Outline
Figures

Volume: 02, Issue: 01, Page: 10-13

CRISPR-dCas9-mediated CpG island editing: A potential game-changer for diabetes treatment

1 Department of Biochemistry and Molecular Biology, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj-8100, Dhaka, Bangladesh

*Corresponding authors

Email address: kkdutta@bsmrstu.edu.bd (Khokon Kumar Dutta)

doi: https://doi.org/10.69517/jber.2025.02.01.0003

Share:

Received:
01 November 2024

Revised:
07 December 2024

Accepted:
14 December 2024

Published:
21 December 2024

Highlights

  • CRISPR-dCas9-mediated CpG island editing of key genes hold the potential for T2DM treatment.
  • Precise modulation of gene expression in the optimal level may restore metabolic balance crucial for T2DM prevention or management.
  • The CpG island hypomethylation in the TXNIP gene is strongly linked with T2DM.
  • CRISPR-dCas9-mediated CpG island editing of TXNIP gene hold the potential to reverse hyperglycemia, enhance insulin sensitivity, and protect beta cells.
  • Initiatives and steps are required to translate the approach for clinical applications for T2DM treatment.

Abstract

Type 2 diabetes mellitus (T2DM) is a chronic metabolic disorder characterized by hyperglycemia. Current therapeutic strategies primarily manage symptoms, leaving a substantial unmet need for curative interventions. This review explores the potential of CRISPR-dCas9-mediated CpG island editing as a promising therapeutic approach for T2DM. CpG islands, DNA regions enriched in cytosine-guanine dinucleotides, play a pivotal role in gene regulation. Their methylation status significantly influences gene expression. By targeting specific CpG islands within genes involved in glucose metabolism and insulin signaling, CRISPR-dCas9 can modulate gene expression and restore metabolic homeostasis. A particular focus is placed on the TXNIP gene, implicated in T2D pathogenesis. Reprogramming TXNIP expression using CRISPR-dCas9 offers potential therapeutic benefits, including protecting pancreatic beta cells, enhancing insulin sensitivity, and mitigating inflammation. While the potential of CRISPR-dCas9-mediated CpG island editing is clear and evident now, further steps are imperative to translate this approach into effective and safe therapies for T2DM patients.

Graphical abstract

Keywords

Type 2 diabetes mellitus, CpG island methylation, CpG island hypermethylation, CpG island hypomethylation, Gene transcription regulation

1. Introduction

Advances in epigenetic editing technology have paved the way for novel therapeutic approaches in various diseases, including diabetes. One promising approach is the CRISPR-dCas9-mediated editing of CpG islands, which could potentially target gene expression through epigenetic modifications. This review explores the role of CpG islands, their involvement in gene regulation, and how CRISPR-dCas9 can be used to edit these regions for potential diabetes treatment, focusing specifically on the TXNIP gene.
CpG islands are short stretches of DNA that are densely packed with cytosine-guanine (CpG) dinucleotides. They are commonly found near gene promoters, playing a critical role in gene regulation due to their influence on transcriptional activity (Bird et al., 1985; Bird, 1986). The unique feature of CpG islands is their high CpG density, their largely unmethylated state, and their strong association with active gene promoters (Gardiner-Garden and Frommer, 1987). Methylation of CpG islands, however, can lead to transcriptional silencing, making these regions key targets in epigenetic regulation.
Several computational tools have been developed to identify CpG islands within genomic sequences, including CpG Island Searcher, IslandPicker, and PromoterScan (Deaton and Bird, 2011). These tools use algorithms that search for high CpG density, appropriate sequence length, high GC content, and an observed/expected CpG ratio greater than a specific threshold (Choy et al., 2010). These features are essential for accurate identification and analysis of CpG islands, particularly in their role in regulating gene expression.
DNA methylation, the addition of methyl groups to the cytosine base in CpG dinucleotides, is a powerful epigenetic modification that silences gene expression when it occurs in gene promoters (Lee and Lee, 2012). Methylation interferes with transcription factor binding and recruits proteins that compact chromatin, thus reducing gene expression. Conversely, unmethylated CpG islands are typically associated with actively transcribed genes (Lee et al., 2015; Yoo et al., 2021). Factors influencing CpG island methylation include DNA methyltransferases (DNMTs), Ten-Eleven Translocation (TET) enzymes, chromatin accessibility, and environmental conditions.
In Dutta et al. (2005), identified a CpG island within the TXNIP gene's promoter region. This CpG Island, rich in cytosine and guanine dinucleotides, is a common target for epigenetic modifications, particularly DNA methylation. DNA methylation, the addition of a methyl group to cytosine residues in CpG dinucleotides, typically silences gene expression by hindering transcription factor binding or promoting chromatin compaction. Aberrant methylation patterns are implicated in various diseases, including diabetes.
Dutta et al. (2005) pioneered the discovery that hypermethylation of this CpG island correlates with decreased TXNIP expression in kidney cancers. Conversely, under normal conditions, the CpG Island is typically hypomethylated, leading to increased TXNIP levels. This dynamic regulation is essential for maintaining balanced cellular proliferation in both normal and cancerous kidney tissues (Dutta et al., 2005; Kim et al., 2021; Zhang et al., 2017).
A similar methylation pattern is observed in diabetic conditions. Hypomethylation of TXNIP correlates with elevated expression and disrupted glucose homeostasis (Zhang et al., 2017; Kim et al., 2021).
The methylation status of the cg19693031 site within the TXNIP gene has been linked to fasting blood glucose regulation in non-diabetic Taiwanese adults (Tsai et al., 2022). The relationship between TXNIP-cg19693031 DNA methylation (DNAm) and type 2 diabetes (T2D) is well-established, with strong correlations to HbA1c, insulin, and fasting glucose levels (Tsai et al., 2022). Hypomethylation at TXNIP-cg19693031 has been robustly associated with T2D, as well as elevated inflammatory biomarkers including VCAM-1, ICAM-1, MMP-2, sRAGE, and P-selectin. Notably, the connection between TXNIP-cg19693031 methylation and T2D persists independently of these inflammatory biomarkers (Xiang et al., 2021).
Further studies have also demonstrated a marked decrease in methylation across five TXNIP loci in individuals with T2D compared to healthy controls, where increasing methylation levels correspond to a reduced T2D risk. Interactions among TXNIP methylation, obesity, and hypertriglyceridemia were identified as contributing factors to T2D onset (Zhang et al., 2020).
Recent research has expanded on these findings, investigating how TXNIP methylation influences T2D risk in detail (Wu et al., 2024; Maeda et al., 2024). Two pivotal studies illustrate these associations, Wu et al. (2024) and Maeda et al. (2024) provide complementary insights into the role of TXNIP DNA methylation in type 2 diabetes (T2D) risk and glycemic regulation. Wu et al. (2024) in a nested case-control study, reported that higher methylation levels at TXNIP CpG sites 2–5 were associated with a 61–87% reduction in T2D risk, highlighting the protective potential of hypermethylation in this region. Similarly, Maeda et al. (2024) in a longitudinal study, found that hypomethylation at cg19693031 was linked to greater increases in fasting plasma glucose (FPG) and hemoglobin A1c (HbA1c) over four years, suggesting that hypomethylation may impair glucose regulation and serve as an early marker of diabetes risk. Together, these findings underscore the importance of TXNIP methylation in diabetes pathogenesis and risk prediction.

2. CRISPR-Cas technology for editing CpG methylation

The discovery of CRISPR-Cas9 system has opened the door and revolutionized genome editing (Doudna and Charpentier, 2014). The catalytically inactive form of dCas9 allows for precise epigenetic editing without altering DNA sequences. By fusing dCas9 to epigenetic effectors like methyltransferases or demethylases, scientists can manipulate CpG island methylation. This offers a potential therapeutic avenue for diseases such as diabetes, where aberrant CpG island methylation affects key genes like TXNIP.

Table 1. A timeline highlighting the key developments in the area of CRISPR-dCas9 Mediated CpG island methylation and demethylation.


3. Current research and future directions

Today, CRISPR/dCas9-based CpG island methylation and demethylation are being explored for treating diseases related to epigenetic dysregulation, such as cancer, neurological disorders, and metabolic diseases. Challenges remain in optimizing delivery, minimizing off-target effects, and achieving long-term, stable modifications (Cano-Rodriguez and Rots, 2016).

3.1 In Vivo examples of CRISPR-dCas9 mediated epigenetic editing

CRISPR/dCas9-mediated CpG island methylation and demethylation in vivo have shown potential in animal models for understanding gene regulation and exploring therapeutic applications. Some prominent examples are presented in Table 2.


Table 2. Key in vivo applications of CRISPR-dCas9-mediated epigenetic editing.


4. CRISPR-dCas9-mediated reprogramming of TXNIP expression.

The CRISPR-dCas9 system can be leveraged to reprogram TXNIP expression by modifying its CpG island methylation status. For instance, CRISPR-dCas9-KRAB has been used to downregulate TXNIP by silencing its promoter region, reducing oxidative stress and improving glucose homeostasis. Alternatively, CRISPR-dCas9-TET1 can reprogram hypermethylated CpG islands in the TXNIP gene, leading to decreased TXNIP expression and improved insulin sensitivity.

5. Therapeutic potential of TXNIP reprogramming in diabetes

The ability to modulate TXNIP expression using CRISPR-dCas9 offers several therapeutic benefits for diabetes management. These include,
-Protecting pancreatic beta cells from apoptosis and oxidative stress.
-Improving insulin sensitivity in peripheral tissues.
-Enhancing glucose uptake and glycemic control.
-Reducing systemic inflammation associated with diabetes.

6. Conclusions

CRISPR-dCas9-mediated CpG island editing represents a novel and promising approach for diabetes treatment. By targeting key genes such as TXNIP, this technology has the potential to reverse hyperglycemia, enhance insulin sensitivity, and protect beta cells. While the potential is immense, further research is required to explore its therapeutic efficacy and address safety concerns.

Acknowledgements

The author would like to express his sincere gratitude to Professor Shinya Toyokuni, MD, PhD, Kyoto University (present location, Nagoya University), Japan, for supporting, guiding and supervising the discovery of Aberrant Methylation Mediated Regulation of TXNIP gene.

Ethical approval

No ethical approval is required for this study.

Declaration by authors

The authors' guidelines were used to generate the manuscript with the assistance of ChatGPT, an artificial intelligence program developed by OpenAI (which included the information mining, drafting and even for verification). However, the authors are responsible for the content and accuracy of the manuscript.

Data availability statement

The raw data are available in corresponding author and ready to submit when ask for it.

Informed consent statement

No informed consent was required to conduct the study.

Conflict of interest

The authors declare no conflict of interest.

Source of funding

This work received no funding from internal or external sources.

Author contributions

Khokon Kumar Dutta contributed to the design and writing of this review. The author critically reviewed the manuscript and agreed to submit final version of the article.

References

Bird A, Taggart M, Frommer M, Miller OJ and Macleod D, 1985. A fraction of the mouse genome that is derived from islands of nonmethylated, CpG-rich DNA. Cell, 40: 91–99. https://doi.org/10.1016/0092-8674(85)90312-5

Bird AP, 1986. CpG-rich islands and the function of DNA methylation. Nature, 321(6067): 209-213. https://doi.org/10.1038/321209a0

Braun CJ, Bruno PM, Horlbeck MA, Gilbert LA, Weissman JS and Hemann MT, 2016. Versatile in vivo regulation of tumor phenotypes by dCas9-mediated transcriptional perturbation. Proceedings of the National Academy of Sciences of the United States of America, 113(27): E3892-900. https://doi.org/10.1073/pnas.1600582113

Cano-Rodriguez D and Rots MG, 2016. Epigenetic editing: On the verge of reprogramming gene expression at will. Current Genetic Medicine Reports, 4(4): 170-179. https://doi.org/10.1007/s40142-016-0104-3

Chen C, Liao Y and Peng G, 2022. Connecting past and present: Single-cell lineage tracing. Protein Cell, 13(11): 790-807 https://doi.org/10.1007/s13238-022-00913-7

Choy JS, Wei S, Lee JY, Tan S, Chu S and Lee TH, 2010. DNA methylation increases nucleosome compaction and rigidity. Journal of the American Chemical Society, 132(6): 1782–1783. https://doi.org/10.1021/ja910264z

Deaton AM and Bird A, 2011. CpG islands and the regulation of transcription. Genes and Development, 25(10): 1010–1022. https://doi.org/10.1101/gad.2037511

Doudna JA and Charpentier E, 2014. Genome editing. The new frontier of genome engineering with CRISPR-Cas9. Science, 346(6213): 1259-1260. https://doi.org/10.1126/science.1258096

Dutta KK, Nishinaka Y, Masutani H, Akatsuka S, Aung TT, Shirase T, Lee WH, Yamada Y, Hiai H, Yodoi J and Toyokuni S, 2005. Two distinct mechanisms for loss of thioredoxin-binding protein-2 in oxidative stress-induced renal carcinogenesis. Laboratory Investigation, 85(6): 798-807. https://doi.org/10.1038/labinvest.3700280

Gardiner-Garden M and Frommer M, 1987. CpG islands in vertebrate genomes. Journal of Molecular Biology, 196(2): 261-282. https://doi.org/10.1016/0022-2836(87)90689-9

Gilbert LA, Larson MH, Morsut L, Liu Z, Brar GA, Torres SE, Stern-Ginossar N, Brandman O, Whitehead EH, Doudna JA, Lim WA, Weissman JS and LS Qi, 2014. CRISPR-mediated modular RNA-guided regulation of transcription in eukaryotes. Cell, 159(3): 647-661. https://doi.org/10.1016/j.cell.2013.06.044

Hanzawa N, Hashimoto K, Yuan X, Kawahori K, Tsujimoto K, Hamaguchi M, Tanaka T, Nagaoka Y, Nishina H, Morita S, Hatada I, Yamada T and Ogawa Y, 2020. Targeted DNA demethylation of the Fgf21 promoter by CRISPR/dCas9-mediated epigenome editing. Scientific Reports, 10: 5181. https://doi.org/10.1038/s41598-020-62035-6

Hilton IB, D’Ippolito AM, Vockley CM, Thakore PI, Crawford GE, Reddy TE and Gersbach CA, 2015. Epigenome editing by a CRISPR-Cas9-based acetyltransferase activates genes from promoters and enhancers. Nature Biotechnology, 33(5): 510-517. https://doi.org/10.1038/nbt.3199

Josipović G, Tadić V, Klasić M, Zanki V, Bečeheli I, Chung F, Ghantous A, Keser T, Madunić J, Bošković M, Lauc G, Herceg Z, Vojta A and Zoldoš V, 2019. Antagonistic and synergistic epigenetic modulation using orthologous CRISPR/dCas9-based modular system. Nucleic Acids Research, 47(18): 9637-9657. https://doi.org/10.1093/nar/gkz709

Kim MJ, Lee HJ, Choi MY, Kang SS, Kim YS, Shin JK and Choi WS, 2021. UHRF1 Induces Methylation of the TXNIP promoter and down-regulates gene expression in cervical cancer. Molecules and Cells, 31: 44(3): 146-159. https://doi.org/10.14348/molcells.2021.0001

Lee JY and Lee TH, 2012. Effects of DNA methylation on the structure of nucleosomes. Journal of the American Chemical Society, 134: 173-175. https://doi.org/10.1021/ja210273w

Lee JY, Lee J, Yue H and Lee TH, 2015. Dynamics of nucleosome assembly and effects of DNA methylation. Journal of Biological Chemistry, 290(7): 4291-303. https://doi.org/10.1074/jbc.M114.619213

Liu XS, Wu H, Ji X, Stelzer Y, Wu X, Czauderna S, Shu J, Dadon D, Young RA and Jaenisch R, 2016. Editing DNA methylation in the mammalian genome. Cell, 167: 233-247.e17. https://doi.org/10.1016/j.cell.2016.08.056

Liu XS, Wu H, Krzisch M, Wu X, Graef J, Muffat J, Hnisz D, Li CH, Yuan B, Xu C, Li Y, Vershkov D, Cacace A, Young RA and Jaenisch R, 2018. Rescue of fragile X syndrome neurons by DNA methylation editing of the FMR1 gene. Cell, 172(5): 979-992.e6. https://doi.org/10.1016/j.cell.2018.01.012

Maeda K, Fujii R, Yamada H, Munetsuna E, Yamazaki M, Ando Y, Mizuno G, Ishikawa H, Ohashi K, Tsuboi Y, Hattori Y, Ishihara Y, Hamajima N, Hashimoto S and Suzuki K, 2024. Association between DNA methylation levels of thioredoxin-interacting protein (TXNIP) and changes in glycemic traits: A longitudinal population-based study. Endocrine Journal, 71 (6): 593-601. https://doi.org/10.1507/endocrj.EJ23-0629

Maeder ML, Linder SJ, Cascio VM, Fu Y, Ho QH and Jounj JK, 2013. CRISPR RNA-guided activation of endogenous human genes. Nature Methods, 10(10): 977-979. https://doi.org/10.1038/nmeth.2598

Mali P, Yang L, Esvelt KM, Aach J, Guell M, DiCarlo JE, Norville JE and Church GM, 2013. RNA-guided human genome engineering via Cas9. Science, 339(6121): 823-826. https://doi.org/10.1126/science.1232033

McCarty NS, Graham AE, Studená L and Ledesma-Amaro R, 2020. Multiplexed CRISPR technologies for gene editing and transcriptional regulation. Nature Communications, 11: 1281. https://doi.org/10.1038/s41467-020-15053-x

Park H, Shin J, Kim Y, Saito T, Saido TC and Kim J, 2022. CRISPR/dCas9-Dnmt3a-mediated targeted DNA methylation of APP rescues brain pathology in a mouse model of Alzheimer’s disease. Translational Neurodegeneration, 11: 41. https://doi.org/10.1186/s40035-022-00314-0

Sapozhnikov DM and Szyf M, 2021. Unraveling the functional role of DNA demethylation at specific promoters by targeted steric blockage of DNA methyltransferase with CRISPR/dCas9. Nature communications, 12:5711. https://doi.org/10.1038/s41467-021-25991-9

Senapedis W, Gallagher KM, Figueroa E,  Farelli JD, Lyng R, Hodgson JG, O’Donnell CW, Newman JV, Pacaro M, Siecinski SK, Chen J and McCauley TG, 2024. Targeted transcriptional downregulation of MYC using epigenomic controllers demonstrates antitumor activity in hepatocellular carcinoma models. Nature Communication, 15: 7875. https://doi.org/10.1038/s41467-024-52202-y

Stepper P, Kungulovski G, Jurkowska RZ, Chandra T, Krueger F, Reinhardt R, Reik W, Jeltsch A and Jurkowski TP, 2017. Efficient targeted DNA methylation with chimeric dCas9–Dnmt3a–Dnmt3L methyltransferase. Nucleic Acids Research, 45(4): 1703–1713. https://doi.org/10.1093/nar/gkw1112

Swiech L, Heidenreich M, Banerjee A, Habib N, Li Y, Trombetta J, Sur M and Zhang F, 2015. In vivo interrogation of gene function in the mammalian brain using CRISPR-Cas9. Nature Biotechnology, 33: 102-6. https://doi.org/10.1038/nbt.3055

Tsai HH, Shen CY, Ho CC, Hsu SY, Tantoh DM, Nfor ON, Chiu SL, Chou YH and Liaw YP, 2022, Interaction between a diabetes-related methylation site (TXNIP cg19693031) and variant (GLUT1 rs841853) on fasting blood glucose levels among non-diabetics. Journal of Translational Medicine, 20: 87. https://doi.org/10.1186/s12967-022-03269-y

Vojta A, Dobrinić P, Tadić V, Bočkor L, Korać P, Julg B, Klasić M and Zoldoš V, 2016. Repurposing the CRISPR-Cas9 system for targeted DNA methylation. Nucleic Acids Research, 44(12): 5615–5628. https://doi.org/10.1093/nar/gkw159

Wu Y, Chen W, Zhao Y, Gu M, Gao Y, Ke Y, Wang L, Wang M, Zhang W, Chen Y, Huo W, Fu X, Li X, Zhang D, Qin P, Hu F, Liu Y, Sun X, Zhang M and Hu D, 2024. Visit to visit transition in TXNIP gene methylation and the risk of type 2 diabetes mellitus: a nested case-control study. Journal of human genetics 69(7): 311-319. https://doi.org/10.1038/s10038-024-01243-8

Xiang Y, Wang Z, Hui Q, Gwinn M, Vaccarino V and Sun YV, 2021. DNA methylation of TXNIP independently associated with inflammation and diabetes mellitus in twins. Twin Research and Human Genetics, 24(5): 273-280. https://doi.org/10.1017/thg.2021.42

Xu X, Tao Y, Gao X, Zhang L, Li X, Zou W, Ruan K, Wang F, Xu GL and Hu R, 2016. A CRISPR-based approach for targeted DNA demethylation. Cell Discovery, 2: 16009. https://doi.org/10.1038/celldisc.2016.9

Yoo J, Park S, Maffeo C, Ha T and Aksimentiev A 2021. DNA sequence and methylation prescribe the inside-out conformational dynamics and bending energetics of DNA minicircles. Nucleic Acids Research, 49: 11459–11475. https://doi.org/10.1093/nar/gkab967

Zhang D, Cheng C, Cao M, Wang T, Chen X, Zhao Y, Wang B, Ren Y, Liu D, Liu L, Chen X, Liu F, Zhou Q, Tian G, Li Q, Guo C, Li H, Wang J, Cheng R, Hu D and Zhang M, 2020. TXNIP hypomethylation and its interaction with obesity and hypertriglyceridemia increase type 2 diabetes mellitus risk: A nested case-control study. Journal of Diabetes, 12(7): 512-520. https://doi.org/10.1111/1753-0407.13021

Zhang P, Gao J, Wang X, Wen W, Yang H, Tian Y, Liu N, Wang Z, Liu H, Zhang Y and Tu Y, 2017. A novel indication of thioredoxin-interacting protein as a tumor suppressor gene in malignant glioma. Oncology Letters, 14(2): 2053-2058. https://doi.org/10.3892/ol.2017.6397

 

How to cite

Dutta KK 2025. CRISPR-dCas9-mediated CpG island editing: A potential game-changer for diabetes treatment. Journal of Bioscience and Environment Research, 2(1): 10-13. https://doi.org/10.69517/jber.2025.02.01.0003

CrossMark Update
CROSSMARK Color horizontal
Article Metrics

Table 2. Key in vivo applications of CRISPR-dCas9-mediated epigenetic editing.

Target/Application

Study

Key findings

Significance

Targeting tumor/cancer related  genes in cancer models

Braun et al. (2016)

Used CRISPR/dCas9 to activate and inactivate the cancer related genes in mice.

Demonstrated the potential to regulate the cancer related genes in vivo.

Manipulating memory-related genes in the brain

Liu et al. (2016)

A CRISPR/dCas9-TET1 fusion methylated CpG islands in the promoters of Bdnf in the adult mouse hippocampus. This increased expression of these genes, critical for memory formation and synaptic plasticity.

Showed that targeted demethylation can enhance gene expression in specific brain regions, suggesting potential treatments for neurodegenerative diseases or cognitive disorders.

Gene reactivation

Liu et al. (2018)

Used CRISPR/dCas9-TET1 to demethylate the FMR1 gene promoter in a Fragile X syndrome mouse model, leading to gene reactivation and partial phenotypic rescue.

Showcased the potential to treat genetic disorders characterized by epigenetic silencing.

Metabolism related gene targeting

Hanzawa et al. (2020)

Used CRISPR/dCas9- TET1CD and CRISPR/dCas9- SunTag to demethylate Fgf21 promoter both in vitro and in vivo.

Demonstrated the ability to edit metabolism related gene promoter through targeted epigenetic modifications.

Targeted repression of oncogenes in liver cancer

Senapedis et al. (2024)

CRISPR/dCas9-DNMT3A methylated CpG islands in the Myc promoter in a hepatocellular carcinoma mouse model. This reduced Myc expression, slowed tumor growth, and improved survival rates.

Demonstrated that targeted CpG methylation in oncogenes can suppress tumor growth, offering a potential therapeutic strategy for cancers with specific gene overexpression.

Epigenetic reversal of age-related memory decline

Swiech et al. (2015)

Used CRISPR/dCas9-TET1 to demethylate genes involved in synaptic plasticity in aged mice, improving cognitive function.

Showcased the potential to reverse age-related epigenetic changes and improve cognitive function.

Alzheimer’s disease

Park et al. (2022)

Reduced expression of full-length APP and C99 in the DG of mouse brain.

Demonstrated the potential of CRISPR-dCas9-mediated epigenetic editing to target specific genes involved in Alzheimer’s disease and reduce disease-related proteins.

 

Table 1. A timeline highlighting the key developments in the area of CRISPR-dCas9 Mediated CpG island methylation and demethylation.

Year

Key milestones and contributions

2013

CRISPR/Cas9 repurposed for targeted epigenetic modifications using dCas9 (Mali etal., 2013; Maeder et al., 2013).

2014

dCas9 fused with transcriptional regulators to control gene expression without DNA sequence alteration (Gilbert et al., 2014).

2015

CRISPR/dCas9 fused with DNMT3A for site-specific CpG methylation, silencing genes, particularly in cancer research (Hilton et al., 2015).

2016

Enhanced DNA methylation with dCas9 and multiple effectors, expanding applications in cancer research and stem cell differentiation (Xu et al., 2016).

2017

dCas9 fused with TET1 for site-specific CpG methylation, reactivating silenced genes (Stepper et al., 2017).

2018

Precise demethylation in endogenous genes, advancing research in neurodevelopmental disorders and cancer (Liu et al., 2018)

2019

Enhanced specificity with dual-function dCas9 constructs for bidirectional CpG methylation control (Josipović et al., 2019).

2020

Multiplexed control of multiple CpG islands, enabling studies of complex gene networks (McCarty et al., 2020).

2021

Development of tools for fine-tuned methylation/demethylation with therapeutic potential in diseases involving aberrant methylation (Sapozhnikov et al., 2021).

2022

Preclinical applications in neurological diseases and cancer; modulation of tumor suppressor genes using dCas9-methylation tools (Chen et al., 2022).

2023

Focus on safer in vivo systems for targeted methylation and demethylation, addressing off-target effects (Vojta et al., 2023).

 



[stm-calc id="1576"]