Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2377 2022-11-25 18:18:18 |
2 Format correction Meta information modification 2377 2022-11-28 03:44:31 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Almeida, R.S.;  Wisnieski, F.;  Karia, B.T.R.;  Smith, M.A.C. Application of CRISPR/CAS9 in Basic Gastric Cancer Research. Encyclopedia. Available online: https://encyclopedia.pub/entry/36636 (accessed on 27 July 2024).
Almeida RS,  Wisnieski F,  Karia BTR,  Smith MAC. Application of CRISPR/CAS9 in Basic Gastric Cancer Research. Encyclopedia. Available at: https://encyclopedia.pub/entry/36636. Accessed July 27, 2024.
Almeida, Renata Sanches, Fernanda Wisnieski, Bruno Takao Real Karia, Marilia Arruda Cardoso Smith. "Application of CRISPR/CAS9 in Basic Gastric Cancer Research" Encyclopedia, https://encyclopedia.pub/entry/36636 (accessed July 27, 2024).
Almeida, R.S.,  Wisnieski, F.,  Karia, B.T.R., & Smith, M.A.C. (2022, November 25). Application of CRISPR/CAS9 in Basic Gastric Cancer Research. In Encyclopedia. https://encyclopedia.pub/entry/36636
Almeida, Renata Sanches, et al. "Application of CRISPR/CAS9 in Basic Gastric Cancer Research." Encyclopedia. Web. 25 November, 2022.
Application of CRISPR/CAS9 in Basic Gastric Cancer Research
Edit

Gastric cancer is the subject of clinical and basic studies due to its high incidence and mortality rates worldwide. Due to the diagnosis occurring in advanced stages and the classic treatment methodologies such as gastrectomy and chemotherapy, they are extremely aggressive and limit the quality of life of these patients. CRISPR/Cas9 is a tool that allows gene editing and has been used to explore the functions of genes related to gastric cancer, in addition to being used in the treatment of this neoplasm, greatly increasing our understanding of cancer genomics.

gastric cancer CRISPR gene editing

1. Introduction

CRISPR/CAs9 is an extremely versatile tool that can be used in the study and understanding of cancer development mechanisms and in the diagnosis and treatment of several diseases, including gastric cancer (GC) [1].
This technique has helped studies in several areas, such as for gastric cancer, where in 2015 the study by Gannom and colleagues used the CRISPR/Cas9 technique for the first time in GC [2]. This pioneering study evaluated the knockdown effect of dual-specificity mitogen-activated protein kinase 1 (MAP2K1) and the relationship of MEK-inhibitory drugs with cancer cell lines, including gastric cancer. This study was important to evidence the RAS/MAPK activation driven by MAP2K1 depletion in gastric cancer.
CRISPR is an important tool that [3] helps us in a simple and practical way to understand the functions of genes that are still poorly described in GC. A collection of relevant GC studies that used CRISPR/Cas9 may be found in Table 1.
Table 1. Studies performed in gastric cells using the CRISPR/Cas9 methodology.

2. Cell Viability and Proliferation

Needless to say, the ability of cells to proliferate rapidly and uncontrollably is a key factor in GC. Therefore, understanding these biological mechanisms and genes is extremely important for the identification of therapeutic targets. The CRISPR/Cas9-mediated knockout of apoptosis-associated speck-like protein containing a C-terminal caspase recruitment domain (ASC) blocked IL18 and augmented apoptosis in human GC cells, helping to reveal a novel pro-tumorigenic ASC/IL18 signaling axis in GC cell survival and a candidate therapeutic target in this disease [14].
Contributing to the findings of mir-21-induced loss of 15-hydroxyprostaglandin dehydrogenase (15-PGDH) in early GC and its phenotypic consequences, another study suppressed 15-PGDH in GC cells using CRISPR/Cas9, which demonstrated increased GC cell proliferation [10]. Therefore, the study suggested that maintaining the 15-PGDH enzyme activity could be a strategy for preventing GC, especially in tubular adenocarcinoma.

3. Cell Cycle Control

The study of genes that influence the cell cycle allows us to identify important cellular processes that differentiate a tumor cell from a healthy cell. Aspects such as unrestrained growth without the influence of external factors, the loss of programmed cell death capacity, and also the repression of tumor suppressor genes, among other properties, are important to identify specific biomarkers and potential therapeutic targets in GC.
To demonstrate the involvement of DNA hypermethylation on the regulation of the tumor suppressor gene REPRIMO and the TP53-dependent G2 arrest mediator homolog (RPRM) in GC, Lai and collaborators [17] used the CRISPR/Cas9 technology to first knockdown DNA methyltransferases (DNMTs). The knockdown of DNMT3A and DNMT3B resulted in a significant increase in RPRM mRNA by decreasing the RPRM promoter methylation in GC cells, suggesting an inverse correlation between DNMT functions and RPRM gene expression. To confirm the tumor suppression role of RPRM, the authors generated RPRM-deficient GC cell lines using CRISPR/Cas9, which were further inoculated in mice. The loss of RPRM enhanced the tumor formation in the in vivo model, confirming the role of RPRM as a tumor suppressor gene in GC. This study provided information regarding the role of RPRM and its regulatory methylation mechanism in GC development with potential application as a therapeutic target.
To regain the tumor suppression gene function, a study reactivated the RPRM expression using CRISPR/dCas9 linked to VP64 and SAM effector domains in GC cell lines, showing a hypermethylated RPMR promoter and expressing very low basal levels of the gene [21]. As a result, a marked reduction in GC proliferation was observed. This result outlined the advantage of this combinatorial epigenome editing approach to reactivate highly methylated tumor suppressor genes as a promising therapy for GC.
Another promising tumor suppressor was described in the study by Hu and collaborators [11], with the knockout of the Morf4-family-associated protein 1 (MRFAP1) gene using CRISPR/Cas9 from the CG human cell lines. The researchers observed that the knockout promoted an interaction of this MLN4924 subtract with suppressed tumor proteins, such as p27, which promoted a decrease in vitality, increased cell cycle arrest, and apoptosis, data that are important to relate the gene–subtract interaction to favor improvements in cancer treatment. The study by Liu et al. [15] used the CRISPR/Cas9-mediated knockout of promoter of CDKN1A antisense DNA damage-activated RNA (PANDAR) and evaluated its effects on the phenotype of GC cell lines. The knockout of PANDAR suppressed the proliferative activity and colony formation in the GC cell line. It was also observed by flow cytometry that the PANDAR knockout blocks the progression of the cell cycle at the G1/S checkpoint. Through this unique pattern of transcriptional modification, PANDAR remarkably facilitated the proliferation of cancer cells, the formation of clones, and resistance to chemotherapy. Another study developed by Zhang and colleagues [12] used the CRISPR/Cas9-mediated knockout of LIM homeobox transcription factor 1 α (LMX1A) in GC cell lines to identify LMX1A as a primary target of miR-9. The authors that demonstrated the knockout of LMX1A increased the cell viability and cell proliferation, reinforcing the role of LMX1A as a tumor suppressor in GC.

4. Invasion and Migration

The basic mechanism of metastasis development involves several important characteristics, such as the ability of cells to detach themselves from the primary tumor site and migrate to colonize more distant sites in the organism [24]. Due to the lack of early diagnosis and effective therapy options for GC, a better understanding of the mechanisms involved in the metastatic process of GC is necessary. In this sense, Zhou and colleagues [8] used the CRISPR/Cas9-mediated knockout of gastric cancer metastasis-associated long non-coding RNA (GMAN) IncRNA and evaluated the phenotype of GC cells. The knockout of the GMAN lncRNA delayed the invasive activity of these cells. This study also showed the overlapping relationship between GMAN and ephrin A1, in which the GMAN knockout induced an ephrin A1 expression reduction. Assessing the effect of ephrin A1 knockout in GC cell lines, it reduced the ability for invasion and metastasis in in vivo experiments. Another important study was developed by Zhu and colleagues [19], in which a GWAS analysis was used to find low-frequency genetic variants associated with the risk of GC. The authors found two genes that contained a variant associated with GC risk, SPOC-domain-containing 1 (SPOCD1) and Butyrophilin subfamily 3 member A2 (BTN3A), and eliminated these genes using CRISPR/Cas9 in GC cell lines. The knockout of SPOCD1 and BTN3A2 inhibited cell proliferation and colony formation. To investigate the effects of SPOCD1 and BTN3A2 knockout on migration and invasion, the authors performed xenograft assays and observed a tumor growth reduction in a rat model associated with SPOCD1 knockout. On the other hand, BTN3A2 was suggested as a susceptibility gene, although no significant changes in the xenograft model were observed. Zhang et al. [7] demonstrated that the CRISPR/Cas9-mediated deletion of SAM-pointed domain-containing Ets transcription factor (PDEF) inhibited the apoptosis, colony formation, migration, and invasion of GC cell lines. These results confirm the involvement of PDEF in the different stages of GC development. The study by Araújo et al. [9] demonstrated that the CRISPR/Cas9-mediated elimination of the Piwi-like protein 1 (PIWIL1) gene decreased GC cell migration and invasion abilities, demonstrating the oncogenic role of PIWIL1. On the other hand, Chen Wei’s study [23] demonstrated that the CRISPR/Ca9-mediated knockout of Somatostatin (SST) significantly promoted the migration and invasion capabilities of GC cell lines. These data are essential for characterizing SST as a potential tumor suppressor in GC.
Another interesting study used the CRISPR/Cas9-mediated knockin of ephrin type-B receptor 2 (EphB2) and evaluated its functions as an independent prognostic marker in patients with GC [22]. The results indicated that the activation of EphB2 in GC cells increased the malignant properties of GC cells, reducing the adhesion but accelerating the migration and invasion capabilities. These results indicate that EphB2 plays a pro-tumor role in GC and has therapeutic potential to be used in this neoplasia.

5. Tumorigenesis Models

Helicobacter pylori is a Gram-negative spiral bacterium that is present in 58% of the global population [25]. Most individuals infected with H. pylori are asymptomatic, and the presence of this bacterium increases the risk of developing ulcers and gastric adenocarcinoma [26]. An interesting study was carried out by Hu and collaborators [8], who sought to demonstrate the mechanisms in which vitamin D3 can assist in the defense of the host by promoting an autophagic reaction in the fight against H. pylori. The results showed that there is a new pathogenic mechanism that H. pylori can survive by hiding inside the autophagosomes in the GC cells by using the CRISPR/Cas9-mediated knockout of protein disulfide-isomerase A3 (PDIA3). From this study, a new vitamin D3 signaling pathway activates the PDIA3-STAT3-MCOLN3-Ca 2+ axis to reactivate the lysosome.
The molecular mechanism by which H. pylori induces peptic ulcers or gastritis cancer is not understood, but it probably involves a combination of host genetic predisposition and bacterial virulence factors (e.g., VacA and CagA proteins) [25]. The vacuolating cytotoxin (VacA) is responsible for several cellular responses, such as cell vacuolization, as well as other processes such as autophagy and necrosis [27]. Foegeding and collaborators [6] inhibited autophagy by using the CRISPR/Cas9-mediated knockout of autophagy-related 16-like 1 (ATG16L1) in HeLa cells. The results showed increased VacA levels or increased vacuolization compared with the control and that the VacA degradation is independent of the autophagic activity. CagA is a virulence factor used for the detection of H. pylori and is considered an important risk factor for severe gastric diseases, including GC. Zhao and coauthors [13] evaluated the effects of integrin receptors and the function of cell adhesion molecule 1 receptors (CEACAM) through the cag-type IV secretion system (cag-T4SS) on the CagA translocation process through a multiple knockout of CEACAM receptors in the GC cell line. The results showed that neither the direct interaction of the components of cag-T4SS with the integrins nor any signaling event mediated by the integrin is necessary for the translocation of CagA. Furthermore, the CRISPR/Cas9 mediated the deletion of miR-30a in H. pylori infected mice, the knockout mice demonstrated that genetic editing did not affect the growth and development of the mice, and little effect was observed on the H. pylori colonization rates of the mice. Increased incidence rates of chronic gastritis, chronic atrophic gastritis, atypical hyperplasia, and other precancerous lesions and manifestations of adenocarcinoma in the antral or gastric mucosa of rats have also been reported. These data demonstrate that miR-30a plays the role of a tumor suppressor in GC [18].

6. Chemotherapy Response

From the development of tumor cells to create resistance to chemotherapeutic-induced cell death, the CRISPR tool has been used to discover new therapeutic targets and drugs for the treatment of GC. Wang and colleagues [16] used CRISPR/Cas9 to mediate the knockout of the gasdermin E (GSDME) gene in a GC cell line to assess the effect of 5-fluoracil on the induction of pyroptosis in these cells. The authors found that the GSDME deficiency changed the pyroptosis induced by 5-FU to apoptosis, characterized by shrinkage, fragmentation in apoptotic bodies, and cell death without lysis. The study by Cui and collaborators [20] used the CRISPR/Cas9-mediated knockout of the legumain (AEP) gene to assess the proliferative capacity of these cells in the presence of different chemotherapeutic agents. This gene has previously been shown to be an oncogene related to invasiveness and metastasis in GC. The authors demonstrated that AEP knockout GC cells caused significantly decreased proliferation after treatment with 5-FU, paclitaxel, docetaxel, and T-DM1. These data demonstrate that AEP is a potential therapeutic target for GC.

References

  1. Zhang, H.; Qin, C.; An, C.; Zheng, X.; Wen, S.; Chen, W.; Liu, X.; Lv, Z.; Yang, P.; Xu, W.; et al. Application of the CRISPR/Cas9-based gene editing technique in basic research, diagnosis, and therapy of cancer. Mol. Cancer 2021, 20, 126.
  2. Gannon, H.S.; Kaplan, N.; Tsherniak, A.; Vazquez, F.; Weir, B.A.; Hahn, W.C.; Meyerson, M. Identification of an "Exceptional Responder" Cell Line to MEK1 Inhibition: Clinical Implications for MEK-Targeted Therapy. Mol. Cancer Res. 2016, 14, 207–215.
  3. Wang, S.-W.; Gao, C.; Zheng, Y.-M.; Yi, L.; Lu, J.-C.; Huang, X.-Y.; Cai, J.-B.; Zhang, P.-F.; Cui, Y.-H.; Ke, A.-W. Current applications and future perspective of CRISPR/Cas9 gene editing in cancer. Mol. Cancer 2022, 21, 57.
  4. Chen, J.; Bell, J.; Lau, B.T.; Whittaker, T.; Stapleton, D.; Ji, H.P. A functional CRISPR/Cas9 screen identifies kinases that modulate FGFR inhibitor response in gastric cancer. Oncogenesis 2019, 8, 33.
  5. Hu, W.; Zhang, L.; Li, M.X.; Shen, J.; Liu, X.D.; Xiao, Z.G.; Wu, D.L.; Ho, I.H.T.; Wu, J.C.Y.; Cheung, C.K.Y.; et al. Vitamin D3 activates the autolysosomal degradation function against Helicobacter pylori through the PDIA3 receptor in gastric epithelial cells. Autophagy 2019, 15, 707–725.
  6. Foegeding, N.J.; Raghunathan, K.; Campbell, A.M.; Kim, S.W.; Lau, K.S.; Kenworthy, A.K.; Cover, T.L.; Ohi, M.D. Intracellular Degradation of Helicobacter pylori VacA Toxin as a Determinant of Gastric Epithelial Cell Viability. Infect. Immun. 2019, 87, e00783-18.
  7. Zhang, Y.-Q.; Pei, J.-H.; Shi, S.-S.; Guo, X.-S.; Cui, G.-Y.; Li, Y.-F.; Zhang, H.-P.; Hu, W.-Q. CRISPR/Cas9-mediated knockout of the PDEF gene inhibits migration and invasion of human gastric cancer AGS cells. Biomed. Pharmacother. 2018, 111, 76–85.
  8. Zhuo, W.; Liu, Y.; Li, S.; Guo, D.; Sun, Q.; Jin, J.; Rao, X.; Li, M.; Sun, M.; Jiang, M.; et al. Long Noncoding RNA GMAN, Up-regulated in Gastric Cancer Tissues, Is Associated with Metastasis in Patients and Promotes Translation of Ephrin A1 by Competitively Binding GMAN-AS. Gastroenterology 2019, 156, 676–691.e11.
  9. Araújo, T.; Khayat, A.; Quintana, L.; Calcagno, D.; Mourão, R.; Modesto, A.; Paiva, J.; Lima, A.; Moreira, F.; Oliveira, E.; et al. Piwi like RNA-mediated gene silencing 1 gene as a possible major player in gastric cancer. World J. Gastroenterol. 2018, 24, 5338–5350.
  10. Park, Y.S.; Lee, J.H.; Jung, D.-B.; Kim, H.-B.; Jung, J.-H.; Pak, S.; Ryu, Y.-M.; Park, H.J.; Park, Y.-Y.; Jung, H.-Y.; et al. MicroRNA-21 induces loss of 15-hydroxyprostaglandin dehydrogenase in early gastric tubular adenocarcinoma. Sci. Rep. 2018, 8, 17717.
  11. Hu, L.; Bai, Z.G.; Ma, X.M.; Bai, N.; Zhang, Z.T. MRFAP1 plays a protective role in neddylation inhibitor MLN4924-mediated gastric cancer cell death. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 8273–8280.
  12. Zhang, X.; Qian, Y.; Li, F.; Bei, S.; Li, M.; Feng, L. microRNA-9 selectively targets LMX1A to promote gastric cancer cell progression. Biochem. Biophys. Res. Commun. 2018, 505, 405–412.
  13. Zhao, Q.; Busch, B.; Jiménez-Soto, L.F.; Ishikawa-Ankerhold, H.; Massberg, S.; Terradot, L.; Fischer, W.; Haas, R. Integrin but not CEACAM receptors are dispensable for Helicobacter pylori CagA translocation. PLoS Pathog. 2018, 14, e1007359.
  14. Deswaerte, V.; Nguyen, P.; West, A.; Browning, A.F.; Yu, L.; Ruwanpura, S.M.; Balic, J.; Livis, T.; Girard, C.; Preaudet, A.; et al. Inflammasome Adaptor ASC Suppresses Apoptosis of Gastric Cancer Cells by an IL18-Mediated Inflammation-Independent Mechanism. Cancer Res. 2018, 78, 1293–1307.
  15. Liu, J.; Ben, Q.; Lu, E.; He, X.; Yang, X.; Ma, J.; Zhang, W.; Wang, Z.; Liu, T.; Zhang, J.; et al. Long noncoding RNA PANDAR blocks CDKN1A gene transcription by competitive interaction with p53 protein in gastric cancer. Cell Death Dis. 2018, 9, 168.
  16. Wang, Y.; Yin, B.; Li, D.; Wang, G.; Han, X.; Sun, X. GSDME mediates caspase-3-dependent pyroptosis in gastric cancer. Biochem. Biophys. Res. Commun. 2018, 495, 1418–1425.
  17. Lai, J.; Wang, H.; Luo, Q.; Huang, S.; Lin, S.; Zheng, Y.; Chen, Q. The relationship between DNA methylation and Reprimo gene expression in gastric cancer cells. Oncotarget 2017, 8, 108610–108623.
  18. Liu, X.; Ji, Q.; Zhang, C.; Liu, X.; Liu, Y.; Liu, N.; Sui, H.; Zhou, L.; Wang, S.; Lihong, Z. miR-30a acts as a tumor suppressor by double-targeting COX-2 and BCL9 in H. pylori gastric cancer models. Sci. Rep. 2017, 7, 7113.
  19. Zhu, M.; Yan, C.; Ren, C.; Huang, X.; Zhu, X.; Gu, H.; Wang, M.; Wang, S.; Gao, Y.; Ji, Y.; et al. Exome Array Analysis Identifies Variants in SPOCD1 and BTN3A2 That Affect Risk for Gastric Cancer. Gastroenterology 2017, 152, 2011–2021.
  20. Cui, Y.; Li, Q.; Li, H.; Wang, Y.; Wang, H.; Chen, W.; Zhang, S.; Cao, J.; Liu, T. Asparaginyl endopeptidase improves the resistance of microtubule-targeting drugs in gastric cancer through IQGAP1 modulating the EGFR/JNK/ERK signaling pathway. OncoTargets Ther. 2017, 10, 627–643.
  21. Garcia-Bloj, B.; Moses, C.; Sgro, A.; Plani-Lam, J.; Arooj, M.; Duffy, C.; Thiruvengadam, S.; Sorolla, A.; Rashwan, R.; Mancera, R.L.; et al. Waking up dormant tumor suppressor genes with zinc fingers, TALEs and the CRISPR/dCas9 system. Oncotarget 2016, 7, 60535–60554.
  22. Yin, J.; Li, Z.; Ye, L.; Birkin, E.; Li, L.; Xu, R.; Chen, G.; Ji, J.; Zhang, Z.; Jiang, W.; et al. EphB2 represents an independent prognostic marker in patients with gastric cancer and promotes tumour cell aggressiveness. J. Cancer 2020, 11, 2778–2787.
  23. Chen, W.; Ding, R.; Tang, J.; Li, H.; Chen, C.; Zhang, Y.; Zhang, Q.; Zhu, X. Knocking Out SST Gene of BGC823 Gastric Cancer Cell by CRISPR/Cas9 Enhances Migration, Invasion and Expression of SEMA5A and KLF2. Cancer Manag. Res. 2020, 12, 1313–1321.
  24. Machlowska, J.; Baj, J.; Sitarz, M.; Maciejewski, R.; Sitarz, R. Gastric Cancer: Epidemiology, Risk Factors, Classification, Genomic Characteristics and Treatment Strategies. Int. J. Mol. Sci. 2020, 21, 4012.
  25. Wang, F.; Meng, W.; Wang, B.; Qiao, L. Helicobacter pylori-induced gastric inflammation and gastric cancer. Cancer Lett. 2014, 345, 196–202.
  26. Hooi, J.K.Y.; Lai, W.Y.; Ng, W.K.; Suen, M.M.Y.; Underwood, F.E.; Tanyingoh, D.; Malfertheiner, P.; Graham, D.Y.; Wong, V.W.S.; Wu, J.C.Y.; et al. Global Prevalence of Helicobacter pylori Infection: Systematic Review and Meta-Analysis. Gastroenterology 2017, 153, 420–429.
  27. Foegeding, N.J.; Caston, R.R.; McClain, M.S.; Ohi, M.D.; Cover, T.L. An Overview of Helicobacter pylori VacA Toxin Biology. oxins 2016, 8, 173.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , ,
View Times: 459
Revisions: 2 times (View History)
Update Date: 28 Nov 2022
1000/1000
Video Production Service