Your browser doesn't support javascript.
loading
Aberrant epigenetic silencing of neuronatin is a frequent event in human osteosarcoma.
Saeed, Haleema; Sinha, Sayantani; Mella, Christine; Kuerbitz, Jeffrey S; Cales, Monica L; Steele, Mark A; Stanke, Jennifer; Damron, Derek; Safadi, Fayez; Kuerbitz, Steven J.
Afiliación
  • Saeed H; Division of Pediatric Hematology/Oncology, Akron Childrens Hospital, Akron, OH, USA.
  • Sinha S; Current affiliation: Shaukat Khanum Cancer Hospital and Research Centre, Lahore, Pakistan.
  • Mella C; Department of Biological Sciences, Kent State University, Kent, OH, USA.
  • Kuerbitz JS; Current affiliation: Department of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
  • Cales ML; Division of Pediatric Hematology/Oncology, Akron Childrens Hospital, Akron, OH, USA.
  • Steele MA; Division of Pediatric Hematology/Oncology, Akron Childrens Hospital, Akron, OH, USA.
  • Stanke J; Current affiliation: Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
  • Damron D; College of Osteopathic Medicine, University of Pikeville, Pikeville, KY, USA.
  • Safadi F; Current affiliation: Penn State Health St. Joseph, Reading, PA, USA.
  • Kuerbitz SJ; Division of Pediatric Hematology/Oncology, Akron Childrens Hospital, Akron, OH, USA.
Oncotarget ; 11(20): 1876-1893, 2020 May 19.
Article en En | MEDLINE | ID: mdl-32499872
ABSTRACT
The paternally imprinted neuronatin (NNAT) gene has been identified as a target of aberrant epigenetic silencing in diverse cancers, but no association with pediatric bone cancers has been reported to date. In screening childhood cancers, we identified aberrant CpG island hypermethylation in a majority of osteosarcoma (OS) samples and in 5 of 6 human OS cell lines studied but not in normal bone-derived tissue samples. CpG island hypermethylation was associated with transcriptional silencing in human OS cells, and silencing was reversible upon treatment with 5-aza-2'-deoxycytidine. Expression of NNAT was detectable in osteoblasts and chondrocytes of human bone, supporting a potential role in bone homeostasis. Enforced expression of NNAT in human OS cells lacking endogenous expression resulted in significant reduction in colony formation and in vitro migration compared to nonexpressor control cells. We next analyzed the effect of NNAT expression on intracellular calcium homeostasis and found that was associated with an attenuated decay of calcium levels to baseline following ATP-induced release of calcium from endoplasmic reticulum (ER) stores. Furthermore, NNAT expression was associated with increased cytotoxicity in OS cells from thapsigargin, an inhibitor of calcium reuptake into ER and an inducer of the ER stress response. These results suggest a possible tumor suppressor role for NNAT in human osteosarcoma. Additional study is needed ascertain sensitization to ER stress-associated apoptosis as a mechanism of NNAT-dependent cytotoxicity. In that case, epigenetic modification therapy to effect NNAT transcriptional derepression may represent a therapeutic strategy potentially of benefit to a majority of osteosarcoma patients.
Palabras clave