CGE_2024v12n1

Cancer Genetics and Epigenetics 2024, Vol.12, No.1, 47-54 http://www.medscipublisher.com/index.php/cge 49 By gaining a deeper understanding of these methylation events, HTS technology can not only provide new insights into disease mechanisms, but also provide a basis for the development of new diagnostic approaches and treatment strategies. 1.2 Histone modification analysis HTS technology also plays an important role in the study of histone modification. By precisely identifying the type and location of histone modifications, the researchers were able to explore how they affect gene expression and chromosome structure. Modification patterns such as acetylation and methylation of histones regulate gene activity by affecting chromatin tightness, which is particularly important in the process of cancer occurrence and cell differentiation. For example, trimethylation at the K27 site of histone H3 (H3K27me3) is an important silent marker, and its abnormal increase in multiple cancers is strongly associated with dysregulation of gene expression and tumor development. While the 2023 study by Zhao et al. focused on one invasive insect, However, Chromatin Immunoprecipitation with high-throughput sequencing (ChIP-seq) revealed that H3K4me3 was related to gene activation, while H3K27me3 was mainly related to transcriptional inhibition. This finding supports the role of H3K27me3 in gene silencing, providing a basis for understanding its role in cancer (Zhao et al., 2023). The 2023 study by Zhou et al. used CUT&Tag technology to analyze histone modifications in early embryonic development in cattle and humans. It was found that H3K9me3 and H3K27me3 co-occupied the genome before embryo gene activation, suggesting a global transcriptional inhibition mechanism. This study reveals the primary role of H3K27me3 in limiting cell potential, providing insights into its function in cancer (Zhou et al., 2023) This study from Yang et al. 2023 focuses on H3K4me3, but it mentions the effect of histone modifications on tumor cell proliferation, migration, and invasion, which echoes the role of H3K27me3, as both may co-participate in cancer development in some contexts (Yang et al. 2023). These studies highlight the critical role of H3K27me3 in cell fate determination and cancer development, particularly in gene silencing and methylation silencing of tumor suppressor genes. By better understanding the role and mechanism of action of H3K27me3, we can provide new strategies and targets for cancer diagnosis and treatment. 1.3 Identification and functional study of epigenetic marks HTS technology has not only made significant progress in identifying epigenetic marks, but has also advanced the understanding of the function of these marks. The latest research, using HTS technology, reveals a direct link between epigenetic marks and disease. For example, by analyzing genome-wide epigenetic maps in different disease states, researchers have been able to identify specific methylation and histone modification patterns that can serve as biomarkers for disease. This study by Whelan et al. 2023 highlights the use of DNA methylation in animal welfare monitoring and, while focusing primarily on animal models, provides a methodological framework for understanding epigenetic markers in human disease. By analyzing animal DNA methylation patterns and techniques, the study demonstrates a key framework for complex DNA methylation biomarkers, DNA methylation clocks, and environment-specific DNA methylation signatures that can provide complex, context-dependent readings of health and disease in disease states (Whelan et al., 2023). The 2023 study by Costa et al. focused on epigenetic reprogramming in cancer, particularly changes in DNA methylation, histone modification, and expression of non-coding RNA. These dynamic epigenetic changes are associated with tumor heterogeneity, infinite self-renewal capacity, and multiseries differentiation, and are a major challenge for treatment and drug resistance. Research has highlighted the ability to restore cancer epigenomes by

RkJQdWJsaXNoZXIy MjQ4ODYzNQ==