1
|
Gao Y, Li Y, Liu Z, Dong Y, Yang S, Wu B, Xiao M, Chen C, Wen Y, Chen L, Jiang H, Yao Y. AHSA1 Regulates Hepatocellular Carcinoma Progression via the TGF-β/Akt-Cyclin D1/CDK6 Pathway. J Hepatocell Carcinoma 2023; 10:2021-2036. [PMID: 38022728 PMCID: PMC10640837 DOI: 10.2147/jhc.s407680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Background Activator of heat shock protein 90 (HSP90) ATPase Activity 1 (AHSA1) regulates proliferation, apoptosis, migration, and invasion of osteosarcoma and hepatocellular carcinoma (HCC). However, the novel mechanism of AHSA1 in the tumor biology of hepatocellular carcinoma (HCC) remains unclear. Methods We analyzed AHSA1 expression in 85 pairs of clinical samples of HCC and the Cancer Genome Atlas database. The role of AHSA1 in HCC was proved by cell proliferation, colony formation, migration, cell cycle analysis in vitro, xenograft models and tumor metastasis assay in vivo, and bioinformatics. Results High AHSA1 expression was demonstrated in HCC and associated with invasive depth, clinical stage, and poor overall survival of patients. Univariate Cox analysis confirmed that AHSA1 was an independent prognostic factor for patients with HCC. Meanwhile, AHSA1 upregulation promoted cell proliferation, colony formation, and cell migration in vitro and tumor cell proliferation and metastasis of HCC cells in vivo. AHSA1 upregulation increased the cell cycle transition from G1 to S phase by increasing the expression of cyclinD1, cyclinD3, and cyclin-dependent kinase 6(CD). Transforming growth factor beta 1 (TGF-β1)-induced protein kinase B (Akt) signaling regulated the expression of downstream targets, including cyclinD1. AHSA1 expression was closely correlated with the expression of TGF-β, Akt, cyclinD1, cyclinD3, and CDK6 using the Gene Expression Profiling Interactive Analysis database. AHSA1 upregulation participated in HCC progression by regulating TGF-β/Akt-cyclinD1/CDK6 signaling. Conclusion AHSA1 might serve as a biomarker for predicting the clinical outcome of patients with HCC. It is vital in tumor metastasis and disease progression of HCC and may facilitate the development of clinical intervention strategies against HCC.
Collapse
Affiliation(s)
- Yanjun Gao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China
- Hubei Provincial Research Center for Precision Medicine of Cancer, Wuhan, 430200, People’s Republic of China
| | - Yingge Li
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China
- Hubei Provincial Research Center for Precision Medicine of Cancer, Wuhan, 430200, People’s Republic of China
| | - Zheming Liu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China
- Hubei Provincial Research Center for Precision Medicine of Cancer, Wuhan, 430200, People’s Republic of China
| | - Yi Dong
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China
- Hubei Provincial Research Center for Precision Medicine of Cancer, Wuhan, 430200, People’s Republic of China
| | - Siqi Yang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China
- Hubei Provincial Research Center for Precision Medicine of Cancer, Wuhan, 430200, People’s Republic of China
| | - Bin Wu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China
- Department of Oncology, Huang-Gang Central Hospital, Huanggang, 438000, People’s Republic of China
| | - Mengxia Xiao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China
- Department of Oncology, Yichun People’s Hospital, Yichun, 336000, People’s Republic of China
| | - Chen Chen
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China
| | - Yingmei Wen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China
- Hubei Provincial Research Center for Precision Medicine of Cancer, Wuhan, 430200, People’s Republic of China
| | - Lei Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China
| | - Haijuan Jiang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, People’s Republic of China
| | - Yi Yao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China
- Hubei Provincial Research Center for Precision Medicine of Cancer, Wuhan, 430200, People’s Republic of China
| |
Collapse
|
2
|
Hou C, Lu M, Lei Z, Dai S, Chen W, Du S, Jin Q, Zhou Z, Li H. HMGB1 Positive Feedback Loop Between Cancer Cells and Tumor-Associated Macrophages Promotes Osteosarcoma Migration and Invasion. J Transl Med 2023; 103:100054. [PMID: 36801636 DOI: 10.1016/j.labinv.2022.100054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 10/11/2022] [Accepted: 12/03/2022] [Indexed: 01/11/2023] Open
Abstract
Numerous studies have demonstrated the key roles of tumor-associated macrophages (TAMs) in osteosarcoma metastasis. Higher levels of high mobility group box 1 (HMGB1) promote osteosarcoma progression. However, whether HMGB1 is involved in the polarization of M2 macrophages into M1 macrophages in osteosarcoma remains largely unknown. Here, HMGB1 and CD206 mRNA expression levels were measured by a quantitative reverse transcription-polymerase chain reaction in osteosarcoma tissues and cells. HMGB1 and receptor for advanced glycation end products (RAGE) protein expression levels were measured by western blotting. Osteosarcoma migration was measured using transwell and wound-healing assays, while a transwell assay determined osteosarcoma invasion. Macrophage subtypes were detected using flow cytometry. HMGB1 expression levels were aberrantly enhanced in osteosarcoma tissues compared with normal tissues and were positively correlated with AJCC III and IV stages, lymph node metastasis, and distant metastasis. Silencing HMGB1 inhibited the migration, invasion, and epithelial-mesenchymal transition (EMT) of osteosarcoma cells. Furthermore, reduced HMGB1 expression levels in conditioned media derived from osteosarcoma cells induced the polarization of M2 TAMs to M1 TAMs. In addition, silencing HMGB1 inhibited the liver and lung metastasis of tumors and reduced the expression levels of HMGB1, CD163, and CD206 in vivo. HMGB1 was found to regulate macrophage polarization through RAGE. Polarized M2 macrophages induced osteosarcoma migration and invasion, activating HMGB1 expression in osteosarcoma cells to form a positive feedback loop. In conclusion, HMGB1 and M2 macrophages enhanced osteosarcoma migration, invasion, and EMT through positive feedback regulation. These findings reveal the significance of tumor cell and TAM interactions in the metastatic microenvironment.
Collapse
Affiliation(s)
- Changhe Hou
- Department of Musculoskeletal Oncology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Ming Lu
- Department of Musculoskeletal Oncology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Zixiong Lei
- Department of Musculoskeletal Oncology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Shuangwu Dai
- Department of Musculoskeletal Oncology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Wei Chen
- Department of Musculoskeletal Oncology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Shaohua Du
- Department of Musculoskeletal Oncology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Qinglin Jin
- Department of Musculoskeletal Oncology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Zhongxin Zhou
- Department of Vascular Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.
| | - Haomiao Li
- Department of Musculoskeletal Oncology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.
| |
Collapse
|
3
|
Chen C, Liu L. Silencing of lncRNA KLF3-AS1 represses cell growth in osteosarcoma via miR-338-3p/MEF2C axis. J Clin Lab Anal 2022; 36:e24698. [PMID: 36250223 DOI: 10.1002/jcla.24698] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Osteosarcoma (OS) is a highly recurrent malignancy occurring among adolescents. The goal of this research was to scrutinize the role and action mechanism of KLF3-AS1 in OS. METHODS Western blotting and quantitative reverse transcription real-time PCR were conducted to ascertain the mRNA expressions of miR-338-3p, KLF3-AS1, and MEF2C in OS cell lines and tissue samples. Colony formation and CCK-8 experiments were done to evaluate the proliferative capacity of the cells. Western blotting was also executed to measure the relative expressions of the proteins Bcl-2 and Bax. RNA immunoprecipitation and dual luciferase reporter experiments were carried out to validate the target relationships among MEF2C, KLF3-AS1, and miR-338-3p. Mouse xenograft models were created to assess the influences of KLF3-AS1 on the growth of tumors in vivo. RESULTS Elevated levels of KLF3-AS1 and MEF2C and reduced amounts of miR-338-3p were identified in OS. KLF3-AS1 targeted miR-338-3p, and miR-338-3p further targeted MEF2C. Silencing KLF3-AS1 induced apoptosis and attenuated proliferation in vitro and repressed the tumor growth in vivo. Inhibiting miR-338-3p inverted the cancer-suppressing effects of KLF3-AS1 silencing. Meanwhile, loss of MEF2C partially eliminated the effects brought about by miR-338-3p downregulation, namely the stimulation of cell growth and suppression of apoptosis. CONCLUSIONS Silencing of KLF3-AS1 could repress the growth of cells and induce apoptosis by regulating miR-338-3p/MEF2C in OS. This suggests that the regulatory axis KLF3-AS1/miR-338-3p/MEF2C is a prospective target for OS treatment.
Collapse
Affiliation(s)
- Chunfa Chen
- Department of Emergency Medicine, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi, China
| | - Liang Liu
- Department of Spinal Surgery, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi, China
| |
Collapse
|
4
|
Tang F, Jiang X, Liao S, Liu Y, He M. Construction of a transcription factor-miRNA-mRNA interactive network elucidates underlying pathogenesis for osteosarcoma and validation by qRT-PCR. Medicine (Baltimore) 2022; 101:e31049. [PMID: 36254052 PMCID: PMC9575767 DOI: 10.1097/md.0000000000031049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
PURPOSE Osteosarcoma is characterized by features of rapid growth and early metastasis with a poor prognosis. The aim of our research is to investigate the potential transcription factor (TF)-miRNA-mRNA regulatory mechanism in osteosarcoma utilizing bioinformatics methods and validate by qRT-PCR. METHODS The microRNA (miRNA) expression profiling datasets (GSE28423 and GSE65071) and mRNA expression profiling dataset GSE33382 were collected from the Gene Expression Omnibus (GEO) database. Differentially expressed miRNAs (DEMs) and differentially expressed genes (DEGs) were screened using the limma package. Then, the TransmiR v2.0, miRDB, and Targetscan 7.2 database were applied for the acquisition of TF-miRNA and miRNA-mRNA interaction relationships, respectively. Finally, we built a TF-miRNA-mRNA interactive network. Furthermore, survival analysis was performed to identify sub-network with prognostic value and validate through qRT-PCR. RESULTS Eight overlapping DEMs and 682 DEGs were identified. Based on bioinformatics methods, 30 TF-miRNA interaction pairs and 25 miRNA-mRNA interaction pairs were screened. Finally, we constructed a TF-miRNA-mRNA regulatory network. Furthermore, laminin subunit gamma 1 (LAMC1) and thrombospondin-1 (THBS1), which involved in the network, were determined to have prognostic value and the corresponding subnetwork was identified. qRT-PCR results showed that LAMC1 mRNA expression was higher in osteosarcoma cells. CONCLUSION Based on the survival analysis, a TF-miRNA-mRNA sub-network, that is TFs (SPI1, HEY1, and CEBPB)-hsa-miR-338-3p-target genes (LAMC1 and THBS1) was established. In conclusion, the construction of a potential TF-related regulatory network will help elucidate the underlying pathological mechanisms of osteosarcoma, and may provide novel insights for the diagnosis and treatment of osteosarcoma.
Collapse
Affiliation(s)
- Fuxing Tang
- Division of Spinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, China
| | - Xiaohong Jiang
- Division of Spinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, China
| | - Shijie Liao
- Division of Spinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, China
| | - Yun Liu
- Division of Spinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, China
| | - Maolin He
- Division of Spinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, China
- *Correspondence: Maolin He, Division of Spinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China (e-mail: )
| |
Collapse
|
5
|
AHSA1 Promotes Proliferation and EMT by Regulating ERK/CALD1 Axis in Hepatocellular Carcinoma. Cancers (Basel) 2022; 14:cancers14194600. [PMID: 36230524 PMCID: PMC9562867 DOI: 10.3390/cancers14194600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/13/2022] [Accepted: 09/17/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Globally, hepatocellular carcinoma is one of the leading causes of cancer-related death. Activator of HSP90 ATPase activity 1(AHSA1) was reported to be involved in regulating the maturation, stability, and degradation of related cancer-promoting proteins. However, the biological function and regulatory mechanism of AHSA1 in hepatocellular carcinoma were unclearly. In this study, we found AHSA1 was upregulated and associated with poor clinical characteristics and prognosis of hepatocellular carcinoma patients. Moreover, AHSA1 promoted proliferation, metastasis, epithelium-mesenchymal transition of hepatocellular carcinoma both in vitro and in vivo by recruiting ERK1/2 and promoting the phosphorylation and inactivation of CALD1. Taken together, our findings provided a mechanistic insight into the role of AHSA1 in hepatocellular carcinoma progression and implied AHSA1 may be a biomarker and therapeutic target for hepatocellular carcinoma patients. Abstract Hepatocellular carcinoma (HCC) is one of the major causes of cancer-related death worldwide. AHSA1 as a chaperone of HSP90 promotes the maturation, stability, and degradation of related cancer-promoting proteins. However, the regulatory mechanism and biological function of AHSA1 in HCC are largely unknown. Actually, we found that AHSA1 was significantly upregulated in HCC tissues and cell lines and was notably correlated with the poor clinical characteristics and prognosis of HCC patients in this study. Furthermore, both in vitro and in vivo, gain- and loss-of-function studies demonstrated that AHSA1 promoted the proliferation, invasion, metastasis, and epithelial-mesenchymal transition (EMT) of HCC. Moreover, the mechanistic study indicated that AHSA1 recruited ERK1/2 and promoted the phosphorylation and inactivation of CALD1, while ERK1/2 phosphorylation inhibitor SCH772984 reversed the role of AHSA1 in the proliferation and EMT of HCC. Furthermore, we demonstrated that the knockdown of CALD1 reversed the inhibition of proliferation and EMT by knocking AHSA1 in HCC. We also illustrated a new molecular mechanism associated with AHSA1 in HCC that was independent of HSP90 and MEK1/2. In summary, AHSA1 may play an oncogenic role in HCC by regulating ERK/CALD1 axis and may serve as a novel therapeutic target for HCC.
Collapse
|
6
|
Chen Z, Li L, Li Z, Wang X, Han M, Gao Z, Wang M, Hu G, Xie X, Du H, Xie Z, Zhang H. Identification of key serum biomarkers for the diagnosis and metastatic prediction of osteosarcoma by analysis of immune cell infiltration. Cancer Cell Int 2022; 22:78. [PMID: 35151325 PMCID: PMC8841093 DOI: 10.1186/s12935-022-02500-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 01/30/2022] [Indexed: 12/15/2022] Open
Abstract
Abstract
Background
The role of circular RNAs (circRNAs) and microRNAs (miRNAs) in osteosarcoma (OS) development has not been fully elucidated. Further, the contribution of the immune response to OS progression is not well defined. However, it is known that circRNAs and miRNAs can serve as biomarkers for the diagnosis, prognosis, and therapy of many cancers. Thus, the aim of this study was to identify novel key serum biomarkers for the diagnosis and metastatic prediction of OS by analysis of immune cell infiltration and associated RNA molecules.
Methods
Human OS differentially expressed circRNAs (DEcircRNAs), differentially expressed miRNAs (DEmiRNAs), and differentially expressed mRNAs (DEmRNAs) were identified by analysis of microarray data downloaded from Gene Expression Omnibus (GEO) datasets. Further, characteristic patterns of OS-infiltrating immune cells were analyzed. On this basis, we identified statistically significant transcription factors. Moreover we performed pathway enrichment analysis, constructed protein–protein interaction networks, and devised competitive endogenous RNA (ceRNA) networks. Biological targets of the ceRNA networks were evaluated and potential OS biomarkers confirmed by RT-qPCR analysis of the patients’ serum.
Results
Seven differentially expressed circRNAs, 166 differentially expressed miRNAs, and 175 differentially expressed mRNAs were identified. An evaluation of cellular OS infiltration identified the highest level of infiltration by M0 macrophages, M2 macrophages, and CD8+ T cells, with M0 macrophages and CD8+ T cells as the most prominent. Significant patterns of tumor-infiltrating immune cells were identified by principal component analysis. Moreover, 185 statistically significant transcription factors were associated with OS. Further, in association with immune cell infiltration, hsa-circ-0010220, hsa-miR-326, hsa-miR-338-3p, and FAM98A were identified as potential novel biomarkers for OS diagnosis. Of these, FAM98A had the most promise as a diagnostic marker for OS and OS metastasis. Most importantly, a novel diagnostic model consisting of these four biomarkers (hsa-circ-0010220, hsa-miR-326, hsa-miR-338-3p, and FAM98A) was established with a 0.928 AUC value.
Conclusions
In summary, potential serum biomarkers for OS diagnosis and metastatic prediction were identified based on an analysis of immune cell infiltration. A novel diagnostic model consisting of these four promising serum biomarkers was established. Taken together, the results of this study provide a new perspective by which to understand immunotherapy of OS.
Collapse
|
7
|
Identifying Differentially Expressed tRNA-Derived Small Fragments as a Biomarker for the Progression and Metastasis of Colorectal Cancer. DISEASE MARKERS 2022; 2022:2646173. [PMID: 35035608 PMCID: PMC8758288 DOI: 10.1155/2022/2646173] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 11/17/2021] [Accepted: 12/06/2021] [Indexed: 12/29/2022]
Abstract
Objectives. The epithelial-to-mesenchymal transition (EMT) is one key step for the invasion and metastasis of colorectal cancer (CRC). Up until now, the underlying mechanism of EMT in CRC is still unpromising. Thus, it is essential to have a better understanding of its carcinogenesis. The transfer RNA-derived small fragments (tsRNAs) are a new group of small noncoding RNAs (sncRNAs), including tRNA-derived stress-induced RNAs (tiRNAs) and tRNA-derived fragments (tRFs), which have been observed to play an important role in many cancers. However, the relationship between tRFs and EMT in CRC is still unknown. Herein, we aimed to investigate the involvement of tRFs in EMT and its contribution to CRC development. Methods. We identified the differentially expressed tsRNAs in colorectal cancer cell line HT29 treated with TGF-β compared with control cells by using high-throughput sequencing and quantitative real-time reverse transcription-polymerase chain reaction (qRT-PCR). QRT-PCR was conducted to validate the differentially expressed fragments in 68 CRC tumor samples (22 women and 46 men) and adjacent nontumor samples. The association of the expression of tRFs with CRC metastasis and clinical stage was analyzed. Meanwhile, the correlation between tRF expression and overall survival (OS) was also analyzed. TargetScan and miRanda and multiple bioinformatic approaches were used to predict the possible target genes of tsRNAs and analyze possible functions of the tRFs. Results. A series of differentially expressed tsRNAs were identified in TGF-β-treated HT29 cells compared with control cells. tRF-phe-GAA-031 and tRF-VAL-TCA-002 were found to be significantly upregulated in CRC tissues compared to adjacent nontumor tissues. They were significantly correlated with distant metastasis and clinical stage. We compared the differences between tumor samples and nontumor tissues from the ROC curves. The area under the ROC curve (AUC) was up to 0.7554 (95% confidence interval: 0.6739 to 0.8369,
) for tRF-Phe-GAA-031 and up to 0.7313 (95% confidence interval: 0.6474 to 0.8151,
) for tRF-VAL-TCA-002. For OS analysis, higher tRF-phe-GAA-031 and tRF-VAL-TCA-002 expressions were associated with shorter survival for CRC patients. Conclusion. A series of differentially expressed tsRNAs are identified in the EMT process of CRC. And tRF-phe-GAA-031 and tRF-VAL-TCA-002 are higher expressed in CRC tissues, and they might play an important role in the metastasis of CRC. Meanwhile, they may be potential biomarkers and intervention targets in the clinical treatment of CRC.
Collapse
|
8
|
Wang Y, Hao W, Wang H. miR-557 suppressed the malignant behaviours of osteosarcoma cells by reducing HOXB9 and deactivating the EMT process. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2021; 49:230-239. [PMID: 33666541 DOI: 10.1080/21691401.2021.1890100] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 02/09/2021] [Indexed: 12/31/2022]
Abstract
MicroRNAs (miRNAs) are vital gene regulators, which play a profound role in the process of forming and developing many diseases, especially tumour. The study intends to excavate the potential regulatory mechanisms of miR-557 and its targeting gene Homeobox B9 (HOXB9) in osteosarcoma. GEO dataset on osteosarcoma was applied to detect the expression of miR-557 and HOXB9. Associations between miR-557 and HOXB9 were speculated by prediction software and verified by dual luciferase assay. Cell proliferation, colony formation and mobility were measured by cell counting kit-8, plate clone formation and transwell assays. Expression of mesenchymal transitions (MTs) related proteins was assessed by western blot analysis. Low expression of miR-557 was presented in osteosarcoma tissues and cell lines. Upregulation of miR-557 restrained osteosarcoma cells proliferation, movement and MT process. HOXB9, served as a target gene of miR-557, was highly expressed in osteosarcoma, and its high expression was associated with poor prognosis in patients with osteosarcoma. In addition, overexpression of HOXB9 attenuated the inhibitory effects of miR-557 on tumour progression by MT process. Overexpression of miR-557 suppressed the growth, metastasis and MT process of osteosarcoma cells by targeting HOXB9, affording novel molecular selection for targeted therapy of osteosarcoma.
Collapse
Affiliation(s)
- Yuanhao Wang
- Department of Spinal Orthopedics, Weihai Municipal Hospital, Weihai City, P. R. China
| | - Wei Hao
- Department of Joint and Sports Medicine, Shandong Provincial Third Hospital affiliated to Shandong University, Jinan City, Shandong Province, P. R. China
| | - Hui Wang
- Department of Orthopaedics, Jining No.1 People's Hospital, Jining City, P. R. China
| |
Collapse
|
9
|
Zhou H, Cao L, Wang C, Fang C, Wu H, Liu C. miR-877-3p inhibits tumor growth and angiogenesis of osteosarcoma through Fibroblast Growth Factor 2 signaling. Bioengineered 2021; 13:8174-8186. [PMID: 34738872 PMCID: PMC9162015 DOI: 10.1080/21655979.2021.1982305] [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] [Indexed: 12/29/2022] Open
Abstract
Osteosarcoma (OS) is the most common high-grade malignant bone tumor in teenagers. MicroRNAs can function as posttranscriptional regulators of gene expression, playing critical roles in cancer dev-877-3p in OS. Quantitative real-time RT-PCR was carried out for detecting miR-877-3p expression in OS. The effects of miR-877-3p on proliferation was analyzed via MTT, colony formation, and flow cytometry assays. Angiogenesis of endothelial cells were investigated by wound healing and tube formation assay. Gene profiling based on PCR array and luciferase reporter assay were conducted to determine target genes of miR-877-3p. In-vivo study was used to determine the effects of miR-877-3p on the tumor growth. The expression of miR-877-3p was markedly downregulated in OS tissues and cell lines. Low expression of miR-877-3p predicts poor prognosis of OS patients. miR-877-3p overexpression was found to inhibit the proliferation of OS cell lines. The angiogenesis assays showed that miR-877-3p attenuated the angiogenesis. Further mechanism studies showed that miR-877-3p can reduce (Fibroblast Growth Factor 2) FGF2 expression in OS cells by binding to the 3’UTR end of FGF2. Moreover, increased expression of miR-877-3p was responsible for the inhibition of tumor growth and angiogenesis. Taken together, our findings indicated that miR-877-3p might exhibit tumor suppressive role by targeting FGF2 signaling, which may serve as potential target for OS.
Collapse
Affiliation(s)
- Hailin Zhou
- Department of Orthopedics, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lei Cao
- Department of Orthopedics, Shanghai Songjiang District Central Hospital, Shanghai, China
| | - Cheng Wang
- Department of Orthopedics, Shanghai Songjiang District Central Hospital, Shanghai, China.,Department of Radiology, Shanghai Songjiang District Central Hospital, Shanghai, China
| | - Chi Fang
- Department of Gynecologic Oncology, Fudan University, Shanghai Cancer Center, Shanghai, China
| | - HaiHui Wu
- Department of Orthopedics, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chao Liu
- Department of Radiology, Shanghai Songjiang District Central Hospital, Shanghai, China
| |
Collapse
|
10
|
A Heterotrinuclear Cluster Complex Attenuates Oral Squamous Cell Carcinoma Development In Vivo and In Vitro. J CLUST SCI 2021. [DOI: 10.1007/s10876-021-02193-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
11
|
Cao R, Shao J, Hu Y, Wang L, Li Z, Sun G, Gao X. Correction to: microRNA-338-3p inhibits proliferation, migration, invasion, and EMT in osteosarcoma cells by targeting activator of 90 kDa heat shock protein ATPase homolog 1. Cancer Cell Int 2021; 21:560. [PMID: 34702261 PMCID: PMC8547048 DOI: 10.1186/s12935-021-02283-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Affiliation(s)
- Riliang Cao
- Department of Pediatric Surgery, Guangdong Women and Children Hospital, Guangzhou, 511400, China
| | - Jianli Shao
- Department of Orthopedic and Traumatology, First Affiliated Hospital, Jinan University, Guangzhou, 510632, China.
| | - Yabin Hu
- Department of Spinal Surgery, The Sixth Affiliated Hospital of Xinjiang Medical University, Ürümqi, 830002, Xinjiang, China.
| | - Liang Wang
- Department of Oncology, First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Zhizhong Li
- Department of Orthopedic and Traumatology, First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Guodong Sun
- Department of Orthopedic and Traumatology, First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Xiaoliang Gao
- Department of Spinal Surgery, The Sixth Affiliated Hospital of Xinjiang Medical University, Ürümqi, 830002, Xinjiang, China
| |
Collapse
|
12
|
Kim D, Moon JW, Min DH, Ko ES, Ahn B, Kim ES, Lee JY. AHA1 regulates cell migration and invasion via the EMT pathway in colorectal adenocarcinomas. Sci Rep 2021; 11:19946. [PMID: 34620942 PMCID: PMC8497578 DOI: 10.1038/s41598-021-99375-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 09/17/2021] [Indexed: 11/09/2022] Open
Abstract
The progression of colorectal cancer (CRC) has been well studied and understood with the development of molecular and genetic techniques. However, specific marker(s) that could be used to predict lymph node (LN) involvement, which is the most important prognostic factor for CRC, have not been identified so far. Our previous study, in which network analysis of LN(+) and LN(-) CRC gene expression was carried out with data obtained from the Cancer Genome Atlas, led to the identification of AHA1. AHA1 is a co-chaperone activator of the Hsp90 ATPase activity. However, the role of AHA1 expression in cancer cells is still unclear. To investigate how AHA1 expression regulates the cancer cell progression and/or metastasis of human CRC, the expression levels of AHA1 and Hsp90 were examined in 105 CRC tissue samples and compared with those in paired normal tissue. The RNA expression levels of AHA1 and Hsp90aa1, but not Hsp90ab, were significantly higher in cancer tissues than in adjacent paired normal tissues (p = 0.032 and p = 0.0002, respectively). In particular, AHA1, but not Hsp90aa1 and Hsp90ab, was closely associated with the TNM stage, LN stage, and tumor metastasis (p = 0.035, p = 0.012, and p = 0.0003, respectively). Moreover, the expression of AHA1 was not only higher in the CRC cell lines than in the normal colon fibroblast cell line but was also associated with the progression of these CRC cell lines. Overexpression of AHA1 in SW480 cells increased, whereas suppression of AHA1 expression in HCT116 cells reduced cell migration and invasion through the regulation of Snail, E-cadherin, pSRC, and pAKT, which are associated with EMT signaling. Taken together, our study suggests that AHA1 contributes to the metastatic advantage of human CRC.
Collapse
Affiliation(s)
- Dasom Kim
- Department of Pathology, Korea University College of Medicine, 73, Goryeodae-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea.,Department of Biomedical Science, Korea University College of Medicine, Seoul, Republic of Korea
| | - Ji Wook Moon
- BK21 FOUR Convergence & Translational Biomedicine Education Research Center, Department of Anatomy, Korea University College of Medicine, Seoul, Republic of Korea
| | - Dong Hwa Min
- Department of Pathology, Korea University College of Medicine, 73, Goryeodae-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea.,Department of Biomedical Science, Korea University College of Medicine, Seoul, Republic of Korea
| | - Eun Sun Ko
- Department of Pathology, Korea University College of Medicine, 73, Goryeodae-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea.,Department of Biomedical Science, Korea University College of Medicine, Seoul, Republic of Korea
| | - Bokyung Ahn
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Eun Sun Kim
- Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Ji-Yun Lee
- Department of Pathology, Korea University College of Medicine, 73, Goryeodae-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea.
| |
Collapse
|
13
|
HIPK3 Circular RNA Promotes Metastases of HCC Through Sponging miR-338-3p to Induce ZEB2 Expression. Dig Dis Sci 2021; 66:3439-3447. [PMID: 33247421 DOI: 10.1007/s10620-020-06688-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 10/21/2020] [Indexed: 01/09/2023]
Abstract
BACKGROUND Upregulation of circHIPK3 has been observed in several kinds of malignancies. However, the mechanisms of circHIPK3 in HCC metastases remains unclear. We investigated the role and the mechanisms of circHIPK3 in the development of HCC. METHODS HCC tissues and paired adjacent non-tumor tissues of surgical patients were used to evaluate circHIPK3 expression. A series of biological experiments had been taken to evaluate the pro-metastatic ability of circHIPK3 during HCC development in vitro and in vivo. The potential mechanisms of circHIPK3 in HCC development were identified by RT-qPCR, Western blot, RIP, and luciferase reporter assays. RESULTS CircHIPK3 expression is significantly upregulated during HCC development. Overexpression of circHIPK3 promotes cell migration, invasion, and metastases in vitro and in vivo. CircHIPK3 promoted HCC metastases by sponging miR-338-3p to regulate EMT-associated proteins E-cadherin, vimentin, and ZEB2 expression. CONCLUSION CircHIPK3 plays a regulatory role in metastatic HCC by sponging miR-338-3p to induce ZEB2 expression, thus promoting EMT procession.
Collapse
|
14
|
Kuai J, Zheng L, Yi X, Liu Z, Qiu B, Lu Z, Jiang Y. ST8SIA6-AS1 promotes the development of hepatocellular carcinoma cells through miR-338-3p/NONO Axis. Dig Liver Dis 2021; 53:1192-1200. [PMID: 33722502 DOI: 10.1016/j.dld.2021.02.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 01/19/2021] [Accepted: 02/09/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Increasing studies have shown a vital fact that long non-coding RNAs (lncRNAs) play a considerable regulatory role in hepatocellular carcinoma (HCC) progression. However, whether ST8 alpha-N-acetyl-neuraminide alpha-2, 8-sialyltransferase 6 antisense RNA 1 (ST8SIA6-AS1) affects the development of HCC is unclear. METHODS The target genes in HCC cell lines were quantified via utilzing quantitative real-time polymerase chain reaction (RT-qPCR) analysis and western blot. Effects of ST8SIA6-AS1 on proliferative, apoptosis and migratory ability of HCC cells were proved by a series of function experiments. The cellular distribution of ST8SIA6-AS1 was examined through fluorescent in situ hybridization (FISH) assay and subcellular fractionation experiments. RNA pulldown assay was implemented to explore the target of ST8SIA6-AS1. RNA Binding Protein Immunoprecipitation (RIP) and luciferase reporter assays were performed to identify the specific relationships between miR-338-3p and ST8SIA6-AS1/ non-POU domain containing octamer binding (NONO). RESULTS The expression of ST8SIA6-AS1 was apparently elevated in HCC cell. Silenced ST8SIA6-AS1 reduced proliferative, migratory and invasive ability of HCC cells. Moreover, ST8SIA6-AS1 targeted miR-338-3p to modulate the expression of NONO in HCC cells. CONCLUSIONS ST8SIA6-AS1 enhances the progression of HCC via miR-338-3p/NONO axis in vitro.
Collapse
Affiliation(s)
- Jinghua Kuai
- Department of Gastroenterology, Qilu Hospital of Shandong University, Qingdao 266000, Shandong, China
| | - Lijie Zheng
- Department of General Surgery, Qilu Hospital of Shandong University, No.758 Hefei Road, Shibei District, Qingdao 266000, Shandong, China.
| | - Xin Yi
- Department of General Surgery, Qilu Hospital of Shandong University, No.758 Hefei Road, Shibei District, Qingdao 266000, Shandong, China
| | - Zengli Liu
- Department of General Surgery, Qilu Hospital of Shandong University, No.758 Hefei Road, Shibei District, Qingdao 266000, Shandong, China
| | - Bo Qiu
- Department of General Surgery, Qilu Hospital of Shandong University, No.758 Hefei Road, Shibei District, Qingdao 266000, Shandong, China
| | - Zhihua Lu
- Department of General Surgery, Qilu Hospital of Shandong University, No.758 Hefei Road, Shibei District, Qingdao 266000, Shandong, China
| | - Yuanhui Jiang
- Department of General Surgery, Qilu Hospital of Shandong University, No.758 Hefei Road, Shibei District, Qingdao 266000, Shandong, China
| |
Collapse
|
15
|
Zhang XR, Shao JL, Li H, Wang L. Silencing of LINC00707 suppresses cell proliferation, migration, and invasion of osteosarcoma cells by modulating miR-338-3p/AHSA1 axis. Open Life Sci 2021; 16:728-736. [PMID: 34316513 PMCID: PMC8285991 DOI: 10.1515/biol-2021-0070] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 04/23/2021] [Accepted: 06/11/2021] [Indexed: 12/18/2022] Open
Abstract
Osteosarcoma is the most common type of primary malignant tumor of the bone, with a high metastatic rate and poor prognosis. Therefore, it is important to further elucidate the molecular mechanisms involved in the development of osteosarcoma and explore new molecular therapeutic targets. Long intergenic nonprotein-coding RNA 707 (LINC00707) is an oncogenic gene in several cancers. In this study, we further clarified its role and regulatory mechanism in osteosarcoma. We found that LINC00707 levels are significantly higher in the osteosarcoma cell lines SW 1353, HOS, U-2 OS, MG-63, and Saos-2 compared to those in human fetal osteoblastic cell line hFOB1.19. LINC00707 silencing suppressed cell proliferation, migration, and invasion of MG-63 and Saos-2 cells. Moreover, LINC00707 can act as a competitive endogenous RNA of miR-338-3p, and miR-338-3p inhibitor and AHSA1 overexpression alleviated the effect of LINC00707 silencing. In conclusion, we demonstrated high expression of LINC00707 in osteosarcoma cell lines and that silencing LINC00707 suppresses cell proliferation, migration, and invasion by targeting the miR-338-3p/AHSA1 axis in MG-63 and Saos-2 cells. These findings suggest that LINC00707 may serve as a potential target for osteosarcoma treatment.
Collapse
Affiliation(s)
- Xiao-Rong Zhang
- Department of Orthopedic and Traumatology, The First Affiliated Hospital of Jinan University, 601 West Huangpu Avenue, Tianhe District, Guangzhou 510630, China
| | - Jian-Li Shao
- Department of Orthopedic and Traumatology, The First Affiliated Hospital of Jinan University, 601 West Huangpu Avenue, Tianhe District, Guangzhou 510630, China
| | - Heng Li
- Department of Orthopedic, Center in Zhanjiang City People's Hospital, Zhanjiang 524045, China
| | - Liang Wang
- Department of Oncology, The First Affiliated Hospital of Jinan University, 601 West Huangpu Avenue, Tianhe District, Guangzhou 510630, China
| |
Collapse
|
16
|
Cao R, Shao J, Zhang W, Lin Y, Huang Z, Li Z. Silencing long intergenic non-protein coding RNA 00987 inhibits proliferation, migration, and invasion of osteosarcoma cells by sponging miR-376a-5p to regulate FNBP1 expression. Discov Oncol 2021; 12:18. [PMID: 35201476 PMCID: PMC8777572 DOI: 10.1007/s12672-021-00412-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 05/24/2021] [Indexed: 01/19/2023] Open
Abstract
High expression of long intergenic non-protein coding RNA 987 (LINC00987) is strongly associated with low overall survival of osteosarcoma; however, its role in osteosarcoma remains unclear. This study explored the biological function and underlying mechanism of LINC00987 in osteosarcoma. In this study, LINC00987 expression in osteosarcoma cells was analyzed using Cancer Cell Line Encyclopedia and qRT-PCR. The proliferation and migration and invasion in osteosarcoma cells were evaluated using Cell Counting Kit-8 and Transwell assays, respectively. Bioinformatic analysis was used to predict the LINC00987-bound miRNAs and miR-376a-5p-bound mRNAs. Dual-luciferase reporter assays were used to assess the interaction between miR-376a-5p, LINC00987, and forming-binding protein 1 (FNBP1). FNBP1 expression was measured by western blotting. LINC00987 was found to be upregulated in osteosarcoma cells. LINC00987 silencing suppressed proliferation, migration, and invasion of osteosarcoma cells. Additionally, miR-376a-5p expression was downregulated in osteosarcoma cells. miR-376a-5p knockdown reversed the effect of LINC00987 silencing on the biological function of osteosarcoma cell. miR-376a-5p was found to target LINC00987 and FNBP1. FNBP1protein level was increased in osteosarcoma cells; however, it was inhibited by silencing LINC00987 and enhanced by silencing miR-376a-5p. In conclusions, this study suggests LINC00987 silencing inhibits osteosarcoma cell proliferation, migration, and invasion by sponging miR-376a-5p to regulate FNBP1 expression. LINC00987 as a potential therapeutic target for osteosarcoma.
Collapse
Affiliation(s)
- Riliang Cao
- Department of Orthopedics, First Affiliated Hospital of Jinan University, 613 W. Huangpu Avenue, Tianhe District, Guangzhou, 510630, China
| | - Jianli Shao
- Department of Orthopedics, First Affiliated Hospital of Jinan University, 613 W. Huangpu Avenue, Tianhe District, Guangzhou, 510630, China
| | - Wencai Zhang
- Department of Orthopedics, First Affiliated Hospital of Jinan University, 613 W. Huangpu Avenue, Tianhe District, Guangzhou, 510630, China
| | - Yongxin Lin
- Department of Orthopedics, First Affiliated Hospital of Jinan University, 613 W. Huangpu Avenue, Tianhe District, Guangzhou, 510630, China
| | - Zerong Huang
- Department of Orthopedics, First Affiliated Hospital of Jinan University, 613 W. Huangpu Avenue, Tianhe District, Guangzhou, 510630, China
| | - Zhizhong Li
- Department of Orthopedics, First Affiliated Hospital of Jinan University, 613 W. Huangpu Avenue, Tianhe District, Guangzhou, 510630, China.
| |
Collapse
|
17
|
Yi Q, Cui H, Liao Y, Xiong J, Ye X, Sun W. A minor review of microRNA-338 exploring the insights of its function in tumorigenesis. Biomed Pharmacother 2021; 139:111720. [PMID: 34243620 DOI: 10.1016/j.biopha.2021.111720] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 12/29/2022] Open
Abstract
MicroRNAs(miRNAs) are small non-coding RNAs which have a critical role in various biological processes via direct binding and post-transcriptionally regulating targeted genes expression. More than one-half of human genes were regulated by miRNAs and their aberrant expression was detected in various human diseases, including cancers. miRNA-338 is a new identified miRNA and increasing evidence show that miRNA-338 participates in the progression of lots of cancers, such as lung cancer, hepatocellular cancer, breast cancer, glioma, and so on. Although a range of targets and signaling pathways such as MACC1 and Wnt/β-catenin signaling pathway were illustrated to be regulated by miRNA-338, which functions in tumor progression are still ambiguous and the underlying molecular mechanisms are also unclear. Herein, we reviewed the latest studies in miRNA-338 and summarized its roles in different type of human tumors, which might provide us new idea for further investigations and potential targeted therapy.
Collapse
Affiliation(s)
- Qian Yi
- Shenzhen Key Laboratory of Tissue Engineering, Department of Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University), Shenzhen 518035, Guangdong, China; Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Hanwei Cui
- The Central Laboratory and Medical Genetics & Molecular Diagnostic Center, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University), Shenzhen 518035, Guangdong, China
| | - Yi Liao
- The Central Laboratory and Medical Genetics & Molecular Diagnostic Center, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University), Shenzhen 518035, Guangdong, China
| | - Jianyi Xiong
- Shenzhen Key Laboratory of Tissue Engineering, Department of Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University), Shenzhen 518035, Guangdong, China.
| | - Xiufeng Ye
- The Central Laboratory and Medical Genetics & Molecular Diagnostic Center, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University), Shenzhen 518035, Guangdong, China.
| | - Weichao Sun
- Shenzhen Key Laboratory of Tissue Engineering, Department of Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University), Shenzhen 518035, Guangdong, China.
| |
Collapse
|
18
|
Cao Y, Xie X, Li M, Gao Y. CircHIPK2 Contributes to DDP Resistance and Malignant Behaviors of DDP-Resistant Ovarian Cancer Cells Both in vitro and in vivo Through circHIPK2/miR-338-3p/CHTOP ceRNA Pathway. Onco Targets Ther 2021; 14:3151-3165. [PMID: 34012271 PMCID: PMC8128508 DOI: 10.2147/ott.s291823] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 04/01/2021] [Indexed: 12/17/2022] Open
Abstract
Background Cisplatin (DDP) is standard-of-care and first-line management for ovarian cancer (OvCa). Circular RNA HIPK2 (circHIPK2) is abnormally upregulated in serum of OvCa patients. However, its role in DDP resistance remains unclear. Methods Expression of cirHIPK2, microRNA (miR)-338-3p and chromatin target of protein arginine methyltransferase (CHTOP) was detected by quantitative reverse transcription PCR and Western blotting. Functional experiments were performed using cell counting kit-8 assay, flow cytometry, transwell assays, Western blotting, and xenograft experiment. The interaction among cirHIPK2, miR-338-3p and CHTOP was confirmed by dual-luciferase reporter assay and RNA pull-down assay. Results Expression of circHIPK2 and CHTOP was upregulated, and miR-338-3p was downregulated in human DDP-resistant OvCa tumors and cells. Blocking circHIPK2 could promote apoptosis and suppress the 50% inhibitory concentration (IC50) of DDP, cell proliferation, cell cycle entrance, migration and invasion in SKOV3/DDP and A2780/DDP cells. Allied with that was decreased B cell lymphoma (Bcl)-2, matrix metalloproteinase 2 (MMP2) and MMP9 levels, and increased Bcl-2-associated X protein (Bax) level. Similarly, overexpression of miR-338-3p functioned suppressive role in SKOV3/DDP and A2780/DDP cells. MiR-338-3p was a target for circHIPK2, and CHTOP was targeted by miR-338-3p, whereas silencing miR-338-3p counteracted the role of circHIPK2 knockdown, and restoring CHTOP either cancelled miR-338-3p role. The growth of A2780/DDP cells in nude mice was restrained by silencing circHIPK2 under DDP treatment or not. Conclusion CircHIPK2 might be a tumor promoter in OvCa and was associated with DDP resistance. Silencing circHIPK2 might suppress DDP-resistant OvCa through regulating miR-338-3p/CHTOP axis.
Collapse
Affiliation(s)
- Yang Cao
- Department of Gynaecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang City, Liaoning Province, People's Republic of China
| | - Xin Xie
- Department of Teaching and Research Center, Liaoning University of Traditional Chinese Medicine, Shenyang City, Liaoning Province, People's Republic of China
| | - Mingzhu Li
- Department of Integrated Traditional Chinese and Western Medicine Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang City, Liaoning Province, People's Republic of China
| | - Yuhua Gao
- Department of Gynaecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang City, Liaoning Province, People's Republic of China
| |
Collapse
|
19
|
Moghbeli M. Molecular interactions of miR-338 during tumor progression and metastasis. Cell Mol Biol Lett 2021; 26:13. [PMID: 33827418 PMCID: PMC8028791 DOI: 10.1186/s11658-021-00257-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 03/25/2021] [Indexed: 02/08/2023] Open
Abstract
Background Cancer, as one of the main causes of human deaths, is currently a significant global health challenge. Since the majority of cancer-related deaths are associated with late diagnosis, it is necessary to develop minimally invasive early detection markers to manage and reduce mortality rates. MicroRNAs (miRNAs), as highly conserved non-coding RNAs, target the specific mRNAs which are involved in regulation of various fundamental cellular processes such as cell proliferation, death, and signaling pathways. MiRNAs can also be regulated by long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs). They are highly stable in body fluids and have tumor-specific expression profiles, which suggest their suitability as efficient non-invasive diagnostic and prognostic tumor markers. Aberrant expression of miR-338 has been widely reported in different cancers. It regulates cell proliferation, migration, angiogenesis, and apoptosis in tumor cells. Main body In the present review, we have summarized all miR-338 interactions with other non-coding RNAs (ncRNAs) and associated signaling pathways to clarify the role of miR-338 during tumor progression. Conclusions It was concluded that miR-338 mainly functions as a tumor suppressor in different cancers. There were also significant associations between miR-338 and other ncRNAs in tumor cells. Moreover, miR-338 has a pivotal role during tumor progression using the regulation of WNT, MAPK, and PI3K/AKT signaling pathways. This review highlights miR-338 as a pivotal ncRNA in biology of tumor cells.
Collapse
Affiliation(s)
- Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
20
|
Wang CQ, Wang XM, Li BL, Zhang YM, Wang L. Arbutin suppresses osteosarcoma progression via miR-338-3p/MTHFD1L and inactivation of the AKT/mTOR pathway. FEBS Open Bio 2020; 11:289-299. [PMID: 33146000 PMCID: PMC7780106 DOI: 10.1002/2211-5463.13024] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/18/2020] [Accepted: 11/02/2020] [Indexed: 12/24/2022] Open
Abstract
Arbutin, a glycoside extracted from the plant Arctostaphylos uva‐ursi, has been previously reported to possess antioxidant, anti‐inflammatory and anticancer effects. Here, we investigated whether arbutin affects the proliferation of the cells of the osteosarcoma (OS) cell lines MG‐63 and SW1353. Arbutin suppressed OS cell viability in a dose‐ and time‐dependent manner, as shown by Cell Counting Kit‐8 assay. Furthermore, arbutin exposure decreased the protein levels of MTHFD1L, CCND1 and phosphorylated‐protein kinase B (AKT)/phosphorylated‐mammalian target of rapamycin (mTOR). Potential upstream miRNAs of MTHFD1L were predicted using TargetScan, PICTAR5, miRanda and miRWalk. We performed luciferase activity assays to show that miR‐338‐3p directly targets and negatively regulates the expression of MTHFD1L. Knockdown of miR‐338‐3p promoted cell invasion, migration and proliferation in arbutin‐treated OS cells via MTHFD1L. In summary, our data suggest that arbutin inhibits OS cell proliferation, migration and invasion via miR‐338‐3p/MTHFD1L and by inactivating the AKT/mTOR pathway.
Collapse
Affiliation(s)
- Cheng-Qun Wang
- Department of Joint Surgery, Affiliated Hospital of Jining Medical University, China
| | - Xiu-Mei Wang
- Electroencephalogram Room, Affiliated Hospital of Jining Medical University, China
| | - Bing-Liang Li
- Department of Joint Surgery, Affiliated Hospital of Jining Medical University, China
| | - Yuan-Min Zhang
- Department of Joint Surgery, Affiliated Hospital of Jining Medical University, China
| | - Lei Wang
- Department of Joint Surgery, Affiliated Hospital of Jining Medical University, China
| |
Collapse
|
21
|
Zhang H, Wang J, Ren T, Huang Y, Yu Y, Chen C, Huang Q, Guo W. LncRNA CASC15 is Upregulated in Osteosarcoma Plasma Exosomes and CASC15 Knockdown Inhibits Osteosarcoma Progression by Regulating miR-338-3p/RAB14 Axis. Onco Targets Ther 2020; 13:12055-12066. [PMID: 33262606 PMCID: PMC7700090 DOI: 10.2147/ott.s282053] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 11/05/2020] [Indexed: 12/16/2022] Open
Abstract
Background Currently, plenty of studies have demonstrated that lncRNAs can act as crucial roles during the progression of various tumors, including osteosarcoma (OS), and emerging evidences indicated that lncRNAs are abundant and stable in exosomes. The objective of this study is to reveal the dysregulated lncRNAs in OS plasma exosomes and explore their functions in OS. Materials and Methods Microarray was performed to analyze dysregulated exosomal lncRNAs. Western blot, qRT-PCR assays, and Dual-luciferase reporter assay were used to verify the interaction among cancer susceptibility 15 (CASC15), miR-338-3p, and RAB14. Cck-8, colony formation assay, and transwell assay were performed to explore and characterize the effects of CASC15 on OS cells. Animal experiments were used to verify the effects of CASC15 in vivo. Results Upregulated CASC15 was observed in OS plasma exosomes compared with control, and the same expression was observed in the OS tissues and cell lines. Further assays indicated that CASC15 knockdown could restrain the proliferation, migration, and invasion of OS cells, and inhibit the growth of OS in xenograft models. Furthermore, our results demonstrated CASC15 regulated OS progression via acting as miR-338-3p sponge, and RAB14 was a direct downstream target of miR-338-3p. Rescue experiments verified CASC15 promotes OS cell growth and metastasis by upregulating RAB14 expression. Conclusion Overall, our findings indicate that CASC15 plays a key role in OS progression by targeting the miR-338-3p/RAB14 axis and can serve as a possible therapeutic target for OS patients.
Collapse
Affiliation(s)
- Hongliang Zhang
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, People's Republic of China.,Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, People's Republic of China
| | - Jun Wang
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, People's Republic of China.,Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, People's Republic of China
| | - Tingting Ren
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, People's Republic of China.,Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, People's Republic of China
| | - Yi Huang
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, People's Republic of China.,Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, People's Republic of China
| | - Yiyang Yu
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, People's Republic of China.,Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, People's Republic of China
| | - Chenglong Chen
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, People's Republic of China.,Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, People's Republic of China
| | - Qingshan Huang
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, People's Republic of China.,Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, People's Republic of China
| | - Wei Guo
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, People's Republic of China.,Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, People's Republic of China
| |
Collapse
|
22
|
Jin B, Jin D, Zhuo Z, Zhang B, Chen K. MiR-1224-5p Activates Autophagy, Cell Invasion and Inhibits Epithelial-to-Mesenchymal Transition in Osteosarcoma Cells by Directly Targeting PLK1 Through PI3K/AKT/mTOR Signaling Pathway. Onco Targets Ther 2020; 13:11807-11818. [PMID: 33235467 PMCID: PMC7680192 DOI: 10.2147/ott.s274451] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 09/23/2020] [Indexed: 12/12/2022] Open
Abstract
Background Osteosarcoma (OS) is one of the most common malignant bone tumors with a poor overall prognosis. MiR-1224-5p plays an important role in cancer, but its function and mechanism in OS have not been studied. Materials and Methods The expression of miR-1224-5p and PLK1 was detected by qRT-PCR in OS cells, adjacent tissues, and cell lines. Dual-luciferase reporter gene assay was used to verify the interaction between miR-1224-5p and PLK1. The expression of miR-1224-5p and PLK1 was intervened by transfection with miR-1224-5p mimic, NC mimic, pc-NC and PLK1, respectively. MTT, colony formation assay, Transwell and flow cytometry were used to observe the cell proliferation, invasion and apoptosis. Western blot was used to detect the expression levels of PLK1, PI3K/AKT/mTOR signaling pathway-related proteins, autophagy-related proteins, and epithelial-mesenchymal transition (EMT)-related proteins in the cells. Results We found that miR-1224-5p was down-regulated and PLK1 expression was up-regulated in OS tissues and cells. On the other hand, it is further confirmed that PLK1 was a target gene of miR-1224-5p. Overexpression of miR-1224-5p inhibited the proliferation, invasion while promoted the apoptosis of OS cells, whereas overexpression of PLK1 promoted the proliferation, invasion and inhibited the apoptosis of OS cells. In the miR-1224-5p group (overexpression of miR-1224-5p), PI3K, AKT, and mTOR protein phosphorylation levels were significantly reduced, while autophagic activity was significantly activated, and the degree of EMT was significantly reduced. But the results in the PLK1 group (overexpression of PLK1) were the opposite. In addition, overexpression of miR-1224-5p reversed the effect of PLK1 upregulation on OS cells. Conclusion MiR-1224-5p targets PLK1 to inhibit PI3K/AKT/mTOR signaling pathway, thus mediating the proliferation, invasion, apoptosis, autophagy and EMT in OS cells.
Collapse
Affiliation(s)
- Bicheng Jin
- Department of Surgery, Guizhou Electric Power Staff Hospital, Guiyang, Guizhou Province, People's Republic of China
| | - Dongfang Jin
- Department of Clinical Laboratory, Jinhua People's Hospital, Jinhua, Zhejiang Province, People's Republic of China
| | - Zhaozhen Zhuo
- Prenatal Diagnosis Center, Guizhou Provincial People's Hospital, Guiyang, Guizhou Province, People's Republic of China
| | - Bo Zhang
- Department of Surgery, Guizhou Electric Power Staff Hospital, Guiyang, Guizhou Province, People's Republic of China
| | - Kun Chen
- Guizhou Provincial People's Hospital Scientific Research Center Laboratory, Guiyang, Guizhou Province, People's Republic of China
| |
Collapse
|
23
|
Song W, Wang K, Yang X, Dai W, Fan Z. Long non‑coding RNA BANCR mediates esophageal squamous cell carcinoma progression by regulating the IGF1R/Raf/MEK/ERK pathway via miR‑338‑3p. Int J Mol Med 2020; 46:1377-1388. [PMID: 32945416 PMCID: PMC7447317 DOI: 10.3892/ijmm.2020.4687] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Accepted: 06/26/2020] [Indexed: 12/14/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is a type of digestive tract malignant tumor that severely threatens human health. The long non‑coding RNA BRAF activated non‑coding RNA (BANCR) and insulin‑like growth factor 1 receptor (IGF1R) are associated with various types of cancer; however, it remains unclear whether BANCR can regulate IGF1R expression in ESCC. In the present study, the expression levels of BANCR, IGF1R mRNA and microRNA‑338‑3p (miRNA/miR‑338‑3p) in ESCC tissues or cells were detected by reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR). The levels of IGF1R, E‑cadherin, N‑cadherin, Vimentin, p‑Raf‑1, p‑MEK1/2 and p‑ERK1/2 were measured by western blot analysis. The proliferation, migration and invasion of ESCC cells were determined by 3‑(4,5‑dimethylthiazol‑2‑yl)‑2,5‑diphenyltetrazolium bromide (MTT) or Transwell assays. The relationship between miR‑338‑3p and BANCR or IGF1R was predicted using starBase2.0 and confirmed by dual‑luciferase reporter assay. The role of BANCR in ESCC in vivo was confirmed through a tumor xenograft assay. It was found that BANCR and IGF1R were upregulated, while miR‑338‑3p was downregulated in ESCC tissues and cells. Both BANCR and IGF1R knockdown suppressed the proliferation, migration, invasion and epithelial‑mesenchymal transition (EMT) of ESCC cells. IGF1R enhancement reversed BANCR knockdown‑mediated effects on the proliferation, migration, invasion and EMT of ESCC cells. BANCR regulated the Raf/MEK/ERK pathway by regulating IGF1R expression. Notably, BANCR regulated IGF1R expression by sponging miR‑338‑3p. Moreover, BANCR silencing inhibited tumor growth in vivo. On the whole, the findings of the present study demonstrate that BANCR inhibition blocks ESCC progression by inactivating the IGF1R/Raf/MEK/ERK pathway by sponging miR‑338‑3p.
Collapse
Affiliation(s)
- Wei Song
- Department of Digestive Endoscopy, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029
- Department of Gastroenterology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Kuangjing Wang
- Department of Digestive Endoscopy, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029
| | - Xiaozhong Yang
- Department of Gastroenterology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Weijie Dai
- Department of Gastroenterology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Zhining Fan
- Department of Digestive Endoscopy, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029
| |
Collapse
|
24
|
Long noncoding RNA XIST knockdown suppresses the growth of colorectal cancer cells via regulating microRNA-338-3p/PAX5 axis. Eur J Cancer Prev 2020; 30:132-142. [PMID: 32826710 DOI: 10.1097/cej.0000000000000596] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Colorectal cancer is one of the most common human cancers worldwide. Long noncoding RNAs (lncRNAs) and microRNAs (miRNAs) have been reported as the regulators in cancers. The purpose of this study was to reveal the functional mechanisms of lncRNA x inactive specific transcript (XIST) and miR-338-3p in colorectal cancer cells. METHODS The transcription level and protein level of genes were assessed by quantitative real-time PCR (qRT-PCR) and western blot assay, respectively. 3-(4, 5-dimethyl-2-thiazolyl)-2, 5-diphenyl-2-H-tetrazolium bromide (MTT) assay and flow cytometry analysis were used to determine cell proliferation ability and apoptosis rate, respectively. In addition, cell migratory ability and invasive ability were measured using transwell assay. Besides, the interaction between miR-338-3p and XIST or paired box 5 (PAX5) was predicted by starBase or TargetScan and then verified by the dual-luciferase reporter assay. RESULTS XIST and PAX5 expression were increased, and miR-338-3p expression was decreased in colorectal cancer tissues and cells. XIST knockdown significantly repressed cell proliferation, migration and invasion, and accelerated apoptosis in colorectal cancer cells. Interestingly, XIST directly downregulated miR-338-3p expression to increase PAX5 level. As expected, XIST knockdown inhibited colorectal cancer cell growth by modulating miR-338-3p expression. Furthermore, miR-338-3p suppressed cell growth via downregulation of PAX5 level in colorectal cancer cells. CONCLUSION Our results demonstrated that the downregulation of XIST inhibited cell proliferation, migration and invasion, and induced apoptosis through modulating miR-338-3p/PAX5 axis in colorectal cancer cells, providing potential target for the prevention and treatment of colorectal cancer.
Collapse
|
25
|
Wang TX, Tan WL, Huang JC, Cui ZF, Liang RD, Li QC, Lu H. Identification of aberrantly methylated differentially expressed genes targeted by differentially expressed miRNA in osteosarcoma. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:373. [PMID: 32355817 PMCID: PMC7186728 DOI: 10.21037/atm.2020.02.74] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background Osteosarcoma (OS) is the most common primary bone tumors diagnosed in children and adolescents. Recent studies have shown a prognostic role of DNA methylation in various cancers, including OS. The aim of this study was to identify the aberrantly methylated genes that are prognostically relevant in OS. Methods The differentially expressed mRNAs, miRNAs and methylated genes (DEGs, DEMs and DMGs respectively) were screened from various GEO databases, and the potential target genes of the DEMs were predicted by the RNA22 program. The protein-protein interaction (PPI) networks were constructed using the STRING database and visualized by Cytoscape software. The functional enrichment and survival analyses of the screened genes was performed using the R software. Results Forty-seven downregulated hypermethylated genes and three upregulated hypomethylated genes were identified that were enriched in cell activation, migration and proliferation functions, and were involved in cancer-related pathways like JAK-STAT and PI3K-AKT. Eight downregulated hypermethylated tumor suppressor genes (TSGs) were identified among the screened genes based on the TSGene database. These hub genes are likely involved in OS genesis, progression and metastasis, and are potential prognostic biomarkers and therapeutic targets. Conclusions TSGs including PYCARD, STAT5A, CXCL12 and CXCL14 were aberrantly methylated in OS, and are potential prognostic biomarkers and therapeutic targets. Our findings provide new insights into the role of methylation in OS progression.
Collapse
Affiliation(s)
- Ting-Xuan Wang
- Department of Orthopedics, The Fifth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Zhuhai 519000, China
| | - Wen-Le Tan
- Department of Orthopedics, Luoding People's Hospital, Luoding 527200, China
| | - Jin-Cheng Huang
- Department of Orthopedics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou 450003, China
| | - Zhi-Fei Cui
- Department of Orthopedics, The Fifth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Zhuhai 519000, China
| | - Ri-Dong Liang
- Department of Orthopedics, Southern Medical University Affiliated Nanhai Hospital, Southern Medical University, Foshan 523800, China
| | - Qing-Chu Li
- Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Academy of Orthopedics, Southern Medical University, Guangzhou 510000, China
| | - Hai Lu
- Department of Orthopedics, The Fifth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Zhuhai 519000, China
| |
Collapse
|
26
|
Niu Q, Liu Z, Gao J, Wang Q. MiR-338-3p Enhances Ovarian Cancer Cell Sensitivity to Cisplatin by Downregulating WNT2B. Yonsei Med J 2019; 60:1146-1156. [PMID: 31769245 PMCID: PMC6881712 DOI: 10.3349/ymj.2019.60.12.1146] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/26/2019] [Accepted: 10/16/2019] [Indexed: 02/07/2023] Open
Abstract
PURPOSE Chemoresistance is a concern in ovarian cancer patients, in whom survival remains. MicroRNA, a novel class of small RNAs, have frequently been found to be dysregulated in human malignancies and to act as negative regulators of gene expression. This study aimed to explore the function of miR-338-3p in cisplatin resistance in ovarian cancer and potential molecular mechanisms thereof. MATERIALS AND METHODS The expression levels of miR-338-3p and WNT2B in ovarian cancer tissues and cells were estimated by real-time quantitative polymerase chain reaction (RT-qPCR). In addition, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazol-3-ium bromide (MTT), transwell, and flow cytometry assays were used to assess biological role of miR-338-3p in vitro. Western blot assay was conducted to measure protein expression of WNT2B, epithelial-mesenchymal transition (EMT)-related proteins, and apoptosis-related proteins. The relationship between miR-338-3p and WNT2B was confirmed by dual-luciferase reporter. Finally, a xenograft tumor model was developed to explore the effects of overexpression of miR-338-3p on tumor growth in ovarian cancer in vivo. RESULTS MiR-338-3p was downregulated in cisplatin resistant ovarian cancer tissues and cells. Mechanistically, high expression of miR-338-3p enhanced cell sensitivity to cisplatin by inhibiting proliferation, motility, and EMT and by promoting apoptosis via targeting WNT2B expression in vitro. Furthermore, overexpression of miR-338-3p increased cisplatin sensitivity among ovarian cancer in an in vivo xenograft tumor model. CONCLUSION MiR-338-3p enhances the sensitivity of ovarian cancer cells to cisplatin by downregulating WNT2B.
Collapse
Affiliation(s)
- Qin Niu
- Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang, China
| | - Zhenghong Liu
- Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang, China.
| | - Jia Gao
- Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang, China
| | - Qiao Wang
- Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang, China
| |
Collapse
|
27
|
Viera GM, Salomao KB, de Sousa GR, Baroni M, Delsin LEA, Pezuk JA, Brassesco MS. miRNA signatures in childhood sarcomas and their clinical implications. Clin Transl Oncol 2019; 21:1583-1623. [PMID: 30949930 DOI: 10.1007/s12094-019-02104-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 03/27/2019] [Indexed: 02/06/2023]
Abstract
Progresses in multimodal treatments have significantly improved the outcomes for childhood cancer. Nonetheless, for about one-third of patients with Ewing sarcoma, rhabdomyosarcoma, or osteosarcoma steady remission has remained intangible. Thus, new biomarkers to improve early diagnosis and the development of precision-targeted medicine remain imperative. Over the last decade, remarkable progress has been made in the basic understanding of miRNAs function and in interpreting the contribution of their dysregulation to cancer development and progression. On this basis, this review focuses on what has been learned about the pivotal roles of miRNAs in the regulation of key genes implicated in childhood sarcomas.
Collapse
Affiliation(s)
- G M Viera
- Ribeirao Preto School of Medicine, University of Sao Paulo, Ribeirao Preto, Brasil
| | - K B Salomao
- Ribeirao Preto School of Medicine, University of Sao Paulo, Ribeirao Preto, Brasil
| | - G R de Sousa
- Ribeirao Preto School of Medicine, University of Sao Paulo, Ribeirao Preto, Brasil
| | - M Baroni
- Ribeirao Preto School of Medicine, University of Sao Paulo, Ribeirao Preto, Brasil
| | - L E A Delsin
- Ribeirao Preto School of Medicine, University of Sao Paulo, Ribeirao Preto, Brasil
| | - J A Pezuk
- Anhanguera University of Sao Paulo, UNIAN/SP, Sao Paulo, Brasil
| | - M S Brassesco
- Faculty of Philosophy, Sciences and Letters at Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brasil.
- Departamento de Biologia, FFCLRP-USP, Av. Bandeirantes, 3900, Bairro Monte Alegre, Ribeirao Preto, SP, CEP 14040-901, Brazil.
| |
Collapse
|
28
|
Wei D, Xin Y, Rong Y, Li Y, Zhang C, Chen Q, Qin S, Wang W, Hao Y. A Mesoporous Gd-MOF with Lewis Basic Sites for 5-Fu Delivery and Inhibition of Human Lung Cancer Cells In Vivo and In Vitro. J Inorg Organomet Polym Mater 2019. [DOI: 10.1007/s10904-019-01305-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
29
|
Huang C, Gan D, Luo F, Wan S, Chen J, Wang A, Li B, Zhu X. Interaction Mechanisms Between the NOX4/ROS and RhoA/ROCK1 Signaling Pathways as New Anti- fibrosis Targets of Ursolic Acid in Hepatic Stellate Cells. Front Pharmacol 2019; 10:431. [PMID: 31130857 PMCID: PMC6510285 DOI: 10.3389/fphar.2019.00431] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 04/04/2019] [Indexed: 01/14/2023] Open
Abstract
Background Studies have shown that both NOX4 and RhoA play essential roles in fibrosis and that they regulate each other. In lung fibrosis, NOX4/ROS is located upstream of the RhoA/ROCK1 signaling pathway, and the two molecules are oppositely located in renal fibrosis. Currently, no reports have indicated whether the above mechanisms or other regulatory mechanisms exist in liver fibrosis. Objectives To investigate the effects of the NOX4/ROS and RhoA/ROCK1 signaling pathways on hepatic stellate cell (HSC)-T6 cells, the interaction mechanisms of the two pathways, and the impact of UA on the two pathways to elucidate the role of UA in the reduction of hepatic fibrosis and potential mechanisms of HSC-T6 cell proliferation, migration, and activation. Methods Stable cell lines were constructed using the lentiviral transduction technique. Cell proliferation, apoptosis, migration, and invasion were examined using the MTS, TdT-mediated dUTP nick-end labeling, cell scratch, and Transwell invasion assays, respectively. The DCFH-DA method was used to investigate the ROS levels in each group. RT-qPCR and western blotting techniques were utilized to assess the mRNA and protein expression in each group. CoIP and the Biacore protein interaction analysis systems were used to evaluate protein interactions. Results The NOX4/ROS and RhoA/ROCK1 signaling pathways promoted the proliferation, migration, and activation of HSCs. UA inhibited cell proliferation, migration, and activation by inhibiting the activation of the two signaling pathways, but the mechanism of apoptosis was independent of these two pathways. The NOX4/ROS pathway was upstream of and positively regulated the RhoA/ROCK1 pathway in HSCs. No direct interaction between the NOX4 and RhoA proteins was detected. Conclusion The NOX4/ROS and RhoA/ROCK1 signaling pathways are two critical signaling pathways in a series of behavioral processes in HSCs, and NOX4/ROS regulates RhoA/ROCK1 through an indirect pathway to control the activation of HSCs. Additionally, NOX4/ROS and RhoA/ROCK1 constitute a new target for UA antifibrosis treatment.
Collapse
Affiliation(s)
- Chenkai Huang
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Dakai Gan
- Department One of Liver Disease, The Ninth Hospital of Nanchang, Nanchang, China
| | - Fangyun Luo
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Sizhe Wan
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jiang Chen
- Digestive Disease Research Institute of Jiangxi Province, Nanchang, China
| | - Anjiang Wang
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Bimin Li
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xuan Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
30
|
Jin H, Liu P, Kong L, Fei X, Gao Y, Wu T, Sun D, Tan X. Identification of RE1-Silencing Transcription Factor as a Promoter of Metastasis in Pancreatic Cancer. Front Oncol 2019; 9:291. [PMID: 31041193 PMCID: PMC6476950 DOI: 10.3389/fonc.2019.00291] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 03/29/2019] [Indexed: 12/24/2022] Open
Abstract
Pancreatic cancer is characterized by its rapid progression and early metastasis. This requires further elucidation of the key promoters for its progression and metastasis. In this study, we identified REST as the hub gene of a gene module which is closely associated with cancer stage by weighted gene correlation network analysis. Validation with the TCGA database, western blot analysis of human pancreatic cancer cell lines (AsPC-1, Capan-2, SW-1990, and PANC-1) and immunohistochemical analysis of paraffin-embedded pancreatic cancer tissue sections showed that REST was enriched in tissue samples of advanced stage and metastatic phenotype cell lines. Survival analysis with the TCGA database and our own follow-up data suggested that patients with higher expression level of REST showed worse overall survival rate. In vitro functional experiments suggested that knockdown of REST suppressed proliferation, migration, invasion and epithelial-mesenchymal transition of AsPC-1 and PANC-1 cells. In vivo experiments (a subcutaneous BALB/c nude mouse model and a superior mesenteric vein injection BALB/c nude mouse model) suggested that knockdown of REST suppressed growth and metastasis of xenograft tumor. Finally, we investigated the underlying molecular mechanism of REST and identified REST as a potential downstream target of MAPK signaling pathway. In conclusion, our results of bioinformatic analysis, in vitro and in vivo functional analysis suggested that REST may serve as a promoter of metastasis in pancreatic cancer.
Collapse
Affiliation(s)
- Haoyi Jin
- Department of Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Peng Liu
- Thyroid and Pancreatic Surgery Ward, Shengjing Hospital of China Medical University, Shenyang, China
| | - Lingming Kong
- Department of Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiang Fei
- Department of Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yang Gao
- Department of Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Tianyu Wu
- Department of Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Defeng Sun
- Department of Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaodong Tan
- Thyroid and Pancreatic Surgery Ward, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
31
|
Yang W, Shan Z, Zhou X, Peng L, Zhi C, Chai J, Liu H, Yang J, Zhang Z. Knockdown of lncRNA GHET1 inhibits osteosarcoma cells proliferation, invasion, migration and EMT in vitro and in vivo. Cancer Biomark 2019; 23:589-601. [PMID: 30475755 DOI: 10.3233/cbm-181863] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Osteosarcoma is the most common primary malignant skeleton tumor that derives from mesenchymal cells. Emerging evidences have identified the vital role of long non-coding RNAs (lncRNAs) in the development of osteosarcoma. In this study, we aimed to investigate the role of lncRNA gastric carcinoma highly expressed transcript 1 (GHET1) in osteosarcoma progression. METHODS The expression levels of relevant genes in clinical samples and cell lines were determined by quantitative real-time PCR. Cell proliferation was determined by CCK8 and cell colony formation assays. Transwell assay was used to detect the invasion and migration of osteosarcoma cells. Cell apoptosis and cell cycle were detected by flow cytometry. Protein levels were detected by western blot. In vivo tumor growth was investigated in the xenograft nude mice model. To determine whether growth inhibition and apoptosis are responsible for antitumor activity of silencing GHET1, immunohistochemistry for proliferation and TUNEL assay was performed in xenograft tissues. In vivo lung metastasis was performed to detect the effect of GHET1 on cell metastasis ability. RESULTS Our results revealed that GHET1 was up-regulated in osteosarcoma tissues compared to normal tissues. GHET1 was also increased in osteosarcoma cell lines compared to normal osteoplastic cell line. The up-regulation of GHET1 was significantly associated with TNM stage, distant metastasis and lymph node metastasis in patients with osteosarcoma. In vitro studies showed that silencing GHET1 in MG-63 and U2OS cells inhibited cell proliferation, cell invasion and migration and epithelial-to-mesenchymal transition (EMT), promoted cell apoptotic rate, and also caused an increase in cell population at G0/G1 phase with a decrease in cell population at S phase. Overexpression of GHET1 promoted the proliferation, invasion and migration of osteosarcoma cells. Importantly, silencing GHET1 inhibited tumor growth and tumor metastasis in mice MG-63-xenograft model in association with changes of EMT-related genes, reduced expression of Ki-67 and promotion of apoptosis. CONCLUSION GHET1 was up-regulated in osteosarcoma tissues and cell lines, inhibited cell apoptosis, promoted cell proliferation, invasion and migration by affecting EMT in vitro, and was correlated with the tumor growth and metastasis in vivo. GHET1 may be a potential therapeutic target of osteosarcoma treatment.
Collapse
Affiliation(s)
- Wei Yang
- Department of General Surgery, Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Zhiming Shan
- Clinical Laboratory, The Affiliated Children Hospital of Zhengzhou University, The Children's Hospital of Henan Province, Zhengzhou Children's Hospital, Zhengzhou, Henan, China
| | - Xinfang Zhou
- Department of General Surgery, Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Liangqun Peng
- Department of General Surgery, Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Chongyang Zhi
- Department of General Surgery, Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Junhui Chai
- Department of General Surgery, Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Hongxing Liu
- Department of General Surgery, Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Junmei Yang
- Clinical Laboratory, The Affiliated Children Hospital of Zhengzhou University, The Children's Hospital of Henan Province, Zhengzhou Children's Hospital, Zhengzhou, Henan, China
| | - Zhandong Zhang
- Department of General Surgery, Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| |
Collapse
|
32
|
Zhang Z, Luo G, Yu C, Yu G, Jiang R, Shi X. MicroRNA-493-5p inhibits proliferation and metastasis of osteosarcoma cells by targeting Kruppel-like factor 5. J Cell Physiol 2019; 234:13525-13533. [PMID: 30773645 DOI: 10.1002/jcp.28030] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 11/27/2018] [Indexed: 01/04/2023]
Abstract
Osteosarcoma, including spinal osteosarcoma, has properties of high degree of malignancy, high rate of recurrence, and high incidence of metastasis. microRNAs can exert oncogenic or tumor suppressive roles in cancer cells. This study explored the effects of microRNA-493-5p (miR-493-5p) on osteosarcoma cell viability, migration, invasion, and apoptosis, as well as the underlying possible mechanism. First, the expression of miR-493-5p in osteosarcoma tissues and cells was detected using quantitative reverse transcription polymerase chain reaction (qRT-PCR). Then, the effects of miR-493-5p overexpression (or suppression) on osteosarcoma cell viability, migration, invasion, and apoptosis, as well as Kruppel-like factor 5 (KLF5) expression, were assessed using the Cell Counting Kit-8 assay, two-chamber transwell assay, Annexin V-FITC/PI apoptosis detection kit, qRT-PCR, and western blotting, respectively. Finally, the roles of KLF5 in miR-493-5p suppression-induced U20S cell viability, migration, and invasion enhancement, as well as the PI3K/AKT pathway activation, were evaluated. We found that miR-493-5p had lower expression in tumor tissues of spinal osteosarcoma and osteosarcoma cells. Overexpression of miR-493-5p inhibited osteosarcoma U20S cell viability, migration, and invasion, but induced cell apoptosis. On the contrary, suppression of miR-493-5p-promoted U20S cell viability, migration, and invasion. KLF5 was a direct target gene of miR-493-5p, which participated in the effects of miR-493-5p on U20S cell viability, migration, invasion, and apoptosis. Furthermore, suppression of the miR-493-5p activated PI3K/AKT pathway in U20S cells by upregulating KLF5. In conclusion, we revealed that miR-493-5p exerted tumor suppressive roles in spinal osteosarcoma and osteosarcoma cells. Overexpression of miR-493-5p inhibited proliferation and metastasis of osteosarcoma cells by downregulating KLF5 and inactivating the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Zaiqing Zhang
- Department of Orthopaedics, Linyi Central Hospital, Yishui, China
| | - Gongzeng Luo
- Department of Orthopaedics, Linyi Central Hospital, Yishui, China
| | - Chuandong Yu
- Department of Orthopaedics, Heze Municipal Hospital, Heze, China
| | - Guisheng Yu
- Department of Orthopaedics, Heze Municipal Hospital, Heze, China
| | - Rui Jiang
- Department of Orthopaedics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xuefeng Shi
- Department of Orthopedics, Trauma & Hand and Foot Surgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| |
Collapse
|
33
|
Zhang X, Pan Y, Fu H, Zhang J. microRNA-205 and microRNA-338-3p Reduces Cell Apoptosis in Prostate Carcinoma Tissue and LNCaP Prostate Carcinoma Cells by Directly Targeting the B-Cell Lymphoma 2 (Bcl-2) Gene. Med Sci Monit 2019; 25:1122-1132. [PMID: 30741252 PMCID: PMC6380162 DOI: 10.12659/msm.912148] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background The inhibitor of apoptosis, B-cell lymphoma 2 (Bcl-2), is encoded by the BCL2 gene. Previous studies have shown that microRNAs are downregulated in prostate cancer. This study aimed to investigate the role of microRNA-205 and microRNA-338-3p and cell apoptosis in prostate carcinoma tissue and the LNCaP human prostate adenocarcinoma cell line by directly targeting the BCL2 gene and Bcl-2 protein expression. Material/Methods Bioinformatics methods predicted the target genes of miR-205 and miR-338-3p, which were validated by a luciferase assay. Immunohistochemistry was used to detect Bcl-2 protein expression in 30 samples of prostate carcinoma tissue and 30 matched samples of normal prostate. The normal prostate epithelial cell line, RWPE-1, and LNCaP human prostate adenocarcinoma cells studied in vitro. BCL2 mRNA expression and Bcl-2 protein expression were determined by quantitative polymerase chain reaction (q-PCR) and Western blot, respectively. Cell apoptosis was measured by flow cytometry using annexin V, fluorescein isothiocyanate, and phycoerythrin (annexin V-FITC/PE). Results TargetScan Human 7.2 predicted that the structures of miR-205 and miR-338-3p had a binding site on the proto-oncogene, BCL2, which was verified by a luciferase assay. The expression of miR-205 and miR-338-3p were significantly downregulated in prostate carcinoma tissues and LNCaP cells when compared with normal controls. BCL2 expression was significantly inhibited by overexpression of miR-205 and miR-338-3p in LNCaP cells. Conclusions The results of this study showed that miR-205 and miR-338-3p downregulated the expression of the BCL2 gene and decreased apoptosis in prostate carcinoma.
Collapse
Affiliation(s)
- Xi Zhang
- Department of Oncology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China (mainland)
| | - Yuliang Pan
- Department of Oncology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China (mainland)
| | - Huiqun Fu
- Department of Oncology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China (mainland)
| | - Juan Zhang
- Department of Oncology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China (mainland)
| |
Collapse
|
34
|
Liu X, Wen J, Wang H, Wang Y. RETRACTED: Long non-coding RNA LINC00460 promotes epithelial ovarian cancer progression by regulating microRNA-338-3p. Biomed Pharmacother 2018; 108:1022-1028. [PMID: 30372802 DOI: 10.1016/j.biopha.2018.09.103] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 09/03/2018] [Accepted: 09/18/2018] [Indexed: 01/09/2023] Open
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/our-business/policies/article-withdrawal). This article has been retracted at the request of the Editor-in-Chief and the authors. Panels from Figures 2C and 2D appear similar to panels from Figures 4D and 4F of the article published by Hai-Xiang Yu, Xiao-Long Wang, Le-Ning Zhang, Ji Zhang and Wei Zhao in Biomedicine & Pharmacotherapy 109 (2019) 751-761 https://doi.org/10.1016/j.biopha.2018.09.110. Panels from Figure 3B appear similar to panels from Figure 3D of the article previously published by Yuanyuan Meng, Qi Li, Lianwei Li and Rong Ma in Biological Chemistry 399 (2017) 93-100 https://doi.org/10.1515/hsz-2017-0199. The ‘GAPDH’ Western Blots from Figure 3C appear similar to the ‘AKT’ Western Blots from Figure 6 of the article previously published by Lan Wu, Bo Shi, Kexin Huang and Guoyu Fan in the Oncology Reports 34 (2015) 2797-2805 https://doi.org/10.3892/or.2015.4251 and the ‘GAPDH’ Western Blots from Figure 4D of the article previously published by Tao Peng, Lixiang Zhou, Ling Zuo and Yongxin Luan in the Oncology Reports 35 (2016) 1057-1064 https://doi.org/10.3892/or.2015.4406.
Collapse
Affiliation(s)
- Xiaoxia Liu
- Department of Gynecology, The First Hospital of Jilin University, Changchun 130021, China
| | - Jihong Wen
- Department of Gynecology, The First Hospital of Jilin University, Changchun 130021, China
| | - Haijiao Wang
- Department of Gynecology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yanli Wang
- Department of Gynecology, The First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
35
|
Jia F, Zhang Z, Zhang X. MicroRNA-338-3p inhibits tumor growth and metastasis in osteosarcoma cells by targeting RUNX2/CDK4 and inhibition of MAPK pathway. J Cell Biochem 2018; 120:6420-6430. [PMID: 30484892 DOI: 10.1002/jcb.27929] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 09/27/2018] [Indexed: 12/19/2022]
Abstract
Osteosarcoma (OS) is one of the most aggressive bone tumors. MicroRNAs (miRNAs) have been found to implicate in the pathogenesis of different types of cancers, including OS. This study aimed to explore the roles of miR-338-3p in OS and investigate the underlying mechanism. Human OS cell lines (MG-63 and U2OS) and osteoblast (hFOB) cell line were used in the study. The expression levels of miR-338-3p, runt-related transcription factor 2 (RUNX2) and cyclin-dependent kinase 4 (CDK4) were altered by transient transfection and determined by quantitative real-time polymerase chain reaction/Western blot analysis. Cell viability, colony numbers, migration, and invasion, and apoptotic cells were measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, colony formation assay, transwell assay, and flow cytometry assay, respectively. Dual luciferase reporter assay was performed to identify the target gene of miR-338-3p. Western blot assay was carried to measure the protein expression levels involved in cell apoptosis, migration, and mitogen-activated protein kinases (MAPK) pathway. We found that the expression of miR-338-3p was downregulated in MG-63 cell and U2OS cells, compared with hFOB cells. MiR-338-3p suppression significantly increased cell viability and colony numbers, promoted cell migration, and invasion, but suppressed cell apoptosis in MG-63 and U2OS cells. Opposite results were observed in the miR-338-3p overexpression. Interestingly, RUNX2 and CDK4 were direct target genes of miR-338-3p. RUNX2 inhibition shared a similar effect of miR-338-3p mimic on MG-63 cells. Furthermore, miR-338-3p inhibited the activation of MAPK pathway in MG-63 cells. To conclude, these findings suggested that miR-338-3p functioned as a tumor suppressor in OS cells by targeting RUNX2 and CDK4, as well as inhibition of the MAPK pathway.
Collapse
Affiliation(s)
- Feng Jia
- Department of Orthopedics and Traumatology, The First Hospital of Zibo City, Zibo, China
| | - Zhen Zhang
- Department of Spine Surgery, The Third Hospital of Jinan, Jinan, China
| | - Xu Zhang
- Department of Orthopedics, Ping An (Hefei) Internet Hospital, Hefei, China
| |
Collapse
|
36
|
miR-338-3p Is Regulated by Estrogens through GPER in Breast Cancer Cells and Cancer-Associated Fibroblasts (CAFs). Cells 2018; 7:cells7110203. [PMID: 30423928 PMCID: PMC6262471 DOI: 10.3390/cells7110203] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 11/02/2018] [Accepted: 11/07/2018] [Indexed: 12/12/2022] Open
Abstract
Estrogens acting through the classic estrogen receptors (ERs) and the G protein estrogen receptor (GPER) regulate the expression of diverse miRNAs, small sequences of non-coding RNA involved in several pathophysiological conditions, including breast cancer. In order to provide novel insights on miRNAs regulation by estrogens in breast tumor, we evaluated the expression of 754 miRNAs by TaqMan Array in ER-negative and GPER-positive SkBr3 breast cancer cells and cancer-associated fibroblasts (CAFs) upon 17β-estradiol (E2) treatment. Various miRNAs were regulated by E2 in a peculiar manner in SkBr3 cancer cells and CAFs, while miR-338-3p displayed a similar regulation in both cell types. By METABRIC database analysis we ascertained that miR-338-3p positively correlates with overall survival in breast cancer patients, according to previous studies showing that miR-338-3p may suppress the growth and invasion of different cancer cells. Well-fitting with these data, a miR-338-3p mimic sequence decreased and a miR-338-3p inhibitor sequence rescued the expression of genes and the proliferative effects induced by E2 through GPER in SkBr3 cancer cells and CAFs. Altogether, our results provide novel evidence on the molecular mechanisms by which E2 may regulate miR-338-3p toward breast cancer progression.
Collapse
|