Abstract

Normal cell function is dependent on the proper maintenance of chromatin structure. Regulation of chromatin structure is controlled by histone modifications that directly influence chromatin architecture and genome function. Specifically, the histone deacetylase (HDAC) family of proteins modulate chromatin compaction and are commonly dysregulated in many tumors, including colorectal cancer (CRC). However, the role of HDAC proteins in early colorectal carcinogenesis has not been previously reported. We found HDAC1, HDAC2, HDAC3, HDAC5, and HDAC7 all to be up-regulated in the field of human CRC. Furthermore, we observed that HDAC2 up-regulation is one of the earliest events in CRC carcinogenesis and observed this in human field carcinogenesis, the azoxymethane-treated rat model, and in more aggressive colon cancer cell lines. The universality of HDAC2 up-regulation suggests that HDAC2 up-regulation is a novel and important early event in CRC, which may serve as a biomarker. HDAC inhibitors (HDACIs) interfere with tumorigenic HDAC activity; however, the precise mechanisms involved in this process remain to be elucidated. We confirmed that HDAC inhibition by valproic acid (VPA) targeted the more aggressive cell line. Using nuclease digestion assays and transmission electron microscopy imaging, we observed that VPA treatment induced greater changes in chromatin structure in the more aggressive cell line. Furthermore, we used the novel imaging technique partial wave spectroscopy (PWS) to quantify nanoscale alterations in chromatin. We noted that the PWS results are consistent with the biological assays, indicating a greater effect of VPA treatment in the more aggressive cell type. Together, these results demonstrate the importance of HDAC activity in early carcinogenic events and the unique role of higher-order chromatin structure in determining cell tumorigenicity.

Highlights

  • Higher-order chromatin structure regulates a number of biological processes on different scales of organization

  • These results indicated that histone deacetylase (HDAC) are a novel marker of human colon field carcinogenesis and support our hypothesis that HDAC inhibitors (HDACIs) target more tumorigenic cell types related to changes in nuclear mass density, chromatin accessibility, and cell viability

  • We found that HDAC2 expression was about 2-fold higher in patients harboring an adenoma elsewhere in the colon compared to the control group (p,0.05, n = 12 subjects; Figure 1C)

Read more

Summary

Introduction

Higher-order chromatin structure regulates a number of biological processes on different scales of organization. Chromatin modulation is well studied at the nucleosome level, which consists of DNA wrapped tightly around a histone octamer composed of the four core histone proteins (H3, H4, H2A, H2B). The core histones are subject to a variety of post-translational modifications on their N-terminal tails, such as methylation, phosphorylation, and acetylation [1]. Given that chromatin structure plays a significant role in gene transcription, dysregulation of proper chromatin structure is present in many diseases. Changes in chromatin structure orchestrate the alterations in tumor suppressor genes or activation of proto-oncogenes needed for neoplastic progression. Epigenetic regulation of gene expression is emerging as an important facet of carcinogenesis, including colorectal cancer (CRC) [2]

Methods
Results
Conclusion
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