1
|
Hazari V, Samali SA, Izadpanahi P, Mollaei H, Sadri F, Rezaei Z. MicroRNA-98: the multifaceted regulator in human cancer progression and therapy. Cancer Cell Int 2024; 24:209. [PMID: 38872210 DOI: 10.1186/s12935-024-03386-2] [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: 12/24/2023] [Accepted: 05/25/2024] [Indexed: 06/15/2024] Open
Abstract
MicroRNA-98 (miR-98) stands as an important molecule in the intricate landscape of oncology. As a subset of microRNAs, these small non-coding RNAs have accompanied a new era in cancer research, underpinning their significant roles in tumorigenesis, metastasis, and therapeutic interventions. This review provides a comprehensive insight into the biogenesis, molecular properties, and physiological undertakings of miR-98, highlighting its double-edged role in cancer progression-acting both as a tumor promoter and suppressor. Intriguingly, miR-98 has profound implications for various aspects of cancer progression, modulating key cellular functions, including proliferation, apoptosis, and the cell cycle. Given its expression patterns, the potential of miR-98 as a diagnostic and prognostic biomarker, especially in liquid biopsies and tumor tissues, is explored, emphasizing the hurdles in translating these findings clinically. The review concludes by evaluating therapeutic avenues to modulate miR-98 expression, addressing the challenges in therapy resistance, and assessing the efficacy of miR-98 interventions. In conclusion, while miR-98's involvement in cancer showcases promising diagnostic and therapeutic avenues, future research should pivot towards understanding its role in tumor-stroma interactions, immune modulation, and metabolic regulation, thereby unlocking novel strategies for cancer management.
Collapse
Affiliation(s)
- Vajihe Hazari
- Department of Obstetrics and Gynecology, School of Medicine, Rooyesh Infertility Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Sahar Ahmad Samali
- Department of Microbiology, Yasooj Branch, Islamic Azad University, Yasooj, Iran
| | | | - Homa Mollaei
- Department of Biology, Faculty of Sciences, University of Birjand, Birjand, Iran
| | - Farzad Sadri
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran.
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran.
| | - Zohreh Rezaei
- Department of Biology, University of Sistan and Baluchestan, Zahedan, Iran.
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran.
| |
Collapse
|
2
|
Zhang Z, Li F, Li Y, Li Z, Jia G. In vitro Anti-malignant Property of PCMT1 Silencing and Identification of the SNHG16/miR-195/PCMT1 Regulatory Axis in Breast Cancer Cells. Clin Breast Cancer 2023; 23:302-316. [PMID: 36639265 DOI: 10.1016/j.clbc.2022.12.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 08/11/2022] [Accepted: 12/19/2022] [Indexed: 12/25/2022]
Abstract
BACKGROUND Protein L-isoaspartate (D-aspartate) O-methyltransferase (PCMT1) is a highly conserved protein repair enzyme that participates in regulating the progression of human cancers. We therefore studied the function and the related mechanisms of PCMT1 in breast cancer cells. METHODS Expression profile and prognostic analysis of PCMT1 in breast cancer patients were analyzed using online databases. PCMT1 expression in breast cancer cells was detected by western blot analysis. Cell proliferation was determined by CCK-8 and colony formation assays. Apoptosis was evaluated using flow cytometry analysis and caspase-3/7 activity assay. Cell invasion was assessed by Transwell invasion assay. The small nucleolar RNA host gene 16 (SNHG16)/miR-195/PCMT1 regulatory axis was identified using bioinformatics analysis. RESULTS PCMT1 expression was increased in breast cancer tissues and cells. High PCMT1 expression was correlated with poor prognosis in breast cancer patients. PCMT1 knockdown suppressed cell proliferation and colony formation ability in breast cancer cells. Moreover, PCMT1 knockdown induced apoptosis and restrained the invasive ability in breast cancer cells. PCMT1 overexpression increased the proliferative and invasive abilities of breast cancer cells. miR-195 was identified as the unique upstream miRNA of PCMT1. SNHG16 was identified as the unique upstream lncRNA of miR-195. SNHG16 knockdown downregulated PCMT1 by increasing miR-195 expression. Breast cancer cell proliferation was regulated by the SNHG16/miR-195/PCMT1 axis. CONCLUSION PCMT1 silencing inhibited cell proliferation and invasion and induced apoptosis in breast cancer cells and the SNHG16/miR-195/PCMT1 regulatory axis might serve as a potential therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Zhongji Zhang
- Department of Thyroid and Breast Surgery, Nanyang First People's Hospital Affiliated to Henan University, Nanyang, China; Key Laboratory of Thyroid Tumor Prevention and Treatment, Nanyang First People's Hospital Affiliated to Henan University, Nanyang, China
| | - Fengbo Li
- Department of Respiratory Medicine, Nanshi Hospital of Nanyang, Nanyang, China
| | - Yan Li
- Department of General Surgery, Nanyang First People's Hospital Affiliated to Henan University, Nanyang, China
| | - Zhong Li
- Department of General Surgery, Nanyang First People's Hospital Affiliated to Henan University, Nanyang, China
| | - Guangwei Jia
- Department of Thyroid and Breast Surgery, Nanyang First People's Hospital Affiliated to Henan University, Nanyang, China.
| |
Collapse
|
3
|
Lv N, Shen S, Chen Q, Tong J. Long noncoding RNAs: glycolysis regulators in gynaecologic cancers. Cancer Cell Int 2023; 23:4. [PMID: 36639695 PMCID: PMC9838043 DOI: 10.1186/s12935-023-02849-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 01/05/2023] [Indexed: 01/15/2023] Open
Abstract
The three most common gynaecologic cancers that seriously threaten female lives and health are ovarian cancer, cervical cancer, and endometrial cancer. Glycolysis plays a vital role in gynaecologic cancers. Several long noncoding RNAs (lncRNAs) are known to function as oncogenic molecules. LncRNAs impact downstream target genes by acting as ceRNAs, guides, scaffolds, decoys, or signalling molecules. However, the role of glycolysis-related lncRNAs in regulating gynaecologic cancers remains poorly understood. In this review, we emphasize the functional roles of many lncRNAs that have been found to promote glycolysis in gynaecologic cancers and discuss reasonable strategies for future research.
Collapse
Affiliation(s)
- Nengyuan Lv
- grid.268505.c0000 0000 8744 8924Department of the Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053 Zhejiang Province People’s Republic of China ,grid.13402.340000 0004 1759 700XDepartment of Obstetrics and Gynecology, Affiliated Hangzhou First People’s Hospital, Zhejiang University of Medicine, Hangzhou, 310006 Zhejiang Province People’s Republic of China
| | - Siyi Shen
- grid.268505.c0000 0000 8744 8924Department of the Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053 Zhejiang Province People’s Republic of China ,grid.13402.340000 0004 1759 700XDepartment of Obstetrics and Gynecology, Affiliated Hangzhou First People’s Hospital, Zhejiang University of Medicine, Hangzhou, 310006 Zhejiang Province People’s Republic of China
| | - Qianying Chen
- grid.268505.c0000 0000 8744 8924Department of the Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053 Zhejiang Province People’s Republic of China ,grid.13402.340000 0004 1759 700XDepartment of Obstetrics and Gynecology, Affiliated Hangzhou First People’s Hospital, Zhejiang University of Medicine, Hangzhou, 310006 Zhejiang Province People’s Republic of China
| | - Jinyi Tong
- grid.268505.c0000 0000 8744 8924Department of the Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053 Zhejiang Province People’s Republic of China ,grid.13402.340000 0004 1759 700XDepartment of Obstetrics and Gynecology, Affiliated Hangzhou First People’s Hospital, Zhejiang University of Medicine, Hangzhou, 310006 Zhejiang Province People’s Republic of China
| |
Collapse
|
4
|
Yuan D, Zhu D, Yin B, Ge H, Zhao Y, Huang A, Wang X, Cao X, Xia N, Qian H. Expression of lncRNA NEAT1 in endometriosis and its biological functions in ectopic endometrial cells as mediated via miR-124-3p. Genes Genomics 2022; 44:527-537. [PMID: 35094286 DOI: 10.1007/s13258-021-01184-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/27/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Endometriosis (EM) is a gynecological disease that poses severe health risks to women, although its pathogenesis has yet to be fully elucidated. It has been shown that long non-coding RNAs (lncRNAs) are closely associated with EM initiation and have a role in the development of this disease. Previous studies exploring the expression of the lncRNA nuclear paraspeckle assembly transcript 1 (NEAT1) have shown that this lncRNA functions as a tumor promoter in endometrial cancer. However, its exact mechanism of action in EM remains unclear. OBJECTIVE This report was designed to illustrate the potential molecular mechanisms of lncRNA NEAT1 on EM. METHODS Endometrial tissues were extracted from EM model rats and patients with EM. Hematoxylin and eosin staining was applied to detect the morphological changes that occurred in rats after construction of the model. Endometrial stromal cells (ESCs) were extracted from either ectopic endometrium (EC) or eutopic endometrium (EU) tissues from patients with EM. LncRNA NEAT1 and miR-124-3p expression in EM tissues and cells were subsequently evaluated by reverse transcription-quantitative (RT-q)PCR analysis. MTT assay, flow cytometric analysis, western blot assay and Transwell assay were then employed to examine the effect of NEAT1 and miR-124-3p on EC-ESC proliferation, apoptosis, migration and invasion, respectively. The targeted relationship between lncRNA NEAT1 and miR-124-3p was subsequently confirmed by dual-luciferase and co-transfection assays. RESULTS MiR-124-3p was identified as a target of NEAT1, and could be negatively regulated by NEAT1 in EC-ESCs. The expression level of NEAT1 was evidently increased, whereas that of miR-124-3p was decreased, in the EM in vivo model, EM tissues and EC-ESCs from patients with EM. The loss-of-function assays further established that silencing of NEAT1 could inhibit EC-ESC proliferation, migration, and invasion, but it led to the promotion of apoptosis via targeting miR-124-3p. CONCLUSIONS NEAT1 is significantly upregulated in EM, promoting malignant behavior in EM through targeting miR-124-3p expression.
Collapse
Affiliation(s)
- Donglan Yuan
- Department of Obstetrics and Gynecology, Taizhou People's Hospital Affiliated to Nantong University, 399 Hailing Road, Taizhou, 225300, People's Republic of China
| | - Dandan Zhu
- Department of Obstetrics and Gynecology, Taizhou People's Hospital Affiliated to Nantong University, 399 Hailing Road, Taizhou, 225300, People's Republic of China
| | - Boyu Yin
- Department of Obstetrics and Gynecology, Taizhou People's Hospital Affiliated to Nantong University, 399 Hailing Road, Taizhou, 225300, People's Republic of China
| | - Hongshan Ge
- Department of Obstetrics and Gynecology, Taizhou People's Hospital Affiliated to Nantong University, 399 Hailing Road, Taizhou, 225300, People's Republic of China
| | - Yinling Zhao
- Department of Obstetrics and Gynecology, Taizhou People's Hospital Affiliated to Nantong University, 399 Hailing Road, Taizhou, 225300, People's Republic of China
| | - Aihua Huang
- Department of Obstetrics and Gynecology, Taizhou People's Hospital Affiliated to Nantong University, 399 Hailing Road, Taizhou, 225300, People's Republic of China
| | - Xiaosu Wang
- Department of Obstetrics and Gynecology, Taizhou People's Hospital Affiliated to Nantong University, 399 Hailing Road, Taizhou, 225300, People's Republic of China
| | - Xiuhong Cao
- Department of Operation, Taizhou People's Hospital Affiliated to Nantong University, Taizhou, 225300, China
| | - Nan Xia
- Department of Obstetrics and Gynecology, Taizhou People's Hospital Affiliated to Nantong University, 399 Hailing Road, Taizhou, 225300, People's Republic of China
| | - Hua Qian
- Department of Obstetrics and Gynecology, Taizhou People's Hospital Affiliated to Nantong University, 399 Hailing Road, Taizhou, 225300, People's Republic of China.
| |
Collapse
|
5
|
Ghafouri-Fard S, Khoshbakht T, Taheri M, Shojaei S. A Review on the Role of Small Nucleolar RNA Host Gene 6 Long Non-coding RNAs in the Carcinogenic Processes. Front Cell Dev Biol 2021; 9:741684. [PMID: 34671603 PMCID: PMC8522957 DOI: 10.3389/fcell.2021.741684] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/09/2021] [Indexed: 01/27/2023] Open
Abstract
Being located on 17q25.1, small nucleolar RNA host gene 6 (SNHG16) is a member of SNHG family of long non-coding RNAs (lncRNA) with 4 exons and 13 splice variants. This lncRNA serves as a sponge for a variety of miRNAs, namely miR-520a-3p, miR-4500, miR-146a miR-16–5p, miR-98, let-7a-5p, hsa-miR-93, miR-17-5p, miR-186, miR-302a-3p, miR-605-3p, miR-140-5p, miR-195, let-7b-5p, miR-16, miR-340, miR-1301, miR-205, miR-488, miR-1285-3p, miR-146a-5p, and miR-124-3p. This lncRNA can affect activity of TGF-β1/SMAD5, mTOR, NF-κB, Wnt, RAS/RAF/MEK/ERK and PI3K/AKT pathways. Almost all studies have reported oncogenic effect of SNHG16 in diverse cell types. Here, we explain the results of studies about the oncogenic role of SNHG16 according to three distinct sets of evidence, i.e., in vitro, animal, and clinical evidence.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tayyebeh Khoshbakht
- Men's Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyedpouzhia Shojaei
- Department of Critical Care Medicine, Imam Hossein Medical and Educational Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Yan LR, Ding HX, Shen SX, Lu XD, Yuan Y, Xu Q. Pepsinogen C expression-related lncRNA/circRNA/mRNA profile and its co-mediated ceRNA network in gastric cancer. Funct Integr Genomics 2021; 21:605-618. [PMID: 34463892 DOI: 10.1007/s10142-021-00803-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/17/2021] [Accepted: 08/22/2021] [Indexed: 11/26/2022]
Abstract
The expression of pepsinogen C (PGC) is considered an ideal negative biomarker of gastric cancer, but its pathological mechanisms remain unclear. This study aims to analyze competing endogenous RNA (ceRNA) networks related to PGC expression at a post-transcriptional level and build an experimental basis for studying the role of PGC in the progression of gastric cancer. RNA sequencing technology was used to detect the differential expression (DE) profiles of PGC-related long non-coding (lnc)RNAs, circular (circ)RNAs, and mRNAs. Ggcorrplot R package and online database were used to construct DElncRNAs/DEcircRNAs co-mediated PGC expression-related ceRNA networks. In vivo and in vitro validations were performed using quantitative reverse transcription-PCR (qRT-PCR). RNA sequencing found 637 DEmRNAs, 698 DElncRNAs, and 38 DEcircRNAs. The PPI network of PGC expression-related mRNAs consisted of 503 nodes and 1179 edges. CFH, PPARG, and MUC6 directly interacted with PGC. Enrichment analysis suggested that DEmRNAs were mainly enriched in cancer-related pathways. Eleven DElncRNAs, 13 circRNAs, and 35 miRNA-mRNA pairs were used to construct ceRNA networks co-mediated by DElncRNAs and DEcircRNAs that were PGC expression-related. The network directly related to PGC was as follows: SNHG16/hsa_circ_0008197-hsa-mir-98-5p/hsa-let-7f-5p/hsa-let-7c-5p-PGC. qRT-PCR validation results showed that PGC, PPARG, SNHG16, and hsa_circ_0008197 were differentially expressed in gastric cancer cells and tissues: PGC positively correlated with PPARG (r = 0.276, P = 0.009), SNHG16 (r = 0.35, P = 0.002), and hsa_circ_0008197 (r = 0.346, P = 0.005). PGC-related DElncRNAs and DEcircRNAs co-mediated complicated ceRNA networks to regulate PGC expression, thus affecting the occurrence and development of gastric cancer at a post-transcriptional level. Of these, the network directly associated with PGC expression was a SNHG16/hsa_circ_0008197-mir-98-5p/hsa-let-7f-5p/hsa-let-7c-5p - PGC axis. This study may form a foundation for the subsequent exploration of the possible regulatory mechanisms of PGC in gastric cancer.
Collapse
Affiliation(s)
- Li-Rong Yan
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Affiliated Hospital of China Medical University, Key Laboratory of Cancer Etiology and Prevention, China Medical University, Liaoning Provincial Education Department, 110001, Shenyang, China
| | - Han-Xi Ding
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Affiliated Hospital of China Medical University, Key Laboratory of Cancer Etiology and Prevention, China Medical University, Liaoning Provincial Education Department, 110001, Shenyang, China
| | - Shi-Xuan Shen
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Affiliated Hospital of China Medical University, Key Laboratory of Cancer Etiology and Prevention, China Medical University, Liaoning Provincial Education Department, 110001, Shenyang, China
| | - Xiao-Dong Lu
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Affiliated Hospital of China Medical University, Key Laboratory of Cancer Etiology and Prevention, China Medical University, Liaoning Provincial Education Department, 110001, Shenyang, China
| | - Yuan Yuan
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Affiliated Hospital of China Medical University, Key Laboratory of Cancer Etiology and Prevention, China Medical University, Liaoning Provincial Education Department, 110001, Shenyang, China.
| | - Qian Xu
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Affiliated Hospital of China Medical University, Key Laboratory of Cancer Etiology and Prevention, China Medical University, Liaoning Provincial Education Department, 110001, Shenyang, China.
| |
Collapse
|
7
|
Li H, Quan F, Zhang P, Shao Y. Long non-coding RNA SNHG16, binding with miR-106b-5p, promoted cell apoptosis and inflammation in allergic rhinitis by up-regulating leukemia inhibitory factor to activate the JAK1/STAT3 signaling pathway. Hum Exp Toxicol 2021; 40:S233-S245. [PMID: 34407675 DOI: 10.1177/09603271211035665] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Allergic rhinitis (AR) is a type I hypersensitive disease. Long non-coding RNA (lncRNA) SNHG16 acts as an oncogene in a variety of tumors and promotes the occurrence of inflammation in many inflammatory diseases. The study aims to investigate the expression of SNHG16 and its potential biological functions in AR. RT-qPCR results showed that the expression of SNHG16 in AR was up-regulated. The AR cell model was constructed by stimulating primary nasal mucosal epithelial cells from AR patients with IL-13. After knocking down the expression of lncRNA SNHG16, cell apoptosis was detected by flow cytometry, and the expression of inflammatory factors was detected by ELISA. The results showed that SNHG16 promoted cell apoptosis and inflammation. Then, bioinformatics analysis was used to screen miRNAs bound with SNHG16. Luciferase reporter gene assay and RNA pull-down experiment were used to verify the relationship. We found that the expression of miR-106b-5p was down-regulated and leukemia inhibitory factor (LIF) expression was up-regulated in the AR cell model. The expression of phospho-Janus kinase 1 and p-signal transducer and activator of transcription 3 (STAT3) were detected by Western blotting. Silencing the expression of LIF could inhibit the activity of JAK1/STAT3 pathway and further inhibit cell apoptosis and the occurrence of inflammation. Then transfected SNHG16 shRNA alone or together with miR-106b-5p antagomir into the AR cell model, we found that silencing the expression of SNHG16 down-regulated the expression of LIF and inhibited the activity of the JAK1/STAT3 pathway, cell apoptosis, and inflammation. However, miR-106b-5p antagomir weakened its inhibitory effects. The role of SNHG16 in AR was further verified by the ovalbumin-induced AR mouse model in vivo. In conclusion, SNHG16 up-regulates LIF expression by binding with miR-106b-5p, thus promoting the activity of JAK1/STAT3 pathway, and promoting the development of AR. These results provide new targets for the treatment of AR and may help reduce the damage caused by AR.
Collapse
Affiliation(s)
- Huajing Li
- Otorhinolaryngology and Head and Neck Surgery Department, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Fang Quan
- Otorhinolaryngology and Head and Neck Surgery Department, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Pengfei Zhang
- Otorhinolaryngology and Head and Neck Surgery Department, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yuan Shao
- Otorhinolaryngology and Head and Neck Surgery Department, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
8
|
Wang Y, Lou N, Zuo M, Zhu F, He Y, Cheng Z, Wang X. STAT3-induced ZBED3-AS1 promotes the malignant phenotypes of melanoma cells by activating PI3K/AKT signaling pathway. RNA Biol 2021; 18:355-368. [PMID: 34241580 DOI: 10.1080/15476286.2021.1950463] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Melanoma is considered as the most frequent primary malignancy occurring in skin. Accumulating studies have suggested that long non-coding RNAs (lncRNAs) play critical parts in multiple cancers. In this study, we explored the molecular mechanism of ZBED3 antisense RNA 1 (ZBED3-AS1) in melanoma. We observed that ZBED3-AS1 expression was remarkably up-regulated in melanoma tissues, and high ZBED3-AS1 level was linked to unsatisfactory survival of melanoma patients. Then, we discovered that ZBED3-AS1 was overexpressed in melanoma cells compared with human epidermal melanocytes. In addition, loss-of-function assays verified that ZBED3-AS1 knockdown restrained cell proliferation, migration, epithelial-mesenchymal transition (EMT), and stemness in melanoma. In addition, signal transducer and activator of transcription 3 (STAT3), which also showed tumour-facilitating functions in melanoma, was confirmed as a transcriptional activator of ZBED3-AS1. Moreover, ZBED3-AS1 enhanced the expression of AT-rich interaction domain 4B (ARID4B) through sequestering miR-381-3p. Importantly, we further confirmed that ZBED3-AS1 promoted the malignant progression of melanoma by regulating miR-381-3p/ARID4B axis to activate the phosphatidylinositol 3-kinase/AKT serine/threonine kinase (PI3K/AKT) signalling pathway. In a word, our research might provide a novel therapeutic target for melanoma.
Collapse
Affiliation(s)
- Yang Wang
- Department of Pathology, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, Shenzhen, Guangdong, China
| | - Nan Lou
- Department of Joint Replacement Surgery, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Min Zuo
- Department of Pathology, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, Shenzhen, Guangdong, China
| | - Fuqiang Zhu
- Department of Pathology, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, Shenzhen, Guangdong, China
| | - Yan He
- Department of Pathology, Longgang Center Hospital of Shenzhen, Guangdong, China
| | - Zhiqiang Cheng
- Department of Pathology, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, Shenzhen, Guangdong, China
| | - Xiaomei Wang
- Department of Pathology, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, Shenzhen, Guangdong, China
| |
Collapse
|
9
|
Xu K, Zhang P, Zhang J, Quan H, Wang J, Liang Y. Identification of potential micro-messenger RNAs (miRNA-mRNA) interaction network of osteosarcoma. Bioengineered 2021; 12:3275-3293. [PMID: 34252359 PMCID: PMC8806609 DOI: 10.1080/21655979.2021.1947065] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Osteosarcoma (OS) is the most common primary malignant tumor in children and adolescents. Numerous studies have reported the importance of miRNA in OS. The purpose of this study is to predict potential biomarkers and new therapeutic targets for OS diagnosis and prognosis by analyzing miRNAs of OS plasma samples from the Gene Expression Omnibus (GEO) database. Data-sets were downloaded from the GEO and analyzed using R software. Different expressions of miRNAs (DE-miRNAs) in plasma and mRNAs (DE-mRNAs) in OS patients were identified. Funrich was used to predict the transcription factors and target genes of miRNAs. By comparing the target mRNAs and DE-mRNAs, the intersection mRNAs were identified. The intersection mRNAs were imported to perform Gene Ontology (GO) functional annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. MiRNA-mRNA regulatory network and a protein-protein interaction (PPI) network were constructed by using Cytoscape. Finally, a total of 164 DE-miRNAs, 256 DE-mRNAs, and 76 intersection mRNAs were identified. The top 10 TF of up- and down-regulated DE-miRNAs were also predicted. In addition, GO and KEGG analyses further revealed the intersection mRNAs. By constructing the miRNA–mRNA networks, we found miR-30d-5p, miR-17-5p, miR-98-5p, miR-301a-3p, and miR-30e-5p were the central hubs. COL1A1, COL1A2, MMP2, CDH11, COL4A1 etc. were predicted to be the key mRNA by constructing the PPI networks. Through a comprehensive bioinformatics analysis of miRNAs and mRNAs in OS, we explored the potential effective biomarkers and novel therapeutic targets for the diagnosis and prognosis of OS.
Collapse
Affiliation(s)
- Keteng Xu
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Pei Zhang
- Department of Orthopedics, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, Hunan, China
| | - Jiale Zhang
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Huahong Quan
- Department of Orthopedics, Dalian Medical University, Dalian, Liaoning, China
| | - Jingcheng Wang
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, China.,Department of Orthopedics, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, Hunan, China
| | - Yuan Liang
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
10
|
Xia W, Liu Y, Cheng T, Xu T, Dong M, Hu X. Extracellular Vesicles Carry lncRNA SNHG16 to Promote Metastasis of Breast Cancer Cells via the miR-892b/PPAPDC1A Axis. Front Cell Dev Biol 2021; 9:628573. [PMID: 34249903 PMCID: PMC8267525 DOI: 10.3389/fcell.2021.628573] [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: 11/12/2020] [Accepted: 04/30/2021] [Indexed: 12/17/2022] Open
Abstract
Breast cancer (BC) represents the most commonly diagnosed malignancy among women. Long non-coding RNAs (lncRNAs) can be transferred by extracellular vesicles (EVs) to participate in BC progression. This study demonstrated that SNHG16 expression was significantly increased in BC tissues and cells. Overexpression of SNHG16 promoted the migration, invasion, and epithelial-mesenchymal transition (EMT) of BC cells. SNHG16 was carried by EVs. Bioinformatics analysis predicted that SNHG16 regulated PPAPDC1A expression by sponging miR-892b, which was confirmed by RNA-fluorescence in situ hybridization (FISH), RT-qPCR, dual-luciferase gene reporter assay, and RNA immunoprecipitation (RIP). MDA-MB-157 and HS578T cells were transfected with pcDNA3.1-SNHG16, miR-892b-mimic, or si-PPAPDC1A for functional rescue experiments in vitro, and the cells were treated with MDA-MB-231 cell-derived EVs. The results confirmed that enhanced miR-892b expression partially eliminated the increase of migration, invasion, and EMT of BC cells mediated by SNHG16 or EVs. The lung metastasis model in nude mice was established by injecting HS578T cells via tail vein. The results showed that si-SNHG16 reduced the metastatic nodules and decreased the vimentin expression. In conclusion, EVs derived from BC cells transferred SNHG16 via the miR-892b/PPAPDC1A axis, thus promoting EMT, migration, and invasion of BC.
Collapse
Affiliation(s)
- Wenfei Xia
- Department of Breast and Thyroid Surgery, Division of General Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yun Liu
- Department of ENT, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Teng Cheng
- Department of Breast and Thyroid Surgery, Division of General Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Xu
- Department of Breast and Thyroid Surgery, Division of General Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Menglu Dong
- Department of Breast and Thyroid Surgery, Division of General Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaopeng Hu
- Department of Breast and Thyroid Surgery, Division of General Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
11
|
Yu Z, Zhang Y, Zheng H, Gao Q, Wang H. LncRNA SNHG16 regulates trophoblast functions by the miR-218-5p/LASP1 axis. J Mol Histol 2021; 52:1021-1033. [PMID: 34110517 DOI: 10.1007/s10735-021-09985-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 05/24/2021] [Indexed: 12/22/2022]
Abstract
Altered placental development and function lead to placental diseases such as preeclampsia (PE) which is mainly characterized by insufficient trophoblast invasion and abnormally invasive placenta disorders. Long noncoding RNAs (lncRNAs) are widely reported to function as crucial players in the pathogenesis of PE. The present investigation clarified the role of lncRNA small nucleolar RNA host gene 16 (SNHG16) in PE. RT-qPCR was used to measure gene expression. The proliferation of trophoblast cells was examined using CCK-8 and EdU assays. Trophoblast migration and invasion were assessed using wound healing and transwell assays. The apoptosis was estimated by flow cytometry. Luciferase reporter and RNA pull-down assays were performed to explore the molecular mechanisms in trophoblast cells. We found that SNHG16 was downregulated in placenta from patients with PE. Moreover, SNHG16 depletion significantly inhibited trophoblast cell proliferation, migration, and invasion and stimulated apoptosis, while SNHG16 overexpression exerted an opposite effect. Subsequently, we confirmed that SNHG16 acted as a competing RNA (ceRNA) of miR-218-5p that was verified to directly target LASP1. Both miR-218-5p depletion and LASP1 upregulation antagonized the effect of SNHG16 knockdown on HTR-8/SVneo cell functions. In conclusion, SNHG16 facilitates trophoblast cell migration and invasion by the miR-218-5p/LASP1 axis.
Collapse
Affiliation(s)
- Zhou Yu
- Department of Obstetrics, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, No. 1 Huanghe West Road, Huaiyin District, Huaian , 223300, Jiangsu, China
| | - Yulei Zhang
- Department of Obstetrics, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, No. 1 Huanghe West Road, Huaiyin District, Huaian , 223300, Jiangsu, China
| | - Haoyu Zheng
- Department of Obstetrics, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, No. 1 Huanghe West Road, Huaiyin District, Huaian , 223300, Jiangsu, China
| | - Qiong Gao
- Department of Obstetrics, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, No. 1 Huanghe West Road, Huaiyin District, Huaian , 223300, Jiangsu, China
| | - Haidong Wang
- Department of Obstetrics, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, No. 1 Huanghe West Road, Huaiyin District, Huaian , 223300, Jiangsu, China.
| |
Collapse
|
12
|
Wu Q, Zhao Y, Shi R, Wang T. LncRNA SNHG16 Facilitates Nasopharyngeal Carcinoma Progression by Acting as ceRNA to Sponge miR-520a-3p and Upregulate MAPK1 Expression. Cancer Manag Res 2021; 13:4103-4114. [PMID: 34045897 PMCID: PMC8147710 DOI: 10.2147/cmar.s305544] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/12/2021] [Indexed: 12/14/2022] Open
Abstract
Background Accumulating evidence shows that lncRNAs are widely involved cellular processes of various tumors. The aim of this study was to explore the potential role and molecular mechanism of lncRNA SNHG16 in nasopharyngeal carcinoma (NPC). Methods SNHG16, miR-520a-3p, and MAPK1 levels were measured by RT-qPCR assay. CCK-8, colony formation, transwell, and flow cytometry assays were adopted to analyze the proliferation, migration, invasion, and apoptosis of NPC cell lines (SUNE1 and 5–8F). Murine xenograft model was used to investigate tumor growth and metastasis in vivo. Immunohistochemical staining was employed to evaluate the levels of Bcl-2, cleaved caspase-3, Bax, and Ki-67. Dual-luciferase reporter assays were conducted to analyze the binding ability between miR-520a-3p and SNHG16 or MAPK1. Results SNHG16 was overexpressed in NPC tissues and cells. High SNHG16 expression indicated a poor prognosis. SNHG16 knockdown could cause significant inhibition on cell proliferation and metastasis, induce cell apoptosis in NPC cells, and repressed tumor growth and metastasis in vivo. Additionally, SNHG16 could directly bind to miR-520a-3p, thus positively regulating MAPK1 expression. Moreover, functional analysis indicated that miR-520a-3p exerted a tumor-suppressing role in NPC progression. Rescue assays demonstrated that MAPK1 upregulation could abrogate the inhibitory effects on NPC cell proliferation and metastasis, as well as the promoting effects on NPC cell apoptosis caused by SNHG16 knockdown. In conclusion, SNHG16 contributed to the proliferation and metastasis of NPC cells by modulating the miR-520a-3p/MAPK1 axis. Conclusion These results suggest that SNHG16 acts as an oncogene in the progression of NPC via modulating the miR-520a-3p/MAPK1 axis.
Collapse
Affiliation(s)
- Qingwei Wu
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yingying Zhao
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Runjie Shi
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Tao Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
13
|
Yuan X, Yan Y, Xue M. Small nucleolar RNA host gene 8: A rising star in the targets for cancer therapy. Biomed Pharmacother 2021; 139:111622. [PMID: 33894626 DOI: 10.1016/j.biopha.2021.111622] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/02/2021] [Accepted: 04/12/2021] [Indexed: 12/12/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are a group of transcripts that have been considered essential participants in cancer pathogenesis and progression over the past few decades. Small nucleolar RNA host gene 8 (SNHG8) is a newly discovered lncRNA that belongs to the SNHG family, a group of transcripts that can be processed into small nucleolar RNAs and exert important biological functions. As an oncogenic factor, SNHG8 is upregulated in multiple cancer types. Herein, we summarize the biological role of SNHG8 in different cancer types and the underlying mechanisms related to the interaction between SNHG8 and microRNAs, mRNAs, and proteins. In addition, this study emphasizes the clinical value of SNHG8 in cancer, hoping to provide new insights into cancer diagnosis, prognosis, and treatment.
Collapse
Affiliation(s)
- Xin Yuan
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yuheng Yan
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Miaomiao Xue
- Department of General Dentistry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
14
|
Bu J, Guo R, Xu XZ, Luo Y, Liu JF. LncRNA SNHG16 promotes epithelial-mesenchymal transition by upregulating ITGA6 through miR-488 inhibition in osteosarcoma. J Bone Oncol 2021; 27:100348. [PMID: 33598394 PMCID: PMC7868993 DOI: 10.1016/j.jbo.2021.100348] [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: 09/02/2020] [Revised: 12/12/2020] [Accepted: 01/05/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Osteosarcoma is a primary cause of cancer-associated death in children and adolescents worldwide. Long non-coding RNAs SNHG16 (lncRNA SNHG16) and integrin subunit-a 6 (ITGA6) are recently reported to be involved in the tumorigenesis of osteosarcoma by multiple mechanisms. However, the correlation between SNHG16 and ITGA6 in osteosarcoma remains undetermined. METHODS Expression of miR-488, SNHG16 and ITGA6, as well as epithelial-mesenchymal transition (EMT) associated markers in osteosarcoma tissues and cell lines were examined by qRT-PCR or Western blotting. Effects of miR-488, SNHG16 and ITGA6 on cell migration, invasion were evaluated by wound-healing assay and transwell assay. Bioinformatics analysis and dual-luciferase reported assays were applied to assess the interaction among miR-488, SNHG16 and ITGA6. RNA immunoprecipitation (RIP) was also used to verify SNHG16 and miR-488 interaction. Finally, animal study was used to detect the effect of SNHG16 on osteosarcoma in vivo. RESULTS SNHG16 and ITGA6 were significantly increased while miR-488 was decreased in osteosarcoma. ITGA6 was screened as a target gene of miR-488, and SNHG16 was sponged by miR-488 in osteosarcoma cells. MiR-488 overexpression and SNHG16 knockdown suppressed migration, invasion and EMT of osteosarcoma cells. Moreover, rescue assays proved that the influences of SNHG16 on osteosarcoma cells migration, invasion and EMT were dependent on miR-488 and ITGA6. In addition, the promotive effects of SNHG16 on osteosarcoma tumor growth and metastasis were further supported by xenograft tumor growth assay. CONCLUSION SNHG16 promoted migration, invasion and EMT of osteosarcoma by sponging miR-488 to release ITGA6.
Collapse
Affiliation(s)
- Jie Bu
- Department of Orthopaedics, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan Province, People's Republic of China
| | - Ru Guo
- Department of Pediatrics, Maternal and Child Health Care Hospital of Hunan Province, Changsha 410008, Hunan Province, People's Republic of China
| | - Xue-Zheng Xu
- Department of Orthopaedics, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan Province, People's Republic of China
| | - Yi Luo
- Department of Orthopaedics, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan Province, People's Republic of China
| | - Jian-Fan Liu
- Department of Orthopaedics, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan Province, People's Republic of China
| |
Collapse
|
15
|
Zhang W, Ren X, Qi L, Zhang C, Tu C, Li Z. The value of lncRNAs as prognostic biomarkers on clinical outcomes in osteosarcoma: a meta-analysis. BMC Cancer 2021; 21:202. [PMID: 33639865 PMCID: PMC7912917 DOI: 10.1186/s12885-021-07882-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 02/05/2021] [Indexed: 02/08/2023] Open
Abstract
Background In recent years, emerging studies have demonstrated critical functions and potential clinical applications of long non-coding RNA (lncRNA) in osteosarcoma. To further validate the prognostic value of multiple lncRNAs, we have conducted this updated meta-analysis. Methods Literature retrieval was conducted by searching PubMed, Web of Science and the Cochrane Library (last update by October 2, 2019). A meta-analysis was performed to explore association between lncRNAs expression and overall survival (OS) of osteosarcoma patients. Relationships between lncRNAs expression and other clinicopathological features were also analyzed respectively. Results Overall, 4351 patients from 62 studies were included in this meta-analysis and 25 lncRNAs were identified. Pooled analyses showed that high expression of 14 lncRNAs connoted worse OS, while two lncRNAs were associated with positive outcome. Further, analysis toward osteosarcoma clinicopathologic features demonstrated that overexpression of TUG1 and XIST indicated poor clinical parameters of patients. Conclusions This meta-analysis has elucidated the prognostic potential of 16 lncRNAs in human osteosarcoma. Evidently, desperate expression and functional targets of these lncRNAs offer new approaches for prognosis and therapy of osteosarcoma. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-07882-w.
Collapse
Affiliation(s)
- Wenchao Zhang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, People's Republic of China.,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Xiaolei Ren
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, People's Republic of China.,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Lin Qi
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, People's Republic of China.,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Chenghao Zhang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, People's Republic of China.,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Chao Tu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, People's Republic of China. .,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China.
| | - Zhihong Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, People's Republic of China. .,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China.
| |
Collapse
|
16
|
Yu B, Zhao J, Dong Y. Circ_0000527 Promotes Retinoblastoma Progression through Modulating miR-98-5p/XIAP Pathway. Curr Eye Res 2021; 46:1414-1423. [PMID: 33629639 DOI: 10.1080/02713683.2021.1891255] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Purpose: Retinoblastoma (RB) is an intraocular malignancy that often occurs in childhood. Circular RNAs (circRNAs) play crucial roles in regulating the malignant phenotypes of various tumors. This study aimed to explore the role and potential mechanism of circ_0000527 in RB.Methods: The levels of circ_0000527, microRNA-98-5p (miR-98-5p) and X-linked inhibitor of apoptosis (XIAP) were determined by qRT-PCR or western blot. Cell proliferation was detected by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) and colony formation assays. Cell apoptosis, migration and invasion were evaluated by flow cytometry, transwell and scratch assays. Xenograft assay was conducted to analyze tumor growth in vivo. The binding relationship between miR-98-5p and circ_0000527 or XIAP was verified by dual-luciferase reporter and RNA immunoprecipitation (RIP) assays.Results: Circ_0000527 and XIAP levels were increased, while miR-98-5p level was reduced in RB tissues and cells. Silencing of circ_0000527 suppressed the proliferation, migration and invasion of RB cells and promoted apoptosis. In addition, knockdown of circ_0000527 inhibited tumor growth in xenograft mice. Besides, circ_0000527 sponged miR-98-5p to regulate RB cell progression, and miR-98-5p targeted XIAP to mediate RB cell development. Moreover, circ_0000527 modulated XIAP expression via sequestering miR-98-5p.Conclusion: Circ_0000527 facilitated RB progression via regulating miR-98-5p/XIAP axis, which provided a promising therapeutic target for RB.
Collapse
Affiliation(s)
- Binke Yu
- Department of Ophthalmology, Xianyang Hospital of Yan'an University, Xianyang, China
| | - Jifei Zhao
- Department of Ophthalmology, The First People's Hospital of Xianyang City, Xianyang, China
| | - Yongxiao Dong
- Department of Ophthalmology, The First People's Hospital of Xianyang City, Xianyang, China
| |
Collapse
|
17
|
Asila A, Yang X, Kaisaer Y, Ma L. SNHG16/miR‐485‐5p/BMP7 axis modulates osteogenic differentiation of human bone marrow‐derived mesenchymal stem cells. J Gene Med 2021; 23:e3296. [PMID: 33179372 DOI: 10.1002/jgm.3296] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 11/06/2020] [Accepted: 11/06/2020] [Indexed: 01/27/2023] Open
Affiliation(s)
- Ailijiang Asila
- Department of Orthopaedics TCM Hospital of Xinjiang Xinjiang China
| | - Xinjun Yang
- Department of Orthopaedics TCM Hospital of Xinjiang Xinjiang China
| | - Yilipan Kaisaer
- Department of Orthopaedics TCM Hospital of Xinjiang Xinjiang China
| | - Lei Ma
- Department of Orthopaedics TCM Hospital of Xinjiang Xinjiang China
| |
Collapse
|
18
|
Zhang Z, Liu J, Wu Y, Zhao X, Hao Y, Wang X, Xue C, Wang Y, Zhang R, Zhang X. Long Noncoding RNA SERTAD2-3 Inhibits Osteosarcoma Proliferation and Migration by Competitively Binding miR-29c. Genet Test Mol Biomarkers 2020; 24:67-72. [PMID: 31999493 DOI: 10.1089/gtmb.2019.0164] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Background: Osteosarcoma (OS) is a malignant tumor disease with high morbidity and mortality in children and adolescents. Recently, attention has been focused on the effects of long noncoding RNAs (lncRNAs) on tumor biology. In this study, we identified the role of lnc-SERTAD2-3 in the development of OS. Materials and Methods: Sixty OS samples and adjacent tissues were collected to determine the relationship between lnc-SERTAD2-3 levels and clinicopathological characteristics. Quantitative real-time PCR (qPCR) was used to measure gene expression levels. A transwell invasion assay, a Cell Counting Kit-8 assay, and flow cytometry were used to measure cell migration, growth, and apoptosis, respectively. The binding site between the lnc-SERTAD2-3 and miR-29c RNAs was evaluated using a luciferase reporter assay. Results: The expression of the lnc-SERTAD2-3 was significantly downregulated in OS samples and three OS cell lines (MG-63, U2OS, and Saos-2) compared to normal tissue. Patients with lower levels of lnc-SERTAD2-3 expression had a more unfavorable prognosis (larger OS size, distant metastasis, and recurrence). Overexpression of lnc-SERTAD2-3 inhibited proliferation and migration, and promoted apoptosis in OS cells. Moreover, we found that lnc-SERTAD2-3 could suppress miR-29c by direct binding. Moreover, reexpression of miR-29c reversed the effect of lnc-SERTAD2-3 on OS cells. Conclusion: Overall, lnc-SERTAD2-3, an OS suppressor, is involved in the inhibition of OS proliferation and migration by targeting miR-29c.
Collapse
Affiliation(s)
- Zhifa Zhang
- Department of Orthopaedics, The PLA General Hospital, Beijing, China
| | - Jiangjun Liu
- Department of Orthopaedics, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Yuezhou Wu
- Department of Emergency Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xuelin Zhao
- Department of Orthopaedics, The PLA General Hospital, Beijing, China
| | - Yongyu Hao
- Department of Orthopaedics, The PLA General Hospital, Beijing, China
| | - Xiangyu Wang
- Department of Orthopaedics, The PLA General Hospital, Beijing, China
| | - Chao Xue
- Department of Orthopaedics, The PLA General Hospital, Beijing, China
| | - Yan Wang
- Department of Orthopaedics, The PLA General Hospital, Beijing, China
| | - Rui Zhang
- Department of Emergency Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xuesong Zhang
- Department of Orthopaedics, The PLA General Hospital, Beijing, China
| |
Collapse
|
19
|
Commentary on: The potency of lncRNA MALAT1/miR-155 in altering asthmatic Th1/Th2 balance by modulation of CTLA4. Biosci Rep 2020; 40:222656. [PMID: 32292999 PMCID: PMC7199447 DOI: 10.1042/bsr20190768] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 04/03/2020] [Accepted: 04/06/2020] [Indexed: 01/14/2023] Open
Abstract
Asthma is a common, allergic respiratory disorder affecting over 350 million people worldwide. One of the key features of asthma is skewing of CD4+ cells toward Th2 responses. This promotes the production of cytokines like IL-4 that induce IgE production resulting in the hypersecretion of mucus and airway smooth muscle contraction. Understanding the factors that favor Th2 expansion in asthma would provide important insights into the underlying pathogenesis of this disorder. Asthma research has focused on signaling pathways that control the transcription of key asthma-related genes. However, increasing evidence shows that post-transcriptional factors also determine CD4+ cell fate and the enhancement of allergic airway responses. A recent paper published by Liang et al. (Bioscience Reports (2020) 40, https://doi.org/10.1042/BSR20190397) highlights a novel role for the long non-coding RNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) in Th2 development in asthma. MALAT1 modulates several biological processes including alternative splicing, epigenetic modification and gene expression. It is one of the most highly expressed lncRNAs in normal tissues and MALAT1 levels correlate with poor clinical outcomes in cancer. The mechanisms of action of MALAT1 in tumor progression and metastasis remain unclear and even less is known about its effects in inflammatory disease states like asthma. The work of Liang et al. demonstrates heightened MALAT1 expression in asthma and further shows that this lncRNA targets miR-155 to promote Th2 differentiation in this disease. This insight sets the stage for future studies to examine how MALAT1 manipulation could deter allergic immune responses in asthmatic airways.
Collapse
|
20
|
Xiao Y, Xiao T, Ou W, Wu Z, Wu J, Tang J, Tian B, Zhou Y, Su M, Wang W. LncRNA SNHG16 as a potential biomarker and therapeutic target in human cancers. Biomark Res 2020; 8:41. [PMID: 32944244 PMCID: PMC7487997 DOI: 10.1186/s40364-020-00221-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/26/2020] [Indexed: 01/27/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) represent an important class of RNAs comprising more than 200 nucleotides, which are produced by RNA polymerase II. Although lacking an open reading framework and protein-encoding activity, lncRNAs can mediate endogenous gene expression by serving as chromatin remodeler, transcriptional or post-transcriptional modulator, and splicing regulator during gene modification. In recent years, increasing evidence shows the significance of lncRNAs in many malignancies, with vital roles in tumorigenesis and cancer progression. Moreover, lncRNAs were also considered potential diagnostic and prognostic markers in cancer. The lncRNA small nuclear RNA host gene 16 (SNHG16), found on chromosome 17q25.1, represents a novel tumor-associated lncRNA. SNHG16 was recently found to exhibit dysregulated expression in a variety of malignancies. There are growing evidence of SNHG16's involvement in characteristics of cancer, including proliferation, apoptosis, together with its involvement in chemoresistance. In addition, SNHG16 has been described as a promising diagnostic and prognostic biomarker in cancer patients. The current review briefly summarizes recently reported findings about SNHG16 and discuss its expression, roles, mechanisms, and diagnostic and prognostic values in human cancers.
Collapse
Affiliation(s)
- Yuhang Xiao
- Thoracic Surgery Department 2, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013 PR China
- Department of Pharmacy, Xiangya Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410001 PR China
| | - Ta Xiao
- Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, Jiangsu 210042 China
| | - Wei Ou
- Department of Pharmacy, The First People’s Hospital of Yue Yang, Yue Yang, PR China
| | - Zhining Wu
- Thoracic Surgery Department 2, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013 PR China
| | - Jie Wu
- Thoracic Surgery Department 2, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013 PR China
| | - Jinming Tang
- Thoracic Surgery Department 2, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013 PR China
| | - Bo Tian
- Thoracic Surgery Department 2, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013 PR China
| | - Yong Zhou
- Thoracic Surgery Department 2, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013 PR China
| | - Min Su
- Thoracic Surgery Department 2, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013 PR China
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Wenxiang Wang
- Thoracic Surgery Department 2, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013 PR China
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
21
|
Liu S, Meng X. LINC00662 Long Non-Coding RNA Knockdown Attenuates the Proliferation, Migration, and Invasion of Osteosarcoma Cells by Regulating the microRNA-15a-5p/Notch2 Axis. Onco Targets Ther 2020; 13:7517-7530. [PMID: 32848412 PMCID: PMC7429411 DOI: 10.2147/ott.s256464] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 07/10/2020] [Indexed: 12/14/2022] Open
Abstract
Purpose Osteosarcoma (OS) is a frequently occurring malignancy in children and adolescents. In this study, we aimed to investigate the effects of the long non-coding RNA (lncRNA) LINC00662 (LINC00662) in OS and the underlying molecular mechanism. Methods The expression of LINC00662, microRNA-15a-5p (miR-15a-5p), and Notch2 in OS was detected by quantitative real-time polymerase chain reaction (qRT-PCR). The proliferation, migration, and invasion of OS cells were analyzed by 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), wound-healing, and transwell assay. The interactions among LINC00662, miR-15a-5p, and Notch2 were determined by dual-luciferase reporter assays. A tumor xenograft model was established in mice for evaluating tumor growth in vivo. Results The expression of LINC00662 and Notch2 was found to be upregulated in OS, but the expression of miR-15a-5p was downregulated. The results demonstrated that LINC00662 knockdown attenuated the proliferation, migration, and invasion of OS cells and suppressed tumor growth in mice. The study further demonstrated that LINC00662 directly interacted with miR-15a-5p, and that Notch2 was a target of miR-15a-5p. The inhibition of miR-15a-5p or Notch2 overexpression markedly reversed the suppressive effect of sh-LINC00662 on the proliferation, migration, and invasion of OS cells. Conclusion The study demonstrated that LINC00662 could be a potential biomarker for OS therapy, and LINC00662 knockdown suppressed the proliferation, migration, and invasion of OS cells by regulating the miR-15a-5p/Notch2 axis.
Collapse
Affiliation(s)
- Shuheng Liu
- Department of Spine Surgery, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan City, Shandong Province, People's Republic of China
| | - Xianghai Meng
- Trauma Center, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan City, Shandong Province, People's Republic of China
| |
Collapse
|
22
|
Jiao R, Jiang W, Wei X, Zhang M, Zhao S, Huang G. Clinicopathological significance and prognosis of long noncoding RNA SNHG16 expression in human cancers: a meta-analysis. BMC Cancer 2020; 20:662. [PMID: 32677912 PMCID: PMC7366298 DOI: 10.1186/s12885-020-07149-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 07/07/2020] [Indexed: 12/31/2022] Open
Abstract
Background Recent studies have highlighted the important role of long non-coding RNA SNHG16 in various human cancers. Here, we conducted a meta-analysis to investigate the effect of SNHG16 expression on clinicopathological features and prognosis in patients with different kinds of human cancers. Methods We performed a systematic search in electronic databases including PubMed, EMBASE, Cochrane Library and Web of Science, to investigate the potential association between SNHG16 expression and prognostic significance and clinical features in cancer patients. Odds ratios (ORs) or hazards ratios (HRs) with corresponding 95% confidence intervals (95% CIs) were pooled to estimate the prognosis value of SNHG16 by StataSE 15.0 software. Results A total of 16 eligible studies with 1299 patients were enrolled in our meta-analysis. The results revealed that increased expression level of SNHG16 was significantly associated with larger tumor size (OR: 3.357; 95% CI: 2.173–5.185; P < 0.001), advanced TNM stage (OR: 2.930; 95% CI: 1.522–5.640; P = 0.001) and poor histological grade (OR: 3.943; 95% CI: 1.955–7.952; P < 0.001), but not correlated with smoking status (P = 0.489), sex (P = 0.932), distant metastasis (P = 0.052), or lymph node metastasis (P = 0.155). Moreover, the pooled HR showed that elevated expression SNHG16 was associated with a significantly poorer overall survival (OS) (HR = 1.866, 95% CI: 1.571–2.216, P < 0.001). For the set of cancer types, high expression of SNHG16 was significantly associated with shorter OS in patients with cancers of the urinary system (HR: 2.523, 95% CI:1.540–4.133; P <0.001), digestive system (HR: 2.406, 95% CI:1.556–3.721; P <0.001), and other cancers (including glioma and non-small cell lung cancer) (HR: 1.786, 95% CI:1.406–2.267; P <0.001). Conclusions LncRNA SNHG16 overexpression might serve as an unfavorable prognostic factor, which provides a basis for medical workers to evaluate the prognosis of patients and to help the decision-making process.
Collapse
Affiliation(s)
- Ruonan Jiao
- Medical Center for Digestive Diseases, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Wei Jiang
- Medical Center for Digestive Diseases, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Xin Wei
- Medical Center for Digestive Diseases, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Mengpei Zhang
- Medical Center for Digestive Diseases, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Si Zhao
- Medical Center for Digestive Diseases, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Guangming Huang
- Medical Center for Digestive Diseases, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China.
| |
Collapse
|
23
|
Zhang Y, Pu Y, Wang J, Li Z, Wang H. Research progress regarding the role of long non-coding RNAs in osteosarcoma. Oncol Lett 2020; 20:2606-2612. [PMID: 32782578 PMCID: PMC7400499 DOI: 10.3892/ol.2020.11807] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 05/29/2020] [Indexed: 12/16/2022] Open
Abstract
Osteosarcoma is a malignant tumor that occurs in children and adolescents. Although treatments for osteosarcoma have improved, the likelihood of survival remains low for most patients with metastasis and recurrence. Elucidating the mechanism underlying the development of osteosarcoma and chemotherapy resistance will be important to improve diagnosis and treatment. Long non-coding RNAs (lncRNAs), which are longer than 200 nucleotides in length and do not encode for proteins, have been shown to play a regulatory role in the occurrence and development of osteosarcoma, and are expected to serve as biomarkers and molecular targets. This review discusses the progress in the study of the role of lncRNAs in osteosarcoma, and highlights the recent developments in this field.
Collapse
Affiliation(s)
- Yanli Zhang
- Department of Orthopedics, Wuwei People's Hospital, Wuwei, Gansu 733000, P.R. China
| | - Yanchuan Pu
- Department of Orthopedics, Wuwei People's Hospital, Wuwei, Gansu 733000, P.R. China
| | - Jin Wang
- Department of Orthopedics, Wuwei People's Hospital, Wuwei, Gansu 733000, P.R. China
| | - Zicai Li
- Department of Orthopedics, Wuwei People's Hospital, Wuwei, Gansu 733000, P.R. China
| | - Hulin Wang
- Department of Orthopedics, Wuwei People's Hospital, Wuwei, Gansu 733000, P.R. China
| |
Collapse
|
24
|
Wang Y, Tong J, Lin H, Ma L, Xu Y. CCHE1 accelerated the initiation of oral squamous cell carcinoma through enhancing PAK2 expression by sponging miR-922. J Oral Pathol Med 2020; 49:636-644. [PMID: 31981240 DOI: 10.1111/jop.12995] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 12/27/2019] [Accepted: 01/20/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) is a normal form of mouth cancer, comprising the majority of oral cancers. A large number of long non-coding RNAs (lncRNAs) have been reported due to their oncogenic function in cancers. Recent studies show that lncRNA CCHE1 is an oncogene in a wide range of cancers. Whether CCHE1 accelerates the progression of OSCC is still undiscovered. METHODS The qRT-PCR analysis was used to determine CCHE1, miR-922, and PAK2 expression levels. CCK8 and colony formation assays were applied to evaluate OSCC cell proliferative ability. Transwell assay was performed to investigate the capability of cell migration and invasion. Cell apoptosis was assessed by flow cytometry analysis. The distribution of CCHE1 in OSCC cells was confirmed via subcellular fractionation assay. Luciferase reporter assay was used to verify the connection between miR-922 and CCHE1 or PAK2. RESULTS qRT-PCR analysis identified the upregulation of CCHE1 in OSCC cells. Knockdown of CCHE1 curbed the proliferation, migration, and invasion and hastened the apoptosis in OSCC cell lines. Moreover, it was found that miR-922 could interact with CCHE1. Besides, PAK2 was identified as the target gene of miR-922 and its expression was negatively modulated by miR-922 and positively regulated by CCHE1. Restoration assay manifested that the suppressing influence of CCHE1 depletion on OSCC progression was rescued by amplified PAK2. CONCLUSIONS CCHE1 increases the expression of PAK2 to promote the progression of OSCC by competitively binding to miR-922 in OSCC cells.
Collapse
Affiliation(s)
- Yongliang Wang
- Department of Prosthodontics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Junjie Tong
- Department of Oral Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Haozhi Lin
- Department of Oral Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lei Ma
- Department of Prosthodontics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yaoxiang Xu
- Department of Oral Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China.,Dental Digital Medicine & 3D Printing Engineering Laboratory of Qingdao University, Qingdao, China
| |
Collapse
|
25
|
Yang X, Huang H, Wang X, Liu H, Liu H, Lin Z. Knockdown of lncRNA SNHG16 suppresses multiple myeloma cell proliferation by sponging miR-342-3p. Cancer Cell Int 2020; 20:38. [PMID: 32025219 PMCID: PMC6998159 DOI: 10.1186/s12935-020-1118-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 01/20/2020] [Indexed: 12/12/2022] Open
Abstract
Background Aberrant expression of long non-coding RNAs (lncRNAs) is closely associated with development and prognosis of human cancers. LncRNA SNHG16 is reportedly involved in human cancer; however, its roles in multiple myeloma (MM) remain unclear. Methods In this study, we investigated the function and molecular mechanisms of SNHG16 in MM. MM cells were transfected with si-SNHG16 or si-NC. SNHG16 expression levels was measured by qRT-PCR. Cell proliferation was monitored using the MTS. Flow cytometry assay was performed to measure the cell cycle and apoptosis. Luciferase reporter assay were performed to confirm the sponged miRNAs of SNHG16. Results SNHG16 expression was up-regulated in MM tissues. SNHG16 knockdown suppressed cell proliferation, arrested cell cycle transition from G1 to S phase, and promoted the apoptosis of MM cells. Moreover, SNHG16 knockdown promoted cleaved-Caspase-3, cleaved-Caspase-9, Foxa3a, and Bax expression, while markedly inhibiting CCND1, Bcl-2, Cyclin D1, PI3K, and p-AKT expression in MM cells. miR-342-3p was a direct target of SNHG16. SNHG16 knockdown significantly increased miR-342-3p expression in MM cells. Overexpression miR-342-3p markedly suppressed cell proliferation, arrested cell cycle transition from G1 to S phase, and promoted apoptosis of MM cells. Overexpression of miR-342-3p markedly promoted cleaved-Caspase-3/-9, Foxa3a, and Bax expression, and inhibited CCND1, Bcl-2, Cyclin D1, PI3K, and p-AKT expression in MM cells. Additionally, repression of miR-342-3p could rescue the effect of SNHG16 knockdown on MM cell proliferation, cycle arrest, apoptosis, and related protein expression. Conclusion Knockdown of lncRNA SNHG16 suppresses MM cell proliferation by sponging miR-342-3p, implicating SNHG16 as a novel therapeutic target for MM.
Collapse
Affiliation(s)
- Xi Yang
- 1Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215006 Jiangsu People's Republic of China.,2Department of Hematology, Affiliated Hospital of NanTong University, No.20 Xishi Road, Nantong, 226001 Jiangsu People's Republic of China
| | - Hongming Huang
- 2Department of Hematology, Affiliated Hospital of NanTong University, No.20 Xishi Road, Nantong, 226001 Jiangsu People's Republic of China
| | - Xinfeng Wang
- 2Department of Hematology, Affiliated Hospital of NanTong University, No.20 Xishi Road, Nantong, 226001 Jiangsu People's Republic of China
| | - Haiyan Liu
- 2Department of Hematology, Affiliated Hospital of NanTong University, No.20 Xishi Road, Nantong, 226001 Jiangsu People's Republic of China
| | - Hong Liu
- 2Department of Hematology, Affiliated Hospital of NanTong University, No.20 Xishi Road, Nantong, 226001 Jiangsu People's Republic of China
| | - Zenghua Lin
- 2Department of Hematology, Affiliated Hospital of NanTong University, No.20 Xishi Road, Nantong, 226001 Jiangsu People's Republic of China
| |
Collapse
|
26
|
A commentary on “Long non-coding RNA MALAT1 as a valuable biomarker for prognosis in osteosarcoma: A systematic review and meta-analysis” (Int J Surg 2019;72:206–213). Int J Surg 2020; 74:27-28. [DOI: 10.1016/j.ijsu.2019.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 11/18/2022]
|
27
|
Qu Z, Li S. Long noncoding RNA LINC01278 favors the progression of osteosarcoma via modulating miR-133a-3p/PTHR1 signaling. J Cell Physiol 2020. [PMID: 31994731 DOI: 10.1002/jcp.29582] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 01/13/2020] [Indexed: 12/12/2022]
Abstract
As one of the most aggressive malignancies, osteosarcoma has high risks of death. Although long noncoding RNAs (lncRNAs) may promote the osteosarcoma progression as verified, the potential molecular mechanism of lncRNAs in osteosarcoma remains unknown. Herein, we analyzed lncRNA microarray of osteosarcoma and selected LINC01278 as the study object. Then, we found that the expression of LINC01278 tested by quantitative reverse-transcription polymerase chain reaction was enhanced in tumor tissues compared with the para-carcinoma tissues and related to clinical stage, distant metastasis in osteosarcoma. In addition, the clinical outcomes were poor in osteosarcoma patients with high LINC01278 level. Moreover, LINC01278 promoted proliferation and restrained apoptosis in osteosarcoma cells. Afterward, mechanistic studies turned out that LINC01278 was a competing endogenous RNA of parathyroid hormone type 1 receptor (PTHR1) in osteosarcoma by sponging miR-133a-3p, which was considered as a tumor inhibitor in osteosarcoma. Furthermore, PTHR1 downregulation restored the impacts of inhibited miR-133a-3p on the processes in osteosarcoma cells. Our findings clarified that the carcinogenic effect of LINC01278 in osteosarcoma was mediated through miR-133a-3p/PTHR1 signaling, creating a novel insight into good targets for the therapy and prognosis of osteosarcoma.
Collapse
Affiliation(s)
- Zhigang Qu
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Shenglong Li
- Department of Bone and Soft Tissue Tumor Surgery, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, China
| |
Collapse
|
28
|
Yang M, Wei W. SNHG16: A Novel Long-Non Coding RNA in Human Cancers. Onco Targets Ther 2019; 12:11679-11690. [PMID: 32021246 PMCID: PMC6942535 DOI: 10.2147/ott.s231630] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 12/13/2019] [Indexed: 01/27/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) have recently been considered as central regulators in diverse biological processes controlling tumorigenesis. Small nucleolar RNA host gene 16 (SNHG16) is an important tumor-associated lncRNA mainly involved in tumorigenesis and progression by competing with endogenous RNA (ceRNA) which sponges tumor-suppressive microRNA (miRNA), and by its recruitment mechanism. SNHG16 is overexpressed in tumor tissues and cell lines of different kinds of cancers, and its presence is associated with a poor clinical prognosis. Reviewing all publications about SNHG16 revealed that it plays a key role in the different hallmarks that define human cancer, including promoting proliferation, activating migration and invasion, inhibiting apoptosis, affecting lipid metabolism and chemoresistance. This review highlights the role that the aberrant expression of SNHG16 plays in the development and progression of cancer, and suggests that SNHG16 may function as a potential biomarker and therapeutic target for human cancers.
Collapse
Affiliation(s)
- Ming Yang
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology and Visual Sciences Key Lab, Beijing Tongren Hospital, Capital Medical University, Beijing100730, People’s Republic of China
| | - Wenbin Wei
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology and Visual Sciences Key Lab, Beijing Tongren Hospital, Capital Medical University, Beijing100730, People’s Republic of China
| |
Collapse
|
29
|
Role of SNHG16 in human cancer. Clin Chim Acta 2019; 503:175-180. [PMID: 31901482 DOI: 10.1016/j.cca.2019.12.023] [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: 09/12/2019] [Revised: 12/27/2019] [Accepted: 12/30/2019] [Indexed: 01/27/2023]
Abstract
A growing body of evidence suggests that long non-coding RNAs (lncRNAs), a novel class of non-coding endogenous single-stranded RNA, play a key role in multiple physiological and pathological processes through transcriptional interference, post-transcriptional regulation, and epigenetic modification. Furthermore, many studies have shown that lncRNAs-as oncogenes or tumour suppressors-play an important role in the occurrence and development of human cancers. Small nucleolar RNA host gene 16 (SNHG16) was initially identified as an oncogenic lncRNA in neuroblastoma, and has since been identified as a carcinogenic regulator of various malignant tumours. Overexpression of SNHG16 is associated with clinical and pathological characteristics of cancer patients, and regulates cell proliferation, apoptosis, invasion and metastasis through a variety of potential mechanisms. Therefore, SNHG16 may be a promising biomarker and therapeutic target for cancers. In this review, we summarize the biological function, related mechanisms and potential clinical significance of SNHG16 in multiple human cancers.
Collapse
|
30
|
Niu X, Yang B, Liu F, Fang Q. LncRNA HOXA11-AS promotes OSCC progression by sponging miR-98-5p to upregulate YBX2 expression. Biomed Pharmacother 2019; 121:109623. [PMID: 31731187 DOI: 10.1016/j.biopha.2019.109623] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/25/2019] [Accepted: 11/01/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) is a type of oral malignancy. Long non-coding RNAs (lncRNAs) have been shown to be related to the occurrence and development of many cancers. Here, we aimed to study the role and molecular mechanism of lncRNA Homeobox A11 antisense RNA (HOXA11-AS) in OSCC. METHODS The expression levels of HOXA11-AS, miR-98-5p and Y box binding protein 2 (YBX2) were detected by quantitative real-time polymerase chain reaction (qRT-PCR). Cell counting kit-8 (CCK-8), flow cytometry and transwell assays were utilized to determine the proliferation, apoptosis, migration and invasion of OSCC cells. Western blot (WB) analysis was conducted to measure the levels of apoptosis, epithelial-mesenchymal transition (EMT), proliferation-related proteins and YBX2 protein. Besides, Dual-luciferase reporter, RNA immunoprecipitation (RIP) and RNA pull down assays were carried out to examine the relationship among HOXA11-AS, miR-98-5p and YBX2. The mice xenograft models were constructed to further determine the effect of HOXA11-AS on OSCC tumor growth in vivo. RESULTS HOXA11-AS was highly expressed in OSCC tissues and cells. Knockdown of HOXA11-AS significantly reduced proliferation, migration, invasion and EMT, while promoted apoptosis of OSCC cells. MiR-98-5p was a target of HOXA11-AS, and its inhibitor could revert the inhibition effect of silenced-HOXA11-AS on the progression of OSCC. Also, YBX2 was a target of miR-98-5p, and its overexpression could invert the suppression effect of miR-98-5p overexpression on the progression of OSCC. YBX2 expression was regulated by HOXA11-AS and miR-98-5p. Furthermore, HOXA11-AS silencing could reduce the tumor growth of OSCC in vivo. CONCLUSION HOXA11-AS plays an active role in the progression of OSCC, and the discovery of HOXA11-AS/miR-98-5p/YBX2 axis provides new therapeutic targets for OSCC.
Collapse
Affiliation(s)
- Xingyu Niu
- Department of Oral Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Bin Yang
- Department of Ophtalmology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Fei Liu
- Department of Oral Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qigen Fang
- Department of Head Neck and Thyroid, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
31
|
Li C, Wang F, Wei B, Wang L, Kong D. LncRNA AWPPH promotes osteosarcoma progression via activation of Wnt/β-catenin pathway through modulating miR-93-3p/FZD7 axis. Biochem Biophys Res Commun 2019; 514:1017-1022. [PMID: 31092328 DOI: 10.1016/j.bbrc.2019.04.203] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 04/30/2019] [Indexed: 01/17/2023]
Abstract
Long noncoding RNAs (lncRNAs) have important regulatory roles in osteosarcoma (OS) progression. Recent researches have shown lncRNA AWPPH promotes lung cancer progression and bladder cancer development. Yet, the function of AWPPH in OS is unknown. In this research, results indicated AWPPH levels were increased in OS tissues in contrast to paracancerous controls. Up-regulated AWPPH was associated with advanced stage, tumor size and metastasis. Besides, AWPPH up-regulation indicated a low survival rate in OS patients. Silencing of AWPPH suppressed proliferation, migration and invasion of OS cells. Mechanistically, AWPPH was demonstrated to sponge miR-93-3p and promote FZD7 expression, causing activation of Wnt/β-catenin. Inhibition of miR-93-3p effectively reversed the effects of AWPPH knockdown on OS cells. Collectively, our findings suggested AWPPH may be a prognostic biomarker and potential therapeutic target. AWPPH enhances FZD7-mediated activation of Wnt/β-catenin by sponging miR-93-3p to promote OS progression.
Collapse
Affiliation(s)
- Chang Li
- Department of VIP Unit, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Fei Wang
- Department of Orthopaedics, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Bo Wei
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Le Wang
- Department of Ophthalmology, The First Affiliated Hospital of Jilin University, China
| | - Daliang Kong
- Department of Orthopaedics, China-Japan Union Hospital of Jilin University, Changchun, 130033, China.
| |
Collapse
|