Erythropoietin Receptor Antagonist Suppressed Ectopic Hemoglobin Synthesis in Xenografts of HeLa Cells to Promote Their Destruction

PLoS One. 2015 Apr 15;10(4):e0122458. doi: 10.1371/journal.pone.0122458. eCollection 2015.

Abstract

The aim of this study is to explore a cause-oriented therapy for patients with uterine cervical cancer that expresses erythropoietin (Epo) and its receptor (EpoR). Epo, by binding to EpoR, stimulates the proliferation and differentiation of erythroid progenitor cells into hemoglobin-containing red blood cells. In this study, we report that the HeLa cells in the xenografts expressed ε, γ, and α globins as well as myoglobin (Mb) to produce tetrameric α2ε2 and α2γ2 and monomeric Mb, most of which were significantly suppressed with an EpoR antagonist EMP9. Western blotting revealed that the EMP9 treatment inhibited the AKT-pAKT, MAPKs-pMAPKs, and STAT5-pSTAT5 signaling pathways. Moreover, the treatment induced apoptosis and suppression of the growth and inhibited the survival through disruption of the harmonized hemoprotein syntheses in the tumor cells concomitant with destruction of vascular nets in the xenografts. Furthermore, macrophages and natural killer (NK) cells with intense HIF-1α expression recruited significantly more in the degenerating foci of the xenografts. These findings were associated with the enhanced expressions of nNOS in the tumor cells and iNOS in macrophages and NK cells in the tumor sites. The treated tumor cells exhibited a substantial number of perforations on the cell surface, which indicates that the tumors were damaged by both the nNOS-induced nitric oxide (NO) production in the tumor cells as well as the iNOS-induced NO production in the innate immune cells. Taken together, these data suggest that HeLa cells constitutively acquire ε, γ and Mb synthetic capacity for their survival. Therefore, EMP9 treatment might be a cause-oriented and effective therapy for patients with squamous cell carcinoma of the uterine cervix.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Apoptosis / drug effects
  • Blotting, Western
  • Cell Proliferation / drug effects
  • Erythropoietin / chemistry
  • Erythropoietin / pharmacology
  • Gene Expression / drug effects
  • HeLa Cells
  • Hemoglobins / biosynthesis*
  • Hemoglobins / genetics
  • Heterografts / drug effects*
  • Heterografts / metabolism
  • Humans
  • Male
  • Mice, Inbred BALB C
  • Mice, Nude
  • Mitogen-Activated Protein Kinases / metabolism
  • Neoplasms, Experimental / genetics
  • Neoplasms, Experimental / metabolism*
  • Neoplasms, Experimental / pathology
  • Peptides / chemical synthesis
  • Peptides / pharmacology*
  • Proto-Oncogene Proteins c-akt / metabolism
  • Receptors, Erythropoietin / antagonists & inhibitors*
  • Receptors, Erythropoietin / genetics
  • Receptors, Erythropoietin / metabolism
  • Reverse Transcriptase Polymerase Chain Reaction
  • STAT5 Transcription Factor / metabolism
  • Signal Transduction / drug effects
  • Transplantation, Heterologous

Substances

  • Hemoglobins
  • Peptides
  • Receptors, Erythropoietin
  • STAT5 Transcription Factor
  • Erythropoietin
  • Proto-Oncogene Proteins c-akt
  • Mitogen-Activated Protein Kinases

Grants and funding

This study was supported by the following grants: Promotion and Mutual Aid-Co-operation for Private Schools of Japan (YY); Grants-in-Aid for Young Scientist (B) (MF: No. 24700998) from the Japan Society for the Promotion of Science; and the University Research Grant (MF: No. SR08) from Kinki University. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.