CRISPR-based Epigenetic editing (Part 53- CRISPR in Gene Editing and Beyond)

Roohi Bansal
Biotechnology by TSB
7 min readOct 5, 2023

Welcome to the 53rd part of the multi-part series on applications of CRISPR in gene editing and beyond.

The second level of genomic regulation after transcription effectors is epigenetic gene regulation. This process involves a series of chemical modifications that results in selective silencing or activation of particular genes without direct altering the DNA primary sequence. Epigenetic modifications can occur within the DNA itself or on the histone proteins that assist in DNA packaging. These modifications play a crucial role in deciding whether genes are activated or suppressed, thereby influencing protein production within cells. This regulatory mechanism helps guarantee that each cell produces solely the proteins that are necessary for its proper function.

A common type of epigenetic modification is DNA methylation. DNA methylation involves the attachment of methyl groups to the C5 position of cytosine residues in CpG dinucleotide sequences of the DNA. The presence of methyl groups in the promoter blocks the binding of transcription factors, and RNA polymerase II; thus, the transcription of the gene is inhibited, causing the gene to be turned off or silenced, and therefore, no protein is produced from that gene.

Histone modification is another prevalent form of epigenetic modification. Histones are structural proteins that aid in the packaging of DNA within the cell nucleus. They can be modified by the addition or removal of chemical groups like methyl or acetyl groups. These chemical groups impact how tightly DNA is wrapped around the histones and whether a given genomic sequence is stretched out and accessible to the transcriptional machinery, affecting a gene to be turned on or off. For instance, the addition of acetyl groups to the lysine residues of histone proteins causes histones to drift away from DNA. This released structure facilitates the binding of transcription factors and RNA polymerase II to the target gene, thus activating its transcription. On the other hand, histone deacetylation, i.e., the removal of acetyl groups, blocks access to transcriptional machinery, thus, repressing the transcription of the target gene.

Also, the addition of methyl groups to the histone proteins can either activate or repress the transcription of the gene, thus turning it on or off. For example, trimethylation of histone H3 at lysine 4 (H3K4me3) in the promoter region activates the transcription of the gene. However, dimethylation of histone H3 at lysine 9 (H3K9me2), is a signal for transcriptional silencing.

Thus, the epigenome, which consists of chemical compounds that modify genomic DNA and histones, acts as the second level of gene expression regulation after the transcription effectors.

To achieve CRISPR-Cas9 mediated epigenetic regulation, dCas9 can be fused to various effectors.

We have already discussed in Part 30 that dead Cas9 or dCas9 is a mutant in which both cleavage domains HNH and RuVC of Cas9 are inactivated. Although dCas9 can no longer cleave DNA, it can still bind target DNA with the same precision when guided by sgRNA.

One such effector is histone acetyltransferase p300, which can be fused to dCas9, creating a dCas9-p300 fusion activator (Hilton et al., 2015; Chen et al., 2017; Kuscu et al., 2019). p300 adds acetyl groups to the histone proteins and facilitates the binding of transcription factors and RNA polymerase II to the target gene, thus significantly activating its transcription. Furthermore, the dCas9-p300 fusion achieves higher activation than dCas9-VP64 alone (discussed in Part 51).

Gene activation with dCas9-p300

Another transcriptional activator is Tet1 demethylase, which, when fused to dCas9, decreases methylation and induces transcription at CpG regions of the promoter of the target gene (Gjaltema and Rots, 2020).

In contrast to transcriptional activators, transcriptional repressors DNA Methyltransferase 3 Alpha, abbreviated as DNMT3A, and Lysine-specific histone demethylase 1 abbreviated as LSD1, repress transcription of the target gene. DNA Methyltransferase 3 Alpha, when fused to dCas9, causes cytosine methylation of the promoter of the target gene, thus repressing its activation. On the other hand, LSD1 is a histone demethylase that, when fused to dCas9, targets histone H3, methylated at lysine 4 residue, and demethylates it, leading to transcriptional repression of the target gene.

This is how dCas9 can regulate the target gene expression.

If you liked this article and want to know more about more about applications of CRISPR in gene editing and beyond, click the below links:

For book lovers:

For video lovers:

https://www.udemy.com/course/crispr-cas-system-applications-in-gene-editing-and-beyond/?referralCode=8ED7BBAEB7AE497D755F

References:

Riedmayr, L. M., Hinrichsmeyer, K. S., Karguth, N., Böhm, S., Splith, V., Michalakis, S., & Becirovic, E. (2022). dCas9-VPR-mediated transcriptional activation of functionally equivalent genes for gene therapy. Nature Protocols, 17(3), 781–818.

McDougald, D. S., Duong, T. T., Palozola, K. C., Marsh, A., Papp, T. E., Mills, J. A., … & Bennett, J. (2019). CRISPR activation enhances in vitro potency of AAV vectors driven by tissue-specific promoters. Molecular Therapy-Methods & Clinical Development, 13, 380–389.

Moreno, A. M., Fu, X., Zhu, J., Katrekar, D., Shih, Y. R. V., Marlett, J., … & Mali, P. (2018). In situ gene therapy via AAV-CRISPR-Cas9-mediated targeted gene regulation. Molecular Therapy, 26(7), 1818–1827.

Lin, S., Ewen-Campen, B., Ni, X., Housden, B. E., & Perrimon, N. (2015). In vivo transcriptional activation using CRISPR/Cas9 in Drosophila. Genetics, 201(2), 433–442.

Chen, M., & Qi, L. S. (2017). Repurposing CRISPR system for transcriptional activation. RNA Activation, 147–157.

Nihongaki, Y., Furuhata, Y., Otabe, T., Hasegawa, S., Yoshimoto, K., & Sato, M. (2017). CRISPR–Cas9-based photoactivatable transcription systems to induce neuronal differentiation. Nature methods, 14(10), 963–966.

Omachi, K., & Miner, J. H. (2021). Comparative analysis and rational design of dCas9-VP64 variants for CRISPR activation. bioRxiv, 2021–08.

Dominguez, A. A., Lim, W. A., & Qi, L. S. (2016). Beyond editing: repurposing CRISPR–Cas9 for precision genome regulation and interrogation. Nature reviews Molecular cell biology, 17(1), 5–15.

Hunt, C., Hartford, S. A., White, D., Pefanis, E., Hanna, T., Herman, C., … & Gong, G. (2021). Tissue-specific activation of gene expression by the Synergistic Activation Mediator (SAM) CRISPRa system in mice. Nature Communications, 12(1), 2770.

Vad-Nielsen, J., Nielsen, A. L., & Luo, Y. (2018). Simple method for assembly of CRISPR synergistic activation mediator gRNA expression array. Journal of biotechnology, 274, 54–57.

Zhang, Y., Yin, C., Zhang, T., Li, F., Yang, W., Kaminski, R., … & Hu, W. (2015). CRISPR/gRNA-directed synergistic activation mediator (SAM) induces specific, persistent and robust reactivation of the HIV-1 latent reservoirs. Scientific reports, 5(1), 16277.

Saayman, S., Ali, S. A., Morris, K. V., & Weinberg, M. S. (2015). The therapeutic application of CRISPR/Cas9 technologies for HIV. Expert opinion on biological therapy, 15(6), 819–830.

Xiong, K., Zhou, Y., Hyttel, P., Bolund, L., Freude, K. K., & Luo, Y. (2016). Generation of induced pluripotent stem cells (iPSCs) stably expressing CRISPR-based synergistic activation mediator (SAM). Stem Cell Research, 17(3), 665–669.

Tanenbaum, M. E., Gilbert, L. A., Qi, L. S., Weissman, J. S., & Vale, R. D. (2014). A protein-tagging system for signal amplification in gene expression and fluorescence imaging. Cell, 159(3), 635–646.

Konermann, S., Brigham, M. D., Trevino, A. E., Joung, J., Abudayyeh, O. O., Barcena, C., … & Zhang, F. (2015). Genome-scale transcriptional activation by an engineered CRISPR-Cas9 complex. Nature, 517(7536), 583–588.

Ji, H., Jiang, Z., Lu, P., Ma, L., Li, C., Pan, H., … & Zhu, H. (2016). Specific reactivation of latent HIV-1 by dCas9-SunTag-VP64-mediated guide RNA targeting the HIV-1 promoter. Molecular Therapy, 24(3), 508–521.

Kazi, T. A., & Biswas, S. R. (2021). CRISPR/dCas system as the modulator of gene expression. Progress in Molecular Biology and Translational Science, 178, 99–122.

Matharu, N., Rattanasopha, S., Tamura, S., Maliskova, L., Wang, Y., Bernard, A., … & Ahituv, N. (2019). CRISPR-mediated activation of a promoter or enhancer rescues obesity caused by haploinsufficiency. Science, 363(6424), eaau0629.

Montefiori, L. E., & Nobrega, M. A. (2019). Gene therapy for pathologic gene expression. Science, 363(6424), 231–232.

Wang, G., Chow, R. D., Bai, Z., Zhu, L., Errami, Y., Dai, X., … & Chen, S. (2019). Multiplexed activation of endogenous genes by CRISPRa elicits potent antitumor immunity. Nature immunology, 20(11), 1494–1505.

Qi, L. S., Larson, M. H., Gilbert, L. A., Doudna, J. A., Weissman, J. S., Arkin, A. P., & Lim, W. A. (2013). Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell, 152(5), 1173–1183.

Gilbert, L. A., Horlbeck, M. A., Adamson, B., Villalta, J. E., Chen, Y., Whitehead, E. H., … & Weissman, J. S. (2014). Genome-scale CRISPR-mediated control of gene repression and activation. Cell, 159(3), 647–661.

Alerasool, N., Segal, D., Lee, H., & Taipale, M. (2020). An efficient KRAB domain for CRISPRi applications in human cells. Nature Methods, 17(11), 1093–1096.

Stoll, G. A., Pandiloski, N., Douse, C. H., & Modis, Y. (2022). Structure and functional mapping of the KRAB‐KAP1 repressor complex. The EMBO Journal, 41(24), e111179.

Brocken, D. J., Tark-Dame, M., & Dame, R. T. (2018). dCas9: a versatile tool for epigenome editing. Current issues in molecular biology, 26(1), 15–32.

Gjaltema, R. A., & Rots, M. G. (2020). Advances of epigenetic editing. Current opinion in chemical biology, 57, 75–81.

Hilton, I. B., D’ippolito, A. M., Vockley, C. M., Thakore, P. I., Crawford, G. E., Reddy, T. E., & Gersbach, C. A. (2015). Epigenome editing by a CRISPR-Cas9-based acetyltransferase activates genes from promoters and enhancers. Nature biotechnology, 33(5), 510–517.

Kuscu, C., Mammadov, R., Czikora, A., Unlu, H., Tufan, T., Fischer, N. L., … & Adli, M. (2019). Temporal and spatial epigenome editing allows precise gene regulation in mammalian cells. Journal of molecular biology, 431(1), 111–121.

Chen, T., Gao, D., Zhang, R., Zeng, G., Yan, H., Lim, E., & Liang, F. S. (2017). Chemically controlled epigenome editing through an inducible dCas9 system. Journal of the American Chemical Society, 139(33), 11337–11340.

Happy learning!

--

--