1
|
Matboli M, Gadallah SH, Rashed WM, Hasanin AH, Essawy N, Ghanem HM, Eissa S. mRNA-miRNA-lncRNA Regulatory Network in Nonalcoholic Fatty Liver Disease. Int J Mol Sci 2021; 22:ijms22136770. [PMID: 34202571 PMCID: PMC8269036 DOI: 10.3390/ijms22136770] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 12/13/2022] Open
Abstract
AIM we aimed to construct a bioinformatics-based co-regulatory network of mRNAs and non coding RNAs (ncRNAs), which is implicated in the pathogenesis of non-alcoholic fatty liver disease (NAFLD), followed by its validation in a NAFLD animal model. MATERIALS AND METHODS The mRNAs-miRNAs-lncRNAs regulatory network involved in NAFLD was retrieved and constructed utilizing bioinformatics tools. Then, we validated this network using an NAFLD animal model, high sucrose and high fat diet (HSHF)-fed rats. Finally, the expression level of the network players was assessed in the liver tissues using reverse transcriptase real-time polymerase chain reaction. RESULTS in-silico constructed network revealed six mRNAs (YAP1, FOXA2, AMOTL2, TEAD2, SMAD4 and NF2), two miRNAs (miR-650 and miR-1205), and two lncRNAs (RPARP-AS1 and SRD5A3-AS1) that play important roles as a co-regulatory network in NAFLD pathogenesis. Moreover, the expression level of these constructed network-players was significantly different between NAFLD and normal control. Conclusion and future perspectives: this study provides new insight into the molecular mechanism of NAFLD pathogenesis and valuable clues for the potential use of the constructed RNA network in effective diagnostic or management strategies of NAFLD.
Collapse
Affiliation(s)
- Marwa Matboli
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Ain Shams University, Cairo 11382, Egypt
- Correspondence: (M.M.); (S.E.)
| | - Shaimaa H. Gadallah
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo 11382, Egypt; (S.H.G.); (H.M.G.)
| | - Wafaa M. Rashed
- Department of Research, Children’s Cancer Hospital-57357, Cairo 11382, Egypt;
| | - Amany Helmy Hasanin
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo 11382, Egypt;
| | - Nada Essawy
- Institut Pasteur, CEDEX 15, 75724 Paris, France;
| | - Hala M. Ghanem
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo 11382, Egypt; (S.H.G.); (H.M.G.)
| | - Sanaa Eissa
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Ain Shams University, Cairo 11382, Egypt
- Correspondence: (M.M.); (S.E.)
| |
Collapse
|
2
|
Yuan G, Xie H, Wei T, Zhu D, Zhang C, Yang Y. Diagnostic potential of extracellular vesicle-associated microRNA-10b and tumor markers for lung adenocarcinoma. Oncol Lett 2021; 22:614. [PMID: 34257722 PMCID: PMC8243083 DOI: 10.3892/ol.2021.12875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 05/21/2021] [Indexed: 11/05/2022] Open
Abstract
MicroRNAs (miRNAs/miRs) in extracellular vesicles (EVs) are potential diagnostic markers. The purpose of the present study was to investigate potential EV miRNA biomarkers for lung adenocarcinoma (LUAD). Potential miRNAs were identified by searching public databases and verified by examining clinical samples. The diagnostic value of EV-associated miR-10b, plasma miR-10b and tumor markers (TMs), including α-fetoprotein (AFP), neuron-specific enolase, carcinoembryonic antigen (CEA), cytokeratin 19 fragment 21-1 (CYFRA211), pro-gastrin-releasing-peptide, carbohydrate antigen (CA)125, CA153, CA199 and CA724, was evaluated via receiver operating characteristic curve analysis. By searching the Gene Expression Omnibus and The Cancer Genome Atlas databases, miR-10b was identified as a potential biomarker. The analysis of clinical samples suggested that EV-associated miR-10b from plasma was significantly differentially expressed between LUAD and control samples. EV-associated miR-10b could function as a diagnostic marker for LUAD, with an AUC of 0.998, which was higher than the AUCs for TMs such as AFP, CEA, CYFRA211, CA125, CA153, CA199, CA724, pro-gastrin-releasing-peptide and neuron-specific enolase. In conclusion, EV-associated miR-10b may be a potential diagnostic biomarker for LUAD that is superior to plasma miR-10b and TMs.
Collapse
Affiliation(s)
- Guangda Yuan
- Department of Thoracic Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215000, P.R. China
| | - Hongya Xie
- Department of Thoracic Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215000, P.R. China
| | - Tengteng Wei
- Department of Thoracic Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215000, P.R. China
| | - Donglin Zhu
- Department of Thoracic Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215000, P.R. China
| | - Chuanyu Zhang
- Department of Thoracic Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215000, P.R. China
| | - Yong Yang
- Department of Thoracic Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215000, P.R. China
| |
Collapse
|
3
|
Xu Q, Xu Z. miR-196b-5p Promotes Proliferation, Migration and Invasion of Lung Adenocarcinoma Cells via Targeting RSPO2. Cancer Manag Res 2021; 12:13393-13402. [PMID: 33402849 PMCID: PMC7778444 DOI: 10.2147/cmar.s274171] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 11/25/2020] [Indexed: 12/24/2022] Open
Abstract
Objective To explore the biological role of miR-196b-5p/RSPO2 in the occurrence and development of lung adenocarcinoma (LUAD) and to provide a basis for finding new therapeutic targets for LUAD. Methods Differentially expressed genes were analyzed based on LUAD microarray, and the target gene of the target miRNA was predicted. qRT-PCR was used to detect the expression levels of miR-196b-5p and RSPO2 mRNA in normal human bronchial epithelial cell line BEAS-2B and LUAD cell lines A549, NCI-H1792 and NCI-H226. Western blot was used to evaluate protein expression. Cell proliferative, migratory and invasive abilities were detected by CCK-8 and transwell assays. Dual-luciferase assay was conducted to verify the targeting relationship between miR-196b-5p and RSPO2. Results The results of qRT-PCR showed that miR-196b-5p was significantly highly expressed in LUAD cells, and the expression level of its downstream target gene RSPO2 was significantly decreased. The results of CCK-8 and transwell assays exhibited that miR-196b-5p promoted proliferation, migration and invasion of LUAD cells, while RSPO2 inhibited the malignant progression of LUAD cells. Dual-luciferase assay confirmed the targeted binding relationship between miR-196b-5p and RSPO2. Overexpression of RSPO2 partially reversed the promotion of miR-196b-5p on proliferation, migration and invasion of LUAD cells. Conclusion miR-196b-5p promoted proliferation, migration and invasion of LUAD cells by targeting and down-regulating RSPO2, which provided ideas for searching new targets for the diagnosis and treatment of LUAD.
Collapse
Affiliation(s)
- Qian Xu
- Department of Oncology Medicine, Fujian Medical University Union Hospital, Fuzhou, 350001, People's Republic of China
| | - Zhenwu Xu
- Department of Thoracic Medical Oncology, Fujian Provincial Cancer Hospital, The Affiliated Hospital of Fujian Medical University, Fuzhou, 350014, People's Republic of China
| |
Collapse
|
4
|
Genome-Wide Characterization of RNA Editing Sites in Primary Gastric Adenocarcinoma through RNA-seq Data Analysis. Int J Genomics 2020; 2020:6493963. [PMID: 33415135 PMCID: PMC7768588 DOI: 10.1155/2020/6493963] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/28/2020] [Accepted: 12/07/2020] [Indexed: 12/15/2022] Open
Abstract
RNA editing is a posttranscriptional nucleotide modification in humans. Of the various types of RNA editing, the adenosine to inosine substitution is the most widespread in higher eukaryotes, which is mediated by the ADAR family enzymes. Inosine is recognized by the biological machinery as guanosine; therefore, editing could have substantial functional effects throughout the genome. RNA editing could contribute to cancer either by exclusive editing of tumor suppressor/promoting genes or by introducing transcriptomic diversity to promote cancer progression. Here, we provided a comprehensive overview of the RNA editing sites in gastric adenocarcinoma and highlighted some of their possible contributions to gastric cancer. RNA-seq data corresponding to 8 gastric adenocarcinoma and their paired nontumor counterparts were retrieved from the GEO database. After preprocessing and variant calling steps, a stringent filtering pipeline was employed to distinguish potential RNA editing sites from SNPs. The identified potential editing sites were annotated and compared with those in the DARNED database. Totally, 12362 high-confidence adenosine to inosine RNA editing sites were detected across all samples. Of these, 12105 and 257 were known and novel editing events, respectively. These editing sites were unevenly distributed across genomic regions, and nearly half of them were located in 3′UTR. Our results revealed that 4868 editing sites were common in both normal and cancer tissues. From the remaining sites, 3985 and 3509 were exclusive to normal and cancer tissues, respectively. Further analysis revealed a significant number of differentially edited events among these sites, which were located in protein coding genes and microRNAs. Given the distinct pattern of RNA editing in gastric adenocarcinoma and adjacent normal tissue, edited sites have the potential to serve as the diagnostic biomarkers and therapeutic targets in gastric cancer.
Collapse
|
5
|
Li Y, Song S, Pizzi MP, Han G, Scott AW, Jin J, Xu Y, Wang Y, Huo L, Ma L, Vellano C, Luo X, MacLeod R, Wang L, Wang Z, Ajani JA. LncRNA PVT1 Is a Poor Prognosticator and Can Be Targeted by PVT1 Antisense Oligos in Gastric Adenocarcinoma. Cancers (Basel) 2020; 12:cancers12102995. [PMID: 33076512 PMCID: PMC7602573 DOI: 10.3390/cancers12102995] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 10/09/2020] [Accepted: 10/13/2020] [Indexed: 12/14/2022] Open
Abstract
Gastric adenocarcinoma (GAC) is inherently resistant or becomes resistant to therapy, leading to a poor prognosis. Mounting evidence suggests that lncRNAs can be used as predictive markers and therapeutic targets in the right context. In this study, we determined the role of lncRNA-PVT1 in GAC along with the value of inhibition of PVT1 using antisense oligos (ASOs). RNA scope in situ hybridization was used to analyze PVT1 expression in tumor tissue microarrays (TMAs) of GAC and paired normal tissues from 792 patients. Functional experiments, including colony formation and invasion assays, were performed to evaluate the effects of PVT1 ASO inhibition of PVT1 in vitro; patient-derived xenograft models were used to evaluate the anti-tumor effects of PVT1 ASOs in vivo. LncRNA-PVT1 was upregulated in GACs compared to the matched adjacent normal tissues in the TMA. LncRNA PVT1 expression was positively correlated with larger tumor size, deeper wall invasion, lymph node metastases, and short survival duration. Inhibition of PVT1 using PVT1 ASOs significantly suppressed tumor cell growth and invasion in vitro and in vivo. PVT1 expression was highly associated with poor prognosis in GAC patients and targeting PVT1 using PVT1 ASOs was effective at curtailing tumor cell growth in vitro and in vivo. Thus, PVT1 is a poor prognosticator as well as therapeutic target. Targeting PVT1 using PVT1 ASOs provides a novel therapeutic strategy for GAC.
Collapse
Affiliation(s)
- Yuan Li
- Department of Gastrointestinal Medical Oncology, Unit 0426, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA; (Y.L.); (S.S.); (M.P.P.); (A.W.S.); (J.J.); (Y.X.); (Y.W.); (L.H.); (L.M.)
- Department of Surgical Oncology and General Surgery, First Hospital of China Medical University, Shenyang 110001, China
| | - Shumei Song
- Department of Gastrointestinal Medical Oncology, Unit 0426, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA; (Y.L.); (S.S.); (M.P.P.); (A.W.S.); (J.J.); (Y.X.); (Y.W.); (L.H.); (L.M.)
| | - Melissa Pool Pizzi
- Department of Gastrointestinal Medical Oncology, Unit 0426, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA; (Y.L.); (S.S.); (M.P.P.); (A.W.S.); (J.J.); (Y.X.); (Y.W.); (L.H.); (L.M.)
| | - Guangchun Han
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (G.H.); (L.W.)
| | - Ailing W. Scott
- Department of Gastrointestinal Medical Oncology, Unit 0426, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA; (Y.L.); (S.S.); (M.P.P.); (A.W.S.); (J.J.); (Y.X.); (Y.W.); (L.H.); (L.M.)
| | - Jiankang Jin
- Department of Gastrointestinal Medical Oncology, Unit 0426, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA; (Y.L.); (S.S.); (M.P.P.); (A.W.S.); (J.J.); (Y.X.); (Y.W.); (L.H.); (L.M.)
| | - Yan Xu
- Department of Gastrointestinal Medical Oncology, Unit 0426, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA; (Y.L.); (S.S.); (M.P.P.); (A.W.S.); (J.J.); (Y.X.); (Y.W.); (L.H.); (L.M.)
- Department of Surgical Oncology and General Surgery, First Hospital of China Medical University, Shenyang 110001, China
| | - Ying Wang
- Department of Gastrointestinal Medical Oncology, Unit 0426, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA; (Y.L.); (S.S.); (M.P.P.); (A.W.S.); (J.J.); (Y.X.); (Y.W.); (L.H.); (L.M.)
| | - Longfei Huo
- Department of Gastrointestinal Medical Oncology, Unit 0426, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA; (Y.L.); (S.S.); (M.P.P.); (A.W.S.); (J.J.); (Y.X.); (Y.W.); (L.H.); (L.M.)
| | - Lang Ma
- Department of Gastrointestinal Medical Oncology, Unit 0426, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA; (Y.L.); (S.S.); (M.P.P.); (A.W.S.); (J.J.); (Y.X.); (Y.W.); (L.H.); (L.M.)
| | - Christopher Vellano
- Center for Co-Clinical Trials, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Xiaolin Luo
- Ionis Pharmaceuticals, Inc., 2855 Gazelle Court, Carlsbad, CA 92010, USA; (X.L.); (R.M.)
| | - Robert MacLeod
- Ionis Pharmaceuticals, Inc., 2855 Gazelle Court, Carlsbad, CA 92010, USA; (X.L.); (R.M.)
| | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (G.H.); (L.W.)
| | - Zhenning Wang
- Department of Surgical Oncology and General Surgery, First Hospital of China Medical University, Shenyang 110001, China
- Correspondence: (Z.W.); (J.A.A.); Tel.: +1-713-792-3685 (Z.W.)
| | - Jaffer A. Ajani
- Department of Gastrointestinal Medical Oncology, Unit 0426, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA; (Y.L.); (S.S.); (M.P.P.); (A.W.S.); (J.J.); (Y.X.); (Y.W.); (L.H.); (L.M.)
- Correspondence: (Z.W.); (J.A.A.); Tel.: +1-713-792-3685 (Z.W.)
| |
Collapse
|
6
|
Sun Y, Tian H, Xu X, Wang L. Low expression of adenomatous polyposis coli 2 correlates with aggressive features and poor prognosis in colorectal cancer. Bioengineered 2020; 11:1027-1033. [PMID: 32951505 PMCID: PMC8291837 DOI: 10.1080/21655979.2020.1820823] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Currently, there are no relevant findings on the diagnostic and prognostic roles of adenomatous polyposis coli 2 (APC2) in colorectal cancer (CRC). This study investigated the clinical value of APC2 dysregulation in the prognosis of CRC. Immunohistochemical scores obtained from tissue microarrays were used to quantify the expression of APC2 protein in 201 CRC tissues and 139 adjacent normal tissues. A chi-squared test was performed to analyze the association between APC2 expression and various clinical characteristics. Differences in 5-year survival between groups were analyzed. A receiver operating characteristic (ROC) curve was generated to investigate the potential association between APC2 and CRC diagnosis. Compared with adjacent normal tissues, APC2 was downregulated in CRC tissues (P = 0.0004). Survival analyses revealed that CRC patients with high APC2 expression (96.74%) obtained better 5-year survival rates than those with low APC2 expression (88.07%). CRC patients with low APC2 expression exhibited obvious lymphovascular invasion (P = 0.010), lymph node metastasis (P = 0.007), and high tumor node metastasis (TNM) stage (P = 0.007). Furthermore, ROC curves confirmed that APC2 was associated with lymphovascular invasion (P = 0.004), lymph node metastasis (P = 0.002), and TNM staging (P = 0.002). In summary, low APC2 expression in CRC tissues was associated with poor prognosis and may be a useful biomarker for the prognosis and clinical classification of CRC.
Collapse
Affiliation(s)
- Yan Sun
- Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University , Guangzhou, China
| | - Hua Tian
- Department of Gastroenterology, Houjie Hospital Affiliated to Guangdong Medical College , Dongguan, China
| | - Xuehu Xu
- Department of General Surgery, The Third Affiliated Hospital of Guangzhou Medical University , Guangzhou, China
| | - Lin Wang
- Department of Oncology, Guangzhou Red Cross Hospital, Medical College, Jinan University , Guangzhou, China
| |
Collapse
|
7
|
Wang G, Zhang H, Li P. Upregulation of hsa_circRNA_102958 Indicates Poor Prognosis and Promotes Ovarian Cancer Progression Through miR-1205/SH2D3A Axis. Cancer Manag Res 2020; 12:4045-4053. [PMID: 32547237 PMCID: PMC7264029 DOI: 10.2147/cmar.s248560] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 05/01/2020] [Indexed: 12/22/2022] Open
Abstract
Background Circular RNA (circRNA) is a recently identified member of noncoding RNAs. It has been demonstrated to regulate gene expression post-transcriptionally and play critical roles in tumorigenesis. However, how circRNA regulates ovarian cancer (OC) progression is poorly understood. Previously, hsa_circRNA_102958 was reported to regulate gastric cancer and colorectal cancer development. This study aims to investigate the role of hsa_circRNA_102958 in OC progression. Materials and Methods qRT-PCR was used to test gene expression. CCK8 and colony formation assays were used to analyze proliferation. Transwell assay was utilized to determine migration and invasion. Luciferase reporter assay was conducted to test the interaction between hsa_circRNA_102958 and miR-1205. Results hsa_circRNA_102958 was upregulated in OC tissues and cell lines. hsa_circRNA_102958 upregulation indicated a poor prognosis in OC patients. Knockdown of hsa_circRNA_102958 significantly suppressed the proliferation, migration and invasion of OC cells and vice versa. hsa_circRNA_102958 was a competing endogenous RNA (ceRNA) for miR-1205. hsa_circRNA_102958 inhibited miR-1205 activity to promote SH2D3A expression. Overexpression of SH2D3A promoted proliferation, migration and invasion of OC cells. Conclusion Our data suggest that hsa_circRNA_102958 promotes OC aggravation through regulation of miR-1205/SH2D3A signaling.
Collapse
Affiliation(s)
- Guan Wang
- Department of Gynecology, Heilongjiang Provincial Hospital, Harbin 150000, People's Republic of China
| | - Huijing Zhang
- Department of Gynecology, Heilongjiang Provincial Hospital, Harbin 150000, People's Republic of China
| | - Peiling Li
- Department of Gynecology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150000, People's Republic of China
| |
Collapse
|
8
|
Reciprocal regulation of miR-1205 and E2F1 modulates progression of laryngeal squamous cell carcinoma. Cell Death Dis 2019; 10:916. [PMID: 31801947 PMCID: PMC6893029 DOI: 10.1038/s41419-019-2154-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 11/01/2019] [Accepted: 11/07/2019] [Indexed: 12/20/2022]
Abstract
The burgeoning functions of many microRNAs (miRs) have been well study in cancer. However, the level and function of miR-1205 in laryngeal squamous cell cancer remains unknown. In the current research, we validated that miR-1205 was notably downregulated in human laryngeal squamous cell carcinoma (LSCC) samples in comparison with tissues adjacent to LSCC, and correlated with T stage, lymph node metastasis, and clinical stage. Using Kaplan–Meier analysis indicates that high expression of miR-1205 has a favorable prognosis for patients with LSCC. Functional assays show that enforced miR-1205 expression attenuates the migration, growth, and invasion of LSCC cells. And E2F1 is verified to be a target of miR-1205, while E2F1 binds to miR-1205 promoter and transcriptionally inhibits miR-1205 expression. Overexpression of E2F1 reverses the inhibitory impacts of miR-1205 on LSCC cells in part. Importantly, E2F1 is abnormally increased in LSCC tissues, and its protein levels were inversely relevant to miR-1205 expression. High E2F1 protein level is in connection with clinical stage, T stage, lymph node metastasis, and poor prognosis. Consequently, reciprocal regulation of miR-1205 and E2F1 plays a crucial role in the progression of LSCC, suggesting a new miR-1205/E2F1-based clinical application for patients of LSCC.
Collapse
|
9
|
Zeng X, Liu Q, Yang Y, Jia W, Li S, He D, Ma R. Placenta-specific protein 8 promotes the proliferation of lung adenocarcinoma PC-9 cells and their tolerance to an epidermal growth factor receptor tyrosine kinase inhibitor by activating the ERK signaling pathway. Oncol Lett 2019; 18:5621-5627. [PMID: 31620204 DOI: 10.3892/ol.2019.10911] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 08/06/2019] [Indexed: 11/06/2022] Open
Abstract
Placenta-specific protein 8 (PLAC8) is a conserved protein with a molecular weight of 12.5 kDa. The specific function of this protein has not been fully elucidated, however, PLAC8 has been found to play an important tumor regulatory role in certain types of cancer, including colon, pancreatic and liver cancer. PLAC8 also participates in the regulation of the cell cycle, autophagy, epithelial-mesenchymal transition and other cellular functions, indicating its potential as a molecular target worth further investigation. The present study investigated the effect of PLAC8 on the proliferation of lung adenocarcinoma PC-9 cells and their sensitivity to gefitinib, an epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI). It was found that the inhibition of PLAC8 expression in PC-9 cells resulted in significantly decreased proliferation, whereas overexpression of PLAC8 significantly increased the proliferation (P<0.05) of PC-9 cells. Furthermore, inhibition of PLAC8 expression resulted in decreased activity of the ERK signaling pathway, while PLAC8 overexpression increased activity of this pathway. Inhibition of the ERK signaling pathway with U0126 reversed the effects induced by inhibiting or overexpressing PLAC8 on cell proliferation. In addition, overexpression of PLAC8 significantly decreased the sensitivity of PC-9 cells to gefitinib, and this effect was reversed by U0126. Overall, these results suggest that PLAC8 is involved in the regulation of proliferation of lung adenocarcinoma PC-9 cells and impacts their sensitivity to an EGFR-TKI. Thus, PLAC8 is a potential novel target in lung adenocarcinoma for future studies.
Collapse
Affiliation(s)
- Xiaofei Zeng
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Qing Liu
- Department of Cardiothoracic Surgery, Chengdu Fifth People's Hospital, Chengdu, Sichuan 611130, P.R. China
| | - Yanhui Yang
- Department of Cardiothoracic Surgery, The First People's Hospital of Neijiang, Sichuan 641000, P.R. China
| | - Weikun Jia
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Shuping Li
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Dongsheng He
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Ruidong Ma
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| |
Collapse
|
10
|
Li MY, Tang XH, Fu Y, Wang TJ, Zhu JM. Regulatory Mechanisms and Clinical Applications of the Long Non-coding RNA PVT1 in Cancer Treatment. Front Oncol 2019; 9:787. [PMID: 31497532 PMCID: PMC6712078 DOI: 10.3389/fonc.2019.00787] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 08/05/2019] [Indexed: 12/24/2022] Open
Abstract
Cancer is the second leading cause of death worldwide, and no obvious decline in incidence and mortality has occurred in recent years. It is imperative to further investigate the mechanisms underlying tumor progression. Long non-coding RNAs have received considerable attention in recent years because of their major regulatory roles in gene expression. Among them, PVT1 is well-studied, and substantial evidence indicates that PVT1 plays critical roles in the onset and development of cancers. Normally, PVT1 acts as an oncogenic factor by promoting cancer cell proliferation, invasion, metastasis, and drug resistance. Herein, we summarize current knowledge regarding the regulatory effects of PVT1 in cancer progression, as well as the related underlying mechanisms, such as interaction with Myc, modulation of miRNAs, and regulation of gene transcription and protein expression. In extracellular fluid, PVT1 mainly promotes cancer initiation, and it normally enhances cellular cancer characteristics in the cytoplasm and cell nucleus. Regarding clinical applications, its role in drug resistance and its potential use as a diagnostic and prognostic marker have received increasing attention. We hope that this review will contribute to a better understanding of the regulatory role of PVT1 in cancer progression, paving the way for the development of PVT1-based therapeutic approaches in cancer treatment.
Collapse
Affiliation(s)
- Meng-Yuan Li
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, China
| | - Xiao-Huan Tang
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Yan Fu
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Tie-Jun Wang
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, China
| | - Jia-Ming Zhu
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|