FOXD1 regulates cell division in clear cell renal cell carcinoma

Document Type

Article

Publication Date

3-24-2021

Institution/Department

MaineHealth Institute for Research, Center for Molecular Medicine, Oncology

Journal Title

BMC cancer

MeSH Headings

Animals; Calcium-Binding Proteins (genetics); Carcinoma, Renal Cell (genetics, mortality, pathology); Cation Transport Proteins (genetics); Cell Division (genetics); Cell Line, Tumor; Female; Forkhead Transcription Factors (genetics, metabolism); G2 Phase Cell Cycle Checkpoints (genetics); Gene Expression Regulation, Neoplastic; Gene Knockout Techniques; Histones (metabolism); Humans; Kaplan-Meier Estimate; Kidney Neoplasms (genetics, mortality, pathology); Male; Mice; Mice, Knockout; Middle Aged; Mitochondrial Membrane Transport Proteins (genetics); Phosphorylation (genetics); RNA-Seq; Up-Regulation; Xenograft Model Antitumor Assays

Abstract

BACKGROUND: Forkhead transcription factors control cell growth in multiple cancer types. Foxd1 is essential for kidney development and mitochondrial metabolism, but its significance in renal cell carcinoma (ccRCC) has not been reported. METHODS: Transcriptome data from the TCGA database was used to correlate FOXD1 expression with patient survival. FOXD1 was knocked out in the 786-O cell line and known targets were analyzed. Reduced cell growth was observed and investigated in vitro using growth rate and Seahorse XF metabolic assays and in vivo using a xenograft model. Cell cycle characteristics were determined by flow cytometry and immunoblotting. Immunostaining for TUNEL and γH2AX was used to measure DNA damage. Association of the FOXD1 pathway with cell cycle progression was investigated through correlation analysis using the TCGA database. RESULTS: FOXD1 expression level in ccRCC correlated inversely with patient survival. Knockout of FOXD1 in 786-O cells altered expression of FOXD1 targets, particularly genes involved in metabolism (MICU1) and cell cycle progression. Investigation of metabolic state revealed significant alterations in mitochondrial metabolism and glycolysis, but no net change in energy production. In vitro growth rate assays showed a significant reduction in growth of 786-O. In vivo, xenografted 786-O showed reduced capacity for tumor formation and reduced tumor size. Cell cycle analysis showed that 786-O had an extended G2/M phase. Investigation of mitosis revealed a deficiency in phosphorylation of histone H3 in 786-O, and increased DNA damage. Genes correlate with FOXD1 in the TCGA dataset associate with several aspects of mitosis, including histone H3 phosphorylation. CONCLUSIONS: We show that FOXD1 regulates the cell cycle in ccRCC cells by control of histone H3 phosphorylation, and that FOXD1 expression governs tumor formation and tumor growth. Transcriptome analysis supports this role for FOXD1 in ccRCC patient tumors and provides an explanation for the inverse correlation between tumor expression of FOXD1 and patient survival. Our findings reveal an important role for FOXD1 in maintaining chromatin stability and promoting cell cycle progression and provide a new tool with which to study the biology of FOXD1 in ccRCC.

First Page

312

Share

COinS