kras protein
Recently Published Documents


TOTAL DOCUMENTS

39
(FIVE YEARS 28)

H-INDEX

8
(FIVE YEARS 3)

2021 ◽  
Vol 20 (1) ◽  
Author(s):  
Gongmin Zhu ◽  
Lijiao Pei ◽  
Hongwei Xia ◽  
Qiulin Tang ◽  
Feng Bi

AbstractColorectal cancer (CRC) is a heterogeneous disease at the cellular and molecular levels. Kirsten rat sarcoma (KRAS) is a commonly mutated oncogene in CRC, with mutations in approximately 40% of all CRC cases; its mutations result in constitutive activation of the KRAS protein, which acts as a molecular switch to persistently stimulate downstream signaling pathways, including cell proliferation and survival, thereby leading to tumorigenesis. Patients whose CRC harbors KRAS mutations have a dismal prognosis. Currently, KRAS mutation testing is a routine clinical practice before treating metastatic cases, and the approaches developed to detect KRAS mutations have exhibited favorable sensitivity and accuracy. Due to the presence of KRAS mutations, this group of CRC patients requires more precise therapies. However, KRAS was historically thought to be an undruggable target until the development of KRASG12C allele-specific inhibitors. These promising inhibitors may provide novel strategies to treat KRAS-mutant CRC. Here, we provide an overview of the role of KRAS in the prognosis, diagnosis and treatment of CRC.


2021 ◽  
Author(s):  
Fiona E Hood ◽  
Yasmina M Sahraoui ◽  
Rosalind E Jenkins ◽  
Ian A Prior

Activating mutations of Ras genes are often observed in cancer. The protein products of the three Ras genes are almost identical. However, for reasons that remain unclear, KRAS is far more frequently mutated than the other Ras isoforms in cancer and RASopathies. We have quantified HRAS, NRAS, KRAS4A and KRAS4B protein abundance across a large panel of cell lines and healthy tissues. We observe consistent patterns of KRAS>NRAS>>HRAS protein expression in cells that correlate with the rank order of Ras mutation frequencies in cancer. Our data provide support for the model of a sweet-spot of Ras dosage mediating isoform-specific contributions to cancer and development. However, they challenge the notion that rare codons mechanistically underpin the predominance of KRAS mutant cancers. Finally, direct measurement of mutant versus wildtype KRAS protein abundance revealed a frequent imbalance that may suggest additional non-gene duplication mechanisms for optimizing oncogenic Ras dosage.


Cancers ◽  
2021 ◽  
Vol 13 (20) ◽  
pp. 5081
Author(s):  
Yuta Adachi ◽  
Ryo Kimura ◽  
Kentaro Hirade ◽  
Hiromichi Ebi

Activating mutations in KRAS are present in 25% of human cancers. When mutated, the KRAS protein becomes constitutively active, stimulating various effector pathways and leading to the deregulation of key cellular processes, including the suppression of apoptosis and enhancement of proliferation. Furthermore, mutant KRAS also promotes metabolic deregulation and alterations in the tumor microenvironment. However, some KRAS mutant cancer cells become independent of KRAS for their survival by activating diverse bypass networks that maintain essential survival signaling originally governed by mutant KRAS. The proposed inducers of KRAS independency are the activation of YAP1 and/or RSK-mTOR pathways and co-mutations in SKT11 (LKB1), KEAP1, and NFE2L2 (NRF2) genes. Metabolic reprogramming, such as increased glutaminolysis, is also associated with KRAS autonomy. The presence or absence of KRAS dependency is related to the heterogeneity of KRAS mutant cancers. Epithelial-to-mesenchymal transition (EMT) in tumor cells is also a characteristic phenotype of KRAS independency. Translationally, this loss of dependence is a cause of primary and acquired resistance to mutant KRAS-specific inhibitors. While KRAS-dependent tumors can be treated with mutant KRAS inhibitor monotherapy, for KRAS-independent tumors, we need an improved understanding of activated bypass signaling pathways towards leveraging vulnerabilities, and advancing therapeutic options for this patient subset.


2021 ◽  
Author(s):  
RUIHONG GONG ◽  
Minting Chen ◽  
Chunhua Huang Huang ◽  
Hoi Leong Xavier Wong ◽  
Hiu Yee Kwan ◽  
...  

Abstract BackgroundKRAS mutation is one of the dominant gene mutations in colorectal cancer (CRC). Up to present, targeting KRAS for CRC treatment remains a clinical challenge. WNT974 (LGK974) is a porcupine inhibitor that interferes Wnt signaling pathway. Artesunate (ART) is a water-soluble semi-synthetic derivative of artemisinin.MethodsThe synergistic effect of ART and WNT974 combination in reducing CRC cell viability was determined by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. RT-PCR was utilized for the mRNA levels of KRAS, CUL7, ANAPC2, UBE2M, RNF123, SYVN1, or β-TrCP. Western blot assay was utilized for the protein levels of KRAS, ANAPC2, β-TrCP, or GSK-3β. Xenograft mouse model assay was performed for the anti-CRC effect of combination of ART and WNT974 in vivo. IHC assay was utilized for the levels of KRAS, β-TrCP, or GSK-3β in tumor tissues. Results Our study shows that the combination of WNT974 and ART exhibits synergistic effect in reducing CRC growth. The combination treatment significantly reduces KRAS protein level and activity in CRC cells. Interestingly, the combination treatment increases E3 ligases ANAPC2 expression. Our data show that overexpression of ANAPC2 significantly reduces KRAS protein levels, which is reversed by MG132. Knockdown of ANAPC2 in CRC abolishes the combination treatment-reduce KRAS expression. Besides, the treatment also increases the expressions of GSK-3β and E3 ligase β-TrCP that is known to degrade GSK-3β-phosphorylated KRAS protein. Knockdown of β-TrCP- and inhibition of GSK-3β abolish the combination treatment-induce KRAS ubiquitination and reduction in expression.ConclusionsOur data clearly show that the combination treatment significantly enhances KRAS protein degradation via the ubiquitination ubiquitin–proteasome pathway, which is also demonstrated in xenograft mouse model. The study provides strong scientific evidence for the development of the combination of WNT974 and ART as KRAS-targeting therapeutics for CRC treatment.


2021 ◽  
Author(s):  
Rui-Hong Gong ◽  
Minting Chen ◽  
Chunhua Huang ◽  
Hoi Leong Xavier Wong ◽  
Hiu Yee Kwan ◽  
...  

Abstract BackgroundKRAS mutation is one of the dominant gene mutations in colorectal cancer (CRC). Up to present, targeting KRAS for CRC treatment remains a clinical challenge. WNT974 (LGK974) is a porcupine inhibitor that interferes Wnt signaling pathway. Artesunate (ART) is a water-soluble semi-synthetic derivative of artemisinin.MethodsThe synergistic effect of ART and WNT974 combination in reducing CRC cell viability was determined by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. RT-PCR was utilized for the mRNA levels of KRAS, CUL7, ANAPC2, UBE2M, RNF123, SYVN1, or β-TrCP. Western blot assay was utilized for the protein levels of KRAS, ANAPC2, β-TrCP, or GSK-3β. Xenograft mouse model assay was performed for the anti-CRC effect of combination of ART and WNT974 in vivo. IHC assay was utilized for the levels of KRAS, β-TrCP, or GSK-3β in tumor tissues.Results Our study shows that the combination of WNT974 and ART exhibits synergistic effect in reducing CRC growth. The combination treatment significantly reduces KRAS protein level and activity in CRC cells. Interestingly, the combination treatment increases E3 ligases ANAPC2 expression. Our data show that overexpression of ANAPC2 significantly reduces KRAS protein levels, which is reversed by MG132. Knockdown of ANAPC2 in CRC abolishes the combination treatment-reduce KRAS expression. Besides, the treatment also increases the expressions of GSK-3β and E3 ligase β-TrCP that is known to degrade GSK-3β-phosphorylated KRAS protein. Knockdown of β-TrCP- and inhibition of GSK-3β abolish the combination treatment-induce KRAS ubiquitination and reduction in expression.ConclusionsOur data clearly show that the combination treatment significantly enhances KRAS protein degradation via the ubiquitination ubiquitin–proteasome pathway, which is also demonstrated in xenograft mouse model. The study provides strong scientific evidence for the development of the combination of WNT974 and ART as KRAS-targeting therapeutics for CRC treatment.


Human Cell ◽  
2021 ◽  
Author(s):  
Sonja Marinović ◽  
Anita Škrtić ◽  
Tina Catela Ivković ◽  
Mirko Poljak ◽  
Sanja Kapitanović

2021 ◽  
Vol 5 (1) ◽  
pp. 28-33
Author(s):  
Merza H. Homady ◽  
Tanya S. Salih ◽  
Mariamm M. Al-Jubori ◽  
Mustafa D. Younus

The study inc1uded 50 tissue blocks embedded in paraffin wax (16 females and 34 males), obtained from a patients group with (CRC) colorectal cancer , as well as 35 Tissue blocks that were embedded in paraffin wax from norma1 co1on (ulcerative co1itis) as controls. A relatively few oncogenes and most prominently tumor-suppressing genes, Kirastien rat sarcoma virus (KRAS), and P53 genes have been mutated into a significant part of CRCs, and a broad collection of mutated genes has been defined in CRC subsets. Current findings showed very significant differences between patients and control subjects in the p53 positive rate (P<0.001). TP53 Pro/Pro genotype positivity was higher in the contro1 group I than in the patient group I and this was a significant difference (Pi<0.001) with an odd ratio of less than one. The genotype Pro/Pro was considered to be protective against colorectal carcinoma preventively fractured 0.767. The positive rate of p53 Arg/Arg genotype in patients was more frequent and statistically significant (P <0.01), because the odd ratio was more than one. The genotype Arg/Arg would be considered a colorectal carcinoma risk factor. We conclude that p53 over expression is used as an indicator of p53 mutation (as identified by immuno-historic chemistry) and KRAS protein expression was negatively impaired for all the patients in the current study.


Author(s):  
Costa V ◽  
◽  
El Achkar VNR ◽  
Ribeiro MP ◽  
Tristao SS ◽  
...  

The aim of this study is to investigate the role of RASSF1A and KRAS protein immunoexpression in Oral Leukoplakia with Epithelial Dysplasia (OLD) and in Oral Squamous Cell Carcinoma (OSCC). Immunohistochemical staining for RASSF1A and KRAS was performed and a semiquantitative analysis was applied to samples of the Control Group (CG, n=20), OLD (n=39), and OSCC (n=100). No significant difference was observed between RASSF1A immunoexpression and OLP and OSCC groups (p>0.05). KRAS expression was higher in OSCC than in OLP and CG (p<0.05). No association was observed between RASSF1A or KRAS expression and alcohol/tobacco use or clinicopathological features (p>0.05) in the OSCC group. Also, patients with OSCC who presented KRAS overexpression had a worse disease-free survival rate (p=0.04). RASSF1A expression was similar in OLD and OSCC groups, suggesting that it plays a critical role in the early stage of OSCC. KRAS expression was higher in OSCC when compared with normal and dysplastic tissues, showing that KRAS expression increases with malignant progression.


2021 ◽  
Author(s):  
Chengyao Chiang ◽  
Tian Xiao ◽  
Songqing Fan ◽  
Hongmei Zheng ◽  
Shuaihu Li ◽  
...  

AbstractOncogenic KRAS mutations are considered to be a key driver for initiation and progression in non-small-cell lung cancer (NSCLC). However, how post-translational modifications (PTMs) of KRAS, especially methylation, modify KRAS activity and downstream signals remain largely unclear. Here, we showed that SET domain containing histone lysine methyltransferase 7 (SETD7) interacts with KRAS and methylates KRAS at lysine 182 and 184. SETD7-mediated methylation of KRAS led to degradation of KRAS and attenuation of the RAS/MEK/ERK signaling cascade, endowing SETD7 with a potent tumor-suppressive role in NSCLC, both in vitro and in vivo. Mechanistically, RABGEF1, a ubiquitin E3 ligase of KRAS, was recruited and promoted KRAS degradation in a K182/K184 methylation-dependent manner. Notably, low SETD7 or RABGEF1 expression was associated with poor prognosis in lung adenocarcinoma patients. Taken together, our results establish a novel connection between lysine methylation and KRAS protein stability, in addition to elucidating a tumor-suppressive function of SETD7 that operates via modulation of oncogenic RAS signaling.


Biology ◽  
2021 ◽  
Vol 10 (1) ◽  
pp. 58
Author(s):  
Shu-Kee Eng ◽  
Ilma Ruzni Imtiaz ◽  
Bey-Hing Goh ◽  
Long Chiau Ming ◽  
Ya-Chee Lim ◽  
...  

Exosomes are cell-derived nanovesicles, and lately, cancer-derived exosomes have been reported to carry KRAS protein, which contributes to the malignancy of many cancers. In this study, farnesylthiosalicylic acid (FTS) was used to inhibit the activities of mutated KRAS in colon cancer SW480 cells to discover the potential link between KRAS activities and cancer-derived exosomes. We observed that FTS inhibits KRAS activity in SW480 cells, but promotes their exosome production. When the exosomal proteins of SW480 cells were profiled, a total of 435 proteins were identified with 16 of them showing significant changes (greater than or equal to two-fold) in response to FTS treatment. Protein network analysis suggests KRAS inhibition may trigger stress in the cells. In addition, a high level of acetyl-coA synthetase family member 4 protein which plays an important role in colon cancer survival was identified in the exosomes secreted by FTS-treated SW480 cells. The uptake of these exosomes suppresses the growth of some cell types, but in general exosomes from FTS-treated cells enhance the recipient cell survival when compared to that of untreated cells. Together our findings suggest that FTS may trigger stress in SW480 cells, and induce more exosomes secretion as the survival messenger to mitigate the impact of KRAS inhibition in colon cancer cells.


Sign in / Sign up

Export Citation Format

Share Document