1
|
Deng H, Zhang Q, Zhao Z, Wang M, Xu Q. SNAI2/FTH1P3/miR-218-5p Positive Feedback Loop Promotes Colorectal Cancer Metastasis. Biochem Genet 2024; 62:2210-2223. [PMID: 37884850 DOI: 10.1007/s10528-023-10546-y] [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: 06/19/2022] [Accepted: 10/05/2023] [Indexed: 10/28/2023]
Abstract
Colorectal cancer (CRC) is a type of intestinal cancer that causes more than 600,000 deaths every year. Overcoming the problems of metastasis requires detailed studies to reveal the potential molecular mechanisms. This study aims to reveal the molecular mechanism of CRC metastasis involving non-coding RNA regulation. The expression profile of FTH1P3 was analyzed based on the data of TCGA-COAD patient cohorts. Q-PCR analysis was performed to validate the expression of FTH1P3 in colorectal cancer cells. JASPR was used to screen transcription factors of FTH1P3. q-ChIP analysis was used to validate the target between FTH1P3 and transcription factor. Scratch assay and transwell assay were used to evaluate the migration and invasion ability of colorectal cancer cells. FTH1P3 is highly expressed in CRC patient cohort. FTH1P3 induced migration and invasion of SW480 cell through regulating epithelial-mesenchymal transition (EMT). In addition, FTH1P3 is a direct target of SNAI2. SNAI2 promotes the expression of FTH1P3. Both FTH1P3 and SNAI2 were directly targeted and repressed by miR-218-5p. Interestingly, ectopic FTH1P3 caused a decreased miR-218-5p level and an elevated nucleic SNAI2 protein expression level. Of note, only ectopic SNAI2 protein resulted in a repressed miR-218-5p and an increased FTH1P3, whereas SNAI2 3'UTR failed to affect the expression of miR-218-5p and FTH1P3. SNAI2 transcriptionally activates FTH1P3 expression. Both SNAI2 and FTH1P3 are targets of miR-218-5p. SNAI2/FTH1P3/miR-218-5p form a positive feedback loop in the regulation of CRC metastasis.
Collapse
Affiliation(s)
- Hong Deng
- Department of General Surgery (Gastrointestinal Surgery), The Affiliated Hospital of Southwest Medical University, Taiping Rd. No 25, Luzhou, 646000, China
| | - Qiang Zhang
- Department of General Surgery (Gastrointestinal Surgery), The Affiliated Hospital of Southwest Medical University, Taiping Rd. No 25, Luzhou, 646000, China
| | - Zhengfei Zhao
- Department of General Surgery (Gastrointestinal Surgery), The Affiliated Hospital of Southwest Medical University, Taiping Rd. No 25, Luzhou, 646000, China
| | - Mingming Wang
- Department of General Surgery (Gastrointestinal Surgery), The Affiliated Hospital of Southwest Medical University, Taiping Rd. No 25, Luzhou, 646000, China
| | - Qin Xu
- Department of General Surgery (Gastrointestinal Surgery), The Affiliated Hospital of Southwest Medical University, Taiping Rd. No 25, Luzhou, 646000, China.
| |
Collapse
|
2
|
Darvish M. LncRNA FTH1P3: A New Biomarker for Cancer-Related Therapeutic Development. Curr Mol Med 2024; 24:576-584. [PMID: 37491858 DOI: 10.2174/1566524023666230724141353] [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: 04/15/2023] [Revised: 06/17/2023] [Accepted: 06/22/2023] [Indexed: 07/27/2023]
Abstract
Cancer is a persistent and urgent health problem that affects the entire world. Not long ago, regulatory biomolecules referred to as long noncoding RNAs (lncRNAs) might have value for their innate abundance and stability. These single-stranded RNAs potentially interfere with several physiological and biochemical cellular processes involved in many human pathological situations, particularly cancer diseases. Ferritin heavy chain1 pseudogene 3 (FTH1P3), a lncRNA that is ubiquitously transcribed and belongs to the ferritin heavy chain (FHC) family, represents a novel class of lncRNAs primarily found in oral squamous cell carcinoma. Further research has shown that FTH1P3 is involved in other malignancies such as uveal melanoma, glioma, esophageal squamous cell carcinoma, non-small cell lung cancer, breast cancer, laryngeal squamous cell carcinoma, and cervical cancer. Accordingly, FTH1P3 significantly enhances cancer symptoms, including cell proliferation, invasion, metastasis, chemoresistance, and inhibition of apoptosis through many specific mechanisms. Notably, the clinical data significantly demonstrated the association of FTH1P3 overexpression with poor prognosis and poor overall survival within the examined samples. Here, we summarize all the research published to date (13 articles) on FTH1P3, focusing on the biological function underlying the regulatory mechanism and its possible clinical relevance.
Collapse
Affiliation(s)
- Maryam Darvish
- Department of Medical Biotechnology, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| |
Collapse
|
3
|
Mousavi SM, Derakhshan M, Baharloii F, Dashti F, Mirazimi SMA, Mahjoubin-Tehran M, Hosseindoost S, Goleij P, Rahimian N, Hamblin MR, Mirzaei H. Non-coding RNAs and glioblastoma: Insight into their roles in metastasis. Mol Ther Oncolytics 2022; 24:262-287. [PMID: 35071748 PMCID: PMC8762369 DOI: 10.1016/j.omto.2021.12.015] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Glioma, also known as glioblastoma multiforme (GBM), is the most prevalent and most lethal primary brain tumor in adults. Gliomas are highly invasive tumors with the highest death rate among all primary brain malignancies. Metastasis occurs as the tumor cells spread from the site of origin to another site in the brain. Metastasis is a multifactorial process, which depends on alterations in metabolism, genetic mutations, and the cancer microenvironment. During recent years, the scientific study of non-coding RNAs (ncRNAs) has led to new insight into the molecular mechanisms involved in glioma. Many studies have reported that ncRNAs play major roles in many biological procedures connected with the development and progression of glioma. Long ncRNAs (lncRNAs), microRNAs (miRNAs), and circular RNAs (circRNAs) are all types of ncRNAs, which are commonly dysregulated in GBM. Dysregulation of ncRNAs can facilitate the invasion and metastasis of glioma. The present review highlights some ncRNAs that have been associated with metastasis in GBM. miRNAs, circRNAs, and lncRNAs are discussed in detail with respect to their relevant signaling pathways involved in metastasis.
Collapse
Affiliation(s)
- Seyed Mojtaba Mousavi
- Department of Neurosciences and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Derakhshan
- Department of Pathology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fatereh Baharloii
- Department of Cardiology, Chamran Cardiovascular Research Education Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fatemeh Dashti
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Mohammad Ali Mirazimi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Maryam Mahjoubin-Tehran
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saereh Hosseindoost
- Brain and Spinal Cord Research Center, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Pouya Goleij
- Department of Genetics, Faculty of Biology, Sana Institute of Higher Education, Sari, Iran
| | - Neda Rahimian
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Department of Internal Medicine, Firoozgar Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
4
|
Liu L, Xu Q, Xiong Y, Deng H, Zhou J. LncRNA LINC01094 contributes to glioma progression by modulating miR-224-5p/CHSY1 axis. Hum Cell 2022; 35:214-225. [PMID: 34716872 DOI: 10.1007/s13577-021-00637-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 05/07/2021] [Indexed: 01/20/2023]
Abstract
Glioma serves as the most common malignancy influencing modern people and is associated with severe morbidity and high mortality. Long non-coding RNAs (lncRNAs) as crucial regulators participate in multiple cancer progression. However, the role of lncRNA LINC01094 in the development of glioma remains unclear. Here, we aimed to explore the effect of lncRNA LINC01094 on the glioma progression and the underlying mechanism. Significantly, we revealed that the expression levels of LINC01094 were elevated in the glioma patient tissues compared to adjacent normal tissues. The LINC01094 expression was enhanced in the glioma cell lines. The depletion of LINC01094 inhibited cell viability and colony formation in the glioma cells. Meanwhile, the migration and invasion of glioma cells were impaired by the depletion of LINC01094. Mechanically, we identified that LINC01094 was able to sponge the miR-224-5p in the glioma cells and miR-224-5p inhibitor could reverse the effect of LINC01094 on glioma progression. In addition, miR-224-5p targeted CHSY1 and LINC01094 up-regulated CHSY1 by targeting miR-224-5p in the glioma cells. LINC01094 promoted glioma progression by the positive regulation of CHSY1. Moreover, tumorigenicity analysis showed that LINC01094 enhanced tumor growth of glioma in vivo. Thus, we conclude that lncRNA LINC01094 promotes glioma progression by modulating miR-224-5p/CHSY1 axis. Our finding provides new insights into the mechanism by which lncRNA LINC01094 contributes to the development of glioma, improving the understanding of lncRNA LINC01094 and glioma. LncRNA LINC01094, miR-224-5p, and CHSY1 may serve as potential targets for glioma.
Collapse
Affiliation(s)
- Luotong Liu
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Qian Xu
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yu Xiong
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Huajiang Deng
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jie Zhou
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China.
| |
Collapse
|
5
|
Stasiak M, Kolenda T, Kozłowska-Masłoń J, Sobocińska J, Poter P, Guglas K, Paszkowska A, Bliźniak R, Teresiak A, Kazimierczak U, Lamperska K. The World of Pseudogenes: New Diagnostic and Therapeutic Targets in Cancers or Still Mystery Molecules? Life (Basel) 2021; 11:life11121354. [PMID: 34947885 PMCID: PMC8705536 DOI: 10.3390/life11121354] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 02/07/2023] Open
Abstract
Pseudogenes were once considered as “junk DNA”, due to loss of their functions as a result of the accumulation of mutations, such as frameshift and presence of premature stop-codons and relocation of genes to inactive heterochromatin regions of the genome. Pseudogenes are divided into two large groups, processed and unprocessed, according to their primary structure and origin. Only 10% of all pseudogenes are transcribed into RNAs and participate in the regulation of parental gene expression at both transcriptional and translational levels through senseRNA (sRNA) and antisense RNA (asRNA). In this review, about 150 pseudogenes in the different types of cancers were analyzed. Part of these pseudogenes seem to be useful in molecular diagnostics and can be detected in various types of biological material including tissue as well as biological fluids (liquid biopsy) using different detection methods. The number of pseudogenes, as well as their function in the human genome, is still unknown. However, thanks to the development of various technologies and bioinformatic tools, it was revealed so far that pseudogenes are involved in the development and progression of certain diseases, especially in cancer.
Collapse
Affiliation(s)
- Maciej Stasiak
- Greater Poland Cancer Centre, Laboratory of Cancer Genetics, Garbary 15, 61-866 Poznan, Poland; (M.S.); (J.K.-M.); (J.S.); (K.G.); (A.P.); (R.B.); (A.T.)
- Greater Poland Cancer Centre, Research and Implementation Unit, Garbary 15, 61-866 Poznan, Poland;
| | - Tomasz Kolenda
- Greater Poland Cancer Centre, Laboratory of Cancer Genetics, Garbary 15, 61-866 Poznan, Poland; (M.S.); (J.K.-M.); (J.S.); (K.G.); (A.P.); (R.B.); (A.T.)
- Greater Poland Cancer Centre, Research and Implementation Unit, Garbary 15, 61-866 Poznan, Poland;
- Correspondence: or (T.K.); or (K.L.)
| | - Joanna Kozłowska-Masłoń
- Greater Poland Cancer Centre, Laboratory of Cancer Genetics, Garbary 15, 61-866 Poznan, Poland; (M.S.); (J.K.-M.); (J.S.); (K.G.); (A.P.); (R.B.); (A.T.)
- Greater Poland Cancer Centre, Research and Implementation Unit, Garbary 15, 61-866 Poznan, Poland;
- Faculty of Biology, Institute of Human Biology and Evolution, Adam Mickiewicz University, Uniwersytetu Poznańskiego 6, 61-614 Poznań, Poland
| | - Joanna Sobocińska
- Greater Poland Cancer Centre, Laboratory of Cancer Genetics, Garbary 15, 61-866 Poznan, Poland; (M.S.); (J.K.-M.); (J.S.); (K.G.); (A.P.); (R.B.); (A.T.)
- Greater Poland Cancer Centre, Research and Implementation Unit, Garbary 15, 61-866 Poznan, Poland;
| | - Paulina Poter
- Greater Poland Cancer Centre, Research and Implementation Unit, Garbary 15, 61-866 Poznan, Poland;
- Greater Poland Cancer Center, Department of Oncologic Pathology and Prophylaxis, Poznan University of Medical Sciences, Garbary 15, 61-866 Poznan, Poland
- Department of Pathology, Pomeranian Medical University, Rybacka 1, 70-204 Szczecin, Poland
| | - Kacper Guglas
- Greater Poland Cancer Centre, Laboratory of Cancer Genetics, Garbary 15, 61-866 Poznan, Poland; (M.S.); (J.K.-M.); (J.S.); (K.G.); (A.P.); (R.B.); (A.T.)
- Greater Poland Cancer Centre, Research and Implementation Unit, Garbary 15, 61-866 Poznan, Poland;
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, 61 Zwirki and Wigury, 02-091 Warsaw, Poland
| | - Anna Paszkowska
- Greater Poland Cancer Centre, Laboratory of Cancer Genetics, Garbary 15, 61-866 Poznan, Poland; (M.S.); (J.K.-M.); (J.S.); (K.G.); (A.P.); (R.B.); (A.T.)
- Greater Poland Cancer Centre, Research and Implementation Unit, Garbary 15, 61-866 Poznan, Poland;
- Faculty of Biology, Adam Mickiewicz University, Umultowska 89, 61-614 Poznan, Poland
| | - Renata Bliźniak
- Greater Poland Cancer Centre, Laboratory of Cancer Genetics, Garbary 15, 61-866 Poznan, Poland; (M.S.); (J.K.-M.); (J.S.); (K.G.); (A.P.); (R.B.); (A.T.)
- Greater Poland Cancer Centre, Research and Implementation Unit, Garbary 15, 61-866 Poznan, Poland;
| | - Anna Teresiak
- Greater Poland Cancer Centre, Laboratory of Cancer Genetics, Garbary 15, 61-866 Poznan, Poland; (M.S.); (J.K.-M.); (J.S.); (K.G.); (A.P.); (R.B.); (A.T.)
- Greater Poland Cancer Centre, Research and Implementation Unit, Garbary 15, 61-866 Poznan, Poland;
| | - Urszula Kazimierczak
- Department of Cancer Immunology, Medical Biotechnology, Poznan University of Medical Sciences, 8 Rokietnicka Street, 60-806 Poznan, Poland;
| | - Katarzyna Lamperska
- Greater Poland Cancer Centre, Laboratory of Cancer Genetics, Garbary 15, 61-866 Poznan, Poland; (M.S.); (J.K.-M.); (J.S.); (K.G.); (A.P.); (R.B.); (A.T.)
- Greater Poland Cancer Centre, Research and Implementation Unit, Garbary 15, 61-866 Poznan, Poland;
- Correspondence: or (T.K.); or (K.L.)
| |
Collapse
|
6
|
Xu K, Jiang X, Ariston Gabriel AN, Li X, Wang Y, Xu S. Evolving Landscape of Long Non-coding RNAs in Cerebrospinal Fluid: A Key Role From Diagnosis to Therapy in Brain Tumors. Front Cell Dev Biol 2021; 9:737670. [PMID: 34692695 PMCID: PMC8529119 DOI: 10.3389/fcell.2021.737670] [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: 07/07/2021] [Accepted: 08/31/2021] [Indexed: 11/23/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are a type of non-coding RNAs that act as molecular fingerprints and modulators of many pathophysiological processes, particularly in cancer. Specifically, lncRNAs can be involved in the pathogenesis and progression of brain tumors, affecting stemness/differentiation, replication, invasion, survival, DNA damage response, and chromatin dynamics. Furthermore, the aberrations in the expressions of these transcripts can promote treatment resistance, leading to tumor recurrence. The development of next-generation sequencing technologies and the creation of lncRNA-specific microarrays have boosted the study of lncRNA etiology. Cerebrospinal fluid (CSF) directly mirrors the biological fluid of biochemical processes in the brain. It can be enriched for small molecules, peptides, or proteins released by the neurons of the central nervous system (CNS) or immune cells. Therefore, strategies that identify and target CSF lncRNAs may be attractive as early diagnostic and therapeutic options. In this review, we have reviewed the studies on CSF lncRNAs in the context of brain tumor pathogenesis and progression and discuss their potential as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Kanghong Xu
- School of Public Health, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Xinquan Jiang
- School of Public Health, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | | | - Xiaomeng Li
- Department of Hematology, Jining First People's Hospital, Jining, China
| | - Yunshan Wang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, China
| | - Shuo Xu
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Key Laboratory of Brain Function Remodeling, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
7
|
Milán-Rois P, Quan A, Slack FJ, Somoza Á. The Role of LncRNAs in Uveal Melanoma. Cancers (Basel) 2021; 13:cancers13164041. [PMID: 34439196 PMCID: PMC8392202 DOI: 10.3390/cancers13164041] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/06/2021] [Accepted: 08/06/2021] [Indexed: 12/13/2022] Open
Abstract
Uveal melanoma (UM) is an intraocular cancer tumor with high metastatic risk. It is considered a rare disease, but 90% of affected patients die within 15 years. Non-coding elements (ncRNAs) such as long non-coding RNAs (lncRNAs) have a crucial role in cellular homeostasis maintenance, taking part in many critical cellular pathways. Their deregulation, therefore, contributes to the induction of cancer and neurodegenerative and metabolic diseases. In cancer, lncRNAs are implicated in apoptosis evasion, proliferation, invasion, drug resistance, and other roles because they affect tumor suppressor genes and oncogenes. For these reasons, lncRNAs are promising targets in personalized medicine and can be used as biomarkers for diseases including UM.
Collapse
Affiliation(s)
- Paula Milán-Rois
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Unidad Asociada al Centro Nacional de Biotecnología (CSIC), 28049 Madrid, Spain;
| | - Anan Quan
- Department of Pathology, Beth Israel Deaconess Medical Center (BIDMC)/Harvard Medical School, Boston, MA 02215, USA; (A.Q.); (F.J.S.)
| | - Frank J. Slack
- Department of Pathology, Beth Israel Deaconess Medical Center (BIDMC)/Harvard Medical School, Boston, MA 02215, USA; (A.Q.); (F.J.S.)
| | - Álvaro Somoza
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Unidad Asociada al Centro Nacional de Biotecnología (CSIC), 28049 Madrid, Spain;
- Correspondence: ; Tel.: +34-91-299-8856
| |
Collapse
|
8
|
Xi S, Cai H, Lu J, Zhang Y, Yu Y, Chen F, Huang Q, Wang F, Chen Z. The pseudogene PRELID1P6 promotes glioma progression via the hnHNPH1-Akt/mTOR axis. Oncogene 2021; 40:4453-4467. [PMID: 34108621 PMCID: PMC8249232 DOI: 10.1038/s41388-021-01854-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 05/06/2021] [Accepted: 05/21/2021] [Indexed: 12/13/2022]
Abstract
Research over the past decade has suggested important roles for pseudogenes in glioma. This study aimed to show that pseudogene PRELI domain-containing 1 pseudogene 6 (PRELID1P6) promotes glioma progression. Aberrant expression of genes was screened using The Cancer Genome Atlas database. We found that mRNA level of PRELID1P6 was highly upregulated in glioma and was associated with a shorter survival time. Functional studies showed that the knockdown of PRELID1P6 decreased cell proliferation, sphere formation, and clone formation ability and blocked the cell cycle transition at G0/G1, while overexpression of PRELID1P6 had the opposite effects. Mechanistically, knockdown of PRELID1P6 changed the cellular localization of heterogeneous nuclear ribonucleoprotein H1 (hnRNPH1) from nucleus to cytoplasm, which promoted ubiquitin-mediated degradation of hnRNPH1. RNA-sequence and gene set enrichment analysis suggested that knockdown of PRELID1P6 regulates the apoptosis signaling pathway. Western blotting showed that PRELID1P6 increased TRF2 expression by hnRNPH1-mediated alternative splicing effect and activated the Akt/mTOR pathway. Furthermore, Akt inhibitor MK2206 treatment reversed the oncogenic function of PRELID1P6. PRELID1P6 was also found to be negatively regulated by miR-1825. Our result showed that PRELID1P6 promotes glioma progression through the hnHNPH1-Akt/mTOR pathway. These findings shed new light on the important role of PRELID1P6 as a novel oncogene for glioma.
Collapse
Affiliation(s)
- Shaoyan Xi
- Department of Pathology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Haiping Cai
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Jiabin Lu
- Department of Pathology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Yu Zhang
- Department of Pathology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Yanjiao Yu
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Furong Chen
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Qitao Huang
- Department of Pathology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Fang Wang
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China.
| | - Zhongping Chen
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China.
| |
Collapse
|
9
|
Di Sanzo M, Quaresima B, Biamonte F, Palmieri C, Faniello MC. FTH1 Pseudogenes in Cancer and Cell Metabolism. Cells 2020; 9:E2554. [PMID: 33260500 PMCID: PMC7760355 DOI: 10.3390/cells9122554] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/24/2020] [Accepted: 11/24/2020] [Indexed: 12/15/2022] Open
Abstract
Ferritin, the principal intracellular iron-storage protein localized in the cytoplasm, nucleus, and mitochondria, plays a major role in iron metabolism. The encoding ferritin genes are members of a multigene family that includes some pseudogenes. Even though pseudogenes have been initially considered as relics of ancient genes or junk DNA devoid of function, their role in controlling gene expression in normal and transformed cells has recently been re-evaluated. Numerous studies have revealed that some pseudogenes compete with their parental gene for binding to the microRNAs (miRNAs), while others generate small interference RNAs (siRNAs) to decrease functional gene expression, and still others encode functional mutated proteins. Consequently, pseudogenes can be considered as actual master regulators of numerous biological processes. Here, we provide a detailed classification and description of the structural features of the ferritin pseudogenes known to date and review the recent evidence on their mutual interrelation within the complex regulatory network of the ferritin gene family.
Collapse
Affiliation(s)
- Maddalena Di Sanzo
- Department of Experimental and Clinical Medicine, “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy; (M.D.S.); (B.Q.); (F.B.)
| | - Barbara Quaresima
- Department of Experimental and Clinical Medicine, “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy; (M.D.S.); (B.Q.); (F.B.)
| | - Flavia Biamonte
- Department of Experimental and Clinical Medicine, “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy; (M.D.S.); (B.Q.); (F.B.)
| | - Camillo Palmieri
- Department of Experimental and Clinical Medicine, “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy; (M.D.S.); (B.Q.); (F.B.)
| | - Maria Concetta Faniello
- Department of Experimental and Clinical Medicine, “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy; (M.D.S.); (B.Q.); (F.B.)
- Research Center of Biochemistry and Advanced Molecular Biology, Department of Experimental and Clinical Medicine, “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy
| |
Collapse
|
10
|
Zhu B, Liu W, Liu H, Xu Q, Xu W. LINC01094 Down-Regulates miR-330-3p and Enhances the Expression of MSI1 to Promote the Progression of Glioma. Cancer Manag Res 2020; 12:6511-6521. [PMID: 32801889 PMCID: PMC7395698 DOI: 10.2147/cmar.s254630] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 06/23/2020] [Indexed: 01/15/2023] Open
Abstract
Background This study aims at probing into the expression, function, and mechanism of LINC01094 and miR-330-3p in glioma. Materials and Methods qRT-PCR was employed to examine LINC01094 and miR-330-3p expressions in gliomas. After gain-of-function and loss-of-function models were constructed, CCK-8 and Transwell assays were used to detect the proliferation, migration and invasion of LN229 and U251 cells, respectively. Additionally, dual luciferase reporter gene assay was utilized to verify the binding site between m4iR-330-3p and LINC01094, miR-330-3p, and the 3ʹUTR of musashi RNA binding protein 1 (MSI1). Then, RNA pull-down, RIP, qRT-PCR and Western blot were employed to detect the regulatory relationships among LINC01094, miR-330-3p, and MSI1. Results The expression of LINC01094 was elevated in glioma tissues and cell lines, and the high expression of LINC01094 was associated with high grade of glioma. In contrast, miR-330-3p was lowly expressed in glioma tissue. Overexpression of LINC01094 or down-regulation of miR-330-3p promoted the proliferation, migration, and invasion of glioma cells, while LINC01094 knockdown or miR-330-3p up-regulation impeded these processes. miR-330-3p was identified as a target miRNA of LINC01094, and it could be negatively regulated by LINC01094. In addition, miR-330-3p antagonized the function of LINC01094 by negatively regulating MSI1. Conclusion LINC01094 promotes the proliferation, migration, and invasion of glioma cells by adsorbing miR-330-3p and up-regulating the expression of MSI1.
Collapse
Affiliation(s)
- Bin Zhu
- Department of Neurosurgery, Huashan North Hospital, Baoshan Branch, Fudan University, Shanghai 200431, People's Republic of China
| | - Wei Liu
- Department of Neurosurgery, Huashan North Hospital, Baoshan Branch, Fudan University, Shanghai 200431, People's Republic of China
| | - Hongliang Liu
- Department of Neurosurgery, Huashan North Hospital, Baoshan Branch, Fudan University, Shanghai 200431, People's Republic of China
| | - Qiang Xu
- Department of Neurosurgery, Huashan North Hospital, Baoshan Branch, Fudan University, Shanghai 200431, People's Republic of China
| | - Wei Xu
- Department of Neurosurgery, Huashan North Hospital, Baoshan Branch, Fudan University, Shanghai 200431, People's Republic of China
| |
Collapse
|
11
|
Su D, Ji Z, Xue P, Guo S, Jia Q, Sun H. Long-Noncoding RNA FGD5-AS1 Enhances the Viability, Migration, and Invasion of Glioblastoma Cells by Regulating the miR-103a-3p/TPD52 Axis. Cancer Manag Res 2020; 12:6317-6329. [PMID: 32848452 PMCID: PMC7425657 DOI: 10.2147/cmar.s253467] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 07/08/2020] [Indexed: 12/12/2022] Open
Abstract
Purpose This study was designed to explore the functional role of FYVE, RhoGEF, and PH domain containing 5 antisense RNA 1 (FGD5-AS1) and the underlying regulatory mechanism in the progression of glioblastoma (GBM). Materials and Methods FGD5-AS1 expression was analyzed in The Cancer Genome Atlas (TCGA), and then detected in GBM tissues and cells by quantitative reverse-transcription polymerase chain reaction. Viability, migration, and invasion of GBM cells were assessed using the MTT, wound healing, and transwell assays, respectively. StarBase/TargetScan analysis and dual-luciferase reporter gene (DLR) assay were performed to investigate the relationship between FGD5-AS1/tumor protein D52 (TPD52) and miR-103a-3p. A xenograft tumor model was established to evaluate the role of FGD5-AS1 in GBM tumorigenesis in vivo. Results FGD5-AS1 was overexpressed in GBM tissues and cells, and silencing of FGD5-AS1 expression resulted in the inhibition of the viability, migration, and invasion of GBM cells. miR-130-3p was a target of FGD5-AS1, and its expression was negatively regulated by FGD5-AS1. Silencing miR-103a-3p expression resulted in the abrogation of the inhibitory effects of si-FGD5-AS1 on the viability, migration, and invasion of GBM cells. TPD52 was a target of miR-103a-3p and suppressed the antitumor effects of FGD5-AS1 silencing on GBM cells. In addition, FGD5-AS1 silencing inhibited the growth of xenograft tumors in vivo by modulating the miR-103a-3p/TPD52 axis. Conclusion Silencing of FGD5-AS1 inhibited the viability, migration, and invasion of GBM cells by regulating the miR-103a-3p/TPD52 axis.
Collapse
Affiliation(s)
- Daoqing Su
- Department of Neurosurgery, Liaocheng People's Hospital and Liaocheng Brain Hospital, Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng City, Shandong Province, People's Republic of China
| | - Zhengang Ji
- Department of Neurosurgery, Liaocheng People's Hospital and Liaocheng Brain Hospital, Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng City, Shandong Province, People's Republic of China
| | - Pengfei Xue
- Department of Neurosurgery, Liaocheng People's Hospital and Liaocheng Brain Hospital, Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng City, Shandong Province, People's Republic of China
| | - Shengfu Guo
- Department of Neurosurgery, Liaocheng People's Hospital and Liaocheng Brain Hospital, Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng City, Shandong Province, People's Republic of China
| | - Qingbin Jia
- Department of Neurosurgery, Liaocheng People's Hospital and Liaocheng Brain Hospital, Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng City, Shandong Province, People's Republic of China
| | - Hanyu Sun
- Department of Neurosurgery, Liaocheng People's Hospital and Liaocheng Brain Hospital, Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng City, Shandong Province, People's Republic of China
| |
Collapse
|
12
|
Lou W, Ding B, Fu P. Pseudogene-Derived lncRNAs and Their miRNA Sponging Mechanism in Human Cancer. Front Cell Dev Biol 2020; 8:85. [PMID: 32185172 PMCID: PMC7058547 DOI: 10.3389/fcell.2020.00085] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 01/30/2020] [Indexed: 12/28/2022] Open
Abstract
Pseudogenes, abundant in the human genome, are traditionally considered as non-functional “junk genes.” However, recent studies have revealed that pseudogenes act as key regulators at DNA, RNA or protein level in diverse human disorders (including cancer), among which pseudogene-derived long non-coding RNA (lncRNA) transcripts are extensively investigated and has been reported to be frequently dysregulated in various types of human cancer. Growing evidence demonstrates that pseudogene-derived lncRNAs play important roles in cancer initiation and progression by serving as competing endogenous RNAs (ceRNAs) through competitively binding to shared microRNAs (miRNAs), thus affecting both their cognate genes and unrelated genes. Herein, we retrospect those current findings about expression, functions and potential ceRNA mechanisms of pseudogene-derived lncRNAs in human cancer, which may provide us with some crucial clues in developing potential targets for cancer therapy in the future.
Collapse
Affiliation(s)
- Weiyang Lou
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Program of Innovative Cancer Therapeutics, Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Key Laboratory of Organ Transplantation, Zhejiang University, Hangzhou, China
| | - Bisha Ding
- Program of Innovative Cancer Therapeutics, Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Key Laboratory of Organ Transplantation, Zhejiang University, Hangzhou, China
| | - Peifen Fu
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
13
|
Silencing of NAMPT leads to up-regulation of insulin receptor substrate 1 gene expression in U87 glioma cells. Endocr Regul 2020; 54:31-42. [DOI: 10.2478/enr-2020-0005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Abstract
Objective. The aim of the present study was to investigate the effect of adipokine NAMPT (nicotinamide phosphoribosyltransferase) silencing on the expression of genes encoding IRS1 (insulin receptor substrate 1) and some other proliferation related proteins in U87 glioma cells for evaluation of the possible significance of this adipokine in intergenic interactions.
Methods. The silencing of NAMPT mRNA was introduced by NAMPT specific siRNA. The expression level of NAMPT, IGFBP3, IRS1, HK2, PER2, CLU, BNIP3, TPD52, GADD45A, and MKI67 genes was studied in U87 glioma cells by quantitative polymerase chain reaction. Anti-visfatin antibody was used for detection of NAMPT protein by Western-blot analysis.
Results. It was shown that the silencing of NAMPT mRNA led to a strong down-regulation of NAMPT protein and significant modification of the expression of IRS1, IGFBP3, CLU, HK2, BNIP3, and MKI67 genes in glioma cells and a strong up-regulation of IGFBP3 and IRS1 and down-regulation of CLU, BNIP3, HK2, and MKI67 gene expressions. At the same time, no significant changes were detected in the expression of GADD45A, PER2, and TPD52 genes in glioma cells treated by siRNA specific to NAMPT. Furthermore, the silencing of NAMPT mRNA suppressed the glioma cell proliferation.
Conclusions. Results of this investigation demonstrated that silencing of NAMPT mRNA with corresponding down-regulation of NAMPT protein and suppression of the glioma cell proliferation affected the expression of IRS1 gene as well as many other genes encoding the proliferation related proteins. It is possible that dysregulation of most of the studied genes in glioma cells after silencing of NAMPT is reflected by a complex of intergenic interactions and that NAMPT is an important factor for genome stability and regulatory mechanisms contributing to the control of glioma cell metabolism and proliferation.
Collapse
|
14
|
Li Z, Wang Y. Long non-coding RNA FTH1P3 promotes the metastasis and aggressiveness of non-small cell lung carcinoma by inducing epithelial-mesenchymal transition. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:3782-3790. [PMID: 31933766 PMCID: PMC6949746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 09/23/2019] [Indexed: 06/10/2023]
Abstract
Long non-coding RNAs (lncRNAs) ferritin heavy chain 1 pseudogene 3 (FTH1P3) has been suggested to act as an oncogene in many types of human malignancy, but its role in non-small cell lung carcinoma (NSCLC) remains unknown. This study aimed to characterize the biologic functions of FTH1P3 in NSCLC and illuminate its clinical significance. The expression levels of FTH1P3 in NSCLC tissues and cell lines were detected by quantitative real-time PCR assay. The relationship of FTH1P3 expression with clinicopathologic features was evaluated by chi-square test, and its correlation with prognosis of NSCLC patients was analyzed by Kaplan-Meier method with log-rank test. Wound healing and transwell invasion assays were applied to evaluate cell migration and invasion abilities, respectively. Western blotwas performed to detect the changes of epithelial-mesenchymal transition (EMT) related protein expression. The results showed that FTH1P3 was highly expressed in NSCLC tumor tissues and NSCLC-derived cell lines, and high expression of FTH1P3 was associated with advanced TNM stage and lymph node metastasis. NSCLC patients with high FTH1P3 expression had a poor overall survival relative to patients with low FTH1P3 expression. Through loss-of-function studies, FTH1P3 inhibition was demonstrated to suppress NSCLC cell migration and invasion in vitro. Notably, FTH1P3 knockdown could decrease expression of N-cadherin, vimentin and Snail protein of NSCLC cells, but promote E-cadherin protein expression, which was in accordance with its effect on cell migration and invasion. To sum up, our data demonstrated that FTH1P3 predicts a poor prognosis and promotes metastasis and aggressiveness in NSCLC by inducing EMT, suggesting FTH1P3 may be a promising target for gene therapy of NSCLC.
Collapse
Affiliation(s)
- Zhengxiong Li
- The Second School of Clinical Medicine, Southern Medical UniversityGuangzhou 510282, Guangdong Province, People’s Republic of China
- Department of Oncology, Shenzhen Longhua District Central HospitalShenzhen 518110, Guangdong Province, People’s Republic of China
- Department of Radiation Therapy, The Seventh Medical Center of PLA General HospitalBeijing 100700, People’s Republic of China
| | - Yadi Wang
- The Second School of Clinical Medicine, Southern Medical UniversityGuangzhou 510282, Guangdong Province, People’s Republic of China
- Department of Radiation Therapy, The Seventh Medical Center of PLA General HospitalBeijing 100700, People’s Republic of China
| |
Collapse
|
15
|
Chen J, Lou W, Ding B, Wang X. Overexpressed pseudogenes, DUXAP8 and DUXAP9, promote growth of renal cell carcinoma and serve as unfavorable prognostic biomarkers. Aging (Albany NY) 2019; 11:5666-5688. [PMID: 31409759 PMCID: PMC6710046 DOI: 10.18632/aging.102152] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 07/31/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND Growing studies have reported that pseudogenes play key roles in multiple human cancers. However, expression and roles of pseudogenes in renal cell carcinoma remains absent. RESULTS 31 upregulated and 16 downregulated pseudogenes were screened. Higher expression of DUXAP8 and DUXAP9 indicated poorer prognosis of kidney cancer. 33 and 5 miRNAs were predicted to potentially binding to DUXAP8 and DUXAP9, respectively. miR-29c-3p was identified as the most potential binding miRNAs of DUXAP8 and DUXAP9 based on expression, survival and correlation analyses. 254 target genes of miR-29c-3p were forecast. 47 hub genes with node degree >= 10 were identified. Subsequent analysis for the top 10 hub genes demonstrated that COL1A1 and COL1A2 may be two functional targets of DUXAP8 and DUXAP9. Expression of DUXAP8, DUXAP9, COL1A1 and COL1A2 were significantly increased in cancer samples compared to normal controls while miR-29c-3p expression was decreased. Luciferase reporter assay revealed that miR-29c-3p could directly bind to DUXAP8, DUXAP9, COL1A1 and COL1A2. Functional experiments showed that DUXAP8 and DUXAP9 enhanced but miR-29c-3p weakened growth of renal cell carcinoma. CONCLUSIONS In conclusion, upregulated DUXAP8 and DUXAP9 promote growth of renal cell carcinoma and serve as two promising prognostic biomarkers. METHODS Dysregulated pseudogenes were obtained by dreamBase and GEPIA. The binding miRNAs of pseudogene and targets of miRNA were predicted using starBase and miRNet. Kaplan-Meier plotter was utilized to perform survival analysis, and Enrichr database was introduced to conduct functional enrichment analysis. Hub genes were identified through STRING and Cytoscape. qRT-PCR, luciferase reporter assay, cell counting assay and colony formation assay were performed to validate in silico analytic results.
Collapse
Affiliation(s)
- Jing Chen
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Zhejiang Province, Hangzhou 313100, China.,First Affiliated Hospital of Jiaxing University, Zhejiang Province, Jiaxing 314000, China
| | - Weiyang Lou
- Department of Surgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 313100, China
| | - Bisha Ding
- Department of Surgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 313100, China
| | - Xian Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Zhejiang Province, Hangzhou 313100, China
| |
Collapse
|
16
|
Ma C, Shi X, Guo W, Niu J, Wang G. miR-107 Enhances the Sensitivity of Breast Cancer Cells to Paclitaxel. Open Med (Wars) 2019; 14:456-466. [PMID: 31206033 PMCID: PMC6555243 DOI: 10.1515/med-2019-0049] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 01/11/2019] [Indexed: 02/06/2023] Open
Abstract
Breast cancer remains the most commonly diagnosed cancer in Chinese women. Paclitaxel (PTX) is a chemotherapy medication used to treat breast cancer patients. However, a side effect of paclitaxel is the severe drug resistance. Previous studies demonstrated that dysregulation of microRNAs could regulate sensitivity to paclitaxel in breast cancer. Here, the present study aimed to lucubrate the underlying mechanisms of miR-107 in regulating the sensitivity of breast cancer cells to PTX. The results demonstrated that miR-107 was down-regulated in breast cancer tumor tissues, while TPD52 was significantly up-regulated compared with the non-tumor adjacent tissues. After confirming that TPD52 may be a major target of miR-107 via a dual-luciferase reporter assay, the western blot and RT-qPCR assays further demonstrated that miR-107 may reduce the expression level of TPD52 as well. In addition, miR-107 may prominently enhance PTX induced reduction of cell viability and the promotion of cell apoptosis in breast cancer, and the variation could be reversed by co-transfected with pcDNA3.1-TPD52. Finally, miR-107 could further reduce the decreased expression of TPD52, Wnt1, β-catenin and cyclin D1 that was induced by PTX in both mRNA and protein levels, which were rescued by pcDNA3.1-TPD52 indicating that miR-107 regulated breast cancer cell sensitivity to PTX may be targeting TPD52 through Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Changpo Ma
- Thoracic Surgery Department, Tianjin Baodi People's Hospital, Tianjin301800, China
| | - Xuejun Shi
- Thoracic Surgery Department, Tianjin Baodi People's Hospital, Tianjin301800, China
| | - Wenchao Guo
- Thoracic Surgery Department, Tianjin Baodi People's Hospital, Tianjin301800, China
| | - Jianxin Niu
- Thoracic Surgery Department, Tianjin Baodi People's Hospital, Tianjin301800, China
| | | |
Collapse
|
17
|
Huang G, Liu J, Yang C, Xiang Y, Wang Y, Wang J, Cao M, Yang W. RNA sequencing discloses the genome‑wide profile of long noncoding RNAs in dilated cardiomyopathy. Mol Med Rep 2019; 19:2569-2580. [PMID: 30720098 PMCID: PMC6423559 DOI: 10.3892/mmr.2019.9937] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 01/17/2019] [Indexed: 12/13/2022] Open
Abstract
Dilated cardiomyopathy (DCM) is a common type of non‑ischemic cardiomyopathy, of which the underlying mechanisms have not yet been fully elucidated. Long noncoding RNAs (lncRNAs) have been reported to serve crucial physiological roles in various cardiac diseases. However, the genome‑wide expression profile of lncRNAs remains to be elucidated in DCM. In the present study, a case‑control study was performed to identify expression deviations in circulating lncRNAs between patients with DCM and controls by RNA sequencing. Partial dysregulated lncRNAs were validated by reverse transcription‑polymerase chain reaction. Gene Ontology, Kyoto Encyclopedia of Genes and Genomes pathway, and lncRNA‑messenger RNA (mRNA) co‑expression network analyses were employed to probe potential functions of these dysregulated lncRNAs in DCM. Comparison between 8 DCM and 8 control samples demonstrated that there were alterations in the expression levels of 988 lncRNAs and 1,418 mRNAs in total. The dysregulated lncRNAs were found to be mainly associated with system development, organ morphogenesis and metabolic regulation in terms of 'biological processes'. Furthermore, the analysis revealed that the gap junction pathway, phagosome, and dilated and hypertrophic cardiomyopathy pathways may serve crucial roles in the development of DCM. The lncRNA‑mRNA co‑expression network also suggested that the target genes of the lncRNAs were different in patients with DCM as compared with those in the controls. In conclusion, the present study revealed the genome‑wide profile of circulating lncRNAs in DCM by RNA sequencing, and explored the potential functions of these lncRNAs in DCM using bioinformatics analysis. These findings provide a theoretical foundation for future studies of lncRNAs in DCM.
Collapse
Affiliation(s)
- Guangyong Huang
- Department of Cardiology, Liaocheng People's Hospital of Shandong University, Liaocheng, Shandong 252000, P.R. China
| | - Jingwen Liu
- Department of Cardiology, Liaocheng People's Hospital of Shandong University, Liaocheng, Shandong 252000, P.R. China
| | - Chuansheng Yang
- Department of Cardiology, Liaocheng People's Hospital of Shandong University, Liaocheng, Shandong 252000, P.R. China
| | - Youzhang Xiang
- Shandong Institute for Endemic Disease Control, Jinan, Shandong 250014, P.R. China
| | - Yuehai Wang
- Department of Cardiology, Liaocheng People's Hospital of Shandong University, Liaocheng, Shandong 252000, P.R. China
| | - Jing Wang
- Shandong Institute for Endemic Disease Control, Jinan, Shandong 250014, P.R. China
| | - Miaomiao Cao
- Department of Cardiology, Liaocheng People's Hospital of Shandong University, Liaocheng, Shandong 252000, P.R. China
| | - Wenbo Yang
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| |
Collapse
|