Your browser doesn't support javascript.
loading
Sex-Specific Expression of Histone Lysine Demethylases (KDMs) in Thyroid Cancer.
Shobab, Leila; Zheng, Hui; Jensen, Kirk; Mendonca-Torres, Maria Cecilia; McCoy, Matthew; Hoperia, Victoria; Rosen, Jennifer; Wartofsky, Leonard; Burman, Kenneth; Vasko, Vasyl.
Affiliation
  • Shobab L; Department of Medicine, Division of Endocrinology, MedStar Washington Hospital Center, Washington, DC 20010, USA.
  • Zheng H; Department of Surgery, MedStar Washington Hospital Center, Washington, DC 20010, USA.
  • Jensen K; Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA.
  • Mendonca-Torres MC; Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA.
  • McCoy M; Innovation Center for Biomedical Informatics, Georgetown University Medical Center, Washington, DC 20007, USA.
  • Hoperia V; Institute of Biology and Medicine, Kyiv National University, 02000 Kyiv, Ukraine.
  • Rosen J; Department of Surgery, MedStar Washington Hospital Center, Washington, DC 20010, USA.
  • Wartofsky L; Department of Medicine, Division of Endocrinology, MedStar Washington Hospital Center, Washington, DC 20010, USA.
  • Burman K; Department of Medicine, Division of Endocrinology, MedStar Washington Hospital Center, Washington, DC 20010, USA.
  • Vasko V; Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA.
Cancers (Basel) ; 16(7)2024 Mar 23.
Article in En | MEDLINE | ID: mdl-38610938
ABSTRACT

BACKGROUND:

The incidence of thyroid cancer in women is 3-4-fold higher than in men. To characterize sex-specific molecular alterations in thyroid cancer, we examined the expression of sex-biased genes in normal thyroids and thyroid tumors.

METHODS:

Ingenuity pathways analysis was used to define sex-biased gene networks using data from the Cancer Genome Atlas (TCGA). Confirmatory studies were performed through the analysis of histone lysine demethylases (KDMs) expression by real-time PCR and immunostaining.

RESULTS:

In normal thyroids, 44 sex-biased genes were comparatively upregulated in male and 28 in female patients. The expressions of 37/72 (51%) sex-biased genes were affected in cancer tissues compared with normal thyroids. Gene network analyses revealed sex-specific patterns in the expressions of KDM5C, KDM5D, and KDM6A. In confirmatory studies, KDM5D mRNA and protein were detected only in males, whereas KDM5C and KDM6A were detected in samples from male and female patients. Nuclear staining with anti-KDMs was found in normal thyroids, but a loss of nuclear expression with a concomitant gain of cytoplasmic staining was observed in cancer tissues.

CONCLUSIONS:

Normal thyroids have a sex-specific molecular signature, and the development of thyroid cancer is associated with a differential expression of sex-biased genes. The sex-specific expression of KDMs, coupled with cancer-related alterations in their intracellular localization, may contribute to mechanisms underlying sex differences in thyroid tumorigenesis.
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Language: En Journal: Cancers (Basel) Year: 2024 Document type: Article Affiliation country: Estados Unidos Country of publication: Suiza

Full text: 1 Collection: 01-internacional Database: MEDLINE Language: En Journal: Cancers (Basel) Year: 2024 Document type: Article Affiliation country: Estados Unidos Country of publication: Suiza