scholarly journals ZIP10 is a negative determinant for anti-tumor effect of mannose in thyroid cancer by activating phosphate mannose isomerase

Author(s):  
Sharui Ma ◽  
Na Wang ◽  
Rui Liu ◽  
Rui Zhang ◽  
Hui Dang ◽  
...  

Abstract Background Mannose, a natural hexose existing in daily food, has been demonstrated to preferentially inhibit the progression of tumors with low expression of phosphate mannose isomerase (PMI). However, its function in thyroid cancer still remains elusive. Methods MTT, colony formation and flow cytometry assays were performed to determine the response of thyroid cancer cells to mannose. Meanwhile, mouse models of subcutaneous xenograft and primary papillary thyroid cancer were established to determine in vivo anti-tumor activity of mannose. The underlying mechanism of mannose selectively killing thyroid cancer cells was clarified by a series of molecular and biochemical experiments. Results Our data demonstrated that mannose selectively suppressed the growth of thyroid cancer cells, and found that enzyme activity of PMI rather than its protein expression was negatively associated with the response of thyroid cancer cells to mannose. Besides, our data showed that zinc ion (Zn2+) chelator TPEN clearly increased the response of mannose-insensitive cells to mannose by inhibiting enzyme activity of PMI, while Zn2+ supplement could effectively reverse this effect. Further studies found that the expression of zinc transport protein ZIP10, which transport Zn2+ from extracellular area into cells, was negatively related to the response of thyroid cancer cells to mannose. Knocking down ZIP10 in mannose-insensitive cells significantly inhibited in vitro and in vivo growth of these cells by decreasing intracellular Zn2+ concentration and enzyme activity of PMI. Moreover, ectopic expression of ZIP10 in mannose-sensitive cells decrease their cellular response to mannose. Mechanistically, mannose exerted its anti-tumor effect by inhibiting cellular glycolysis; however, this effect was highly dependent on expression status of ZIP10. Conclusion The present study demonstrate that mannose selectively kills thyroid cancer cells dependent on enzyme activity of PMI rather than its expression, and provide a mechanistic rationale for exploring clinical use of mannose in thyroid cancer therapy.

2016 ◽  
Vol 2016 ◽  
pp. 1-8 ◽  
Author(s):  
Jun Zhou ◽  
Li-Li He ◽  
Xiao-Fei Ding ◽  
Qiu-Qi Yuan ◽  
Jian-Xin Zhang ◽  
...  

Background.mTOR signaling would be a promising target for thyroid cancer therapy. However, in clinical trials, objective response rate with mTOR inhibitor monotherapy in most cancer types was modest. A new focus on development of combinatorial strategies with rapalogs is increasing.Objective.Investigating the combinatorial antitumor effect of rapamycin andβ-elemene in follicular thyroid cancer cells.Methods.MTT assay was used to determine the FTC-133 cell proliferation after culturing with rapamycin and/orβ-elemene. To analyze their combinatorial effect, immunoblotting was performed to analyze the activation status of AKT. Moreover,β-elemene attenuated rapamycin-induced immunosuppression was tested in mice.Results.Combination of rapamycin andβ-elemene exerted significant synergistic antiproliferative effects in FTC-133 cell linesin vitro, based on inhibiting the AKT feedback activation induced by rapamycin.In vivo, theβ-elemene could attenuate rapamycin-induced immunosuppression via reversing imbalance of Treg/Th17, with the underlying mechanism needed to be declared.Conclusions. We demonstrate that the novel combination of mTOR inhibitor withβ-elemene synergistically attenuates tumor cell growth in follicular thyroid cancer, which requires additional preclinical validation.


2021 ◽  
Vol 10 ◽  
Author(s):  
Xiaoli Liu ◽  
Qingfeng Fu ◽  
Xuehai Bian ◽  
Yantao Fu ◽  
Jingwei Xin ◽  
...  

The principal issue derived from thyroid cancer is its high propensity to metastasize to the lymph node. Aberrant exprssion of long non-coding RNAs have been extensively reported to be significantly correlated with lymphatic metastasis of thyroid cancer. However, the clinical significance and functional role of lncRNA-MAPK8IP1P2 in lymphatic metastasis of thyroid cancer remain unclear. Here, we reported that MAPK8IP1P2 was downregulated in thyroid cancer tissues with lymphatic metastasis. Upregulating MAPK8IP1P2 inhibited, while silencing MAPK8IP1P2 enhanced anoikis resistance in vitro and lymphatic metastasis of thyroid cancer cells in vivo. Mechanistically, MAPK8IP1P2 activated Hippo signaling by sponging miR-146b-3p to disrupt the inhibitory effect of miR-146b-3p on NF2, RASSF1, and RASSF5 expression, which further inhibited anoikis resistance and lymphatic metastasis in thyroid cancer. Importantly, miR-146b-3p mimics reversed the inhibitory effect of MAPK8IP1P2 overexpression on anoikis resistance of thyroid cancer cells. In conclusion, our findings suggest that MAPK8IP1P2 may serve as a potential biomarker to predict lymphatic metastasis in thyroid cancer, or a potential therapeutic target in lymphatic metastatic thyroid cancer.


2021 ◽  
Vol 5 (2) ◽  
pp. 143-154
Author(s):  
Le Xiong ◽  
Xiao-Min Lin ◽  
Jun-Hua Nie ◽  
Hai-Shan Ye ◽  
Jia Liu

2021 ◽  
Author(s):  
Shuwang Peng ◽  
Luyang Chen ◽  
Zhengtai Yuan ◽  
Shanshan Duan

Abstract Background: Long non-coding RNAs (lncRNAs) possess pivotal roles in human cancers, including thyroid cancer. In this study, we sought to elucidate the precise action of MIR31HG in the functional behaviors of thyroid cancer cells.Methods: MIR31HG, microRNA (miR)-761 and mitogen-activated protein kinase 1 (MAPK1) were quantified by quantitative real-time PCR (qRT-PCR) or immunoblotting analysis. Cell viability, proliferation, apoptosis, invasion, and migration abilities were evaluated by MTS, 5-Ethynyl-2’-Deoxyuridine (EdU), flow cytometry, transwell and wound-healing assays, respectively. Dual-luciferase reporter assays were performed to validate the relationship between miR-761 and MIR31HG or MAPK1. Mouse xenografts were formed to assess the effect of MIR31HG on tumor growth.Results: MIR31HG expression was at high levels in thyroid cancer. Suppression of MIR31HG impeded cell proliferation, invasion, and migration, as well as promoted cell apoptosis in vitro, and diminished the growth of xenograft tumors in vivo. Mechanistically, MIR31HG targeted and regulated miR-761. Moreover, the effects of MIR31HG suppression was partly dependent on increased abundance of miR-761. MAPK1 was established as a direct and functional target of miR-761. Furthermore, MIR31HG involved the modulation of MAPK1 expression through competitively binding to miR-761 by the shared binding sequence.Conclusion: Our findings demonstrate the workings of an undescribed regulatory network, in which MIR31HG targets miR-761 to regulate MAPK1 expression, leading to the alteration of the functional behaviors of thyroid cancer cells.


Sign in / Sign up

Export Citation Format

Share Document