scholarly journals Tetraspanin 3, as a Novel Regulator of Intracellular β1 Integrin Recycling, Promotes Non-Small Cell Lung Cancer Cell Proliferation

Author(s):  
Hongbo Su ◽  
Yitong Xu ◽  
Yao Zhang ◽  
Xizi Jiang ◽  
Qingfu Zhang ◽  
...  

Abstract Background: The role of tetraspanins in cancer development has been widely reported. However, the expression and roles of tetraspanin 3 (TSPAN3) in solid tumors, including non-small cell lung cancer (NSCLC), have not yet been extensively investigated. In this study, we explored the role of TSPAN3 in NSCLC and its potential role in the trafficking of integrin β1, which is highly expressed in this type of cancer.Methods: The expression of TSPAN3 in NSCLC tissue and NSCLC cell lines was evaluated, and the correlation between TSPAN3 expression and disease progression was assessed. Furthermore, loss- and gain-of-expression studies were conducted to determine the biological function of TSPAN3 both in vivo and vitro. The regulatory role of TSPAN3 in β1 integrin expression and intracellular recycling was studied through western blot, RT-PCR, and immunofluorescence analyses.Results: TSPAN3 was found to be highly expressed in lung cancer cells and tissues. Moreover, high levels of TSPAN3 positively correlated with poor differentiation, lymph node involvement, advanced pathological tumor-node-metastasis stage, and poor prognosis in NSCLC patients. TSPAN3 showed the potential to promote the proliferation of NSCLC cells in vitro and in vivo. Specifically, TSPAN3 was found to interact with β1 integrin and Rab11a, thereby facilitating the sorting of β1 integrin into Rab11a endosomes and promoting β1 integrin recycling and upregulation. Conclusions: These findings reveal a novel role for TSPAN3 in the regulation of intracellular recycling of β1 integrin. Hence, TSPAN3 may represent a potentially valuable therapeutic target for NSCLC.

2021 ◽  
Vol 19 (1) ◽  
Author(s):  
Xiaoyang Liu ◽  
Yang Cheng ◽  
Yan Wang ◽  
Yinhong Zhang

Abstract Background Accumulating evidence demonstrated that circular RNAs (circRNAs) play pivotal regulatory roles in the pathology of cancers. Disclosing the roles and molecular mechanisms of circRNAs in tumorigenesis and development is essential to identify novel diagnostic and therapeutic targets. In this study, we explored the role of circVAPA in non-small-cell lung cancer (NSCLC) progression and its associated mechanism. Methods The expression level of RNA was analyzed by real-time quantitative polymerase chain reaction (RT-qPCR). Cell proliferation was assessed by MTT assay and colony-forming assay. Cell apoptosis was analyzed by flow cytometry. Cell migration and invasion were assessed by transwell assays. Dual-luciferase reporter, RNA pull-down, and RNA immunoprecipitation (RIP) assays were used to test the intermolecular interactions. The role of circVAPA was assessed in vivo. And xenograft tumor tissues were analyzed by immunohistochemistry (IHC) staining. Results CircVAPA expression was upregulated in NSCLC tissues and cell lines, and a high level of circVAPA was associated with a poor prognosis of NSCLC patients. CircVAPA silencing suppressed the proliferation, migration, and invasion and induced the apoptosis of NSCLC cells. CircVAPA served as a molecular sponge for microRNA-342-3p (miR-342-3p). miR-342-3p interference largely reversed circVAPA knockdown-mediated anti-tumor effects in NSCLC cells. Zinc finger E-box-binding homeobox 2 (ZEB2) was a target of miR-342-3p, and miR-342-3p overexpression suppressed the malignant behaviors of NSCLC cells largely by downregulating ZEB2. CircVAPA silence repressed xenograft tumor growth in vivo, and IHC assay confirmed that circVAPA silence restrained the proliferation and metastasis but induced the apoptosis of NSCLC cells in vivo. Conclusion CircVAPA contributes to the progression of NSCLC by binding to miR-342-3p to upregulate ZEB2. CircVAPA/miR-342-3p/ZEB2 axis might be a novel potential target for NSCLC treatment.


2020 ◽  
Author(s):  
Zhihong Zhang ◽  
Zhenxiu Shan ◽  
Rubin Chen ◽  
Xiaorong Peng ◽  
Bin Xu ◽  
...  

AbstractNon-small cell lung cancer (NSCLC) is a leading threat to human lives with high incidence and mortality. Circular RNAs (circRNAs) were reported to play important roles in human cancers. The purpose of this study was to investigate the role of circ_0005962 and explore the underlying functional mechanisms. The expression of circ_0005962, miR-382-5p and pyruvate dehydrogenase kinase 4 (PDK4) was detected by quantitative real-time polymerase chain reaction (qRT-PCR). Cell proliferation and cell apoptosis were assessed by cell counting kit-8 (CCK-8) assay and flow cytometry assay, respectively. The protein levels of Beclin 1, light chain3 (LC3-II/LC3-I), PDK4, Cleaved Caspase 3 (C-caspase 3) and proliferating cell nuclear antigen (PCNA) were examined using western blot analysis. Glycolysis was determined according to the levels of glucose consumption and lactate production. The interaction between miR-382-5p and circ_0005962 or PDK4 was predicted by the online tool CircInteractome or starbase and verified by dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay. Xenograft model was constructed to investigate the role of circ_0005962 in vivo. circ_0005962 expressed with a high level in NSCLC tissues and cells. Circ_0005962 knockdown inhibited proliferation, autophagy, and glycolysis but promoted apoptosis in NSCLC cells. MiR-382-5p was targeted by circ_0005962, and its inhibition reversed the role of circ_0005962 knockdown. Besides, PDK4, a target of miR-382-5p, was regulated by circ_0005962 through miR-382-5p, and its overexpression abolished the effects of miR-382-5p reintroduction. Circ_0005962 knockdown suppressed tumor growth in vivo. Circ_0005962 knockdown restrained cell proliferation, autophagy, and glycolysis but stimulated apoptosis through modulating the circ_0005962/miR-382-5p/PDK4 axis. Our study broadened the insights into understanding the mechanism of NSCLC progression.


2020 ◽  
Vol 18 (1) ◽  
Author(s):  
Hang Li ◽  
Jun Che ◽  
Mian Jiang ◽  
Ming Cui ◽  
Guoxing Feng ◽  
...  

Abstract Introduction Radioresistance is a major challenge in lung cancer radiotherapy, and new radiosensitizers are urgently needed. Estrogen receptor β (ERβ) is involved in the progression of non-small cell lung cancer (NSCLC), however, the role of ERβ in the response to radiotherapy in lung cancer remains elusive. In the present study, we investigated the mechanism underlying ERβ-mediated transcriptional activation and radioresistance of NSCLC cells. Methods Quantitative real-time PCR, western blot and immunohistochemistry were used to detect the expression of CLPTM1L, ERβ and other target genes. The mechanism of CLPTM1L in modulation of radiosensitivity was investigated by chromatin immunoprecipitation assay, luciferase reporter gene assay, immunofluorescence staining, confocal microscopy, coimmunoprecipitation and GST pull-down assays. The functional role of CLPTM1L was detected by function assays in vitro and in vivo. Results CLPTM1L expression was negatively correlated with the radiosensitivity of NSCLC cell lines, and irradiation upregulated CLPTM1L in radioresistant (A549) but not in radiosensitive (H460) NSCLC cells. Meanwhile, IR induced the translocation of CLPTM1L from the cytoplasm into the nucleus in NSCLC cells. Moreover, CLPTM1L induced radioresistance in NSCLC cells. iTRAQ-based analysis and cDNA microarray identified irradiation-related genes commonly targeted by CLPTM1L and ERβ, and CLPTM1L upregulated ERβ-induced genes CDC25A, c-Jun, and BCL2. Mechanistically, CLPTM1L coactivated ERβ by directly interacting with ERβ through the LXXLL NR (nuclear receptor)-binding motif. Functionally, ERβ silencing was sufficient to block CLPTM1L-enhanced radioresistance of NSCLC cells in vitro. CLPTM1L shRNA treatment in combination with irradiation significantly inhibited cancer cell growth in NSCLC xenograft tumors in vivo. Conclusions The present results indicate that CLPTM1L acts as a critical coactivator of ERβ to promote the transcription of its target genes and induce radioresistance of NSCLC cells, suggesting a new target for radiosensitization in NSCLC therapy.


2021 ◽  
pp. 1-9
Author(s):  
Li-Na Pan ◽  
Yun-Fang Ma ◽  
Jia-An Hu ◽  
Zhi-Hong Xu

Circular RNA (circRNA) has been shown to participate in various tumors, including lung cancer. In the present study, we explored the expression and functional relevance of hsa_circ_0003288 in human non-small cell lung cancer (NSCLC). We verified that hsa_circ_0003288 expression was upregulated in lung cancer tissues and cell lines. Overexpression of hsa_circ_0003288 dramatically promoted lung cancer cell proliferation, colony formation, inhibited apoptosis, and increased cell migration and invasion in vitro. Xenograft experiments showed that hsa_circ_0003288 overexpression accelerated tumor growth in vivo. Mechanistically, hsa_circ_0003288 negatively regulated miR-145 to exert the oncogenic role in lung cancer. Overexpression of miR-145 decreased cell proliferation, induced apoptosis, and suppressed migration and invasion in lung cancer. Additionally, miR-145 co-transfection abolished the oncogenic role of hsa_circ_0003288. Collectively, these findings identified a novel regulatory role of hsa_circ_0003288/miR-145 axis in the progression of NSCLC.


2020 ◽  
Vol 72 (2) ◽  
pp. 243-252
Author(s):  
Xin Zhang ◽  
Zhonghua Sun ◽  
Yuepei Zou

In this study, we aimed to elucidate the role of long non-coding RNA nuclear enriched abundant transcript 1 (lncRNA NEAT1) in non-small cell lung cancer (NSCLC). Quantitative real-time polymerase chain reaction (qRT-PCR) was employed to detect the abundance of NEAT1, microRNA-582-5p (miR-582-5p) and eukaryotic translation initiation factor 4 gamma 2 (EIF4G2). Proliferation, apoptosis, metastasis and glycolytic metabolism were determined by 3-(4,5-dimethylthiazol- 2-yl)-2,5-diphenyltetrazolium bromide (MTT) flow cytometry, transwell assays and fluorescence-based glucose and lactate assay kits. The targets of NEAT1 and miR-582-5p were predicted by the starBase website, and dual-luciferase reporter and RNA immunoprecipitation (RIP) assays were performed to verify these predictions. Western blot analysis was conducted to detect the protein expression of EIF4G2. A xenograft tumor model was built to clarify the role of NEAT1 in vivo. Results showed that NEAT1 interference inhibited proliferation, metastasis and glycolysis, and facilitated the apoptosis of NSCLC cells. MiR-582-5p was a functional target of NEAT1, and the biological influence of NEAT1 intervention on NSCLC cells was alleviated by transfection with anti-miR-582-5p. MiR-582-5p could bind to EIF4G2 messenger RNA (mRNA); it exerted its antitumor role in NSCLC cells by inhibiting EIF4G2. EIF4G2 was regulated by NEAT1/miR-582- 5p signaling. NEAT1 accelerated NSCLC tumor growth via the miR-582-5p/EIF4G2 axis in vivo. In conclusion, NEAT1 affected NSCLC by elevating their malignant potential via the miR-582-5p/EIF4G2 axis.


2021 ◽  
Author(s):  
wen yuan ◽  
Wenhui Zeng ◽  
Haiyan Tan ◽  
Muhammad Jamal ◽  
Tian Xie ◽  
...  

Abstract BackgroundCell Division Cycle Associated 7 (CDCA7) was first identified as a direct target gene of c-Myc and dysregulated in various types of human cancer. However, it has limited implication in non-small Cell Lung Cancer (NSCLC). We aimed to explore the critical role of CDCA7 in NSCLC.Methods In this study, we identified CDCA7 upregulation and association with the prognosis of NSCLC by integrating analysis of 3 Gene Expression Omnibus (GEO) databases. Real-time PCR and immunohistochemistry (IHC) were used to determine collected clinical NSCLC samples. Chi-square test was used to examine possible correlations between CDCA7 expression and clinicopathological factors. Univariate and multivariate Cox proportional hazards regression analysis were performed to determine whether CDCA7 is an independent risk factor for overall survival (OS). The effect of CDCA7 expression on proliferation, cell cycle and apoptosis ability of NSCLC cells was detected by cell counting kit-8 (CCK-8) and flow cytometry. CDCA7 stably knocking down cell line was established and Western blotting assay was applied to measure relevant protein expression. Xenograft models were used to examine the role of CDCA7 on tumorigenicity of NSCLC cells.Results Analysis of clinical samples confirmed the CDCA7 high expression in tumor tissues compared with adjacent non-tumor tissues and predicted shorter OS time. COX proportional risk model analysis showed that the expression levels of CDCA7 was independent prognostic factors. We observed that CDCA7 silencing efficiently affect the proliferation, apoptosis and cycle distribution of NSCLC cells in vitro. Further results demonstrated that the expression of CDCA7 in A549/DDP cells was significantly higher than that in A549 cells, CDCA7 silencing efficiently down regulation cisplatin sensitivity in A549/DDP cells. Importantly, the depletion of CDCA7 strongly reduced the tumorigenicity of NSCLC cells in vivo. Furthermore, depletion of CDCA7 expression markedly affected the expression of cell division protein kinase 6 (CDK6) and caspase7 both in vitro and in vivo. In vitro study, we showed that CDCA7 silencing promotes A549 apoptosis via extracellular regulated protein kinases (ERK) pathway.ConclusionHighly expressed CDCA7 plays a crucial role in the pathogenesis of NSCLC and might be a potential prognostic factor and therapeutic target in NSCLC.


Author(s):  
Lillian J. Eichner ◽  
Stephanie D. Curtis ◽  
Sonja N. Brun ◽  
Joshua T. Baumgart ◽  
Debbie S. Ross ◽  
...  

SummaryTranscriptional deregulation is a common feature of many cancers, which is often accompanied by changes in epigenetic controls. These findings have led to the development of therapeutic agents aimed at broad modulation and reprogramming of transcription in a variety of cancers. Histone Deacetylase 3, HDAC3, is one of the main targets of HDAC inhibitors currently in clinical development as cancer therapies, yet the in vivo role of HDAC3 in solid tumors is unknown. Here, we define the role of HDAC3 in two genetic engineered models of the most common subtypes of Kras-driven Non-Small Cell Lung Cancer (NSCLC), KrasG12D, STK11−/− (KL) and KrasG12D, p53−/− (KP), where we found that HDAC3 is strongly required for tumor growth of both genotypes in vivo. Transcriptional profiling and mechanistic studies revealed that HDAC3 represses p65 RelA/NF-kB-mediated induction of the Senescence Associated Secretory Program (SASP). Additionally, HDAC3 was found to cooperate with the lung cancer lineage transcription factor NKX2-1 to mediate expression of a common set of target genes. Leveraging observations about one HDAC3/NKX2-1 common target, FGFR1, we identified that an HDAC3-dependent transcriptional cassette becomes hyperactivated as Kras mutant cancer cells develop resistance to the MEK inhibitor Trametinib, and this can be rescued by treatment with the Class I HDAC inhibitor Entinostat. These unexpected findings reveal new roles for HDAC3 in proliferation control in tumors in vivo and identify specific therapeutic contexts for the utilization of HDAC3 inhibitors, whose ability to mechanistically induce SASP may be harnessed therapeutically.


PLoS ONE ◽  
2015 ◽  
Vol 10 (12) ◽  
pp. e0144825 ◽  
Author(s):  
Nibedita Chattopadhyay ◽  
Allison J. Berger ◽  
Erik Koenig ◽  
Bret Bannerman ◽  
James Garnsey ◽  
...  

2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Furui Zhang ◽  
Ruirui Cheng ◽  
Ping Li ◽  
Chunya Lu ◽  
Guojun Zhang

Abstract Background Non-small cell lung cancer (NSCLC) is a threat to human health. Circular RNAs (circRNAs) have been proved to function in NSCLC development. In this study, the role of circRNA hsa_circ_0010235 in NSCLC progression and the possible molecular mechanism were explored. Methods Expression of hsa_circ_0010235, miRNA (miR)-433-3p and TOR signaling pathway regulator-like (TIPRL) was examined by quantitative real-time PCR (qRT-PCR). Cell viability and clonogenicity were detected by cell counting kit-8 (CCK-8) assay and colony formation assay, respectively. Flow cytometry was performed to monitor cell apoptosis and cell cycle distribution. Western blot assay was employed to evaluate the protein levels of TIPRL, light chain 3 (LC3)-II/I and p62. Cell metastasis was assessed by Transwell and wound healing assays. The targeted relationship between miR-433-3p and hsa_circ_0010235 or TIPRL was confirmed by dual-luciferase reporter and RNA immunoprecipitation (RIP) assays. Furthermore, the role of hsa_circ_0010235 in vivo was investigated by xenograft assay. Results Hsa_circ_0010235 and TIPRL were highly expressed in NSCLC tissues and cells, while miR-433-3p was downregulated. Depletion of hsa_circ_0010235 or gain of miR-433-3p repressed proliferation and autophagy but promoted apoptosis in NSCLC cells. Hsa_circ_0010235 sponged miR-433-3p to upregulate TIPRL expression, so as to affect NSCLC development. Hsa_circ_0010235 knockdown also blocked tumor growth in vivo. Conclusion Hsa_circ_0010235 knockdown suppressed NSCLC progression by regulating miR-433-3p/TIPRL axis, affording a novel mechanism of NSCLC progression.


2021 ◽  
pp. 1-14
Author(s):  
Bin Zhang ◽  
Zhou Jin ◽  
Hao Zhang

BACKGROUND: The disorder of LINC01207 has a significant regulatory effect on cancers, nevertheless its role in non-small cell lung cancer (NSCLC) have not been illustrated. This study investigated the regulatory effect of LINC01207 on NSCLC and clarify its molecular mechanism. METHODS: Bioinformatics analysis was used to find the target lncRNA, miRNA and mRNA. LncBase and TargetScan databases predicted the relationship between LINC01207, miR-525-5p and ARHGAP11A. Dual-luciferase reporter gene assay and RNA binding protein immunoprecipitation assay were used to verify the binding relationship between genes. Fluorescence in situ hybridization assay was used to localize the expression of LINC01207 in NSCLC tissue. qRT-PCR and Western blot assays were used to measure the expression of LINC01207, miR-525-5p and ARHGAP11A. CCK-8 assay, Transwell assay and flow cytometry assay were used to detect NSCLC cell abilities. Mouse xenograft models further determined the effect of LINC01207 on the growth of NSCLC in vivo. RESULTS: LINC01207 was up-regulated in NSCLC tissue and cells, which was mainly localized in the cytoplasm. LINC01207 knockdown could inhibit the proliferation, migration and invasion of cancer cells and induce cell apoptosis. In addition, silencing LINC01207 could suppress tumor growth in vivo. LINC01207 could sponge and inhibit the expression of miR-525-5p in NSCLC cells, and inhibiting LINC01207 and miR-525-5p simultaneously could reverse the effect of miR-525-5p on the progression of NSCLC cells. Further study on downstream target genes showed that miR-525-5p could restrain the expression of ARHGAP11A, and then affect the progression of NSCLC. LINC01207 acting as a competing endogenous RNA (ceRNA) could regulate the expression of ARHGAP11A by competitively binding with miR-525-5p. CONCLUSION: LINC01207 regulates the progression of NSCLC by regulating the miR-525-5p/ARHGAP11A axis as a ceRNA and plays a carcinogenic role. In conclusion, our study elucidates the mechanism of LINC01207 regulating the progression of NSCLC, and provides a new idea for the diagnosis and treatment of NSCLC guided by lncRNA.


Sign in / Sign up

Export Citation Format

Share Document