Abstract

Simple SummaryThe transcription factor p53 is a crucial tumor suppressor that regulates diverse cellular responses to protect against cancer development. Deactivating p53 signaling either by altering p53 regulators or by p53 mutations occurs frequently in human colorectal carcinoma (CRC). Forty-three percent of CRCs harbor p53 mutations that reduce wild-type p53 tumor suppressor activity and often provide neo-morphic functions, which contribute to tumorigenesis. In this review, we summarize wild-type p53 signaling, how it can be deregulated in CRC, and the functional and phenotypical effects of p53 mutations. We also discuss current therapeutic strategies of targeting p53.The transcription factor p53 functions as a critical tumor suppressor by orchestrating a plethora of cellular responses such as DNA repair, cell cycle arrest, cellular senescence, cell death, cell differentiation, and metabolism. In unstressed cells, p53 levels are kept low due to its polyubiquitination by the E3 ubiquitin ligase MDM2. In response to various stress signals, including DNA damage and aberrant growth signals, the interaction between p53 and MDM2 is blocked and p53 becomes stabilized, allowing p53 to regulate a diverse set of cellular responses mainly through the transactivation of its target genes. The outcome of p53 activation is controlled by its dynamics, its interactions with other proteins, and post-translational modifications. Due to its involvement in several tumor-suppressing pathways, p53 function is frequently impaired in human cancers. In colorectal cancer (CRC), the TP53 gene is mutated in 43% of tumors, and the remaining tumors often have compromised p53 functioning because of alterations in the genes encoding proteins involved in p53 regulation, such as ATM (13%) or DNA-PKcs (11%). TP53 mutations in CRC are usually missense mutations that impair wild-type p53 function (loss-of-function) and that even might provide neo-morphic (gain-of-function) activities such as promoting cancer cell stemness, cell proliferation, invasion, and metastasis, thereby promoting cancer progression. Although the first compounds targeting p53 are in clinical trials, a better understanding of wild-type and mutant p53 functions will likely pave the way for novel CRC therapies.

Highlights

  • Colorectal cancer (CRC) comprises two common tumor types, namely, colon cancer and rectal cancer, which make up 71% and 29% of CRC cases, respectively [1]

  • Missense mutations of p53 usually abrogate all or most of the cellular responses mediated by wild-type p53 and frequently show a dominantnegative effect (DNE) over wild-type p53 by forming mixed tetramers, thereby impairing the function of a remaining p53 wild-type allele

  • CRCs are often classified by their genetic and epigenetic aberrations into three subtypes: chromosomal instability (CIN), CpG island methylator phenotype (CIMP), and microsatellite instability (MSI), which are not mutually exclusive [72,73]. p53 mutations are some of the most frequent mutations in CIN-positive CRCs, which usually develop via the so-called canonical pathway following a step-wise progression from early adenoma via late adenoma to adenocarcinoma through acquiring a series of somatic mutations

Read more

Summary

Introduction

Colorectal cancer (CRC) comprises two common tumor types, namely, colon cancer and rectal cancer, which make up 71% and 29% of CRC cases, respectively [1]. The major risk factor for developing CRC is age: most CRCs are diagnosed in patients older than 50 In addition to this inherent risk factor, there are other main risk factors, which are lifestyle dependent. These bio-behavioral risk factors include obesity, a sedentary lifestyle, high intakes of red and processed meat, alcohol consumption, and smoking [3]. These risk factors are associated with socioeconomic development and this is reflected in the global incidence and mortality rates, with the majority of CRC cases occurring in countries with high or very high human development indexes. We discuss how the acquisition of p53 mutations and the deregulation of p53 upstream regulators disrupt p53 signaling in CRC, and how mutant p53 could be exploited as a potential therapeutic target in CRC therapy

The p53 Pathway
Control of p53 Levels
Functional Effects of p53 Mutations in CRC
Gain-of-function of p53 Mutants in CRC
Alterations of p53 Regulators in Colorectal Cancer
Activation of p53 Signaling in p53 Wild-Type Cancer Cells
Findings
Conclusions and Future Perspectives
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call