Skip to main content
Science & Technology

Genome-Wide CRISPR Screens: A Promising Tool to Investigate Essential Cistromes

By 7th February 2020No Comments

The following study was conducted by Scientists from Dana-Farber Cancer Institute, Boston; Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston; Harvard T.H. Chan School of Public Health, Boston; Center for Genetic Medicine Research, Children’s National Hospital, Washington; The George Washington School of Medicine and Health Sciences, Washington; Harvard School of Public Health, Boston; University of Virginia, Charlottesville, USA; Northeastern University, People’s Republic of China. Study is published in Proceedings of the National Academy of Sciences Journal as detailed below.

Proceedings of the National Academy of Sciences; 116(50): 25186-25195

Deciphering Essential Cistromes Using Genome-Wide CRISPR Screens

Significance

Systematically dissecting the function of a large set of cis-regulatory elements or transcription factor binding sites (cistromes) has been technically challenging. Using genome-wide CRISPR screens, we profiled over 10,000 FOXA1 and CTCF binding sites for their roles in regulating the fitness of breast and prostate cancer cells, and accordingly developed a model to predict essentiality for cis-elements. These efforts not only reveal how the key transcription factors and their cistromes regulate cell essentiality in hormone-dependent cancers but also highlight an efficient approach to investigate the functions of noncoding regions of the genome.

Abstract

Although millions of transcription factor binding sites, or cistromes, have been identified across the human genome, defining which of these sites is functional in a given condition remains challenging. Using CRISPR/Cas9 knockout screens and gene essentiality or fitness as the readout, we systematically investigated the essentiality of over 10,000 FOXA1 and CTCF binding sites in breast and prostate cancer cells. We found that essential FOXA1 binding sites act as enhancers to orchestrate the expression of nearby essential genes through the binding of lineage-specific transcription factors. In contrast, CRISPR screens of the CTCF cistrome revealed 2 classes of essential binding sites. The first class of essential CTCF binding sites act like FOXA1 sites as enhancers to regulate the expression of nearby essential genes, while a second class of essential CTCF binding sites was identified at topologically associated domain (TAD) boundaries and display distinct characteristics. Using regression methods trained on our screening data and public epigenetic profiles, we developed a model to predict essential cis-elements with high accuracy. The model for FOXA1 essentiality correctly predicts noncoding variants associated with cancer risk and progression. Taken together, CRISPR screens of cis-regulatory elements can define the essential cistrome of a given factor and can inform the development of predictive models of cistrome function.

Source:

Proceedings of the National Academy of Sciences

URL: https://www.pnas.org/content/116/50/25186

Citation:

Fei, T., W. Li, et al. (2019). “Deciphering essential cistromes using genome-wide CRISPR screens.” Proceedings of the National Academy of Sciences 116(50): 25186-25195.