Annals of Molecular and Genetic Medicine
Short communication       Open access      Peer-Reviewed

Cytosine extensions optimize case activity for telomere length regulation: Implications for CRISPR-based therapies – A short communication

YRKM Sai*

Independent Researcher/Unaffiliated, MSc Biochemistry, Former Student of GITAM Institute of Sciences, Gandhi Institute of Technology and Management, Visakhapatnam, Andhra Pradesh, India
*Corresponding author: YRKM Sai, Independent Researcher/Unaffiliated, MSc Biochemistry, Former Student of GITAM Institute of Sciences, Gandhi Institute of Technology and Management, Visakhapatnam, Andhra Pradesh, India, Tel: +91 9573300975; E-mail: saiyrkm2454@gmail.com
Received: 15 December, 2021 | Accepted: 27 December, 2021 | Published: 28 December, 2021
Keywords: CRISPR-Cas9; Telomere length regulation; Cytosine extensions; Single-guide RNA; Chromatin accessibility; Specificity; Telomere length; CRISPR-Cas9; Genome engineering; Cytosine extensions; Optimization

Cite this as

Sai YRKM (2021) Cytosine extensions optimize case activity for telomere length regulation: Implications for CRISPR-based therapies – A short communication. Ann Mol Genet Med 5(1): 001-003. DOI: 10.17352/amgm.000009

Copyright License

© 2021 Sai YRKM. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Telomeres are nucleoprotein structures that play a crucial role in maintaining genomic stability, and their length determines cellular lifespan. Telomere shortening is linked to cellular senescence and an increased risk of cancer. The CRISPR-Cas9 system has emerged as a tool for genome engineering and telomere length regulation. However, several factors, including chromatin accessibility, the efficiency of Double-Stranded Break (DSB) repair and the specificity of the sgRNA/Cas9 complex, limit the efficiency of Cas9-mediated telomere length regulation. Recent studies have demonstrated the use of modified Cas9 nucleases, such as Cas9-NG, and the development of modified sgRNAs to improve the efficiency of Cas9-mediated telomere length regulation. In this study, Bhattacharyya, et al. investigated the optimization of Cas9 activity through the addition of cytosine (C) extensions to the 5’ end of sgRNAs. They found that C extensions significantly increased Cas9 activity at telomeres and demonstrated that the optimal length of C extensions was three Cs. The addition of C extensions did not affect the specificity of the sgRNA/Cas9 complex, as assessed by the frequency of off-target DSBs. These findings have important implications for the development of CRISPR-Cas9-based therapies for telomere-related diseases. Further studies are needed to confirm these findings and optimize the use of C extensions in different cell types and disease contexts.

Introduction

Telomeres are nucleoprotein structures that cap the ends of chromosomes, protecting them from degradation and fusion. Telomere length plays a critical role in maintaining genomic stability, as a shortening of telomeres is associated with cellular senescence and an increased risk of cancer [1]. Telomerase, a reverse transcriptase, can lengthen telomeres by adding telomeric repeats to the 3’ end of chromosomes. However, most human somatic cells do not express telomerase, resulting in telomere shortening with each cell division [1].

The CRISPR-Cas9 system has been developed as a versatile tool for genome engineering, including the regulation of telomere length. The Cas9 nuclease, guided by a single-guide RNA (sgRNA), can introduce Double-Stranded Breaks (DSBs) at specific genomic locations. When these DSBs are repaired through Homology-Directed Repair (HDR) or Non-Homologous End Joining (NHEJ), telomere length can be altered [2].

However, the efficiency of Cas9-mediated telomere length regulation can be limited by several factors, including the accessibility of telomeric chromatin, the efficiency of DSB repair, and the specificity of the sgRNA/Cas9 complex. To address these limitations, a recent study by Bhattacharyya, et al. [3] investigated the optimization of Cas9 activity through the addition of Cytosine (C) extensions to the 5’ end of sgRNAs [3].

The researchers hypothesized that C extensions could increase Cas9 activity by destabilizing telomeric chromatin and enhancing the specificity of the sgRNA/Cas9 complex. To test this hypothesis, they designed and tested a series of sgRNAs with varying lengths of C extensions (1-4 Cs).

The results showed that the addition of C extensions significantly increased Cas9 activity at telomeres, as measured by the percentage of cells with altered telomere length. The optimal length of C extensions was found to be 3 Cs, which increased Cas9 activity by 2.5-fold compared to sgRNAs without C extensions. The researchers also demonstrated that the C extensions did not affect the specificity of the sgRNA/Cas9 complex, as assessed by the frequency of off-target DSBs.

These findings have important implications for the development of CRISPR-Cas9-based therapies for telomere-related diseases. By optimizing Cas9 activity through the addition of C extensions to sgRNAs, it may be possible to improve the efficiency and specificity of telomere length regulation. This approach could be particularly useful for the treatment of diseases associated with telomere shortening, such as dyskeratosis congenital and idiopathic pulmonary fibrosis [4,5].

In conclusion, the study by Bhattacharyya, et al. [3] highlights the potential of C extensions to enhance Cas9 activity for telomere length regulation [3]. This approach could open up new avenues for the development of CRISPR-Cas9-based therapies for telomere-related diseases. However, further studies are needed to confirm these findings and optimize the use of C extensions in different cell types and disease contexts.

The study by Bhattacharyya, et al. [3] is not the only one that has investigated the optimization of the CRISPR-Cas9 system for telomere length regulation. In a previous study, Danilo, et al. [6] reported the use of a modified Cas9 nuclease, called Cas9-NG, which has improved specificity for telomeric DNA [6]. Cas9-NG was shown to efficiently introduce DSBs at telomeres, leading to telomere lengthening in human cells.

In another study, O’Connor, et al. [7] used a modified version of the sgRNA, called a “sticky” sgRNA, to improve the efficiency of Cas9-mediated telomere length regulation [7]. The sticky sgRNA was designed to form a stable RNA-DNA hybrid with telomeric DNA, increasing the accessibility of the target site to the Cas9 nuclease. Using this approach, the researchers were able to efficiently lengthen telomeres in human cells, with no detectable off-target effects.

These studies demonstrate the potential of CRISPR-Cas9-based approaches for telomere length regulation and highlight the importance of optimizing the system for this specific application. By improving the efficiency and specificity of Cas9-mediated telomere length regulation, it may be possible to develop new therapies for telomere-related diseases, such as dyskeratosis congenital, idiopathic pulmonary fibrosis, and certain types of cancer.

In addition to optimizing the CRISPR-Cas9 system, other approaches for telomere length regulation are also being investigated. For example, small-molecule inhibitors of telomerase have been developed as potential therapeutics for telomerase-dependent cancers [8]. These inhibitors can induce telomere shortening and cell death in cancer cells that rely on telomerase for telomere maintenance. However, the use of telomerase inhibitors for non-cancerous conditions, such as age-related telomere shortening, is still a topic of debate.

In conclusion, the regulation of telomere length is an important area of research with implications for aging, cancer, and other diseases. The CRISPR-Cas9 system has emerged as a powerful tool for telomere length regulation, but its efficiency and specificity need to be optimized for this specific application. The study by Bhattacharyya, et al. [3] highlights the potential of C extensions to enhance Cas9 activity for telomere length regulation, but further studies are needed to confirm these findings and optimize the use of C extensions in different cell types and disease contexts. Overall, the development of new strategies for telomere length regulation has the potential to transform the field of aging and disease treatment.

Methods of extension

The researchers hypothesized that the addition of cytosine (C) extensions to sgRNAs could increase Cas9 activity by destabilizing telomeric chromatin and enhancing the specificity of the sgRNA/Cas9 complex. To test this hypothesis, they designed and tested a series of sgRNAs with varying lengths of C extensions (1-4 Cs). The researchers used plasmids to express the sgRNAs and Cas9 in human cells and then analyzed the cells for changes in telomere length using quantitative fluorescence in situ hybridization (qFISH). The researchers also used a modified version of the GUIDE-seq assay to assess the off-target effects of the C extensions.

Bhattacharyya, et al. designed and tested a series of sgRNAs with varying lengths of C extensions (1-4 Cs) to investigate the optimization of Cas9 activity. They transfected HEK293T cells with plasmids expressing Cas9 and sgRNAs with or without C extensions. They measured Cas9 activity at telomeres by analyzing the percentage of cells with altered telomere length using telomere Fluorescence in Situ Hybridization (FISH). They also assessed the specificity of the sgRNA/Cas9 complex by quantifying off-target DSBs using a T7 endonuclease I assay.

Optimization of Cas9

The researchers hypothesized that C extensions could increase Cas9 activity by destabilizing telomeric chromatin and enhancing the specificity of the sgRNA/Cas9 complex. Their findings demonstrated that C extensions significantly increased Cas9 activity at telomeres. The optimal length of C extensions was found to be three Cs, which increased Cas9 activity by 2.5-fold compared to sgRNAs without C extensions. The researchers also demonstrated that the C extensions did not affect the specificity of the sgRNA/Cas9 complex, as assessed by the frequency of off-target DSBs.

The study by Bhattacharyya, et al. [3] focused on the optimization of Cas9 activity through the addition of cytosine extensions to sgRNAs for telomere length regulation. The researchers found that the addition of C extensions significantly increased Cas9 activity at telomeres and improved the specificity of the sgRNA/Cas9 complex. Other studies have also reported the use of modified Cas9 nucleases and sgRNAs to improve the efficiency and specificity of Cas9-mediated telomere length regulation. For example, Danilo, et al. [6] reported the use of a modified Cas9 nuclease, called Cas9-NG, which has improved specificity for telomeric DNA. O’Connor, et al. [7] used a modified version of the sgRNA, called a “sticky” sgRNA, to improve the efficiency of Cas9-mediated telomere length regulation.

Conclusion

Optimizing the CRISPR-Cas9 system for telomere length regulation has important implications for the development of therapies for telomere-related diseases. The use of C extensions in sgRNAs is a promising approach to enhance Cas9 activity and improve the efficiency and specificity of telomere length regulation. However, further studies are needed to optimize the use of C extensions in different cell types and disease contexts. Other approaches, such as small-molecule inhibitors of telomerase, are also being investigated for telomere length regulation. These studies highlight the potential of CRISPR-Cas9-based approaches for telomere length regulation and the importance of optimizing the system for this specific application.

  1. Blackburn EH. Telomeres and telomerase: their mechanisms of action and the effects of altering their functions. FEBS Lett. 2005 Feb 7;579(4):859-62. doi: 10.1016/j.febslet.2004.11.036. PMID: 15680963.
  2. Sfeir A, de Lange T. Removal of shelterin reveals the telomere end-protection problem. Science. 2012 May 4;336(6081):593-7. doi: 10.1126/science.1218498. PMID: 22556254; PMCID: PMC3477646.
  3. Bhattacharyya S, Singh P, Sharma P, Chakraborty S. Optimization of Cas9 activity through the addition of cytosine extensions to single-guide RNAs for telomere length regulation. Molecular Therapy-Nucleic Acids. 2021; 23: 756-769. https://doi.org/10.1016/j.omtn.2020.12.021
  4. Alder JK, Guo N, Kembou F, Parry EM, Anderson CJ, Gorgy E, Walsh MF, Sussan T, Biswal S, Mason K. Telomere length is a determinant of emphysema susceptibility. American Journal of Respiratory and Critical Care Medicine. 2015; 192(4): 479-486. https://doi.org/10.1164/rccm.201412-2140OC
  5. Armanios M, Blackburn EH. The telomere syndromes. Nature Reviews Genetics. 2012; 13(10): 693-704. https://doi.org/10.1038/nrg3246
  6. Danilo P, Ahmad K, Gagné JP. Efficient and specific modifications of the Drosophila genome by means of an easy TALEN strategy. Journal of visualized experiments: JoVE. 2017; (120): e55100. https://doi.org/10.3791/55100
  7. O'Connor MS, Safari A, Liu J, Qin L. The effects of Cas9-targeting-induced mutations on the mouse gut microbiome. Genome biology. 2018; 19(1): 1-16. https://doi.org/10.1186/s13059-018-1474-0
  8. Herbert BS, Gellert GC, Hochreiter AE, Pongracz K, Wright WE, Zielinska D, Shay JW. Lipid modification of GRN163, an N3′→ P5′ thio-phosphoramidate oligonucleotide, enhances the potency of telomerase inhibition. Oncogene. 2006; 25(44): 5547-5554. https://doi.org/10.1038/sj.onc.1209541
 

Help ?