Akt-mediated Ephexin1–Ras interaction promotes oncogenic Ras signaling and colorectal and lung cancer cell proliferation

Autor: Sung-Chul Lim, Joohyun Ryu, In-Youb Chang, Jeeho Kim, Ho Jin You, Jung-Hee Lee, Young Jin Jeon
Jazyk: angličtina
Rok vydání: 2021
Předmět:
Male
Cancer Research
Lung Neoplasms
MAP Kinase Signaling System
Immunology
Mice
Nude

GTPase
Biology
medicine.disease_cause
Models
Biological

Article
Cellular and Molecular Neuroscience
Phosphoserine
Targeted therapies
Protein Domains
Cell Line
Tumor

medicine
Animals
Guanine Nucleotide Exchange Factors
Humans
Amino Acid Sequence
RNA
Messenger

Phosphorylation
Cancer models
Cell Proliferation
Mice
Inbred BALB C

QH573-671
Kinase
Erythropoietin-producing hepatocellular (Eph) receptor
Cell Biology
Oncogenes
Prognosis
Up-Regulation
Gene Expression Regulation
Neoplastic

HEK293 Cells
Fibroblast growth factor receptor
Cancer cell
Cancer research
ras Proteins
Guanine nucleotide exchange factor
Signal transduction
Carcinogenesis
Colorectal Neoplasms
Cytology
Proto-Oncogene Proteins c-akt
Protein Binding
Zdroj: Cell Death and Disease, Vol 12, Iss 11, Pp 1-14 (2021)
Cell Death & Disease
ISSN: 2041-4889
Popis: Oncogenic Ras mutations are frequently observed in solid tumor cells and are present in pancreatic cancer, colorectal cancer (CRC), and lung cancer (LC) [1, 2]. Mutational activation of K-Ras in these tissues is sufficient to initiate neoplasia in mice [3–5]. Thus, understanding the mechanisms behind Ras-induced oncogenesis is an important goal in cancer therapy. A significant consequence of Ras-mediated signal transduction is the altered expression of a large number of genes. It is well established that canonical Ras signaling is coupled to transcriptional regulation through the activation of the MAPK cascade, which involves the sequential phosphorylation and activation of the serine/threonine kinases RAF, MEK1/2, and ERK1/2 [6–12]. More recently, new regulatory factors that directly interact with K-Ras were identified and shown to play a crucial role in the full range of K-Ras oncogenic phenotypes [13–19]. Despite decades of effort, many aspects of the molecular mechanism underlying Ras-induced tumorigenesis and possible therapeutic targeting remain difficult to identify. Ephexin1 is a member of the DbI family of guanine nucleotide exchange factors (GEFs) and serves as a direct link between Eph receptors and the Rho-family of GTPases [20, 21]. Ephexin1 is highly expressed in the nervous system during development and is involved in many neurophysiological events [22–26]. It is phosphorylated by the Src family of kinases and by fibroblast growth factor receptor signaling, leading to the regulation of GEF activity, which in turn regulates actin cytoskeletal dynamics [22, 27]. Through extensive analysis of the gene expression patterns in cells with activated forms of Ras and Rho families, it was found that Ephexin1 was highly upregulated by H-RasG12V in NIH3T3 cells [28]. In humans, Ephexin1 is upregulated in papillary thyroid cancers (PTC) and has been identified as the most reliable marker for PTC diagnosis [29]. Therefore, the implication of these results is that Ephexin1 may be involved in tumor proliferation and progression, particularly in cancer cells with oncogenic Ras mutations. However, the functional consequences of Ephexin1 had not yet been addressed. In the present study, we show that Ephexin1 is overexpressed in both CRC and LC tissues and is associated with a poor prognosis for both cancers, and provide evidence for the functional and clinical significance of Ephexin1 and identify a potential therapeutic target for the case of CRC and LC caused by Ras mutations.
Databáze: OpenAIRE