1
|
Alimohammadi M, Fooladi AAI, Mafi A, Alavioun SM, Cho WC, Reiter RJ, Khormizi FZ, Yousefi T, Farahani N, Khoshnazar SM, Hushmandi K. Long noncoding RNAs and HPV-related cervical cancer: Uncovering molecular mechanisms and clinical applications. Transl Oncol 2025; 55:102363. [PMID: 40121995 PMCID: PMC11982485 DOI: 10.1016/j.tranon.2025.102363] [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: 10/30/2024] [Revised: 03/08/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025] Open
Abstract
Cervical cancer (CC) is the primary cause of cancer-related mortality among women in developing countries and is the most prevalent disease linked to human papillomavirus (HPV). Over 70 % of CC cases result from persistent infections with high-risk HPV types. The virus typically targets the mucocutaneous epithelium, generating viral particles in mature epithelial cells, which leads to disruptions in normal cell-cycle regulation and promotes uncontrolled cellular proliferation. This unchecked cell division results in the accumulation of genetic damage, contributing to the pathogenesis of CC. While HPV infection is a key etiological factor, the disease's progression also necessitates the involvement of genetic and epigenetic influences. One of the epigenetic regulators, long noncoding RNAs (lncRNAs), are characterized by transcripts exceeding 200 nucleotides. These molecules play crucial roles in various cellular processes, including transcription regulation, RNA metaboli35 per 100,000sm, and apoptosis. Investigating the specific roles of lncRNAs in modulating gene expression related to the oncogenic mechanisms of CC, particularly in the context of high-risk HPV infections, may provide valuable insights for diagnostic and therapeutic advancements. Herein, we first review key molecular mechanisms by which lncRNAs interfere with CC-related HPV development. Then, diagnostic, prognostic, and therapeutic potentials of these lncRNA molecules will be highlighted in depth. The focus of this article is on the role of lncRNAs associated with HPV-related CC, emphasizing the investigation of signaling pathways and their underlying molecular mechanisms. Furthermore, we explore the therapeutic potential and diagnostic relevance of the most significant lncRNAs in the context of CC, thereby highlighting their importance in advancing treatment strategies and improving patient outcomes.
Collapse
Affiliation(s)
- Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Ali Imani Fooladi
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Alireza Mafi
- Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran; Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Seyedeh Mana Alavioun
- Department of Basic sciences, Faculty of Veterinary Medicine, Urmia university, Urmia, Iran
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, Long School of Medicine, San Antonio, TX, USA
| | | | - Tooba Yousefi
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Seyedeh Mahdieh Khoshnazar
- Gastroenterology and Hepatology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran.
| | - Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Elimam H, Abdel Mageed SS, Hatawsh A, Moussa R, Radwan AF, Elfar N, Alhamshry NAA, Abd-Elmawla MA, Mohammed OA, Zaki MB, Doghish AS. Unraveling the influence of LncRNA in gastric cancer pathogenesis: a comprehensive review focus on signaling pathways interplay. Med Oncol 2024; 41:218. [PMID: 39103705 DOI: 10.1007/s12032-024-02455-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 07/16/2024] [Indexed: 08/07/2024]
Abstract
Gastric cancers (GCs) are among the most common and fatal malignancies in the world. Despite our increasing understanding of the molecular mechanisms underlying GC, further biomarkers are still needed for more in-depth examination, focused prognosis, and treatment. GC is one among the long non-coding RNAs, or lncRNAs, that have emerged as key regulators of the pathophysiology of cancer. This comprehensive review focuses on the diverse functions of long noncoding RNAs (lncRNAs) in the development of GC and their interactions with important intracellular signaling pathways. LncRNAs affect GC-related carcinogenic signaling cascades including pathways for EGFR, PI3K/AKT/mTOR, p53, Wnt/β-catenin, JAK/STAT, Hedgehog, NF-κB, and hypoxia-inducible factor. Dysregulated long non-coding RNA (lncRNA) expression has been associated with multiple characteristics of cancer, such as extended growth, apoptosis resistance, enhanced invasion and metastasis, angiogenesis, and therapy resistance. For instance, lncRNAs such as HOTAIR, MALAT1, and H19 promote the development of GC via altering these pathways. Beyond their main roles, GC lncRNAs exhibit potential as diagnostic and prognostic biomarkers. The overview discusses CRISPR/Cas9 genome-modifying methods, antisense oligonucleotides, small molecules, and RNA interference as potential therapeutic approaches to regulate the expression of long noncoding RNAs (lncRNAs). An in-depth discussion of the intricate functions that lncRNAs play in the development of the majority of stomach malignancies is provided in this review. It provides the groundwork for future translational research in lncRNA-based whole processes toward GC by highlighting their carcinogenic effects, regulatory roles in significant signaling cascades, and practical scientific uses as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Hanan Elimam
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, 32897, Egypt.
| | - Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Abdulrahman Hatawsh
- Biotechnology School, Nile University, 26th of July Corridor, Sheikh Zayed City, 12588, Giza, Egypt
| | - Rewan Moussa
- Faculty of Medicine, Helwan University, Cairo, 11795, Egypt
| | - Abdullah F Radwan
- Department of Biochemistry, Faculty of Pharmacy, Egyptian Russian University, Badr City, 11829, Cairo, Egypt
| | - Nourhan Elfar
- School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation, New Administrative Capital, 11578, Cairo, Egypt
- Egyptian Drug Authority (EDA), Ministry of Health and Population, Cairo, 11567, Egypt
| | - Nora A A Alhamshry
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, 32897, Egypt
| | - Mai A Abd-Elmawla
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, 61922, Bisha, Saudi Arabia
| | - Mohamed Bakr Zaki
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, 32897, Egypt
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt.
- Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231, Cairo, Egypt.
| |
Collapse
|
3
|
Mohammadi R, Zareh A, Rabani E, Kheirandish Zarandi P, Khoncheh A, Heiat M. Expression of Pivotal Long Non-coding RNAs Implicated in Gastric Cancer: A Bioinformatic and Clinical Study. Biochem Genet 2024; 62:3111-3135. [PMID: 38070023 DOI: 10.1007/s10528-023-10586-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 11/03/2023] [Indexed: 07/31/2024]
Abstract
Gastric cancer (GC) is a prominent public health issue and ranks as the third most prevalent cause of cancer-related mortality on a global scale. The role of long non-coding RNAs (lncRNAs) in cancer is not yet fully understood, particularly in relation to GC development. The objective of this study was to examine the expression levels of lncRNAs in GC tissues using a bioinformatics-based ranking approach. A bioinformatics methodology was employed to prioritize lncRNAs that are hypothesized to play a role in GC tumorigenesis. Moreover, a selection was made for experimental validation of the highest-ranked lncRNAs, which include HCG18, OIP5-AS1, FGD5-AS1, and NORAD. Additionally, quantitative real-time polymerase chain reaction (qRT-PCR) was employed to confirm the results obtained from bioinformatics analysis in a total of 35 GC samples and their corresponding adjacent non-tumoral samples. Receiver operating characteristic (ROC) curves and the corresponding area under the ROC curve (AUC) were utilized to evaluate the diagnostic efficacy of the lncRNAs. The bioinformatics analysis revealed that the lncRNA HCG18 is the highest-ranked lncRNA associated with GC. Furthermore, the expression levels of HCG18, OIP5-AS1, FGD5-AS1, and NORAD were found to be significantly elevated in GC samples when compared to adjacent non-tumoral samples. The calculated values for the AUC of HCG18, OIP5-AS1, FGD5-AS1, and NORAD were 0.80, 0.74, 0.73, and 0.71, respectively. The results of the study indicate that the lncRNAs HCG18, OIP5-AS1, FGD5-AS1, and NORAD may play a role in the development of GC. Additionally, the present study revealed that utilizing bioinformatic techniques can prove to be a highly effective strategy in identifying potential lncRNAs pertinent to the progression of GC.
Collapse
Affiliation(s)
- Ramtin Mohammadi
- Department of Biology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Ali Zareh
- Baqiyatallah Research Center for Gastroenterology and Liver Diseases (BRCGL), Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Elmira Rabani
- Department of Biology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Peyman Kheirandish Zarandi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
- Cancer Biology Signaling Pathway Interest Group (CBSPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Ahmad Khoncheh
- Baqiyatallah Research Center for Gastroenterology and Liver Diseases (BRCGL), Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Heiat
- Baqiyatallah Research Center for Gastroenterology and Liver Diseases (BRCGL), Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Li K, Wang H, Jiang B, Jin X. TRIM28 in cancer and cancer therapy. Front Genet 2024; 15:1431564. [PMID: 39100077 PMCID: PMC11294089 DOI: 10.3389/fgene.2024.1431564] [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: 05/12/2024] [Accepted: 07/01/2024] [Indexed: 08/06/2024] Open
Abstract
TRIM28 (tripartite motif protein 28) was initially believed to be a transcription inhibitor that plays an important role in DNA damage repair (DDR) and in maintaining cancer cellular stemness. As research has continued to deepen, several studies have found that TRIM28 not only has ubiquitin E3 ligase activity to promote degradation of substrates, but also can promote SUMOylation of substrates. Although TRIM28 is highly expressed in various cancer tissues and has oncogenic effects, there are still a few studies indicating that TRIM28 has certain anticancer effects. Additionally, TRIM28 is subject to complex upstream regulation. In this review, we have elaborated on the structure and regulation of TRIM28. At the same time, highlighting the functional role of TRIM28 in tumor development and emphasizing its impact on cancer treatment provides a new direction for future clinical antitumor treatment.
Collapse
Affiliation(s)
- Kailang Li
- Department of Oncology and Hematology, Beilun District People’s Hospital, Ningbo, China
| | - Haifeng Wang
- Department of Oncology and Hematology, Beilun District People’s Hospital, Ningbo, China
| | - Bitao Jiang
- Department of Oncology and Hematology, Beilun District People’s Hospital, Ningbo, China
| | - Xiaofeng Jin
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathphysiology, Medical School of Ningbo University, Ningbo, China
| |
Collapse
|
5
|
Saeinasab M, Atlasi Y, M Matin M. Functional role of lncRNAs in gastrointestinal malignancies: the peculiar case of small nucleolar RNA host gene family. FEBS J 2024; 291:1353-1385. [PMID: 36282516 DOI: 10.1111/febs.16668] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/18/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
Abstract
Long noncoding RNAs (lncRNAs) play crucial roles in normal physiology and are often de-regulated in disease states such as cancer. Recently, a class of lncRNAs referred to as the small nucleolar RNA host gene (SNHG) family have emerged as important players in tumourigenesis. Here, we discuss new findings describing the role of SNHGs in gastrointestinal tumours and summarize the three main functions by which these lncRNAs promote carcinogenesis, namely: competing with endogenous RNAs, modulating protein function, and regulating epigenetic marking. Furthermore, we discuss how SNHGs participate in different hallmarks of cancer, and how this class of lncRNAs may serve as potential biomarkers in cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Morvarid Saeinasab
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Iran
| | - Yaser Atlasi
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, UK
| | - Maryam M Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Iran
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Iran
| |
Collapse
|
6
|
Zhang X, Shao R. LncRNA SNHG8 upregulates MUC5B to induce idiopathic pulmonary fibrosis progression by targeting miR-4701-5p. Heliyon 2024; 10:e23233. [PMID: 38163156 PMCID: PMC10756985 DOI: 10.1016/j.heliyon.2023.e23233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 01/03/2024] Open
Abstract
Long noncoding RNAs (lncRNAs) play a critical role in idiopathic pulmonary fibrosis (IPF); however, the underlying molecular mechanisms are unclear. Our study demonstrated that lncRNA small nucleolar RNA host gene 8 (SNHG8) was increased in bleomycin (BLM)-induced A549 cells. LncRNA SNHG8 overexpression further elevated fibrosis-related factors monocyte chemotactic protein 1 (MCP1), CC motif chemokine ligand 18 (CCL18), and α-smooth muscle actin (α-SMA), as well as increased collagen type I alpha-1 chain (COL1A1) and collagen type III alpha-1 chain (COL3A1). Meanwhile, lncRNA SNHG8 knockdown exhibited an opposite role in reducing BLM-induced pulmonary fibrosis. With regard to the mechanism, SNHG8 was then revealed to act as a competing endogenous RNA (ceRNA) for microRNA (miR)-4701-5p in regulating Mucin 5B (MUC5B) expression. Furthermore, the interactions between SNHG8 and miR-4701-5p, between miR-4701-5p and MUC5B, and between SNHG8 and MUC5B on the influence of fibrosis-related indicators were confirmed, respectively. In addition, SNHG8 overexpression enhanced the levels of transforming growth factor (TGF)-β1 and phosphorylation Smad2/3 (p-Smad2/3), which was suppressed by SNHG8 knockdown in BLM-induced A549 cells. Moreover, miR-4701-5p inhibitor-induced elevation of TGF-β1 and p-Smad2/3 was significantly suppressed by SNHG8 knockdown. In conclusion, SNHG8 knockdown attenuated pulmonary fibrosis progression by regulating miR-4701-5p/MUC5B axis, which might be associated with the modulation of TGF-β1/Smad2/3 signaling. These findings reveal that lncRNA SNHG8 may become a potential target for the treatment of IPF.
Collapse
Affiliation(s)
- Xiaoping Zhang
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450014, China
| | - Runxia Shao
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450014, China
| |
Collapse
|
7
|
Liu S, Rong Y, Tang M, Zhao Q, Li C, Gao W, Yang X. The Functions and Mechanisms of Long Non-coding RNA SNHGs in Gastric Cancer. Comb Chem High Throughput Screen 2024; 27:2639-2653. [PMID: 37842903 DOI: 10.2174/0113862073268591230928100922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/08/2023] [Accepted: 09/01/2023] [Indexed: 10/17/2023]
Abstract
Gastric cancer (GC) is one of the most common malignancies worldwide. Despite significant advancements in surgical and adjuvant treatments, patient prognosis remains unsatisfactory. Long non-coding RNAs (lncRNAs) are a class of RNA molecules that lack protein-coding capacity but can engage in the malignant biological behaviors of tumors through various mechanisms. Among them, small nucleolar host genes (SNHGs) represent a subgroup of lncRNAs. Studies have revealed their involvement not only in gastric cancer cell proliferation, invasion, migration, epithelial- mesenchymal transition (EMT), and apoptosis but also in chemotherapy resistance and tumor stemness. This review comprehensively summarizes the biological functions, molecular mechanisms, and clinical significance of SNHGs in gastric cancer. It provides novel insights into potential biomarkers and therapeutic targets for the exploration of gastric cancer.
Collapse
Affiliation(s)
- Songhua Liu
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou 730000, China
| | - Yao Rong
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou 730000, China
| | - Mingzheng Tang
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou 730000, China
| | - Qiqi Zhao
- College of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, China
| | - Chunyan Li
- The First Clinical Medical School of Lanzhou University, Lanzhou, 730000, China
| | - Wenbin Gao
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou 730000, China
| | - Xiaojun Yang
- The First Clinical Medical School of Lanzhou University, Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, 730000, China
- Gansu key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology, Gansu Provincial Hospital, Lanzhou, 730000, China
- Gansu Research Center of Prevention and Control Project for Digestive Oncology, Gansu Provincial Hospital, Lanzhou, 730000, China
- Key Laboratory of Gastrointestinal Tumor Diagnosis and Treatment of National Health and Health Commission, Lanzhou, 730000, China
| |
Collapse
|
8
|
Matsuoka T, Yashiro M. Molecular Insight into Gastric Cancer Invasion-Current Status and Future Directions. Cancers (Basel) 2023; 16:54. [PMID: 38201481 PMCID: PMC10778111 DOI: 10.3390/cancers16010054] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/15/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Gastric cancer (GC) is one of the most common malignancies worldwide. There has been no efficient therapy for stage IV GC patients due to this disease's heterogeneity and dissemination ability. Despite the rapid advancement of molecular targeted therapies, such as HER2 and immune checkpoint inhibitors, survival of GC patients is still unsatisfactory because the understanding of the mechanism of GC progression is still incomplete. Invasion is the most important feature of GC metastasis, which causes poor mortality in patients. Recently, genomic research has critically deepened our knowledge of which gene products are dysregulated in invasive GC. Furthermore, the study of the interaction of GC cells with the tumor microenvironment has emerged as a principal subject in driving invasion and metastasis. These results are expected to provide a profound knowledge of how biological molecules are implicated in GC development. This review summarizes the advances in our current understanding of the molecular mechanism of GC invasion. We also highlight the future directions of the invasion therapeutics of GC. Compared to conventional therapy using protease or molecular inhibitors alone, multi-therapy targeting invasion plasticity may seem to be an assuring direction for the progression of novel strategies.
Collapse
Affiliation(s)
| | - Masakazu Yashiro
- Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka 5458585, Japan;
| |
Collapse
|
9
|
Huldani H, Gandla K, Asiri M, Romero-Parra RM, Alsalamy A, Hjazi A, Najm MAA, Fawaz A, Hussien BM, Singh R. A comprehensive insight into the role of small nucleolar RNAs (snoRNAs) and SNHGs in human cancers. Pathol Res Pract 2023; 249:154679. [PMID: 37567032 DOI: 10.1016/j.prp.2023.154679] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/04/2023] [Accepted: 07/07/2023] [Indexed: 08/13/2023]
Abstract
Long non-coding RNAs (lncRNAs), which comprise most non-coding RNAs (ncRNAs), have recently become a focus of cancer research. How many functional ncRNAs exist is still a matter of debate. Although insufficient evidence supports that most lncRNAs function as transcriptional by-products, it is widely known that an increasing number of lncRNAs play essential roles in cells. Small nucleolar RNAs (snoRNAs), 60-300 nucleotides in length, have been better studied than long non-coding RNAs (lncRNAs) and are predominantly present in the nucleolus. Most snoRNAs are encoded in introns of protein- and non-protein-coding genes called small nucleolar RNA host genes (SNHGs). In this article, we explore the biology and characteristics of SNHGs and their role in developing human malignancies. In addition, we provide an update on the ability of these snoRNAs to serve as prognostic and diagnostic variables in various forms of cancer.
Collapse
Affiliation(s)
- Huldani Huldani
- Department of Physiology, Faculty of Medicine, Lambung Mangkurat University, Banjarmasin, South Kalimantan, Indonesia
| | - Kumaraswamy Gandla
- Department of Pharmaceutical Analysis, Chaitanya Deemed to be University, Hanamkonda, India.
| | - Mohammed Asiri
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | | | - Ali Alsalamy
- College of Medical Technology, Imam Ja'afar Al-Sadiq University, Al-Muthanna 66002, Iraq
| | - Ahmed Hjazi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Mazin A A Najm
- Pharmaceutical Chemistry Department, College of Pharmacy, Al-Ayen University, Thi-Qar, Iraq
| | - Albab Fawaz
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | - Beneen M Hussien
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Rajesh Singh
- Uttaranchal Institute of Technology, Uttaranchal University, Dehradun 248007, India
| |
Collapse
|
10
|
Ghafouri-Fard S, Harsij A, Hussen BM, Taheri M, Ayatollahi SA. A review on the role of SNHG8 in human disorders. Pathol Res Pract 2023; 245:154458. [PMID: 37043963 DOI: 10.1016/j.prp.2023.154458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 04/01/2023] [Accepted: 04/07/2023] [Indexed: 04/14/2023]
Abstract
Small nucleolar RNA host gene 8 (SNHG8) is a long non-coding RNA that has physiological roles in epithelial and muscle satellite cells. This lncRNA has been reported to be over-expressed in a variety of cancer cell lines. Its silencing has attenuated tumor growth in animal models of cancers. SNHG8 can be served as a molecular sponge for some miRNAs to regulate their target genes. miR-634/ZBTB20, miR-335-5p/PYGO2, miR588/ATG7, miR-152/c-MET, miR-1270/BACH1, miR-491/PDGFRA, miR-512-5p/TRIM28, miR-149-5p/PPM1F, miR-542-3p/CCND1/CDK6, miR-656-3p/SERBP1, miR-656-3p/SATB1, miR-1270/S100A11 and miR-384/HOXB7 are examples of molecular axes being regulated by SNHG8 in the context of cancer. Moreover, it can affect pathogenesis of atherosclerosis, chronic cerebral ischemia, acute gouty arthritis, ischemic stroke and myocardial infarction through modulation of a number of molecular axes such as SNHG8/miR-384/Hoxa13/FAM3A and miR-335/RASA1 as well as NF-κB signaling pathway. The current review aims at summarization of the role of SNHG8 in diverse human disorders.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atefeh Harsij
- Men's Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bashdar Mahmud Hussen
- Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Kurdistan, Erbil, Iraq
| | - Mohammad Taheri
- Institute of Human Genetics, Jena University Hospital, Jena, Germany; Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | | |
Collapse
|
11
|
Hashemi M, Rashidi M, Hushmandi K, Ten Hagen TLM, Salimimoghadam S, Taheriazam A, Entezari M, Falahati M. HMGA2 regulation by miRNAs in cancer: affecting cancer hallmarks and therapy response. Pharmacol Res 2023; 190:106732. [PMID: 36931542 DOI: 10.1016/j.phrs.2023.106732] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 03/17/2023]
Abstract
High mobility group A 2 (HMGA2) is a protein that modulates the structure of chromatin in the nucleus. Importantly, aberrant expression of HMGA2 occurs during carcinogenesis, and this protein is an upstream mediator of cancer hallmarks including evasion of apoptosis, proliferation, invasion, metastasis, and therapy resistance. HMGA2 targets critical signaling pathways such as Wnt/β-catenin and mTOR in cancer cells. Therefore, suppression of HMGA2 function notably decreases cancer progression and improves outcome in patients. As HMGA2 is mainly oncogenic, targeting expression by non-coding RNAs (ncRNAs) is crucial to take into consideration since it affects HMGA2 function. MicroRNAs (miRNAs) belong to ncRNAs and are master regulators of vital cell processes, which affect all aspects of cancer hallmarks. Long ncRNAs (lncRNAs) and circular RNAs (circRNAs), other members of ncRNAs, are upstream mediators of miRNAs. The current review intends to discuss the importance of the miRNA/HMGA2 axis in modulation of various types of cancer, and mentions lncRNAs and circRNAs, which regulate this axis as upstream mediators. Finally, we discuss the effect of miRNAs and HMGA2 interactions on the response of cancer cells to therapy. Regarding the critical role of HMGA2 in regulation of critical signaling pathways in cancer cells, and considering the confirmed interaction between HMGA2 and one of the master regulators of cancer, miRNAs, targeting miRNA/HMGA2 axis in cancer therapy is promising and this could be the subject of future clinical trial experiments.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, 4815733971, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, 4815733971, Iran.
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| | - Timo L M Ten Hagen
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus MC, Rotterdam, the Netherlands.
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mojtaba Falahati
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus MC, Rotterdam, the Netherlands.
| |
Collapse
|
12
|
Islam Khan MZ, Law HKW. Suppression of small nucleolar RNA host gene 8 (SNHG8) inhibits the progression of colorectal cancer cells. Noncoding RNA Res 2023; 8:224-232. [PMID: 36860208 PMCID: PMC9969251 DOI: 10.1016/j.ncrna.2023.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/12/2023] [Accepted: 02/13/2023] [Indexed: 02/16/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common gastrointestinal malignancies around the world with high mortality. Accumulating evidences demonstrate that long non-coding RNAs (lncRNAs) play critical roles in CRC tumorigenesis by regulating different pathways of carcinogenesis. SNHG8 (small nucleolar RNA host gene 8), a lncRNA, is highly expressed in several cancers and acts as an oncogene that promotes cancer progression. However, the oncogenic role of SNHG8 in CRC carcinogenesis and the underlying molecular mechanisms remain unknown. In this study, we explored the role of SNHG8 in CRC cell lines by performing a series of functional experiments. Similar to the data reported in the Encyclopedia of RNA Interactome, our RT-qPCR results showed that SNHG8 expression was significantly upregulated in CRC cell lines (DLD-1, HT-29, HCT-116, and SW480) compared to the normal colon cell line (CCD-112CoN). We performed dicer-substrate siRNA transfection to knockdown the expression of SNHG8 in HCT-116 and SW480 cell lines which were expressing high levels of SNHG8. SNHG8 knockdown significantly reduced CRC cell growth and proliferation by inducing autophagy and apoptosis pathways through the AKT/AMPK/mTOR axis. We performed wound healing migration assay and demonstrated that SNHG8 knockdown significantly increased migration index in both cell lines, indicating reduced migration abilities of cells. Further investigation showed that SNHG8 knockdown suppresses epithelial to mesenchymal transition and reduces cellular migratory properties of CRC cells. Taken together, our study suggests that SNHG8 acts as an oncogene in CRC through the mTOR-dependent autophagy, apoptosis, and EMT pathways. Our study provides a better understanding the role of SNHG8 in CRC at molecular level and SNHG8 might be used as novel therapeutic target for CRC management.
Collapse
|
13
|
Zhu W, Tan L, Ma T, Yin Z, Gao J. Long noncoding RNA SNHG8 promotes chemoresistance in gastric cancer via binding with hnRNPA1 and stabilizing TROY expression. Dig Liver Dis 2022; 54:1573-1582. [PMID: 35354542 DOI: 10.1016/j.dld.2022.02.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 02/21/2022] [Accepted: 02/22/2022] [Indexed: 12/30/2022]
Abstract
AIMS To determine SNHG8's function and potential mechanisms in gastric cancer (GC) chemoresistance. METHODS We assessed SNHG8 expression in GC cell lines, GC/CDDP cell lines (cell lines treated with cisplatin), and 42 GC tissues and SNHG8 levels in the lncRNA microarray analysis of AGS/CDDP and AGS cell lines. We also examined GC cell viability in vivo and in vitro and its apoptosis level with Flow cytometry assays. SNHG8 was localized in subcells using fluorescence in situ hybridization (FISH) and cell fraction assays, hnRNPA1's link to SNHG8 was determined utilizing RNA immunoprecipitation (RIP) and FISH assays, gene expression profiles were assessed employing RNA transcriptome sequencing, and hnRNPA1's relationship with TROY was ascertained with the RIP assay. RESULTS SNHG8 increased significantly in GC cell lines and GC tissues. However, a decrease in its expression promoted sensitivity to chemotherapy and inhibited DNA damage repair in vitro and in vivo. SNHG8 appeared to regulate TROY expression via linking with hnRNPA1. Reducing TROY levels considerably stimulated GC cell chemosensitivity, whereas heightening them partially rescued the rate of chemoresistance caused by downregulating SNHG8. CONCLUSION In summary, the "SNHG8/hnRNPA1-TROY" axis is crucial to GC chemoresistance.
Collapse
Affiliation(s)
- Wenzhong Zhu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lulu Tan
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tiantian Ma
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhijie Yin
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jinbo Gao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
14
|
Shetty A, Venkatesh T, Kabbekodu SP, Tsutsumi R, Suresh PS. LncRNA-miRNA-mRNA regulatory axes in endometrial cancer: a comprehensive overview. Arch Gynecol Obstet 2022; 306:1431-1447. [PMID: 35182183 DOI: 10.1007/s00404-022-06423-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 01/24/2022] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Recent research on tumorigenesis and progression has opened up an array of novel molecular mechanisms in the form of interactions between cellular non-coding RNAs (long non-coding RNA[lncRNA]/microRNA [miRNA]) and coding transcripts that regulate health and disease. Endometrial cancer (EC) is a prominent gynecological malignancy with a high incidence rate and poorly known etiology and prognostic factors that hinder the success of disease management. The emerging role of lncRNA-miRNA-mRNA interactions and their dysregulation in the pathophysiology of EC has been elucidated in many recent studies. METHODS A thorough literature review was conducted to explore information about lncRNA-miRNA-mRNA axes in EC. RESULTS Several lncRNAs act as molecular sponges that sequester various tumor suppressor miRNAs to inhibit their function, leading to the dysregulation of their target mRNA transcripts that contribute to the EC regulation. CONCLUSIONS This review summarizes these networks of molecular mechanisms and their contribution to different aspects of endometrial carcinogenesis, leading to a better conceptualization of the molecular pathways that underlie the disease and helping establish novel diagnostic biomarkers and therapeutic intervention points to aid the curative intent of EC.
Collapse
Affiliation(s)
- Abhishek Shetty
- Department of Biosciences, Mangalore University, Mangalagangothri, Mangalore, 574 199, Karnataka, India
| | - Thejaswini Venkatesh
- Department of Biochemistry and Molecular Biology, Central University of Kerala, Kasargod, 671316, Kerala, India
| | - Shama Prasada Kabbekodu
- Department of Cell and Molecular Biology, School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Rie Tsutsumi
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15, Kuramoto-cho, Tokushima City, 770-8503, Japan
| | - Padmanaban S Suresh
- School of Biotechnology, National Institute of Technology, Calicut, 673601, Kerala, India.
| |
Collapse
|
15
|
Lv F, Li X, Wang Y. Lycorine inhibits angiogenesis by docking to PDGFRα. BMC Cancer 2022; 22:873. [PMID: 35948939 PMCID: PMC9364594 DOI: 10.1186/s12885-022-09929-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 07/22/2022] [Indexed: 11/29/2022] Open
Abstract
Lycorine (Lyc) is a natural alkaloid derived from medicinal plants of the Amaryllidaceae family. Lyc has been reported to inhibit the recurrence and metastasis of different kinds of tumors. However, Lyc’s effect on angiogenesis and its specific mechanism are still not clear. This study was designed to test the antiangiogenesis effect of Lyc and to explore the possible mechanisms. We performed cell experiments to confirm Lyc’s inhibitory effect on angiogenesis and employed sunitinib as a positive control. Moreover, the synergistic effect of Lyc and sunitinib was also explored. Next, we conducted bioinformatics analyses to predict the potential targets of Lyc and verified them by western blotting and immunofluorescence. Molecular docking, kinase activity assays, Biacore assays and cellular thermal shift assays (CETSAs) were applied to elucidate the mechanism by which Lyc inhibited target activity. Lyc inhibited angiogenesis in human umbilical vein endothelial cells (HUVECs). Employing bioinformatics, we found that Lyc’s target was PDGFRα and that Lyc attenuated PDGFRα phosphorylation. We also found that Lyc inhibited PDGFRα activation by docking to it to restrain its activity. Additionally, Lyc significantly inhibited PDGF-AA-induced angiogenesis. This study provides new insights into the molecular functions of Lyc and indicates its potential as a therapeutic agent for tumor angiogenesis.
Collapse
Affiliation(s)
- Fei Lv
- Department of Oncology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110000, Liaoning Province, China
| | - XiaoQi Li
- Department of Oncology III, People's Hospital of Liaoning Provinve, Shenyang, , Liaoning, China
| | - Ying Wang
- Department of Oncology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110000, Liaoning Province, China.
| |
Collapse
|
16
|
Sadri F, Hosseini SF, Aghayei A, Fereidouni M, Rezaei Z. The Tumor Suppressor Roles and Mechanisms of MiR-491 in Human Cancers. DNA Cell Biol 2022; 41:810-823. [PMID: 35914029 DOI: 10.1089/dna.2022.0274] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
MicroRNAs (miRNAs) are short non-coding RNAs that bind to the 3' untranslated region (3'' UTR) of target mRNAs to control gene expression post-transcriptionally. Recent indications have highlighted their important roles in a variety of pathophysiological conditions as well as human malignancies. Dysregulated miRNAs act as tumor suppressor genes or oncogenes in a variety of cancers. MiR-491 has been shown to have a major effect on tumorigenesis in multiple malignancies through binding to specific genes and signaling cascades, thereby preventing cancer progression. This review provides an overview of miR-491 expression in regulatory mechanisms and biological procedures of tumor cells, as well as the prospective possible treatment effects of various types of human cancers.
Collapse
Affiliation(s)
- Farzad Sadri
- Student Research Committee, Department of Molecular Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Seyede Fatemeh Hosseini
- Department of Nursing, Tabas School of Nursing, Birjand University of Medical Sciences, Birjand, Iran
| | - Atena Aghayei
- Department of Biology, Faculty of Science, Yazd University, Yazd, Iran
| | - Mohammad Fereidouni
- Department of Medical Immunology, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran.,Cellular and Molecular Research Center, Department of Immunology, Birjand University of Medical Sciences, Birjand, Iran
| | - Zohreh Rezaei
- Cellular and Molecular Research Center, Department of Immunology, Birjand University of Medical Sciences, Birjand, Iran.,Department of Biology, University of Sistan and Baluchestan, Zahedan, Iran
| |
Collapse
|
17
|
Low Expression of miR-491-3p Is Correlated with Lymph Node Metastasis in Gastric Cancer. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:7807956. [PMID: 35815280 PMCID: PMC9262502 DOI: 10.1155/2022/7807956] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/13/2022] [Accepted: 06/17/2022] [Indexed: 02/03/2023]
Abstract
Objective MiR-491-3p, as a tumor suppressor miRNA, was found decreased in many solid tissues. In this study, we aim to investigate miR-491-3p expression in gastric cancer with or without lymph node metastasis (LNM). Methods GSE173215 dataset from Gene Expression Omnibus (GEO) was used to measure miRNA expression from tissue samples of gastric cancer patients. Moreover, gastric tumor tissues (non-LNM: n = 78; LNM: n = 68) were obtained to detect the miR-491-3p expression. Receiver operating characteristic (ROC) curve and Kaplan–Meier (KM) survival analysis, as well as Cox regression analysis, were performed to reveal the role of miR-491-3p in diagnosis and prognosis of gastric cancer. Results According to GSE173215 datasets (t = −11.25, adjust P value = 1.30E-06) and our clinical results (0.390 ± 0.193 vs. 0.562 ± 0.166, P < 0.005), the gastric cancer patients with LNM showed lower miR-491-3p expression than those without LNM, demonstrating a high diagnostic efficiency (sensitivity: 74.36%; specificity: 69.12%). In addition, both LNM and low miR-491-3p expression were correlated with the poor prognosis of gastric cancer. Furthermore, the LNM patient with low expression of miR-491-3p had the worse prognosis, but the non-LNM patient with high expression of miR-491-3p had the best prognosis. MiR-491-3p expression (HR = 0.003, 95%CI: 3.35E-04∼0.028) and LNM (HR = 2.326, 95%CI: 1.046∼5.173) were independent risk factors for gastric cancer. Conclusion Downregulated miR-491-3p expression was found in gastric cancer, being a high diagnostic efficiency and an independent risk factor for gastric cancer, especially in those having LNM.
Collapse
|
18
|
Huang Y, Jiang L, Liu Y, Liu L, Wang J, Shi L. Long non-coding RNAs in virus-related cancers. Rev Med Virol 2022; 32:e2364. [PMID: 35607835 DOI: 10.1002/rmv.2364] [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: 08/10/2021] [Revised: 03/30/2022] [Accepted: 05/06/2022] [Indexed: 12/12/2022]
Abstract
Some viral infections lead to tumourigenesis explained by a variety of underlying molecular mechanisms. Long non-coding RNAs (lncRNAs) have the potential to be added to this list due to their diverse mechanisms in biological functions and disease processes via gene alternation, transcriptional regulation, protein modification, microRNA sponging and interaction with RNA/DNA/proteins. In this review, we summarise the dysregulation and mechanism of lncRNAs in virus-related cancers focussing on Hepatitis B virus, Epstein-Barr virus, Human Papillomavirus. We will also discuss the potential implications of lncRNAs in COVID-19.
Collapse
Affiliation(s)
- Yushan Huang
- School of Public Health, Lanzhou University, Lanzhou, China
| | - Longchang Jiang
- Department of Vascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yang Liu
- Gansu Provincial People's Hospital, Lanzhou, China
| | - Lixian Liu
- Shijiangzhuang City Zoo, Shijiazhuang, China
| | - Junling Wang
- School of Public Health, Lanzhou University, Lanzhou, China
| | - Lei Shi
- School of Public Health, Lanzhou University, Lanzhou, China
| |
Collapse
|
19
|
Shan B, Qu S, Lv S, Fan D, Wang S. YY1-induced long non-coding RNA small nucleolar RNA host gene 8 promotes the tumorigenesis of melanoma via the microRNA-656-3p/SERPINE1 mRNA binding protein 1 axis. Bioengineered 2022; 13:4832-4843. [PMID: 35156513 PMCID: PMC8973976 DOI: 10.1080/21655979.2022.2034586] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Long non-coding (lnc) RNA serves a vital role in the cellular processes of carcinoma. This study aimed to explore the accurate mechanism underlying lncRNA small nucleolar RNA host gene 8 (SNHG8) in melanoma. In this study, lncRNA SNHG8 expression were upregulated in melanoma tissues and cells, and lncRNA SNHG8 knockdown reduced melanoma cell viability, migration and invasion. Moreover, lncRNA SNHG8 expression could be induced by transcription factor YY1. In addition, we found that miR-656 could directly bind to lncRNA SNHG8 and SERPINE1 mRNA binding protein 1 (SERBP1). Rescue assays indicated that miR-656 overexpression inhibited the aforementioned cellular activities in melanoma cells, which were reversed by SERBP1 overexpression. In conclusion, this work elucidated that YY1-induced upregulation of lncRNA SNHG8 boosted the development of melanoma via the miR-656-3p/SERBP1 axis, providing a novel therapeutic strategy for melanoma treatment.
Collapse
Affiliation(s)
- Baihui Shan
- Department of Dermatology, The Second Hospital of Jilin University, China
| | - Shengming Qu
- Department of Dermatology, The Second Hospital of Jilin University, China
| | - Sha Lv
- Department of Dermatology, The Second Hospital of Jilin University, China
| | - Dandan Fan
- Department of Dermatology, Jilin Province People’s Hospital, China
| | - Shu Wang
- Department of Radio Therapy, The Second Hospital of Jilin University, China
| |
Collapse
|
20
|
Nie K, Liu L, Peng L, Zhang M, Zhang C, Xiao B, Xia Z, Huang W. Effects of Meranzin Hydrate On the LncRNA-miRNA-mRNA Regulatory Network in the Hippocampus of a Rat Model of Depression. J Mol Neurosci 2022; 72:910-922. [PMID: 35099722 DOI: 10.1007/s12031-022-01971-6] [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: 06/02/2021] [Accepted: 01/10/2022] [Indexed: 10/19/2022]
Abstract
Meranzin hydrate (MH) is a frequently used antidepressant drug in China; however it underlying mechanism remains unknown. In this study, we aimed to explore whether MH could ameliorate depression-like behavior in rats by regulating the competitive endogenous RNA (ceRNA) network. We developed a depression-like rat model using an unpredictable chronic mild stress (UCMS) protocol, and the differentially expressed lncRNAs, miRNAs, and mRNAs were identified between the model group and MH group. Then, a ceRNA network responding to MH treatment was constructed by their corresponding relationships in the databases. Finally, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were conducted to explore molecular mechanisms associated with MH treatment. The study indicated that rats in the model group showed loss of weight and deteriorated behavior in behavior tests compared with rats in the normal group. A total of 826 lncRNAs, 121 miRNAs, and 954 mRNAs were differentially expressed in the hippocampus of UCMS rats after MH treatment. In addition, 13 miRNAs were selected, and 12 of them were validated in the hippocampus by qRT-PCR. Then, we predicted upstream lncRNAs and downstream mRNAs of the validated miRNAs and interacted with the results of microarrays. Eventually, a lncRNA-miRNA-mRNA regulatory network, responding to MH treatment, was constructed based on the 314 lncRNAs, 11 miRNAs, and 221 mRNAs. KEGG pathways suggested that these genes may be highly related to Wnt signaling, axon guidance, and MAPK signaling pathways. All these results suggest that MH may be a potential representative compound for the treatment of depression, and its mechanism of action is related to the ceRNA modification.
Collapse
Affiliation(s)
- Kechao Nie
- Department of Integrated Traditional Chinese & Western Internal Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410001, Hunan, China
| | - Lin Liu
- The First Hospital of Hunan University of Chinese Medicine, Changsha, 410021, Hunan, China
| | - Luqi Peng
- Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Mei Zhang
- Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Chunhu Zhang
- Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Bo Xiao
- Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Zian Xia
- Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Wei Huang
- Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
21
|
Luan Q, Yang R, Lin L, Li X. SNHG8 promotes cell proliferation, migration, and invasion of nasopharyngeal carcinoma cells as an oncogene through miR-588/HMGA2 axis. Can J Physiol Pharmacol 2022; 100:158-166. [PMID: 35080988 DOI: 10.1139/cjpp-2021-0149] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Nasopharyngeal carcinoma (NC) poses a threat to the life of patients. Long non-coding RNA (LncRNA) is a novel kind of non-coding RNA, which plays a pivotal role through sponge microRNA (miRNA). Abnormal expression of small nucleolar RNA host gene 8 (SNHG8) is involved in various tumors; however, the role of SNHG8 in NC remains unknown. Quantitative real-time PCR (qRT-PCR) and Western blotting was employed to detect the expression levels of SNHG8, miR-588, and high mobility group A2 (HMGA2). Cell proliferation, migration, and invasion were analyzed by CCK-8 and transwell assays. miR-588 binding sites in SNHG8 were predicted by LncBase analysis. Luciferase reporter and RNA pull-down assay were used to confirm the interaction of SNHG8 and miR-588. SNHG8 was highly expressed in NC cells. The prognosis of the patients with NC in the high expression levels of SNHG8 was poorer than that in the low expression levels. The expression of SNHG8 was closely related to tumor size, TNM stage, and distal metastasis. Knockdown of SNHG8 inhibited cell proliferation, migration, and invasion of NC. SNHG8 targeted miR-588. Inhibition of miR-588 could partially reverse the knockdown of SNHG8 in NC cells, and miR-588 targeted HMGA2. In conclusion, SNHG8 promotes proliferation, migration, and invasion of NC cells through miR-588/HMGA2 in NC as an oncogene.
Collapse
Affiliation(s)
- Qiang Luan
- Department of Otolaryngology, Traditional Chinese Medicine Hospital of Muping District of Yantai City, No. 505 Government Street, Yantai, 264100, Shangdong province, China
| | - Ruifang Yang
- Department of Medical Laboratory, Yantai Affiliated Hospital of Binzhou Medical College, No. 717 Jinbu Street, Yantai, 264100, Shangdong province, China
| | - Lejun Lin
- Department of Nuclear Medicine, Yantai Yuhuangding Hospital, No. 20 Yuhuangding East Road, Yantai, 264100, Shangdong province, China
| | - Xuecheng Li
- Department of General Surgery, Traditional Chinese Medicine Hospital of Muping District of Yantai City, No. 505 Government Street, Yantai, 264100, Shangdong province, China
| |
Collapse
|
22
|
Wu Y, Liang Y, Li M, Zhang H. Knockdown of long non-coding RNA SNHG8 suppresses the progression of esophageal cancer by regulating miR-1270/BACH1 axis. Bioengineered 2022; 13:3384-3394. [PMID: 35067159 PMCID: PMC8974072 DOI: 10.1080/21655979.2021.2021064] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The emerging evidence showed that lncRNAs (long non-coding RNAs) could regulate the progression and affect the malignant behaviors of cancers. LncRNA SNHG8 (small nucleolar RNA host gene 8) has been reported to participate in most cancers development. Here in this study, the role of lncRNA SNHG8 in esophageal cancer was uncovered by a series of functional experiments. The expression pattern of SNHG8 in tumor tissues or cells was first detected by qRT-PCR. Using a lentivirus knockdown shRNA is to repress the expression of SNHG8. Subsequently, the in vitro and in vivo experiments were utilized to evaluate whether the malignant behaviors of esophageal cancer were influenced by knockdown SNHG8. The results indicated that lncRNA SNHG8 should be a cancer-promoting factor with a relatively high expression level in esophageal cancer. Moreover, knockdown SNHG8 inhibited the cell viability and induced cell apoptosis in KYSE30 and TE-1 cells. In addition, based on the results of the binding site analysis and the luciferase reporter system, SNHG8 functions by the miR-1270/BACH1 axis. The follow-up experiments verified that lncRNA SNHG8 could down-regulate the expression of miR-1270 to increase the BACH1 expression. Finally, we confirmed that knockdown SNHG8 retarded the progression of esophageal cancer with a xenograft model. To sum up, our findings suggested that lncRNA SNHG8 is a cancer-promoting factor in esophageal cancer. Knockdown SNHG8 could suppress the progression of esophageal cancer, which implies SNHG8 could be used as a therapeutic target in esophageal cancer.
Collapse
Affiliation(s)
- Yonghong Wu
- Department of Medical Insurance and Price, Xiangyang No. 1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
| | - Yan Liang
- Hematology Department, Xiangyang No. 1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
| | - Min Li
- Gastroenterology Department, Xiangyang No. 1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
| | - Haidong Zhang
- Oncology Department, Xiangyang No. 1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
| |
Collapse
|
23
|
Gu H, Zhong Y, Liu J, Shen Q, Wei R, Zhu H, Zhang X, Xia X, Yao M, Ni M. The Role of miR-4256/HOXC8 Signaling Axis in the Gastric Cancer Progression: Evidence From lncRNA-miRNA-mRNA Network Analysis. Front Oncol 2022; 11:793678. [PMID: 35111675 PMCID: PMC8801578 DOI: 10.3389/fonc.2021.793678] [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: 10/12/2021] [Accepted: 12/21/2021] [Indexed: 12/14/2022] Open
Abstract
Gastric cancer is a deadly human malignancy and the molecular mechanisms underlying gastric cancer pathophysiology are very complicated. Thus, further investigations are warranted to decipher the underlying molecular mechanisms. With the development of high-throughput screening and bioinformatics, gene expression profiles with large scale have been performed in gastric cancer. In the present study, we mined The Cancer Genome Atlas (TCGA) database and analyzed the gene expression profiles between gastric cancer tissues and normal gastric tissues. A series of differentially expressed lncRNAs, miRNAs and mRNAs between gastric cancer tissues and normal gastric tissues were identified. Based on the differentially expressed genes, we constructed miRNA-mRNA network, lncRNA-mRNA network and transcriptional factors-mRNA-miRNA-lncRNA network. Furthermore, the Kaplan survival analysis showed that high expression levels of EVX1, GBX2, GCM1, HOXC8, HOXC9, HOXC10, HOXC11, HOXC12 and HOXC13 were all significantly correlated with shorter overall survival of the patients with gastric cancer. On the other hand, low expression level of HOXA13 was associated with shorter overall survival of patients with gastric cancer. Among these hub genes, we performed the in vitro functional studies of HOXC8 in the gastric cancer cells. Knockdown of HOXC8 and overexpression of miR-4256 both significantly repressed the gastric cancer cell proliferation and migration, and miR-4256 repressed the expression of HOXC8 via targeting its 3' untranslated region in gastric cancer cells. Collectively, our results revealed that a complex interaction networks of differentially expressed genes in gastric cancer, and further functional studies indicated that miR-4256/HOXC8 may be an important axis in regulating gastric cancer progression.
Collapse
Affiliation(s)
- Haijuan Gu
- Department of Pharmacy, Tumor Hospital Affiliated to Nantong University, Nantong, China
| | - Yuejiao Zhong
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, China
| | - Jibin Liu
- Institute of Oncology, Tumor Hospital Affiliated to Nantong University, Nantong, China
| | - Qian Shen
- Department of Oncology, Tumor Hospital Affiliated to Nantong University, Nantong, China
| | - Rong Wei
- Department of Pharmacy, Tumor Hospital Affiliated to Nantong University, Nantong, China
| | - Haixia Zhu
- Clinical Laboratory, Tumor Hospital Affiliated to Nantong University, Nantong, China
| | - Xunlei Zhang
- Department of Oncology, Tumor Hospital Affiliated to Nantong University, Nantong, China
| | - Xianxian Xia
- Department of Pharmacy, Tumor Hospital Affiliated to Nantong University, Nantong, China
| | - Min Yao
- Department of Pharmacy, Tumor Hospital Affiliated to Nantong University, Nantong, China
| | - Meixin Ni
- Department of Pharmacy, Tumor Hospital Affiliated to Nantong University, Nantong, China
| |
Collapse
|
24
|
Shafabakhsh R, Arianfar F, Vosough M, Mirzaei HR, Mahjoubin-Tehran M, Khanbabaei H, Kowsari H, Shojaie L, Azar MEF, Hamblin MR, Mirzaei H. Autophagy and gastrointestinal cancers: the behind the scenes role of long non-coding RNAs in initiation, progression, and treatment resistance. Cancer Gene Ther 2021; 28:1229-1255. [PMID: 33432087 DOI: 10.1038/s41417-020-00272-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 10/06/2020] [Accepted: 11/23/2020] [Indexed: 02/07/2023]
Abstract
Gastrointestinal (GI) cancers comprise a heterogeneous group of complex disorders that affect different organs, including esophagus, stomach, gallbladder, liver, biliary tract, pancreas, small intestine, colon, rectum, and anus. Recently, an explosion in nucleic acid-based technologies has led to the discovery of long non-coding RNAs (lncRNAs) that have been found to possess unique regulatory functions. This class of RNAs is >200 nucleotides in length, and is characterized by their lack of protein coding. LncRNAs exert regulatory effects in GI cancer development by affecting different functions such as the proliferation and metastasis of cancer cells, apoptosis, glycolysis and angiogenesis. Over the past few decades, considerable evidence has revealed the important role of autophagy in both GI cancer progression and suppression. In addition, recent studies have confirmed a significant correlation between lncRNAs and the regulation of autophagy. In this review, we summarize how lncRNAs play a behind the scenes role in the pathogenesis of GI cancers through regulation of autophagy.
Collapse
Affiliation(s)
- Rana Shafabakhsh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Farzaneh Arianfar
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Massoud Vosough
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 1665659911, Iran
| | - Hamid Reza Mirzaei
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Mahjoubin-Tehran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hashem Khanbabaei
- Medical Physics Department, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hamed Kowsari
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Layla Shojaie
- Research Center for Liver Diseases, Keck School of Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028, South Africa.
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
25
|
Dang Y, Zhou Y, Ou X, Wang Q, Wei D, Xie F. lncRNA AC007207.2 Promotes Malignant Properties of Osteosarcoma via the miR-1306-5p/SIRT7 Axis. Cancer Manag Res 2021; 13:7277-7288. [PMID: 34584454 PMCID: PMC8464591 DOI: 10.2147/cmar.s318975] [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: 05/07/2021] [Accepted: 09/08/2021] [Indexed: 12/12/2022] Open
Abstract
Background Long non-coding RNAs (lncRNAs) have been implicated in initiation and development of numerous cancers. In the present study, we explored the role of lncRNAs AC007207.2 in osteosarcoma (OS). Methods Gene expression data of OS tissues was downloaded from the TARGET database. All the experiments were repeated at least three times. Data were analyzed using Perl, R, SPSS v12.0 and GraphPad Prism 8 software. Results We found lncRNA AC007207.2 was over-expressed in OS tissues and cell lines, and this phenomenon was associated with the worse prognosis of OS. Moreover, we found that AC007207.2 promotes proliferation and metastasis of OS cells via the miR-1306-5p/SIRT7 axis. Meanwhile, we found miR-1306-5p remarkably inhibits the malignant behavior of OS cells. Conclusion lncRNA AC007207.2 promotes progression of OS by upregulating SIRT7 expression through miR-1306-5p sponging. Thus, lncRNA AC007207.2/miR-1306-5p/SIRT7 axis is a promising therapeutic target for OS treatment.
Collapse
Affiliation(s)
- Youting Dang
- Department of Pediatric Orthopedics, Honghui Hospital, Xi'an Jiao Tong University, Xi'an, Shaanxi, 710054, People's Republic of China
| | - Yunping Zhou
- Department of Hand Surgery, Honghui Hospital, Xi'an Jiao Tong University, Xi'an, Shaanxi, 710054, People's Republic of China
| | - Xuehai Ou
- Department of Hand Surgery, Honghui Hospital, Xi'an Jiao Tong University, Xi'an, Shaanxi, 710054, People's Republic of China
| | - Qiang Wang
- Department of Hand Surgery, Honghui Hospital, Xi'an Jiao Tong University, Xi'an, Shaanxi, 710054, People's Republic of China
| | - Dengke Wei
- Department of Hand Surgery, Honghui Hospital, Xi'an Jiao Tong University, Xi'an, Shaanxi, 710054, People's Republic of China
| | - Fei Xie
- Department of Hand Surgery, Honghui Hospital, Xi'an Jiao Tong University, Xi'an, Shaanxi, 710054, People's Republic of China
| |
Collapse
|
26
|
Wang S, Li J, Chen A, Song H. Differentiated expression of long non-coding RNA-small nucleolar RNA host gene 8 in atherosclerosis and its molecular mechanism. Bioengineered 2021; 12:7167-7176. [PMID: 34558393 PMCID: PMC8806704 DOI: 10.1080/21655979.2021.1979441] [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] [Indexed: 11/01/2022] Open
Abstract
Atherosclerosis (AS) is one of the most common cardiovascular diseases, and the incidence is increasing year by year. Many studies have shown that long non-coding RNA plays a vital role in the pathogenesis of AS. This study aimed to explore the role and mechanism of lncRNA-small nucleolar RNA host gene 8 (SNHG8) in AS. The expressions of serum lncSNHG8 and miR-224-3p were determined by quantitative real-time polymerase chain reaction (qRT-PCR). The diagnostic meaning of lncSNHG8 in AS was estimated by Receiver operating characteristic (ROC) curve. The correlation between lncSNHG8 and various clinical indicators, as well as miR-244-3p was evaluated by Pearson correlation coefficient analysis. Cell proliferation and migration were estimated by cell counting kit-8 (CCK-8) and Transwell assay. The interaction between lncSNHG8 and miR-224-3p was proved by luciferase reporter gene assay. The expression level of lncSNHG8 was increased in AS patients, while miR-224-3p expression was decreased. The ROC curve indicated that lncSNHG8 with high serum expression had the ability to distinguish AS. Pearson correlation coefficient exhibited that the level of miR-224-3p was negatively correlated with the level of lncSNHG8. The results of cell experiments indicated that inhibition of the expression of lncSNHG8 significantly inhibited the proliferation and migration of vascular smooth muscle cells (VSMCs). Luciferase reporter gene experiments confirmed that there was a target relationship between lncSNHG8 and miR-224-3p. In conclusion, lncSNHG8 had high diagnostic value for AS. It promoted the proliferation and migration of VSMCs by adsorption and inhibition of miR-224-3p.
Collapse
Affiliation(s)
- Shuang Wang
- Department of Emergency Neurology, Yidu Central Hospital of Weifang, Weifang, Shandong China
| | - Jianchao Li
- Department of Emergency Neurology, Yidu Central Hospital of Weifang, Weifang, Shandong China
| | - Aimei Chen
- Department of Traditional Chinese Medicine, Yidu Central Hospital of Weifang, Weifang, Shandong China
| | - He Song
- Department of Emergency, Yidu Central Hospital of Weifang, Weifang, Shandong China
| |
Collapse
|
27
|
Zhao L, Jia Y, Liu Y, Han B, Wang J, Jiang X. Integrated Bioinformatics Analysis of DNA Methylation Biomarkers in Thyroid Cancer Based on TCGA Database. Biochem Genet 2021; 60:629-639. [PMID: 34387764 DOI: 10.1007/s10528-021-10117-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 08/02/2021] [Indexed: 12/24/2022]
Abstract
Previous studies have reported a cluster of aberrant promoter methylation changes associated with silencing of tumor suppressor genes in thyroid cancer (TC), but these results of individual genes are far from enough. In this work, we aimed to investigate the onset and pattern of methylation changes during the progression of TC by informatics analysis. We downloaded the DNA methylation and RNA sequencing datasets from The Cancer Genome Atlas focusing on TC. Abnormally methylated differentially expressed genes (DEGs) were sorted and pathways were analyzed. The KEGG and GO were then used to perform enrichment and functional analysis of identified pathways and genes. Gene-drug interaction network and human protein atlas were applied to obtain feature DNA methylation biomarkers. In total, we identified 2170 methylation-driven DEGs, including 1054 hypermethylatedlow-expression DEGs and 1116 hypomethylated-high-expression DEGs at the screening step. Further analysis screened total of eight feature DNA methylation biomarkers (RXRG, MET, PDGFRA, FCGR3A, VEGFA, CSF1R, FCGR1A and C1QA). Pathway analysis showed that aberrantly methylated DEGs mainly associated with transcriptional misregulation in cancer, MAPK signaling, and intrinsic apoptotic signaling in TC. Taken together, we have identified novel aberrantly methylated genes and pathways linked to TC, which might serve as novel biomarkers for precision diagnosis and disease treatment.
Collapse
Affiliation(s)
- Lifeng Zhao
- Department of Endocrinology, Tianjin First Center Hospital, No. 24, Fu-Kang Road, Nankai District, Tianjin, 300192, China.
| | - Yuanyuan Jia
- Department of Endocrinology, Tianjin First Center Hospital, No. 24, Fu-Kang Road, Nankai District, Tianjin, 300192, China
| | - Ying Liu
- Department of Endocrinology, Tianjin First Center Hospital, No. 24, Fu-Kang Road, Nankai District, Tianjin, 300192, China
| | - Baoling Han
- Department of Endocrinology, Tianjin First Center Hospital, No. 24, Fu-Kang Road, Nankai District, Tianjin, 300192, China
| | - Jian Wang
- Department of Endocrinology, Tianjin First Center Hospital, No. 24, Fu-Kang Road, Nankai District, Tianjin, 300192, China
| | - Xia Jiang
- Department of Endocrinology, Tianjin First Center Hospital, No. 24, Fu-Kang Road, Nankai District, Tianjin, 300192, China
| |
Collapse
|
28
|
Qian J, Lei X, Sun Y, Zheng L, Li J, Zhang S, Zhang L, Li W, Shi J, Jia W, Tang T. Long non-coding RNA SNHG8 enhances triple-negative breast cancer cell proliferation and migration by regulating the miR-335-5p/PYGO2 axis. Biol Direct 2021; 16:13. [PMID: 34362407 PMCID: PMC8349079 DOI: 10.1186/s13062-021-00295-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 06/28/2021] [Indexed: 12/30/2022] Open
Abstract
Background Growing evidence has demonstrated that long non-coding RNAs (lncRNAs) can function as modulators in the development of triple-negative breast cancer (TNBC). However, the function of lncRNA small nucleolar RNA host gene 8 (SNHG8) in TNBC remains unclear. Therefore, our study aimed at investigating the role of SNHG8 in the proliferation and migration of TNBC cells. Methods SNHG8 expression was evaluated using RT-qPCR assay. Cell proliferation and migration were assessed by EdU, colony formation and Transwell assays. The levels of proteins related to EMT process were examined by western blot assay. The interaction among SNHG8, miR-335-5p and pygopus family PHD finger 2 (PYGO2) was detected by RIP assay, RNA pull down assay and luciferase reporter assay. Results SNHG8 expression was significantly up-regulated in TNBC cells. SNHG8 silencing obviously inhibited TNBC cell proliferation, migration and EMT process. Moreover, SNHG8 acted as a sponge to sequester miR-335-5p in TNBC cells. Besides, PYGO2 was proven as a target gene of miR-335-5p, and SNHG8 promoted TNBC cell proliferation, migration and EMT process through regulating miR-335-5p and PYGO2. Conclusions Totally, our study indicated that SNHG8 promoted TNBC cell proliferation and migration by regulating the miR-335-5p/PYGO2 axis.
Collapse
Affiliation(s)
- Jintao Qian
- Department of General Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
| | - Xinhan Lei
- Department of General Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
| | - Yue Sun
- Department of General Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
| | - Lu Zheng
- Department of General Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
| | - Jia Li
- Department of General Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
| | - Shuai Zhang
- Department of General Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
| | - Lei Zhang
- Department of General Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
| | - Wanwan Li
- Department of General Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
| | - Jianing Shi
- Department of General Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
| | - Wenjun Jia
- Department of General Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China.
| | - Tong Tang
- Department of General Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China.
| |
Collapse
|
29
|
Chen C, Chen Q, Wu J, Zou H. H3K27ac-induced FOXC2-AS1 accelerates tongue squamous cell carcinoma by upregulating E2F3. J Oral Pathol Med 2021; 50:1018-1030. [PMID: 34358374 DOI: 10.1111/jop.13232] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/09/2021] [Accepted: 07/13/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND The important roles of lncRNAs have been reported in cancers, including tongue squamous cell carcinoma (TSCC). Here, we investigated the functional role and molecular mechanisms of lncRNA FOXC2-AS1 in TSCC. METHODS The expression level of FOXC2-AS1 in TSCC was determined by RT-qPCR. Its biological role was evaluated through colony formation assay, flow cytometry, wound healing, transwell, and Western blot analyses. The interactions among gene were tested by mechanistic investigations. RESULTS FOXC2-AS1 expression was high in TSCC tissues and cells. Functional assays in vitro showed that silencing FOXC2-AS1 restrained cell proliferation, cell cycle, migration, invasion, and EMT. In the mechanism, it was verified that H3K27 acetylation (H3K27ac) triggered an increase in FOXC2-AS1 expression. Furthermore, FOXC2-AS1 was identified as a cytoplasmic lncRNA and served as a ceRNA to upregulate E2F3 expression via sponging miR-6868-5p. CONCLUSION H3K27ac-induced FOXC2-AS1 exhibits carcinogenic property in TSCC by the miR-6868-5p/E2F3 axis.
Collapse
Affiliation(s)
- Cuiting Chen
- Department of Stomatology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Qiongyu Chen
- Department of Stomatology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jian Wu
- Department of Stomatology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Han Zou
- Department of Stomatology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
30
|
Hu Y, Zhang Y, Ding M, Xu R. Long noncoding RNA TMPO-AS1/miR-126-5p/BRCC3 axis accelerates gastric cancer progression and angiogenesis via activating PI3K/Akt/mTOR pathway. J Gastroenterol Hepatol 2021; 36:1877-1888. [PMID: 33295056 DOI: 10.1111/jgh.15362] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 10/30/2020] [Accepted: 11/23/2020] [Indexed: 01/01/2023]
Abstract
BACKGROUND AND AIM Gastric cancer (GC) is an aggressive tumor featured by uncontrolled cell proliferation and metastasis. In recent years, long noncoding RNAs (lncRNAs) act as crucial regulators and biological markers in multiple cancers. LncRNA TMPO-AS1 has been revealed to be an oncogene in some cancers. Nevertheless, there is little known about the biological role of TMPO-AS1 in GC. METHODS Reverse transcription-quantitative polymerase chain reaction analysis was used to examine the expression level of TMPO-AS1 in GC tissues and cells. Cell Counting Kit-8, colony formation, wound healing assays, and western blot analysis were performed to determine the role of TMPO-AS1 in GC cells. RNA pull-down, luciferase reporter, and RNA immunoprecipitation assays were used to test the interaction among TMPO-AS1, miR-126-5p, and BRCC3. RESULTS TMPO-AS1 was highly expressed in GC tissues and cells. Upregulated TMPO-AS1 was closely associated with adverse prognosis of GC patients. Functional assays showed that TMPO-AS1 promoted GC cell proliferation, migration, and angiogenesis. Furthermore, it was found that TMPO-AS1 acted as a competing endogenous RNA for miR-126-5p to upregulate BRCC3 expression. Rescue assays revealed that TMPO-AS1 facilitated cellular progression of GC by sponging miR-126-5p and upregulating BRCC3. In addition, we found that the effects of the TMPO-AS1/miR-126-5p/BRCC3 axis on GC cell progression were related to the PI3K/Akt/mTOR pathway. CONCLUSIONS Our study demonstrated that the TMPO-AS1/miR-126-5p/BRCC3 axis was involved in GC progression via the regulation of PI3K/Akt/mTOR pathway, which might provide a potential therapeutic strategy for GC.
Collapse
Affiliation(s)
- Yu Hu
- Department of Pathology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Ying Zhang
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Meng Ding
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Ruisi Xu
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
31
|
Liu Y, Zhou P, Wang F, Zhang X, Yang D, Hong L, Ruan D. Inhibition of lncRNA SNHG8 plays a protective role in hypoxia-ischemia-reoxygenation-induced myocardial injury by regulating miR-335 and RASA1 expression. Mol Med Rep 2021; 24:597. [PMID: 34165173 PMCID: PMC8240175 DOI: 10.3892/mmr.2021.12236] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 05/12/2021] [Indexed: 12/24/2022] Open
Abstract
Long non-coding (lnc)RNAs serve a role in a number of diseases, including different types of cancer and acute myocardial infarction. The aim of the present study was to investigate the protective role of lncRNA small nucleolar RNA host gene 8 (SNHG8) in hypoxia-ischemia-reoxygenation (HI/R)-induced myocardial injury and its potential mechanism of action. Cell viability, proliferation, creatine kinase myocardial band, cell apoptosis and protein expression levels were determined by Cell Counting Kit-8 assay, EdU assay, ELISA, flow cytometry and western blotting, respectively. The association between SNHG8 and microRNA (miR)-335 was confirmed using a dual-luciferase reporter gene assay. The effects of the miR-335 inhibitor transfections had on increasing apoptosis and decreasing H9C2 cell viability were reversed in cells co-transfected with SNHG8 small interfering (si)RNA. Furthermore, it was found that miR-335 could regulate RAS p21 protein activator 1 (RASA1) expression and that transfection with SNHG8 siRNA downregulated RASA1 expression. Silencing of RASA1 protected against HI/R-induced H9C2 cell injury. However, SNHG8 siRNA did not further reduce apoptosis, demonstrating that SNHG8 may act through RASA1, and RASA1 may mediate the protection of SNHG8 siRNA in HI/R myocardial injury. Thus, inhibition of lncRNA SNHG8 alleviated HI/R-induced myocardial damage by regulating miR-335 and RASA1.
Collapse
Affiliation(s)
- Yanfeng Liu
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Ping Zhou
- Department of Cardiology, Yifeng County People's Hospital, Yichun, Jiangxi 336300, P.R. China
| | - Fengxiao Wang
- Department of Cardiology, Jiangxi Huimin Hospital, Nanchang, Jiangxi 330046, P.R. China
| | - Xuehong Zhang
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Dongmei Yang
- Department of Cardiovascular Surgery, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Lang Hong
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Dongyun Ruan
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
32
|
Mesquita FP, Lucena da Silva E, Souza PFN, Lima LB, Amaral JL, Zuercher W, Albuquerque LM, Rabenhorst SHB, Moreira-Nunes CA, Amaral de Moraes ME, Montenegro RC. Kinase inhibitor screening reveals aurora-a kinase is a potential therapeutic and prognostic biomarker of gastric cancer. J Cell Biochem 2021; 122:1376-1388. [PMID: 34160883 DOI: 10.1002/jcb.30015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 05/09/2021] [Accepted: 05/14/2021] [Indexed: 12/17/2022]
Abstract
Gastric cancer is one of the most common and deadly types of cancer in the world, and poor prognosis with treatment failure is widely reported in the literature. In this context, kinases have been considered a relevant choice for targeted therapy in gastric cancer. Here, we explore the antiproliferative and antimigratory effects of the AURKA inhibitor and the prognostic and therapeutic value as a biomarker of gastric cancer. A total of 145 kinase inhibitors were screened to evaluate the cytotoxic or cytostatic effects in the gastric cancer cell line. Using the Alamar Blue assay, flow cytometry, quantitative polymerase chain reaction, and observation of caspase 3/7 activity and cell migration, we investigated the antiproliferative, proapoptotic, and antimigratory effects of the AURKA inhibitor. Moreover, AURKA overexpression was evaluated in the gastric cell lines and the gastric tumor tissue. Out of the 145 inhibitors, two presented the highest antiproliferative effect. Both molecules can induce apoptosis by the caspases 3/7 pathway in addition to inhibiting cancer cell migration, mainly the AURKA inhibitor. Moreover, molecular docking analysis revealed that GW779439X interacts in the active site of the AURKA enzyme with similar energy as a well-described inhibitor. Our study identified AURKA overexpression in the gastric cancer cell line and gastric tumor tissue, revealing that its overexpression in patients with cancer is correlated with low survival. Therefore, it is feasible to suggest AURKA as a potential marker of gastric cancer, besides providing robust information for diagnosis and estimated survival of patients. AURKA can be considered a new molecular target used in the prognosis and therapy of gastric cancer.
Collapse
Affiliation(s)
- Felipe P Mesquita
- Department of Physiology and Pharmacology, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, Brazil
| | - Emerson Lucena da Silva
- Department of Physiology and Pharmacology, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, Brazil
| | - Pedro F N Souza
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, Brazil
| | - Luina B Lima
- Department of Physiology and Pharmacology, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, Brazil
| | - Jackson L Amaral
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, Brazil
| | - William Zuercher
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Louise M Albuquerque
- Department of Pathology and Forensic Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Silvia H B Rabenhorst
- Department of Pathology and Forensic Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Caroline A Moreira-Nunes
- Department of Physiology and Pharmacology, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, Brazil
| | - Maria E Amaral de Moraes
- Department of Physiology and Pharmacology, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, Brazil
| | - Raquel C Montenegro
- Department of Physiology and Pharmacology, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, Brazil
| |
Collapse
|
33
|
He C, Fu Y, Chen Y, Li X. Long non-coding RNA SNHG8 promotes autophagy as a ceRNA to upregulate ATG7 by sponging microRNA-588 in colorectal cancer. Oncol Lett 2021; 22:577. [PMID: 34122628 DOI: 10.3892/ol.2021.12838] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 02/04/2021] [Indexed: 12/21/2022] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer worldwide. Long non-coding RNA (lncRNA) small nucleolar RNA host gene 8 (SNHG8) acts as an oncogene in different types of cancer, including prostate, breast and ovarian cancer. SNHG8 promotes the tumorigenesis of CRC; however, its underlying molecular mechanism remains unclear. The present study aimed to explore the mechanism of SNHG8 on CRC development via various assays, including western blot, pull-down, PCR and immunofluorescence assays. The results of the present study demonstrated that SNHG8 expression was substantially upregulated in primary tumor tissues from The Cancer Genome Atlas dataset. Western blot and immunofluorescence analyses demonstrated that SNHG8 facilitated cell proliferation and autophagy in CRC cells. Notably, the function of SNHG8 in enhancing autophagy was dependent on autophagy-related gene 7 (ATG7). In addition, western blot analysis indicated that the effect of SNHG8 on autophagy in CRC cells was dependent on the miR-588/ATG7 axis. Taken together, the results of the present study suggest that SNHG8 promotes autophagy in CRC cells.
Collapse
Affiliation(s)
- Chi He
- Department of General Surgery, Shenyang Anorectal Hospital, Shenyang, Liaoning 110054, P.R. China
| | - Yi Fu
- Department of General Surgery, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116000, P.R. China
| | - Yan Chen
- Department of General Surgery, Daqing Oilfield General Hospital, Daqing, Heilongjiang 163000, P.R. China
| | - Xiquan Li
- Department of General Surgery, Shenyang Anorectal Hospital, Shenyang, Liaoning 110054, P.R. China
| |
Collapse
|
34
|
Li L, Lai Q, Zhang M, Jia J. Long non-coding RNA DLGAP1-AS1 promotes the progression of gastric cancer via miR-515-5p/MARK4 axis. ACTA ACUST UNITED AC 2021; 54:e10062. [PMID: 34037089 PMCID: PMC8148880 DOI: 10.1590/1414-431x2020e10062] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 03/18/2021] [Indexed: 12/24/2022]
Abstract
Long non-coding RNA (lncRNA) is an essential regulator of carcinogenesis and cancer progression. In the study, we explored the role of lncRNA DLGAP1-AS1 in gastric cancer (GC). qRT-PCR was carried out to detect DLGAP1-AS1 expression in GC tissues and cell lines. CCK-8 assay, EdU assay, and transwell experiments were employed to detect the malignant biological behaviors of GC cells with DLGAP1-AS1 knockdown or overexpression. Bioinformatics and dual-luciferase report assay were used to confirm the binding relationship between DLGAP1-AS1 and miR-515-5p. MARK4 expression was detected by western blot after DLGAP1-AS1/miR-515-5p was selectively regulated. DLGAP1-AS1 was up-regulated in GC tissues and cell lines, and its high expression was closely associated with larger tumor size, higher TNM stage, and lymph node metastasis. Furthermore, DLGAP1-AS1 overexpression enhanced cell proliferation, migration, and invasion, and miR-515-5p could reverse these effects. DLGAP1-AS1 participated in the regulation of the MARK4 signaling pathway by targeting miR-515-5p. DLGAP1-AS1 promoted GC progression through miR-515-5p/MARK4 signaling pathway.
Collapse
Affiliation(s)
- Liping Li
- Department of Medical Oncology, Dongguan People's Hospital, Dongguan, Guangdong, China
| | - Qingjun Lai
- Department of Medical Oncology, Dongguan People's Hospital, Dongguan, Guangdong, China
| | - Manman Zhang
- Department of Medical Oncology, Dongguan People's Hospital, Dongguan, Guangdong, China
| | - Jun Jia
- Department of Medical Oncology, Dongguan People's Hospital, Dongguan, Guangdong, China
| |
Collapse
|
35
|
Long Noncoding RNA HCG11 Acts as a Tumor Suppressor in Gastric Cancer by Regulating miR-942-5p/BRMS1 Axis. JOURNAL OF ONCOLOGY 2021; 2021:9961189. [PMID: 34054958 PMCID: PMC8131154 DOI: 10.1155/2021/9961189] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 04/30/2021] [Indexed: 12/29/2022]
Abstract
The functions of long noncoding RNAs (lncRNAs) have been widely investigated in human cancers, including gastric cancer (GC). The purpose of this study was to elucidate the role of lncRNA HCG11 in GC. In this study, mRNA and protein expressions were detected by quantitative real-time polymerase chain reaction assays (RT-qPCR) and Western blot analysis. The proliferation ability of GC cells was examined by (3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyl Tetrazolium Bromide) MTT assays. The invasion and migration abilities of GC cells were evaluated by Transwell assays. The binding sites between miR-942-5p and HCG11/BRMS1 were confirmed by dual-luciferase reporter assays. Results showed that LncRNA HCG11 was downregulated in GC cells. Functionally, overexpression of HCG11 inhibited GC cell proliferation, migration, and invasion. In addition, lncRNA HCG11 was found to act as a molecular sponge of miR-942-5p. Furthermore, miR-942-5p promoted GC progression by suppressing lncRNA HCG11 expression. Besides that, BRMS1 was confirmed as a direct target of miR-942-5p. More importantly, breast cancer metastasis suppressor 1 (BRMS1) inhibited GC progression by upregulating lncRNA HCG11 and downregulating miR-942-5p. In conclusion, LncRNA HCG11 inhibited cell proliferation, migration, and invasion in GC by sponging miR-942-5p and upregulating BRMS1.
Collapse
|
36
|
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: 2.8] [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
|
37
|
Xu X, Xie Q, Xie M, Zeng Y, Liu Q. LncRNA SNHG8 Serves as an Oncogene in Breast Cancer Through miR-634/ZBTB20 Axis. Cancer Manag Res 2021; 13:3017-3028. [PMID: 33854372 PMCID: PMC8039051 DOI: 10.2147/cmar.s270128] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 12/16/2020] [Indexed: 12/14/2022] Open
Abstract
Background Small nucleolus RNA Host Gene 8 (SNHG8) belongs to a subgroup with long non-coding RNAs. LncRNA SNHG8 presents up-regulated in miscellaneous cancers, like gastric cancer, liver cancer, and esophageal squamous cell cancer. Nevertheless, the expression pattern and the pathological function of lncRNA SNHG8 in breast cancer remain obscure. Methods We examined the expression levels of lncRNA SNHG8 in the tissue samples and cell lines from breast cancer via RT-qPCR in the present study. The functions of lncRNA SNHG8 on the progression of breast cancer cell were examined by CCK-8, EdU, Transwell chamber assays, and flow cytometry analyses. The expression of proteins was assessed using Western blot assay. Results We found that proliferation, migration, and invasion of breast cancer cells were significantly inhibited due to knockdown of lncRNA SNHG8, while inducing apoptosis of these cells. Mechanistically, SNHG8 functioned as an inhibitor of miR-634 in tumor tissues. Conclusion LncRNA SNHG8 sponged the miR-634 to increase the expression level of ZBTB20, thus further aggravating the malignancy of breast cancer. Hence, the lncRNA SNHG8-miR-634-ZBTB20 axis may be a promising therapeutic target to treat breast cancers.
Collapse
Affiliation(s)
- Xianyun Xu
- Key Laboratory of Prevention and treatment of cardiovascular and cerebrovascular diseases of Ministry of Education, Jiangxi Provincial Clinical Research Center for Vascular Anomalies, Basic Medical School, Gannan Medical University, Ganzhou, Jiangxi, 341000, People's Republic of China
| | - Qiongjun Xie
- Basic Medical School, Gannan Medical University, Ganzhou, Jiangxi, 341000, People's Republic of China
| | - Mingfeng Xie
- Department of Pediatric Surgery, Jiangxi Provincial Clinical Research Center for Vascular Anomalies, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, The First Affiliate Hospital of Gannan Medical University, Ganzhou, Jiangxi, 341000, People's Republic of China
| | - Yong Zeng
- Department of Pediatric Surgery, Jiangxi Provincial Clinical Research Center for Vascular Anomalies, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, The First Affiliate Hospital of Gannan Medical University, Ganzhou, Jiangxi, 341000, People's Republic of China
| | - Qian Liu
- Key Laboratory of Prevention and treatment of cardiovascular and cerebrovascular diseases of Ministry of Education, Jiangxi Provincial Clinical Research Center for Vascular Anomalies, Basic Medical School, Gannan Medical University, Ganzhou, Jiangxi, 341000, People's Republic of China
| |
Collapse
|
38
|
Tian X, Liu Y, Wang Z, Wu S. lncRNA SNHG8 promotes aggressive behaviors of nasopharyngeal carcinoma via regulating miR-656-3p/SATB1 axis. Biomed Pharmacother 2020; 131:110564. [PMID: 32920509 DOI: 10.1016/j.biopha.2020.110564] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/21/2020] [Accepted: 07/25/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Long non-coding RNA (lncRNA) has been proposed to regulate tumorigenesis, however, the role of small nucleolar RNA host gene 8 (SNHG8) in nasopharyngeal carcinoma (NPC) remains unclear. METHODS Levels of SNHG8 in NPC tissues and cells were analyzed with real-time quantitative PCR method. Cell counting kit-8 assay, colony formation assay, wound-healing assay, and transwell invasion assay were performed to detect cell viability, migration, and invasion. Luciferase activity assay and RIP assay were performed to explore relationships among SNHG8, microRNA-656-3p (miR-656-3p), and special AT-rich sequence-binding protein 1 (SATB1). RESULTS We found SNHG8 level was increased expression in NPC tissues and cells.In vitro assays revealed that SNHG8 stimulates NPC cell proliferation, colony formation, cell migration, and cell invasion. In vivo assay confirmed knockdown of SNHG8 could hamper tumor growth. Furthermore, we showed SNHG8 serves as a sponge for miR-656-3p to regulate SATB1 expression, and participated in NPC progression. CONCLUSIONS In summary, our work indicated the importance of SNHG8 in NPC progression, which provided novel treatment methods for NPC.
Collapse
Affiliation(s)
- Xiaoyan Tian
- Department of Otorhinolaryngology Head and Neck Surgery, Second Affiliated Hospital of Nanchang University, No.1 Minde Street, Nanchang 330006, China
| | - Yuehui Liu
- Department of Otorhinolaryngology Head and Neck Surgery, Second Affiliated Hospital of Nanchang University, No.1 Minde Street, Nanchang 330006, China.
| | - Zhi Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Second Affiliated Hospital of Nanchang University, No.1 Minde Street, Nanchang 330006, China
| | - Shuhong Wu
- Department of Otorhinolaryngology Head and Neck Surgery, Second Affiliated Hospital of Nanchang University, No.1 Minde Street, Nanchang 330006, China
| |
Collapse
|
39
|
Meng Q, Wang X, Xue T, Zhao Q, Wang W, Zhao K. Long noncoding RNA MIR99AHG promotes gastric cancer progression by inducing EMT and inhibiting apoptosis via miR577/FOXP1 axis. Cancer Cell Int 2020; 20:414. [PMID: 32874129 PMCID: PMC7457246 DOI: 10.1186/s12935-020-01510-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 08/19/2020] [Indexed: 12/12/2022] Open
Abstract
Background Long non-coding RNAs (lncRNAs) play a vital role in the genesis and development of human cancer. LncRNA MIR99AHG has been reported to be upregulated in acute myeloid leukemia (AML); however, its function in gastric cancer (GC) is still not clear. Here we were aiming to screen the prognostic lncRNA candidates and to explore the function of MIR99AHG in GC. Methods We have preliminarily screened some candidate lncRNA biomarkers in GC tissues through analyzing microarray datasets. The expression level of MIR99AHG in GC cell lines and tissues was monitored via qPCR. Survival analysis was performed with the patients of our hospital and TCGA database cases. CCK-8 assay, trans-well assay and flow cytometry were performed to determine cell proliferation, invasion, migration and apoptosis. Meanwhile, a target of MIR99AHG was predicted and identified by luciferase reporter gene detection experiments. Results MIR99AHG was strongly up-regulated in human GC and contributed to cancer progression. Kaplan–Meier analysis revealed that up-regulating MIR99AHG expression was positively correlated with unfavorable overall survival (P < 0.01) of patients from our hospital and TCGA database. Knockdown of MIR99AHG expression inhibited cell proliferation, invasion, migration and promoted cell apoptosis. Moreover, MIR99AHG worked as an oncogenic gene though competing for endogenous RNA (ceRNA) of miR-577. Conclusions Our findings suggested that MIR99AHG contributes to malignant phenotypes of GC and may become a promising therapeutic target.
Collapse
Affiliation(s)
- Qingyang Meng
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiangjun Wang
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Tongqing Xue
- Department of Oncology, Huaian Hospital of Huaian City, No.161 Zhenhuailou East Road, Huai'an, 223200 Jiangsu China
| | - Qiangfang Zhao
- Department of Oncology, Huaian Hospital of Huaian City, No.161 Zhenhuailou East Road, Huai'an, 223200 Jiangsu China
| | - Wei Wang
- Department of Oncology, Huaian Hospital of Huaian City, No.161 Zhenhuailou East Road, Huai'an, 223200 Jiangsu China
| | - Kun Zhao
- Department of Oncology, Huaian Hospital of Huaian City, No.161 Zhenhuailou East Road, Huai'an, 223200 Jiangsu China
| |
Collapse
|
40
|
Zhang Y, Bian Y. Long Non-Coding RNA SNHG8 Plays a Key Role in Myocardial Infarction Through Affecting Hypoxia-Induced Cardiomyocyte Injury. Med Sci Monit 2020; 26:e924016. [PMID: 32772038 PMCID: PMC7437243 DOI: 10.12659/msm.924016] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background The objective of the study was to explore the role of long non-coding RNA SNHG8 (lncRNA SNHG8) in myocardial infarction (MI) and the related mechanism of action. Material/Methods In vitro model of MI was established by hypoxia induction in cardiomyocyte line H9c2 cells. H9c2 cells were transfected with control-plasmid, SNHG8-plasmid, control-shRNA and SNHG8-shRNA. Quantitative real-time polymerase chain reaction (qRT-PCR) assay was performed to measure transfection efficiency. Creatine kinase-muscle/brain (CK-MB) release, cardiac troponin 1 (cTnI) release and mitochondria viability were detected by using related detection kits. MTT (3-(45)-dimethylthiahiazo (-z-y 1)-35-diphenytetrazoliumromide) assay was used to detect cell viability and flow cytometry analysis was used to detect cell apoptosis. Western blot assay was performed to measure protein expression of cleaved-Caspase3, p-p65 and p65. Enzyme-linked immunosorbent assay (ELISA) and qRT-PCR assay were performed to detect expression of interleukin (IL)-1β, tumor necrosis factor (TNF)-α and IL-6. Results LncRNA SNHG8 was overexpressed in hypoxia-induced cardiomyocytes. SNHG8-plasmid increased lncRNA SNHG8 expression, CK-MB release, cTnI release, and mitochondria viability in hypoxia-induced H9c2 cells. In addition, SNHG8-plasmid reduced cell viability, induced cell apoptosis, and increased expression of cleaved-caspase3, IL-1β, TNF-α, IL-6, and p-p65 in hypoxia-induced H9c2 cells, while the effects of SNHG8-shRNA were opposite. Conclusions We demonstrated that lncRNA SNHG8 affected myocardial infarction by affecting hypoxia-induced cardiomyocyte injury via regulation of the NF-κB pathway.
Collapse
Affiliation(s)
- Yue Zhang
- Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| | - Yunfei Bian
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| |
Collapse
|
41
|
A novel ceRNA axis involves in regulating immune infiltrates and macrophage polarization in gastric cancer. Int Immunopharmacol 2020; 87:106845. [PMID: 32763781 DOI: 10.1016/j.intimp.2020.106845] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 07/05/2020] [Accepted: 07/26/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Increasing evidence suggests that the lncRNA-miRNA-mRNA regulatory network is highly correlated with gastric cancer (GC) development. However, a prognosis-associated lncRNA-miRNA-mRNA network remains to be identified in GC. METHODS Differentially expressed genes (DEGs) were screened by integrating 6 microarray datasets using the RRA method. Hub genes were identified by analysing their degrees in a PPI (protein-protein interaction) network. Upstream miRNAs and lncRNAs of hub genes were predicted by miRTarBase and miRNet, respectively. Key genes, miRNAs and lncRNAs were identified by evaluating their expression and prognosis in GEPIA and Kaplan-Meier plotter, respectively. A key lncRNA-miRNA-mRNA network was constructed in Cytoscape, and the correlations were analysed in the ENCORI database. We also evaluated the mRNA expression of ceRNA axes in the TIMER and Oncomine databases and their correlation with prognosis in GC patients with different clinical features using Kaplan-Meier plotter. In addition, correlations between mRNA and immune infiltrating cells in GC were investigated by the TIMER database. Finally, several experiments were conducted to verify our analyses. RESULTS Forty-two upregulated and 86 downregulated DEGs were obtained from the "RRA" integrated analysis. Eight of the 20 hub genes were identified as key genes by analysing their expression and prognosis. Seventeen miRNAs were predicted to target key genes, and low expression of 4 miRNAs suggested poor outcome in GC. Furthermore, 155 lncRNAs were predicted to target 4 key miRNAs, and only 5 lncRNAs were highly expressed, suggesting poor outcomes in patients with GC. Then, the H19-miR-29a-3p-COL1A2 axis was constructed by correlation analysis. In addition, COL1A2 was positively correlated with lymphatic metastasis, immune infiltrating cell levels, markers of monocytes, tumour-associated macrophages (TAMs), and M2 macrophages but not M1 macrophages in GC. The experimental results revealed that the H19-miR-29a-3p-COL1A2 axis may promote macrophage polarization from M1 to M2 in GC. CONCLUSIONS A novel lncRNA-miRNA-mRNA axis was identified and may be involved in regulating immune cell infiltration and macrophage polarization, which may provide new treatment strategies for GC.
Collapse
|
42
|
Li F, Chen ZH, Tan BB, Li Y. Long non-coding RNAs as potential markers for occurrence, progression, and prognosis of gastric cancer. Shijie Huaren Xiaohua Zazhi 2020; 28:544-552. [DOI: 10.11569/wcjd.v28.i13.544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In recent years, more and more attention has been paid to the relationship between long non-coding RNAs (lncRNAs) and tumor. Abnormal expression of lncRNAs plays an oncogenic or tumor-suppressing role in gastric cancer (GC) by participating in the biological behaviors of GC cells, such as proliferation, invasion, and migration. By summarizing the relevant literature, this paper discusses the research status, detection technology, and mechanism of action of lncRNAs in GC, as well as their potential as markers for occurrence, progression, prognosis, and drug resistance of GC. It is expected that lncRNAs can play an important role in early detection, early treatment, and effective improvement of chemotherapy resistance of GC to achieve personalized precise treatment of this malignancy.
Collapse
Affiliation(s)
- Fang Li
- Department of Pathology, Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei Province, China
| | - Zi-Hao Chen
- Third Department of Surgery, Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei Province, China
| | - Bi-Bo Tan
- Third Department of Surgery, Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei Province, China
| | - Yong Li
- Third Department of Surgery, Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei Province, China
| |
Collapse
|
43
|
Liu X, Sun R, Chen J, Liu L, Cui X, Shen S, Cui G, Ren Z, Yu Z. Crosstalk Mechanisms Between HGF/c-Met Axis and ncRNAs in Malignancy. Front Cell Dev Biol 2020; 8:23. [PMID: 32083078 PMCID: PMC7004951 DOI: 10.3389/fcell.2020.00023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 01/13/2020] [Indexed: 12/24/2022] Open
Abstract
Several lines of evidence have confirmed the magnitude of crosstalk between HGF/c-Met axis (hepatocyte growth factor and its high-affinity receptor c-mesenchymal-epithelial transition factor) and non-coding RNAs (ncRNAs) in tumorigenesis. Through activating canonical or non-canonical signaling pathways, the HGF/c-Met axis mediates a range of oncogenic processes such as cell proliferation, invasion, apoptosis, and angiogenesis and is increasingly becoming a promising target for cancer therapy. Meanwhile, ncRNAs are a cluster of functional RNA molecules that perform their biological roles at the RNA level and are essential regulators of gene expression. The expression of ncRNAs is cell/tissue/tumor-specific, which makes them excellent candidates for cancer research. Many studies have revealed that ncRNAs play a crucial role in cancer initiation and progression by regulating different downstream genes or signal transduction pathways, including HGF/c-Met axis. In this review, we discuss the regulatory association between ncRNAs and the HGF/c-Met axis by providing a comprehensive understanding of their potential mechanisms and roles in cancer development. These findings could reveal their possible clinical applications as biomarkers for therapeutic interventions.
Collapse
Affiliation(s)
- Xin Liu
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Laboratory of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ranran Sun
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Laboratory of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianan Chen
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Laboratory of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Liwen Liu
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Laboratory of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xichun Cui
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Laboratory of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shen Shen
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Laboratory of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guangying Cui
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Laboratory of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhigang Ren
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Laboratory of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zujiang Yu
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Laboratory of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|