AI Article Synopsis

  • Dysfunction of histone methylation, particularly involving the enzyme SMYD2, is linked to cancer progression, and this study focuses on its role in high-grade serous ovarian carcinomas (HGSOCs).
  • Researchers found that SMYD2 is significantly overexpressed in HGSOC tissues and demonstrated that inhibiting SMYD2 led to reduced cell growth and increased apoptosis in HGSOC cells.
  • Additionally, the selective SMYD2 inhibitor LLY-507 showed potential as a standalone treatment and in combination with the PARP inhibitor olaparib, indicating a promising therapeutic strategy for patients with HGSOC.

Article Abstract

Dysfunction of histone methylation is known to be related to cancer progression. The histone methyltransferase SMYD2 methylates histone protein H3 and non-histone proteins, including poly ADP ribose polymerase 1 (PARP1). There have been reports of SMYD2 overexpression in several types of cancers. However, there are no reports regarding its role in high-grade serous ovarian carcinomas (HGSOCs). Therefore, we investigated the expression profile and conducted functional analysis on SMYD2 in HGSOC cells. In addition, we verified whether SMYD2 inhibition increases the susceptibility of HGSOC cells to PARP inhibitors. We analyzed the expression of histone methyltransferase SMYD2 by quantitative real-time polymerase chain reaction and immunohistochemistry using HGSOC clinical tissues (n = 35). We performed functional analyses, including cell proliferation assay, cell cycle analysis, and immunoblotting, after treatment with SMYD2 siRNAs and SMYD2 selective inhibitor LLY-507 in HGSOC cells. We also performed colony-formation assay after combination treatment with LLY-507 and PARP inhibitor olaparib in HGSOC cells. The expression profiles of SMYD2 showed significant overexpression of SMYD2 in HGSOC clinical tissues. The knockdown or inhibition of SMYD2 by siRNAs or LLY-507, respectively, suppressed cell growth by increasing the proportion of apoptotic cells. LLY-507 showed additive effect with olaparib in the colony-formation assay. These findings suggest that LLY-507 can be used alone or in combination with a PARP inhibitor for the treatment of patients with HGSOC.

Download full-text PDF

Source
http://dx.doi.org/10.1016/j.bbrc.2019.03.155DOI Listing

Publication Analysis

Top Keywords

hgsoc cells
16
histone methyltransferase
12
methyltransferase smyd2
12
smyd2
11
smyd2 selective
8
selective inhibitor
8
inhibitor lly-507
8
lly-507 combination
8
poly adp
8
adp ribose
8

Similar Publications

Ovarian cancer is the deadliest gynecologic cancer, and with the majority of patients dying within the first five years of diagnosis, new therapeutic options are required. The small guanosine triphosphatase (GTPase) Ras-related nuclear protein (Ran) has been reported to be highly expressed in high-grade serous ovarian cancers (HGSOCs) and associated with poor outcomes. Blocking Ran function or preventing its expression were shown to be promising treatment strategies, however, there are currently no small molecule inhibitors available to specifically inhibit Ran function.

View Article and Find Full Text PDF

High-grade serous ovarian carcinoma (HGSOC) remains the most common and deadly form of ovarian cancer. However, available cell lines usually fail to appropriately represent its complex molecular and histological features. To overcome this drawback, we established OVAR79, a new cell line derived from the ascitic fluid of a patient with a diagnosis of HGSOC, which adds a unique set of properties to the study of ovarian cancer.

View Article and Find Full Text PDF

Background: Ovarian cancer (OC) is a globally prevalent malignancy with significant morbidity and mortality, yet its heterogeneity poses challenges in treatment and prognosis. Recognizing the crucial role of the tumor microenvironment (TME) in OC progression, this study leverages integrative multi-omics and machine learning to uncover TME-associated prognostic biomarkers, paving the way for more personalized therapeutic interventions.

Methods: Employing a rigorous multi-omics approach, this study analyzed single-cell RNA sequencing (scRNA-seq) data from OC and normal tissue samples, including high-grade serous OC (HGSOC) from the Gene Expression Omnibus (GEO: GSE184880) and The Cancer Genome Atlas (TCGA) OC cohort, utilizing the Seurat package to annotate 700 TME-related genes.

View Article and Find Full Text PDF

New prognostic index for neoadjuvant chemotherapy outcome in patients with advanced high-grade serous ovarian cancer.

BMC Cancer

December 2024

Department of Gynecologic Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China.

Background: A validated prognostic index for the outcome of patients with advanced high-grade serous ovarian cancer (HGSOC) undergoing neoadjuvant chemotherapy (NACT) remains elusive. To address this need, we developed an ovarian neoadjuvant chemotherapy prognostic index (ONCPI) to improve predictive accuracy.

Methods: We encompassed an analysis of the clinicopathological characteristics of patients with advanced HGSOC who were administered platinum-based NACT.

View Article and Find Full Text PDF
Article Synopsis
  • The study investigates the limitations of immunotherapies in treating high-grade serous ovarian cancer (HGSOC) and explores the potential of combining immunotherapy with PARP inhibitors, emphasizing the roles of BRCAness and the tumor microenvironment in treatment response.
  • Researchers performed detailed immunogenomic analyses and machine learning to identify a 24-gene signature predicting BRCAness, revealing high immune cell infiltration and an association with immunosuppressive cells in samples receiving combination therapies.
  • Findings indicate that PARP inhibitors can activate immune-related pathways similarly to genetic BRCAness, and the team has developed a web app for analyzing ovarian cancer samples and providing a vulnerability score for patient stratification.
View Article and Find Full Text PDF

Want AI Summaries of new PubMed Abstracts delivered to your In-box?

Enter search terms and have AI summaries delivered each week - change queries or unsubscribe any time!