scholarly journals Genome-scale CRISPR-Cas9 knockout screening in hepatocellular carcinoma with lenvatinib resistance

2021 ◽  
Vol 7 (1) ◽  
Author(s):  
Yonggang Lu ◽  
Haoming Shen ◽  
Wenjie Huang ◽  
Sha He ◽  
Jianlin Chen ◽  
...  

AbstractLenvatinib is the first target drug approved for advanced hepatocellular carcinoma (HCC). However, the development of drug resistance is common, and the mechanisms of lenvatinib resistance and resistant targets in HCC are poorly understood. By using CRISPR/Cas9 library screening, we screened out two key resistance genes, neurofibromin 1(NF1), and dual specificity phosphatase 9 (DUSP9), as critical drivers for lenvatinib resistance in HCC. With RNAi knockdown and CRISPR/Cas9 knockout models, we further clarified the mechanisms by which NF1 loss reactivates the PI3K/AKT and MAPK/ERK signaling pathways, while DUSP9 loss activates the MAPK/ERK signaling pathways, thereby inactivating FOXO3, followed by degradation of FOXO3, finally induced lenvatinib resistance. We also screened out trametinib, a small molecule pathway inhibitor for MEK, that can be used to reverse resistance induced by NF1 and DUSP9 loss in HCC cells. Trametinib was still able to halt HCC growth even when NF1 was knocked out in mice. Collectively, the findings indicate that NF1 and DUSP9 takes critical role in lenvatinib resistance and may be novel specific targets and predictive markers for lenvatinib resistance in HCC.

2019 ◽  
Vol 10 (1) ◽  
Author(s):  
Lai Wei ◽  
Derek Lee ◽  
Cheuk-Ting Law ◽  
Misty Shuo Zhang ◽  
Jialing Shen ◽  
...  

Abstract Sorafenib is the standard treatment for advanced hepatocellular carcinoma (HCC). However, the development of drug resistance is common. By using genome-wide CRISPR/Cas9 library screening, we identify phosphoglycerate dehydrogenase (PHGDH), the first committed enzyme in the serine synthesis pathway (SSP), as a critical driver for Sorafenib resistance. Sorafenib treatment activates SSP by inducing PHGDH expression. With RNAi knockdown and CRISPR/Cas9 knockout models, we show that inactivation of PHGDH paralyzes the SSP and reduce the production of αKG, serine, and NADPH. Concomitantly, inactivation of PHGDH elevates ROS level and induces HCC apoptosis upon Sorafenib treatment. More strikingly, treatment of PHGDH inhibitor NCT-503 works synergistically with Sorafenib to abolish HCC growth in vivo. Similar findings are also obtained in other FDA-approved tyrosine kinase inhibitors (TKIs), including Regorafenib or Lenvatinib. In summary, our results demonstrate that targeting PHGDH is an effective approach to overcome TKI drug resistance in HCC.


Author(s):  
Hui Li ◽  
Huicheng Chen ◽  
Haibin Wang ◽  
Yilong Dong ◽  
Min Yin ◽  
...  

Hepatocellular carcinoma (HCC) is a disease with poor prognosis rates and ineffective therapeutic options. Previous studies have reported the involvement of mitogen-inducible gene 6 (MIG-6) as a negative regulator in tumor formation. MicroRNAs (miRNAs) play crucial roles in the development of different types of cancer. However, the underlying mechanisms of miRNAs in HCC are poorly understood. This study was aimed to investigate the role of miR-374a in HCC and its role in the regulation of expression of MIG-6. The results showed that MIG-6 overexpression significantly inhibited cell viability of HepG2 cells after 4 days posttransfection. Moreover, MIG-6 was a direct target of miR-374a, and the expression of MIG-6 was remarkably downregulated by the overexpression of miR-374a in HepG2 cells. Furthermore, we found that overexpression of miR-374a promoted cell viability; however, the protective effect was abolished by MIG-6 overexpression. In addition, overexpression of miR-374a activated the EGFR and AKT/ERK signaling pathways by regulation of MIG-6. Our findings suggest that miR-374a could promote cell viability by targeting MIG-6 and activating the EGFR and AKT/ERK signaling pathways. These data provide a promising therapeutic strategy for HCC treatment.


Cancers ◽  
2021 ◽  
Vol 13 (2) ◽  
pp. 243
Author(s):  
Yan Liao ◽  
Yue Yang ◽  
Di Pan ◽  
Youxiang Ding ◽  
Heng Zhang ◽  
...  

As one of the most common malignancies worldwide, Hepatocellular carcinoma (HCC) has been treated by Sorafenib, which is the first approved target drug by FDA for advanced HCC. However, drug resistance is one of the obstacles to its application. As a typical characteristic of most solid tumors, hypoxia has become a key cause of resistance to chemotherapy and radiotherapy. It is important to elucidate the underlying mechanisms of Sorafenib resistance under hypoxia. In this study, the morphological changes of hepatocellular carcinoma cells were observed by Live Cell Imaging System and Transmission Electron Microscope; Sorafenib was found to induce necroptosis in liver cancer. Under hypoxia, the distribution of necroptosis related proteins was changed, which contributed to Sorafenib resistance. HSP90α binds with the necrosome complex and promotes chaperone-mediated autophagy (CMA) degradation, which leads necroptosis blocking and results in Sorafenib resistance. The patient-derived tumor xenograft (PDX) model has been established to investigate the potential therapeutic strategies to overcome Sorafenib resistance. 17-AAG inhibited HSP90α and presented obvious reversal effects of Sorafenib resistance in vivo and in vitro. All the results emphasized that HSP90α plays a critical role in Sorafenib resistance under hypoxia and 17-AAG combined with Sorafenib is a promising therapy for hepatocellular carcinoma.


2021 ◽  
Author(s):  
Sha She ◽  
Min Yang ◽  
Shi Ying Li ◽  
Huai Dong Hu ◽  
YiXuan Yang ◽  
...  

Abstract Background C-reactive protein (CRP), an acute phase protein, has been increasingly implicated in various tumors, and the role of CRP is positively correlated with invasion and metastasis in hepatocellular carcinoma cells. However, the mechanism of CRP affecting HCC progression remains poorly investigated. The present study investigated the role of CRP in HCC and the underlying mechanisms. Methods In the current study, CRP overexpression and suppression expression experiments were used to evaluate the effect of CRP on malignant biological behavior of liver cancer cells in vitro. Then iTRAQ-mass spectrometry analysis was used to identify CRP co-immunoprecipitation complexes. Detecting the interaction between CRP and Eph receptor B3 (EphB3) by co-precipitation. Moreover, immunofluorescence colocalization and co-precipitation, and Western Blot, in vivo model were applied to study the molecular mechanism of CRP affecting the development of Hepatocellular Carcinoma. Results We first found that CRP was significantly upregulated in HCC tissues and HCC cells, the expression level correlated with the metastatic ability of HCC cells. Knockdown of CRP significantly suppresses migration and invasion capacity in HCC cells. Through a proteomic analysis of CRP co-immunoprecipitation complexes, the EphB3 was identified as a new CRP interactor. Then we found that the expression and functions of EphB3 were consistent with CRP in HCC. In addition, co-immunoprecipitation and immunofluorescence assays suggested that EphB3 was able to interact with MAPK/ERK to activate MAPK/ERK signaling pathways. Furthermore, we showed that CRP can induce the phosphorylation of MAPK/ERK by binding EphB3. CRP also significantly stimulated MMP-9 expression, mainly by activating HIF-1α via the MAPK/ERK pathways. Conclusions Our findings showed that CRP increased HCC cells migration and invasion by binding EphB3 to activate MAPK/ERK signaling pathways. It suggested that CRP may become a prognostic factor and a potential therapeutic target for liver cancer.


2021 ◽  
Vol 11 ◽  
Author(s):  
Xin Lu ◽  
Panpan Ma ◽  
Lingyu Kong ◽  
Xi Wang ◽  
Yaqi Wang ◽  
...  

Our previous studies have proved that 17β-hydroxysteroid dehydrogenase 4 (HSD17B4) is a novel proliferation-promoting protein. The overexpression of HSD17B4 promotes hepatocellular carcinoma (HCC) cell proliferation. Vitamin K2 (VK2), a fat-soluble vitamin, has the function of promoting coagulation and can inhibit the progression of liver cancer. A previous study demonstrated that VK2 could bind to HSD17B4 in HepG2 cells. However, the mechanism of VK2 in inhibiting HCC cell proliferation is not clear. In this study, we investigate whether VK2 can inhibit the proliferation of HCC cell induced by HSD17B4 and the possible mechanism. We detected the effect of VK2 on HSD17B4-induced HCC cell proliferation, and the activation of STAT3, AKT, and MEK/ERK signaling pathways. We measured the effect of HSD17B4 on the growth of transplanted tumor and the inhibitory effect of VK2. Our results indicated that VK2 directly binds to HSD17B4, but does not affect the expression of HSD17B4, to inhibit the proliferation of HCC cells by inhibiting the activation of Akt and MEK/ERK signaling pathways, leading to decreased STAT3 activation. VK2 also inhibited the growth of HSD17B4-induced transplanted tumors. These findings provide a theoretical and experimental basis for possible future prevention and treatment of HCC using VK2.


2009 ◽  
Vol 47 (01) ◽  
Author(s):  
H Schulze-Bergkamen ◽  
A Weinmann ◽  
M Wörns ◽  
PR Spies ◽  
A Teufel ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document