophil immunophenotypes in childhood B‑cell precursor acute lymphoblastic leukemia. Oncotarget. 2016; 7(17): 24664–24676.
  36. Cho MC, Chung Y, Jang S, et al. Prognostic impact of germinal center B-cell-like and non-germinal center B-cell-like subtypes of bone marrow involvement in patients with diffuse large B-cell lymphoma treated with R-CHOP. Medicine (Baltimore). 2018; 97(45): e13046.
  37. Lu TX, Miao Yi, Wu JZ, et al. The distinct clinical features and prognosis of the CD10⁺MUM1⁺ and CD10⁻Bcl6⁻MUM1⁻ diffuse large B-cell lymphoma. Sci Rep. 2016; 6: 20465.
  38. Ohshima K, Kawasaki C, Muta H, et al. CD10 and Bcl10 expression in diffuse large B-cell lymphoma: CD10 is a marker of improved prognosis. Histopathology. 2001; 39(2): 156–162.
  39. Peng F, Guo L, Yao WK, et al. Identification of prognostic factors in patients with diffuse large B-cell lymphoma. Indian J Pathol Microbiol. 2017; 60(1): 87–91.
  40. Meyer PN, Fu K, Greiner TC, et al. Immunohistochemical methods for predicting cell of origin and survival in patients with diffuse large B-cell lymphoma treated with rituximab. J Clin Oncol. 2011; 29(2): 200–207.
  41. Barcus ME, Karageorge LS, Veloso YL, et al. CD10 expression in follicular lymphoma versus reactive follicular hyperplasia: evaluation in paraffin-embedded tissue. Appl Immunohistochem Mol Morphol. 2000; 8(4): 263–266.
  42. Ohnishi N, Takata K, Miyata-Takata T, et al. CD10 down expression in follicular lymphoma correlates with gastrointestinal lesion involving the stomach and large intestine. Cancer Sci. 2016; 107(11): 1687–1695.
  43. McGowan P, Nelles N, Wimmer J, et al. Differentiating between Burkitt lymphoma and CD10+ diffuse large B-cell lymphoma: the role of commonly used flow cytometry cell markers and the application of a multiparameter scoring system. Am J Clin Pathol. 2012; 137(4): 665–670.
  44. Pilkington GR, Pallesen G. Phenotypic characterization of non-haemopoietic small cell tumours of childhood with monoclonal antibodies to leucocytes, epithelial cells and cytoskeletal proteins. Histopathology. 1989; 14(4): 347–357.
  45. Chu P, Arber DA. Paraffin-section detection of CD10 in 505 nonhematopoietic neoplasms. Frequent expression in renal cell carcinoma and endometrial stromal sarcoma. Am J Clin Pathol. 2000; 113(3): 374–382.
  46. Cohen AJ, Bunn PA, Franklin W, et al. Neutral endopeptidase: variable expression in human lung, inactivation in lung cancer, and modulation of peptide-induced calcium flux. Cancer Res. 1996; 56(4): 831–839.
  47. Yada K, Kashima K, Daa T, et al. Expression of CD10 in basal cell carcinoma. Am J Dermatopathol. 2004; 26(6): 463–471.
  48. Notohara K, Hamazaki S, Tsukayama C, et al. Solid-pseudopapillary tumor of the pancreas: immunohistochemical localization of neuroendocrine markers and CD10. Am J Surg Pathol. 2000; 24(10): 1361–1371.
  49. Borscheri N, Roessner A, Röcken C. Canalicular immunostaining of neprilysin (CD10) as a diagnostic marker for hepatocellular carcinomas. Am J Surg Pathol. 2001; 25(10): 1297–1303.
  50. Makretsov NA, Hayes M, Carter BA, et al. Stromal CD10 expression in invasive breast carcinoma correlates with poor prognosis, estrogen receptor negativity, and high grade. Mod Pathol. 2007; 20(1): 84–89.
  51. Huang WB, Zhou XJ, Chen JY, et al. CD10-positive stromal cells in gastric carcinoma: correlation with invasion and metastasis. Jpn J Clin Oncol. 2005; 35(5): 245–250.
  52. Terauchi M, Kajiyama H, Shibata K, et al. Anti-progressive effect of neutral endopeptidase 24.11 (NEP/CD10) on cervical carcinoma in vitro and in vivo. Oncology. 2005; 69(1): 52–62.
  53. Bircan S, Candir O, Kapucuoglu N, et al. CD10 expression in urothelial bladder carcinomas: a pilot study. Urol Int. 2006; 77(2): 107–113.
  54. Tokuhara T, Adachi M, Hashida H, et al. Neutral endopeptidase/CD10 and aminopeptidase N/CD13 gene expression as a prognostic factor in non-small cell lung cancer. Jpn J Thorac Cardiovasc Surg. 2001; 49(8): 489–496.
  55. Deschamps L, Handra-Luca A, O'Toole D, et al. CD10 expression in pancreatic endocrine tumors: correlation with prognostic factors and survival. Hum Pathol. 2006; 37(7): 802–808.
  56. Fujita S, Taniguchi H, Yao T, et al. Multi-institutional study of risk factors of liver metastasis from colorectal cancer: correlation with CD10 expression. Int J Colorectal Dis. 2010; 25(6): 681–686.
  57. Piattelli A, Fioroni M, Iezzi G, et al. CD10 expression in stromal cells of oral cavity squamous cell carcinoma: a clinic and pathologic correlation. Oral Dis. 2006; 12(3): 301–304.
  58. Kuniyasu H, Luo Y, Fujii K, et al. CD10 enhances metastasis of colorectal cancer by abrogating the anti-tumoural effect of methionine-enkephalin in the liver. Gut. 2010; 59(3): 348–356.
  59. Fukusumi T, Ishii H, Konno M, et al. CD10 as a novel marker of therapeutic resistance and cancer stem cells in head and neck squamous cell carcinoma. Br J Cancer. 2014; 111(3): 506–514.
  60. Uehara K, Ikehara F, Tanabe Y, et al. CD10 expression in the neuroendocrine carcinoma component of endometrial mixed carcinoma: association with long survival. Diagn Pathol. 2016; 11: 16.
  61. Kajiyama H, Shibata K, Terauchi M, et al. Neutral endopeptidase 24.11/CD10 suppresses progressive potential in ovarian carcinoma in vitro and in vivo. Clin Cancer Res. 2005; 11(5): 1798–1808.
  62. Abdou AG. CD10 expression in tumour and stromal cells of bladder carcinoma: an association with bilharziasis. APMIS. 2007; 115(11): 1206–1218.
  63. Hu M, Yao J, Carroll DK, et al. Regulation of in situ to invasive breast carcinoma transition. Cancer Cell. 2008; 13(5): 394–406.
  64. Erhuma M, Köbel M, Mustafa T, et al. Expression of neutral endopeptidase (NEP/CD10) on pancreatic tumor cell lines, pancreatitis and pancreatic tumor tissues. Int J Cancer. 2007; 120(11): 2393–2400.
  65. Smollich M, Götte M, Yip GW, et al. On the role of endothelin-converting enzyme-1 (ECE-1) and neprilysin in human breast cancer. Breast Cancer Res Treat. 2007; 106(3): 361–369.
  66. Fleischmann A, Schlomm T, Huland H, et al. Distinct subcellular expression patterns of neutral endopeptidase (CD10) in prostate cancer predict diverging clinical courses in surgically treated patients. Clin Cancer Res. 2008; 14(23): 7838–7842.
  67. Freedland SJ, Seligson DB, Liu AY, et al. Loss of CD10 (neutral endopeptidase) is a frequent and early event in human prostate cancer. Prostate. 2003; 55(1): 71–80.
  68. Fleischmann A, Rocha C, Saxer-Sekulic N, et al. High CD10 expression in lymph node metastases from surgically treated prostate cancer independently predicts early death. Virchows Arch. 2011; 458(6): 741–748.
  69. Cui R, Yuan F, Wang Y, et al. Clinicopathological characteristics and treatment strategies for patients with low-grade endometrial stromal sarcoma. Medicine (Baltimore). 2017; 96(15): e6584.
  70. Oliva E. Cellular mesenchymal tumors of the uterus: a review emphasizing recent observations. Int J Gynecol Pathol. 2014; 33(4): 374–384.
  71. Tomoda C, Kushima R, Takeuti E, et al. CD10 expression is useful in the diagnosis of follicular carcinoma and follicular variant of papillary thyroid carcinoma. Thyroid. 2003; 13(3): 291–295.
  72. Mokhtari M, Ameri F. Diagnostic value of CD-10 marker in differentiating of papillary thyroid carcinoma from benign thyroid lesions. Adv Biomed Res. 2014; 3: 206.



Hematology in Clinical Practice