1
|
Razavinia A, Razavinia A, Jamshidi Khalife Lou R, Ghavami M, Shahri F, Tafazoli A, Khalesi B, Hashemi ZS, Khalili S. Exosomes as novel tools for renal cell carcinoma therapy, diagnosis, and prognosis. Heliyon 2024; 10:e32875. [PMID: 38948044 PMCID: PMC11211897 DOI: 10.1016/j.heliyon.2024.e32875] [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: 09/12/2023] [Revised: 03/06/2024] [Accepted: 06/11/2024] [Indexed: 07/02/2024] Open
Abstract
Background Renal Cell Carcinoma (RCC) stands as a formidable challenge within the field of oncology, despite considerable research endeavors. The advanced stages of this malignancy present formidable barriers to effective treatment and management. Objective This review aims to explore the potential of exosomes in addressing the diagnostic and therapeutic challenges associated with RCC. Specifically, it investigates the role of exosomes as biomarkers and therapeutic vehicles in the context of RCC management. Methods For this review article, a comprehensive literature search was conducted using databases such as PubMed, employing relevant keywords to identify research articles pertinent to the objectives of the review. Initially, 200 articles were identified, which underwent screening to remove duplicates and assess relevance based on titles and abstracts, followed by a detailed examination of full texts. From the selected articles, relevant data were extracted and synthesized to address the review's objectives. The conclusions were drawn based on a thorough analysis of the findings. The quality was ensured through independent review and resolution of discrepancies among multiple reviewers. Results Exosomes demonstrate potential as diagnostic tools for early detection, prognosis, and treatment monitoring in RCC. Their ability to deliver various therapeutic agents, such as small interfering RNAs, lncRNAs, chemotherapeutic drugs, and immune-stimulating agents, allows for a personalized approach to RCC management. By leveraging exosome-based technologies, precision and efficacy in treatment strategies can be significantly enhanced. Conclusion Despite the promising advancements enabled by exosomes in the management of RCC, further research is necessary to refine exosome-based technologies and validate their efficacy, safety, and long-term benefits through rigorous clinical trials. Embracing exosomes as integral components of RCC diagnosis and treatment represents a significant step towards improving patient outcomes and addressing the persistent challenges posed by this malignancy in the field of oncology.
Collapse
Affiliation(s)
- Amir Razavinia
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Abazar Razavinia
- Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Roya Jamshidi Khalife Lou
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mahlegha Ghavami
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS, Canada
| | - Forouzan Shahri
- Department of Chemistry, Faculty of Sciences, University of Guilan, Iran
| | - Aida Tafazoli
- Department of Bacterial and Virology, Shiraz medical school, Shiraz, Iran
| | - Bahman Khalesi
- Department of Research and Production of Poultry Viral Vaccine, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization, Karaj 3197619751, Iran
| | - Zahra Sadat Hashemi
- ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Saeed Khalili
- Department of Biology Sciences, Shahid Rajaee Teacher Training University, Tehran, Iran
| |
Collapse
|
2
|
Yang L, Zang Y, Liu P, Xing X, Mou Z. A two-layer circuit cascade-based DNA machine for highly sensitive miRNA imaging in living cells. Analyst 2024; 149:2925-2931. [PMID: 38587246 DOI: 10.1039/d4an00277f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Sensitive detection of microRNA (miRNA), one of the most promising biomarkers, plays crucial roles in cancer diagnosis. However, the low expression level of miRNA makes it extremely urgent to develop ultrasensitive and highly selective strategies for quantification of miRNA. Herein, a DNA machine is rationally constructed for amplified detection and imaging of low-abundance miRNA in living cells based on the toehold-mediated strand displacement reaction (TMSDR). The isothermal and enzyme-free DNA machine with low background leakage is fabricated by integrating two DNA circuits into a cascade system, in which the output of one circuit serves as the input of the other one. Once the DNA machine is transfected into breast cancer cells, the overexpressed miRNA-203 initiates the first-layer circuit through TMSDR, leading to the concentration variation of fuel strands, which further influences the assembly of hairpin DNA in the second-layer circuit and the occurrence of fluorescence resonance energy transfer (FRET) for fluorescence imaging. Benefiting from the cascade of the two-layer amplification reaction, the proposed DNA machine acquires a detection limit down to 4 fM for quantification of miR-203 and a 10 000-fold improvement in amplification efficiency over the single circuit. Therefore, the two-layer circuit cascade-based DNA machine provides an effective platform for amplified analysis of low-abundance miRNA with high sensitivity, which holds great promise in biomedical and clinical research.
Collapse
Affiliation(s)
- Lin Yang
- School of Chemical Engineering, Shandong Institute of Petroleum and Chemical Technology, Dongying 257061, P. R. China.
| | - Yan Zang
- School of Chemical Engineering, Shandong Institute of Petroleum and Chemical Technology, Dongying 257061, P. R. China.
| | - Peng Liu
- School of Chemical Engineering, Shandong Institute of Petroleum and Chemical Technology, Dongying 257061, P. R. China.
| | - Xin Xing
- School of Chemical Engineering, Shandong Institute of Petroleum and Chemical Technology, Dongying 257061, P. R. China.
| | - Zhenxin Mou
- School of Nursing, Shandong Shengli Vocational College, Dongying 257061, P. R. China.
| |
Collapse
|
3
|
Imran M, Abida, Eltaib L, Siddique MI, Kamal M, Asdaq SMB, Singla N, Al-Hajeili M, Alhakami FA, AlQarni AF, Abdulkhaliq AA, Rabaan AA. Beyond the genome: MALAT1's role in advancing urologic cancer care. Pathol Res Pract 2024; 256:155226. [PMID: 38452585 DOI: 10.1016/j.prp.2024.155226] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/07/2024] [Accepted: 02/21/2024] [Indexed: 03/09/2024]
Abstract
Urologic cancers (UCs), which include bladder, kidney, and prostate tumors, account for almost a quarter of all malignancies. Long non-coding RNAs (lncRNAs) are tissue-specific RNAs that influence cell growth, death, and division. LncRNAs are dysregulated in UCs, and their abnormal expression may allow them to be used in cancer detection, outlook, and therapy. With the identification of several novel lncRNAs and significant exploration of their functions in various illnesses, particularly cancer, the study of lncRNAs has evolved into a new obsession. MALAT1 is a flexible tumor regulator implicated in an array of biological activities and disorders, resulting in an important research issue. MALAT1 appears as a hotspot, having been linked to the dysregulation of cell communication, and is intimately linked to cancer genesis, advancement, and response to treatment. MALAT1 additionally operates as a competitive endogenous RNA, binding to microRNAs and resuming downstream mRNA transcription and operation. This regulatory system influences cell growth, apoptosis, motility, penetration, and cell cycle pausing. MALAT1's evaluation and prognosis significance are highlighted, with a thorough review of its manifestation levels in several UC situations and its association with clinicopathological markers. The investigation highlights MALAT1's adaptability as a possible treatment target, providing fresh ways for therapy in UCs as we integrate existing information The article not only gathers current knowledge on MALAT1's activities but also lays the groundwork for revolutionary advances in the treatment of UCs.
Collapse
Affiliation(s)
- Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia.
| | - Abida
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| | - Lina Eltaib
- Department of Pharmaceutics, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| | - Muhammad Irfan Siddique
- Department of Pharmaceutics, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| | - Mehnaz Kamal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | | | - Neelam Singla
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur 302017, India
| | - Marwan Al-Hajeili
- Department of Medicine, King Abdulaziz University, Jeddah 23624, Saudi Arabia
| | - Fatemah Abdulaziz Alhakami
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Jazan University, Saudi Arabia
| | - Ahmed Farhan AlQarni
- Histopathology Laboratory, Najran Armed Forces Hospital, Najran 66251, Saudi Arabia
| | - Altaf A Abdulkhaliq
- Department of Biochemistry, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Ali A Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran 31311, Saudi Arabia; College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; Department of Public Health and Nutrition, The University of Haripur, Haripur 22610, Pakistan
| |
Collapse
|
4
|
Ramanathan K, Fekadie M, Padmanabhan G, Gulilat H. Long noncoding RNA: An emerging diagnostic and therapeutic target in kidney diseases. Cell Biochem Funct 2024; 42:e3901. [PMID: 38100151 DOI: 10.1002/cbf.3901] [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: 09/01/2023] [Revised: 11/13/2023] [Accepted: 11/29/2023] [Indexed: 01/26/2024]
Abstract
Long noncoding RNAs (lncRNAs) have critical roles in the development of many diseases including kidney disease. An increasing number of studies have shown that lncRNAs are involved in kidney development and that their dysregulation can result in distinct disease processes, including acute kidney injury, chronic kidney disease, and renal cell carcinoma. Understanding the roles of lncRNAs in kidney disease may provide new diagnostic and therapeutic opportunities in the clinic. This review provides an overview of lncRNA characteristics, and biological function and discusses specific studies that provide insight into the function and potential application of lncRNAs in kidney disease treatment.
Collapse
Affiliation(s)
- Kumaresan Ramanathan
- Department of Biomedical Sciences, Faculty of Medical Sciences, Institute of Health, Jimma University, Jimma, Ethiopia
| | - Minale Fekadie
- Department of Biomedical Sciences, Faculty of Medical Sciences, Institute of Health, Jimma University, Jimma, Ethiopia
| | | | - Henok Gulilat
- Department of Biomedical Sciences, Faculty of Medical Sciences, Institute of Health, Jimma University, Jimma, Ethiopia
| |
Collapse
|
5
|
Anbiyaee O, Moalemnia A, Ghaedrahmati F, Shooshtari MK, Khoshnam SE, Kempisty B, Halili SA, Farzaneh M, Morenikeji OB. The functions of long non-coding RNA (lncRNA)-MALAT-1 in the pathogenesis of renal cell carcinoma. BMC Nephrol 2023; 24:380. [PMID: 38124072 PMCID: PMC10731893 DOI: 10.1186/s12882-023-03438-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023] Open
Abstract
Renal cell carcinoma (RCC), a prevalent form of renal malignancy, is distinguished by its proclivity for robust tumor proliferation and metastatic dissemination. Long non-coding RNAs (lncRNAs) have emerged as pivotal modulators of gene expression, exerting substantial influence over diverse biological processes, encompassing the intricate landscape of cancer development. Metastasis-associated lung adenocarcinoma transcript 1 (MALAT-1), an exemplar among lncRNAs, has been discovered to assume functional responsibilities within the context of RCC. The conspicuous expression of MALAT-1 in RCC cells has been closely linked to the advancement of tumors and an unfavorable prognosis. Experimental evidence has demonstrated the pronounced ability of MALAT-1 to stimulate RCC cell proliferation, migration, and invasion, thereby underscoring its active participation in facilitating the metastatic cascade. Furthermore, MALAT-1 has been implicated in orchestrating angiogenesis, an indispensable process for tumor expansion and metastatic dissemination, through its regulatory influence on pro-angiogenic factor expression. MALAT-1 has also been linked to the evasion of immune surveillance in RCC, as it can regulate the expression of immune checkpoint molecules and modulate the tumor microenvironment. Hence, the potential utility of MALAT-1 as a diagnostic and prognostic biomarker in RCC emerges, warranting further investigation and validation of its clinical significance. This comprehensive review provides an overview of the diverse functional roles exhibited by MALAT-1 in RCC.
Collapse
Affiliation(s)
- Omid Anbiyaee
- Cardiovascular Research Center, School of Medicine, Namazi Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Arash Moalemnia
- Faculty of Medicine, Dezful University of Medical Sciences, Dezful, Iran
| | - Farhoodeh Ghaedrahmati
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maryam Khombi Shooshtari
- Chronic Renal Failure Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyed Esmaeil Khoshnam
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Bartosz Kempisty
- Department of Human Morphology and Embryology Division of Anatomy, Wrocław Medical University, Wrocław, Poland
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University, Torun, Poland
- Physiology Graduate Faculty North, Carolina State University, Raleigh, NC, 27695, US
- Center of Assisted Reproduction Department of Obstetrics and Gynecology, University Hospital and Masaryk University, Brno, Czech Republic
| | - Shahla Ahmadi Halili
- Department of Internal Medicine, School of Science, Chronic Renal Failure Research Center, Ahvaz Jundishapur University of Medical Science, Ahvaz, Iran
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Olanrewaju B Morenikeji
- Division of Biological and Health Sciences, University of Pittsburgh at Bradford, Bradford, PA, USA.
| |
Collapse
|
6
|
Gao Y, Wan L, Li M, Wang B, Ma Y. NRF2/HO-1 axis, BIRC5, and TP53 expression in ESCC and its correlation with clinical pathological characteristics and prognosis. Int J Biol Markers 2023; 38:174-184. [PMID: 37312528 DOI: 10.1177/03936155231176571] [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] [Indexed: 06/15/2023]
Abstract
BACKGROUND Many types of cancer exhibit high nuclear factor erythroid 2-related factor 2 (NRF2), which is effective in resisting drugs and radiation. However, the role of NRF2 gene expression in predicting the prognosis of esophageal squamous cell carcinoma (ESCC) remains unclear. METHODS The association between NRF2, heme oxygenase-1 (HO-1), baculovirus IAP repeat 5 (BIRC5), P53 gene expression levels and their relationship to immune-infiltrating cells were assessed using the Cancer Genome Atlas dataset, the Human Protein Atlas and the TISDB database. The expression of NRF2, HO-1, BIRC5, and TP53 in 118 ESCC patients was detected by immunohistochemistry, and the relationship between their expression level and clinicopathological parameters and prognosis was analyzed. RESULTS In ESCC, NRF2 overexpression was significantly associated with Han ethnicity, lymph node metastasis, and distant metastasis. HO-1 overexpression was significantly associated with differentiation, advanced clinical staging, lymph node metastasis, nerve invasion, and distant metastasis. BIRC5 overexpression was significantly associated with Han ethnicity and lymph node metastasis. TP53 overexpression was significantly associated with Han ethnicity and T staging. The NRF2/HO-1 axis expression was positively correlated with BIRC5 and TP53. Kaplan-Meier and multivariate Cox regression analysis showed that NRF2, BIRC5, and TP53 genes co-expression was an independent prognostic risk factor. TISIDB dataset analysis showed that immune-infiltrating cells were significantly negatively correlated with NRF2 and BIRC5. CONCLUSION NRF2, BIRC5, and TP53 axis gene expressions are predictors of poor prognosis for ESCC. The overexpression of the NRF2/HO-1/BIRC5 axis may not be related to immune-infiltrating cells.
Collapse
Affiliation(s)
- Yongmei Gao
- Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Li Wan
- Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Mengyan Li
- Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Bo Wang
- Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yuqing Ma
- Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
7
|
Liu K, Gao Y, Zhang Q. Prognostic significance of MALAT1 in clear cell renal cell carcinoma based on TCGA and GEO. Medicine (Baltimore) 2023; 102:e35249. [PMID: 37713833 PMCID: PMC10508397 DOI: 10.1097/md.0000000000035249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/24/2023] [Indexed: 09/17/2023] Open
Abstract
Long noncoding RNAs metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) can regulate tumorigenesis and progression of various cancers. However, there is little known about the tumor biology and regulatory mechanism of MALAT1 in clear cell renal cell carcinoma (ccRCC). The objective of this study was to evaluate the prognostic value and potential functions of MALAT1 in ccRCC based on the cancer genome atlas. Through bioinformatics research, we analyzed the expression of MALAT1 in ccRCC, and the relationship with clinicopathological features, overall survival and infiltration of immune cells, and established the prognostic models. The results showed that MALAT1 was highly expressed in ccRCC tissues and predicted poor ccRCC patient outcome. The expression level of MALAT1 was significantly correlated with histologic grade, pathologic grade, T stage, M stage. ROC curve showed that MALAT1 had a good diagnostic accuracy, area under the curve of 0.752. The univariate and multivariate cox regression analysis showed that high MALAT1 expression was an independent prognostic factor for overall survival in the cancer genome atlas (hazard ratio = 2.271, 95% confidence interval: 1.435-3.593, P < .001). Gene set enrichment analysis revealed that MALAT1 expression was associated with the DNA methylation, epigenetic regulation of gene expression signaling pathway. In addition, the prognostic models were established to predict 1-, 3- and 5-year survival. This study showed that high expression of MALAT1 might be a potential diagnostic and prognostic biomarker.
Collapse
Affiliation(s)
- Kai Liu
- Department of Pathology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Yingxue Gao
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Quanwu Zhang
- Department of Pathology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| |
Collapse
|
8
|
Sweef O, Zaabout E, Bakheet A, Halawa M, Gad I, Akela M, Tousson E, Abdelghany A, Furuta S. Unraveling Therapeutic Opportunities and the Diagnostic Potential of microRNAs for Human Lung Cancer. Pharmaceutics 2023; 15:2061. [PMID: 37631277 PMCID: PMC10459057 DOI: 10.3390/pharmaceutics15082061] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/12/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023] Open
Abstract
Lung cancer is a major public health problem and a leading cause of cancer-related deaths worldwide. Despite advances in treatment options, the five-year survival rate for lung cancer patients remains low, emphasizing the urgent need for innovative diagnostic and therapeutic strategies. MicroRNAs (miRNAs) have emerged as potential biomarkers and therapeutic targets for lung cancer due to their crucial roles in regulating cell proliferation, differentiation, and apoptosis. For example, miR-34a and miR-150, once delivered to lung cancer via liposomes or nanoparticles, can inhibit tumor growth by downregulating critical cancer promoting genes. Conversely, miR-21 and miR-155, frequently overexpressed in lung cancer, are associated with increased cell proliferation, invasion, and chemotherapy resistance. In this review, we summarize the current knowledge of the roles of miRNAs in lung carcinogenesis, especially those induced by exposure to environmental pollutants, namely, arsenic and benzopyrene, which account for up to 1/10 of lung cancer cases. We then discuss the recent advances in miRNA-based cancer therapeutics and diagnostics. Such information will provide new insights into lung cancer pathogenesis and innovative diagnostic and therapeutic modalities based on miRNAs.
Collapse
Affiliation(s)
- Osama Sweef
- Division of Cancer Biology, Department of Medicine, MetroHealth Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH 44109, USA
- Department of Zoology, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Elsayed Zaabout
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ahmed Bakheet
- Division of Cancer Biology, Department of Medicine, MetroHealth Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH 44109, USA
| | - Mohamed Halawa
- Department of Pharmacology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Ibrahim Gad
- Department of Statistics and Mathematics, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Mohamed Akela
- Department of Biology, College of Science and Humanities in Al-Kharj, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Ehab Tousson
- Department of Zoology, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Ashraf Abdelghany
- Biomedical Research Center of University of Granada, Excellence Research Unit “Modeling Nature” (MNat), University of Granada, 18016 Granada, Spain
| | - Saori Furuta
- Division of Cancer Biology, Department of Medicine, MetroHealth Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH 44109, USA
| |
Collapse
|
9
|
Gan L, Xiao Q, Zhou Y, Fu Y, Tang M. Role of anoikis-related gene PLK1 in kidney renal papillary cell carcinoma: a bioinformatics analysis and preliminary verification on promoting proliferation and migration. Front Pharmacol 2023; 14:1211675. [PMID: 37456749 PMCID: PMC10339314 DOI: 10.3389/fphar.2023.1211675] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/20/2023] [Indexed: 07/18/2023] Open
Abstract
Background: Kidney renal papillary cell carcinoma (KIRP) is a rare malignancy with a very poor prognosis. Anoikis is a specific form of apoptosis involved in carcinogenesis, but the role of anoikis in KIRP has not been explored. Methods: Anoikis-related genes (ARGs) were obtained from the GeneCards database and Harmonizome database and were used to identify different subtypes of KIRP and construct a prognostic model of KIRP. In addition, we also explored the immune microenvironment and enrichment pathways among different subtypes by consensus clustering into different subtypes. Drug sensitivity analysis was used to screen for potential drugs. Finally, we verified the mRNA and protein expression of the independent prognostic gene PLK1 in patient tissues and various cells and further verified the changes in relevant prognostic functions after constructing a PLK1 stable knockdown model using ShRNA. Results: We identified 99 differentially expressed anoikis-related genes (DEGs) associated with KIRP survival, and selected 3 genes from them to construct a prognostic model, which can well predict the prognosis of KIRP patients. Consensus clustering divided KIRP into two subtypes, and there was a significant difference in survival rates between the two subtypes. Immune profiling revealed differing immune statuses between the two subtypes, and functional analysis reveals the differential activity of different functions in different subtypes. Drug sensitivity analysis screened out 15 highly sensitive drugs in the high-risk group and 11 highly sensitive drugs in the low-risk group. Univariate and multivariate Cox regression analysis confirmed that PLK1 was an independent prognostic factor in KIRP, and its mRNA and protein expression levels were consistent with gene differential expression levels, both of which were highly expressed in KIRP. Functional verification of PLK1 in KIRP revealed significant results. Specifically, silencing PLK1 inhibited cell proliferation, clonogenicity, and migration, which indicated that PLK1 plays an important role in the proliferation and migration of KIRP. Conclusion: The prognosis model constructed by ARGs in this study can accurately predict the prognosis of KIRP patients. ARGs, especially PLK1, play an important role in the development of KIRP. This research can help doctors provide individualized treatment plans for KIRP patients and provide researchers with new research ideas.
Collapse
Affiliation(s)
- Li Gan
- Department of Anesthesiology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Qiyu Xiao
- Department of Nuclear Medicine, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Yusong Zhou
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Ying Fu
- Department of Nuclear Medicine, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Mengjie Tang
- Department of Pathology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
10
|
Liu Y, Cheng X, Xi P, Zhang Z, Sun T, Gong B. Bioinformatic analysis highlights SNHG6 as a putative prognostic biomarker for kidney renal papillary cell carcinoma. BMC Urol 2023; 23:54. [PMID: 37004005 PMCID: PMC10067223 DOI: 10.1186/s12894-023-01218-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 03/20/2023] [Indexed: 04/03/2023] Open
Abstract
PURPOSE Kidney renal papillary cell carcinoma (KIRP) is a highly heterogeneous malignancy and current systemic therapeutic strategies are difficult to achieve a satisfactory outcome for advanced disease. Meanwhile, there is a lack of effective biomarkers to predict the prognosis of KIRP. METHODS Using TCGA, GTEx, UALCAN, TIMER, TIMER 2.0 and STRING databases, we analyzed the relationship of SNHG6 with KIRP subtypes, tumor-infiltrating immune cells and potential target mRNAs. Based on TCGA data, ROC curves, Kaplan-Meier survival analysis and COX regression analysis were performed to evaluate the diagnostic and prognostic value of SNHG6 in KIRP. Nomogram was used to predict 3- and 5-year disease-specific survival in KIRP patients. In addition, with the help of Genetic ontology and Gene set enrichment analysis, the biological processes and signalling pathways that SNHG6 may be involved in KIRP were initially explored. RESULTS In patients with KIRP, SNHG6 was significantly upregulated and associated with a more aggressive subtype (lymph node involvement, pathological stage IV, CIMP phenotype) and poor prognosis. The ROC curve showed good diagnostic efficacy (AUC value: 0.828) and the C-index of the Nomogram for predicting DSS at 3 and 5 years was 0.920 (0.898-0.941). In the immune microenvironment of KIRP, SNHG6 expression levels were negatively correlated with macrophage abundance and positively correlated with cancer-associated fibroblasts. Furthermore, SNHG6 may promote KIRP progression by regulating the expression of molecules such as AURKB, NDC80, UBE2C, NUF2, PTTG1, CENPH, SPC25, CDCA3, CENPM, BIRC5, TROAP, EZH2. Last, GSEA suggests that SNHG6 may be involved in the regulation of the PPAR signalling pathway and the SLIT/ROBO signalling pathway. CONCLUSIONS Our analysis suggests that a high SNHG6 expression status in KIRP is associated with a poorer prognosis for patients, and also elucidates some potential mechanisms contributing to this poorer outcome. This may provide new insights into the treatment and management of KIRP in the foreseeable future.
Collapse
Affiliation(s)
- Yifu Liu
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
- Jiangxi Institute of Urology, Nanchang, 330006, Jiangxi, China
| | - Xiaofeng Cheng
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
- Jiangxi Institute of Urology, Nanchang, 330006, Jiangxi, China
| | - Ping Xi
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
- Jiangxi Institute of Urology, Nanchang, 330006, Jiangxi, China
| | - Zhicheng Zhang
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
- Jiangxi Institute of Urology, Nanchang, 330006, Jiangxi, China
| | - Ting Sun
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
- Jiangxi Institute of Urology, Nanchang, 330006, Jiangxi, China
| | - Binbin Gong
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China.
- Jiangxi Institute of Urology, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
11
|
Emerging RNA-Based Therapeutic and Diagnostic Options: Recent Advances and Future Challenges in Genitourinary Cancers. Int J Mol Sci 2023; 24:ijms24054601. [PMID: 36902032 PMCID: PMC10003365 DOI: 10.3390/ijms24054601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 02/15/2023] [Accepted: 02/23/2023] [Indexed: 03/02/2023] Open
Abstract
Renal cell carcinoma, bladder cancer, and prostate cancer are the most widespread genitourinary tumors. Their treatment and diagnosis have significantly evolved over recent years, due to an increasing understanding of oncogenic factors and the molecular mechanisms involved. Using sophisticated genome sequencing technologies, the non-coding RNAs, such as microRNAs, long non-coding RNAs, and circular RNAs, have all been implicated in the occurrence and progression of genitourinary cancers. Interestingly, DNA, protein, and RNA interactions with lncRNAs and other biological macromolecules drive some of these cancer phenotypes. Studies on the molecular mechanisms of lncRNAs have identified new functional markers that could be potentially useful as biomarkers for effective diagnosis and/or as targets for therapeutic intervention. This review focuses on the mechanisms underlying abnormal lncRNA expression in genitourinary tumors and discusses their role in diagnostics, prognosis, and treatment.
Collapse
|
12
|
Zhang Q, Ren H, Ge L, Zhang W, Song F, Huang P. A review on the role of long non-coding RNA and microRNA network in clear cell renal cell carcinoma and its tumor microenvironment. Cancer Cell Int 2023; 23:16. [PMID: 36732762 PMCID: PMC9893571 DOI: 10.1186/s12935-023-02861-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 01/28/2023] [Indexed: 02/04/2023] Open
Abstract
Renal cell carcinoma (RCC) is the second lethal urogenital malignancy with the increasing incidence and mortality in the world. Clear cell renal cell carcinoma (ccRCC) is one major subtype of RCC, which accounts for about 70 to 80% of all RCC cases. Although many innovative therapeutic options have emerged during the last few decades, the efficacy of these treatments for ccRCC patients is very limited. To date, the prognosis of patients with advanced or metastatic ccRCC is still poor. The 5-year survival rate of these patients remains less than 10%, which mainly attributes to the complexity and heterogeneity of the tumor microenvironment (TME). It has been demonstrated that long non-coding RNAs (lncRNAs) perform an indispensable role in the initiation and progression of various tumors. They mostly function as sponges for microRNAs (miRNAs) to regulate the expression of target genes, finally influence the growth, metastasis, apoptosis, drug resistance and TME of tumor cells. However, the role of lncRNA/miRNA/mRNA axis in the TME of ccRCC remains poorly understood. In this review, we summarized the biological function of lncRNA/miRNA/mRNA axis in the pathogenesis of ccRCC, then discussed how lncRNA/miRNA/mRNA axis regulate the TME, finally highlighted their potential application as novel biomarkers and therapeutic targets for ccRCC.
Collapse
Affiliation(s)
- Qi Zhang
- grid.469325.f0000 0004 1761 325XDepartment of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China ,Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Hao Ren
- grid.469325.f0000 0004 1761 325XDepartment of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China ,Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Luqi Ge
- grid.469325.f0000 0004 1761 325XDepartment of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China ,Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Wen Zhang
- grid.469325.f0000 0004 1761 325XDepartment of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Feifeng Song
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China ,Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China
| | - Ping Huang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China ,Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China
| |
Collapse
|
13
|
Yao H, Lyu F, Ma J, Sun F, Tang G, Wu J, Zhou Z. PIMREG is a prognostic biomarker involved in immune microenvironment of clear cell renal cell carcinoma and associated with the transition from G1 phase to S phase. Front Oncol 2023; 13:1035321. [PMID: 36776322 PMCID: PMC9909346 DOI: 10.3389/fonc.2023.1035321] [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: 09/02/2022] [Accepted: 01/12/2023] [Indexed: 01/28/2023] Open
Abstract
Background Clear cell renal cell carcinoma (ccRCC) is one of the most common tumors in the world and affects human health seriously. PIMREG is a mitotic regulator which is essential to the metaphase-to-anaphase transition in cell cycle. Although PIMREG plays a crucial role in the malignant progression of tumors, there are few reports on its role in ccRCC. Methods The transcriptional expression profile and clinical data of PIMREG were downloaded from TCGA database and verified by qRT-PCR. Kaplan-Meier plotter was used to analyze the effect of PIMREG on overall survival (OS), disease specific survival (DSS) and progression-free interval (PFI) of patients with ccRCC. Univariable and multivariable Cox regression analysis were used to determine the independent prognostic factors of ccRCC. The effects of PIMREG on cell migration and invasion were detected by wound healing assay and transwell invasion assay, and CCK-8 assay, colony formation assay and cell cycle assay were used to detect the effect of PIMREG on cell proliferation. In addition, the changes in cell cycle related proteins were detected by western blot. Results PIMREG was highly expressed in human ccRCC and was positively correlated with pathologic stage, TNM stage and histologic grade. In addition, patients with high expression of PIMREG had a poor prognosis. Univariable and multivariable Cox regression analysis identified that PIMREG was an independent prognostic factor of ccRCC. Additionally, PIMREG was also closely related to immune cell infiltration. Experiments in vitro identified that the knockdown of PIMREG could significantly inhibit the proliferation, migration and invasion abilities of ccRCC. The expression of cyclin D1, CDK4 and CDK6 was also significantly reduced after PIMREG knockdown. Conclusions PIMREG plays a vital role in the development of ccRCC and may become a potential therapeutic target in the future.
Collapse
Affiliation(s)
- Huibao Yao
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Feifei Lyu
- Department of Traditional Chinese Medicine, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Jian Ma
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Fengze Sun
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Gonglin Tang
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Jitao Wu
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China,*Correspondence: Zhongbao Zhou, ; Jitao Wu,
| | - Zhongbao Zhou
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China,Department of Urology, Beijing TianTan Hospital, Capital Medical University, Beijing, China,*Correspondence: Zhongbao Zhou, ; Jitao Wu,
| |
Collapse
|
14
|
Li J, Wang N, Wen X, Huang LY, Cui RQ, Zhang J. Serum miRNA-203 as a Novel Biomarker for the Early Prediction of Acute ST-elevation Myocardial Infarction. J Cardiovasc Transl Res 2022; 15:1406-1413. [PMID: 35507256 DOI: 10.1007/s12265-022-10269-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 04/27/2022] [Indexed: 12/16/2022]
Abstract
Existing markers of myocardial infarction have limited diagnostic value for infarction, so it is necessary to identify new markers of infarction. To study the predictive value of serum miRNA-203 for acute ST-elevation myocardial infarction. Seventy patients with STEMI who were diagnosed in Hefei Second People's Hospital from December 2020 to December 2021 were selected, and 35 patients with transient chest pain who were hospitalized for other diseases in the Cardiology Department of our hospital during the same period were selected as the control group. The sera of the two groups of patients were collected, and a miRNA-203 semiquantitative experiment was performed. The miRNA-203 level in the STEMI group was higher than that in the control group. The AUC area of miRNA-203 in predicting STEMI was 0.912. Logistic regression analysis showed that miRNA-203 and white blood cell counts were independent risk factors for STEMI (P<0.05), and their ORs (95% CI) were 3.913 (1.574-9.728) and 2.13 (1.247-3.641), respectively. The present study reveals that miRNA-203 could be a possible candidate for a novel biomarker in the early prediction of STEMI.
Collapse
Affiliation(s)
- Jun Li
- Department of Cardiology, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, 230011, Anhui, China
- Anhui Medical University, Hefei, 230000, Anhui, China
| | - Na Wang
- Department of Obstetrics and Gynecology, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, 230011, Anhui, China
- Bengbu Medical College, Bengbu, 233000, Anhui, China
| | - Xiang Wen
- Department of Cardiology, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, 230011, Anhui, China
| | - Lu-Yao Huang
- Department of Cardiology, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, 230011, Anhui, China
| | - Rui-Qing Cui
- Department of Cardiology, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, 230011, Anhui, China
| | - Jing Zhang
- Department of Cardiology, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, 230011, Anhui, China.
| |
Collapse
|
15
|
Chen L, Shen M. LncRNA VPS9D1-AS1 Sponging miR-520a-5p Contributes to the Development of Uterine Corpus Endometrial Carcinoma by Enhancing BIRC5 Expression. Mol Biotechnol 2022; 64:1328-1339. [PMID: 35619019 DOI: 10.1007/s12033-022-00510-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 05/09/2022] [Indexed: 12/31/2022]
Abstract
The pattern of VPS9D1-AS1 expression and its effects on uterine corpus endometrial carcinoma (UCEC) remained unclear. VPS9D1-AS1, miR-520a-5p, and BIRC5 mRNA levels were quantified by qRT-PCR. Bax, Bcl-2, N-cadherin, E-cadherin, and BIRC5 protein levels were analyzed through western blotting. Cell migration, invasion, proliferation, as well as apoptosis of cells were checked after performing assay for wound-healing, Transwell, cell-counting kit-8 (CCK-8) assay, and western blotting. VPS9D1-AS1 effects on UCEC were observed in nude mice. Through bioinformatics tools, we analyzed the association present among miR-520a-5p, BIRC5, and VPS9D1-AS1 along with RNA immunoprecipitation, and Dual-Luciferase verification reporter analysis. Our findings suggested VPS9D1-AS1 gene expression was up-regulated in both tissues as well as cells of UCEC. VPS9D1-AS1 knockdown suppressed migration, invasion, epithelial-mesenchymal transition (EMT) along with proliferation of UCEC cells, caused in vitro cell apoptosis initiation, and in vivo reduction of tumor growth. Mechanistically, it was verified that VPS9D1-AS1 targeted BIRC5 and caused miR-520a-5p sponging. Inhibitor of miR-520-5p markedly reversed the anti-tumor effects of VPS9D1-AS1 knockdown or BIRC5 knockdown on UCEC progression. Our studies revealed that VPS9D1-AS1 contributed to the UCEC development and progression by binding to miR-520a-5p competitively and inducing BIRC5 expression, indicating that VPS9D1-AS1 might act as a therapeutic target to develop new therapies for UCEC patients.
Collapse
Affiliation(s)
- Lu Chen
- Gynaecology and Obstetrics, The Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan University, No. 168, Hongkong Road, Jiang'an District, Wuhan, 430015, Hubei, China
| | - Meng Shen
- Gynaecology and Obstetrics, The Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan University, No. 168, Hongkong Road, Jiang'an District, Wuhan, 430015, Hubei, China.
| |
Collapse
|
16
|
Chen D, Zhang C, Zang Y, Wang W, Zhang J. Identification of an immune-related gene prognostic index for predicting survival and immunotherapy efficacy in papillary renal cell carcinoma. Front Genet 2022; 13:970900. [PMID: 36159976 PMCID: PMC9499392 DOI: 10.3389/fgene.2022.970900] [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: 06/16/2022] [Accepted: 07/07/2022] [Indexed: 12/02/2022] Open
Abstract
Despite considerable progress has been made in the understanding of the genetics and molecular biology of renal cell carcinoma (RCC), therapeutic options of patients with papillary renal cell carcinoma (PRCC) are limited. Immunotherapy based on immune checkpoint inhibitors (ICIs) has become a hot point in researching new drug for tumor and been tested in a number of human clinical trials. In this study, an immune-related gene prognostic index (IRGPI) was developed and provided a comprehensive and systematic analysis of distinct phenotypic and molecular portraits in the recognition, surveillance, and prognosis of PRCC. The reliability of the IRGPI was evaluated using independent datasets from GEO database and the expression levels of the genes in the IRGPI detected by real-time PCR. Collectively, the currently established IRGPI could be used as a potential biomarker to evaluate the response and efficacy of immunotherapy in PRCC.
Collapse
Affiliation(s)
- Dongshan Chen
- Department of Urology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
- Department of Urology, Tongji Hospital, Tongji Medical College, Wuhan, China
- Department of Urology, Beijing Chaoyang Hospital Affiliated Capital Medical University, Beijing, China
| | - Chen Zhang
- School of Life Science and Engineering, Handan University, Handan, China
| | - Yuanwei Zang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Wei Wang
- Department of Urology, Beijing Chaoyang Hospital Affiliated Capital Medical University, Beijing, China
| | - Jiandong Zhang
- Department of Urology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
- Department of Urology, Tongji Hospital, Tongji Medical College, Wuhan, China
- Department of Urology, Beijing Chaoyang Hospital Affiliated Capital Medical University, Beijing, China
| |
Collapse
|
17
|
Matuszyk J. MALAT1-miRNAs network regulate thymidylate synthase and affect 5FU-based chemotherapy. Mol Med 2022; 28:89. [PMID: 35922756 PMCID: PMC9351108 DOI: 10.1186/s10020-022-00516-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/22/2022] [Indexed: 12/12/2022] Open
Abstract
Background The active metabolite of 5-Fluorouracil (5FU), used in the treatment of several types of cancer, acts by inhibiting the thymidylate synthase encoded by the TYMS gene, which catalyzes the rate-limiting step in DNA replication. The major failure of 5FU-based cancer therapy is the development of drug resistance. High levels of TYMS-encoded protein in cancerous tissues are predictive of poor response to 5FU treatment. Expression of TYMS is regulated by various mechanisms, including involving non-coding RNAs, both miRNAs and long non-coding RNAs (lncRNAs). Aim To delineate the miRNAs and lncRNAs network regulating the level of TYMS-encoded protein. Main body Several miRNAs targeting TYMS mRNA have been identified in colon cancers, the levels of which can be regulated to varying degrees by lncRNAs. Due to their regulation by the MALAT1 lncRNA, these miRNAs can be divided into three groups: (1) miR-197-3p, miR-203a-3p, miR-375-3p which are downregulated by MALAT1 as confirmed experimentally and the levels of these miRNAs are actually reduced in colon and gastric cancers; (2) miR-140-3p, miR-330-3p that could potentially interact with MALAT1, but not yet supported by experimental results; (3) miR-192-5p, miR-215-5p whose seed sequences do not recognize complementary response elements within MALAT1. Considering the putative MALAT1-miRNAs interaction network, attention is drawn to the potential positive feedback loop causing increased expression of MALAT1 in colon cancer and hepatocellular carcinoma, where YAP1 acts as a transcriptional co-factor which, by binding to the TCF4 transcription factor/ β-catenin complex, may increase the activation of the MALAT1 gene whereas the MALAT1 lncRNA can inhibit miR-375-3p which in turn targets YAP1 mRNA. Conclusion The network of non-coding RNAs may reduce the sensitivity of cancer cells to 5FU treatment by upregulating the level of thymidylate synthase.
Collapse
Affiliation(s)
- Janusz Matuszyk
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 12 R. Weigla Street, 53-114, Wroclaw, Poland.
| |
Collapse
|
18
|
Wang D, Zhu X, Siqin B, Ren C, Yi F. Long non-coding RNA CYTOR modulates cancer progression through miR-136-5p/MAT2B axis in renal cell carcinoma. Toxicol Appl Pharmacol 2022; 447:116067. [PMID: 35597301 DOI: 10.1016/j.taap.2022.116067] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 04/30/2022] [Accepted: 05/13/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND To explore the role of long noncoding RNAs (lncRNAs) cytoskeleton regulator RNA (CYTOR) in renal cell carcinoma (RCC). METHODS The levels of CYTOR in RCC tissues and cell lines were detected by RT-qPCR. 786-O and Caki-1 cells were transfected with CYTOR-shRNA or pcDNA-CYTOR respectively, or co-transfected with CYTOR-shRNA and miR-136-5p inhibitor, or co-transfected with miR-136-5p mimic and pcDNA-MAT2B. MTT assay, Transwell assay and flow cytometry were used to evaluate cell proliferation, invasion and apoptosis. The relationship between lncRNA CYTOR and miRNA-136-5p was detected by dual luciferase reporter gene and RNA pull down assays, and the targeted relationship between miRNA-136-5p and MAT2B was verified by dual luciferase reporter gene assay. The interaction between MAT2B and BAG3 protein was verified by co-IP experiment. The role of lncRNA CYTOR in vivo was also examined. RESULTS LncRNA CYTOR was up-regulated in RCC tissues and cell lines, and miR-136-5p was down-regulated in renal carcinoma cell lines and tissues. Downregulation of CYTOR inhibited cell proliferation and invasion and promoted apoptosis. miR-136-5p was sponged by lncRNA CYTOR, which negatively regulated the development of RCC. MAT2B was a target gene of miR-136-5p. MAT2B protein interacted directly with BAG3 protein to affect the proliferation, invasion and apoptosis of RCC cells. In vivo experiments showed that the expression level of miR-136-5p was increased, and MAT2B expression was decreased after CYTOR knockdown, thereby inhibiting the development of RCC. CONCLUSIONS LncRNA CYTOR promoted the progression of RCC by targeting miR-136-5p to regulate the target gene MAT2B, which interacted with BAG3 protein.
Collapse
Affiliation(s)
- Dan Wang
- Department of Urology Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China
| | - Xiaojun Zhu
- Department of Urology Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China
| | - Buhe Siqin
- Department of Urology Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China
| | - Chao Ren
- Department of Urology Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China
| | - Faxian Yi
- Department of Urology Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China.
| |
Collapse
|
19
|
Wang Y, Li X, Wang H, Zhang G. CircCAMSAP1 promotes non-small cell lung cancer proliferation and inhibits cell apoptosis by sponging miR-1182 and regulating BIRC5. Bioengineered 2022; 13:2428-2439. [PMID: 35132928 PMCID: PMC8974160 DOI: 10.1080/21655979.2021.2011639] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Recently, various studies have suggested that circular RNAs (circRNAs) are ubiquitous in various malignant events, including non-small cell lung cancer (NSCLC) and are closely related to cell proliferation and apoptosis. Unfortunately, the molecular functions involved in this action still have little overlap. Therefore, this study aimed to identify a novel circCAMSAP1 role in NSCLC. Overexpression of circCAMSAP1 has been demonstrated in NSCLC lung tissues and cell lines. Sequencing and RNase R experiments were planned to determine whether circCAMSAP1 is looped and exists in NSCLC. We also found that downregulated circCAMSAP1 repressed cell proliferation and increased apoptosis of NSCLC cells in vitro and suppressed xenograft tumor growth in vivo. Furthermore, a luciferase assay revealed that circCAMSAP1 could regulate baculoviral inhibitor of apoptosis protein (IAP) repeat containing 5 (BIRC5, also known as survivin) expression by directly binding to miR-1182. However, BIRC5 without 3ʹ untranslated regions (3ʹUTR) could reverse the influence of downregulated circCAMSAP1 on proliferation and apoptosis in NSCLC. Together, our findings reveal a novel mechanism by which the circCAMSAP1/miR-1182/BIRC5 axis promotes NSCLC progression.
Collapse
Affiliation(s)
- Yunfei Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaobo Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Huaqi Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Guojun Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
20
|
Wang Z, Yang X, Gui S, Yang F, Cao Z, Cheng R, Xia X, Li C. The Roles and Mechanisms of lncRNAs in Liver Fibrosis. Front Pharmacol 2021; 12:779606. [PMID: 34899344 PMCID: PMC8652206 DOI: 10.3389/fphar.2021.779606] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 11/02/2021] [Indexed: 12/12/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) can potentially regulate all aspects of cellular activity including differentiation and development, metabolism, proliferation, apoptosis, and activation, and benefited from advances in transcriptomic and genomic research techniques and database management technologies, its functions and mechanisms in physiological and pathological states have been widely reported. Liver fibrosis is typically characterized by a reversible wound healing response, often accompanied by an excessive accumulation of extracellular matrix. In recent years, a range of lncRNAs have been investigated and found to be involved in several cellular-level regulatory processes as competing endogenous RNAs (ceRNAs) that play an important role in the development of liver fibrosis. A variety of lncRNAs have also been shown to contribute to the altered cell cycle, proliferation profile associated with the accelerated development of liver fibrosis. This review aims to discuss the functions and mechanisms of lncRNAs in the development and regression of liver fibrosis, to explore the major lncRNAs involved in the signaling pathways regulating liver fibrosis, to elucidate the mechanisms mediated by lncRNA dysregulation and to provide new diagnostic and therapeutic strategies for liver fibrosis.
Collapse
Affiliation(s)
- Zhifa Wang
- Department of Rehabilitation Medicine, Chaohu Hospital of Anhui Medical University, Hefei Anhui, China
| | - Xiaoke Yang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Siyu Gui
- Department of Ophthalmology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fan Yang
- The First Clinical Medical College, Anhui Medical University, Hefei, China
| | - Zhuo Cao
- The First Clinical Medical College, Anhui Medical University, Hefei, China
| | - Rong Cheng
- Department of Gastroenterology, Anhui Provincial Children's Hospital, Hefei, China
| | - Xiaowei Xia
- Department of Gastroenterology, Anhui Provincial Children's Hospital, Hefei, China
| | - Chuanying Li
- Department of Gastroenterology, Anhui Provincial Children's Hospital, Hefei, China
| |
Collapse
|
21
|
Shi Y, Liu JB, Deng J, Zou DZ, Wu JJ, Cao YH, Yin J, Ma YS, Da F, Li W. The role of ceRNA-mediated diagnosis and therapy in hepatocellular carcinoma. Hereditas 2021; 158:44. [PMID: 34758879 PMCID: PMC8582193 DOI: 10.1186/s41065-021-00208-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 10/12/2021] [Indexed: 01/27/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related death worldwide due to its high degree of malignancy, high incidence, and low survival rate. However, the underlying mechanisms of hepatocarcinogenesis remain unclear. Long non coding RNA (lncRNA) has been shown as a novel type of RNA. lncRNA by acting as ceRNA can participate in various biological processes of HCC cells, such as tumor cell proliferation, migration, invasion, apoptosis and drug resistance by regulating downstream target gene expression and cancer-related signaling pathways. Meanwhile, lncRNA can predict the efficacy of treatment strategies for HCC and serve as a potential target for the diagnosis and treatment of HCC. Therefore, lncRNA serving as ceRNA may become a vital candidate biomarker for clinical diagnosis and treatment. In this review, the epidemiology of HCC, including morbidity, mortality, regional distribution, risk factors, and current treatment advances, was briefly discussed, and some biological functions of lncRNA in HCC were summarized with emphasis on the molecular mechanism and clinical application of lncRNA-mediated ceRNA regulatory network in HCC. This paper can contribute to the better understanding of the mechanism of the influence of lncRNA-mediated ceRNA networks (ceRNETs) on HCC and provide directions and strategies for future studies.
Collapse
Affiliation(s)
- Yi Shi
- College of Life Sciences and Chemistry, Hunan University of Technology, Zhuzhou, 412007, Hunan, China.,Cancer Institute, Affiliated Tumor Hospital of Nantong University, Nantong, 226631, China.,National Engineering Laboratory for Deep Process of Rice and Byproducts, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, 410004, Hunan, China
| | - Ji-Bin Liu
- Cancer Institute, Affiliated Tumor Hospital of Nantong University, Nantong, 226631, China
| | - Jing Deng
- National Engineering Laboratory for Deep Process of Rice and Byproducts, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, 410004, Hunan, China
| | - Da-Zhi Zou
- Department of Spine Surgery, Longhui County People's Hospital, Longhui, 422200, Hunan, China
| | - Jian-Jun Wu
- Nantong Haimen Yuelai Health Centre, Haimen, 226100, China
| | - Ya-Hong Cao
- Department of Respiratory, Nantong Traditional Chinese Medicine Hospital, Nantong, 226019, Jiangsu Province, China
| | - Jie Yin
- Department of General Surgery, Haian people's Hospital, Haian, 226600, Jiangsu, China
| | - Yu-Shui Ma
- Cancer Institute, Affiliated Tumor Hospital of Nantong University, Nantong, 226631, China.
| | - Fu Da
- Cancer Institute, Affiliated Tumor Hospital of Nantong University, Nantong, 226631, China. .,National Engineering Laboratory for Deep Process of Rice and Byproducts, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, 410004, Hunan, China.
| | - Wen Li
- College of Life Sciences and Chemistry, Hunan University of Technology, Zhuzhou, 412007, Hunan, China. .,National Engineering Laboratory for Deep Process of Rice and Byproducts, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, 410004, Hunan, China.
| |
Collapse
|
22
|
Chu Q, Gu X, Zheng Q, Guo Z, Shan D, Wang J, Zhu H. Long noncoding RNA SNHG4: a novel target in human diseases. Cancer Cell Int 2021; 21:583. [PMID: 34717631 PMCID: PMC8557547 DOI: 10.1186/s12935-021-02292-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 10/21/2021] [Indexed: 12/21/2022] Open
Abstract
Recently, long noncoding RNAs (lncRNAs) have attracted great attention from researchers. LncRNAs are non-protein-coding RNAs of more than 200 nucleotides in length. Multiple studies have been published on the relationship between lncRNA expression and the progression of human diseases. LncRNA small nucleolar RNA host gene 4 (SNHG4), a member of the lncRNA SNHG family, is abnormally expressed in a variety of human diseases, including gastric cancer, renal cell carcinoma, glioblastoma, neuroblastoma, prostate cancer, colorectal cancer, osteosarcoma, cervical cancer, liver cancer, lung cancer, non-small-cell lung cancer, neonatal pneumonia, diabetic retinopathy, neuropathic pain, acute cerebral infarction, acute myeloid leukaemia, and endometriosis. In this paper, the structure of SNHG4 is first introduced, and then studies in humans, animal models and cells are summarized to highlight the expression and function of SNHG4 in the above diseases. In addition, the specific mechanism of SNHG4 as a competing endogenous RNA (ceRNA) is discussed. The findings indicate that SNHG4 can be used as a biomarker for disease prognosis evaluation and as a potential target for disease diagnosis and treatment.
Collapse
Affiliation(s)
- Qingfei Chu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, NO. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Xinyu Gu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, NO. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Qiuxian Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, NO. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Zixuan Guo
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, NO. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Dandan Shan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, NO. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Jing Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, NO. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Haihong Zhu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, NO. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China.
| |
Collapse
|
23
|
LncRNAs in the Regulation of Genes and Signaling Pathways through miRNA-Mediated and Other Mechanisms in Clear Cell Renal Cell Carcinoma. Int J Mol Sci 2021; 22:ijms222011193. [PMID: 34681854 PMCID: PMC8539140 DOI: 10.3390/ijms222011193] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/05/2021] [Accepted: 10/08/2021] [Indexed: 12/13/2022] Open
Abstract
The fundamental novelty in the pathogenesis of renal cell carcinoma (RCC) was discovered as a result of the recent identification of the role of long non-coding RNAs (lncRNAs). Here, we discuss several mechanisms for the dysregulation of the expression of protein-coding genes initiated by lncRNAs in the most common and aggressive type of kidney cancer-clear cell RCC (ccRCC). A model of competitive endogenous RNA (ceRNA) is considered, in which lncRNA acts on genes through the lncRNA/miRNA/mRNA axis. For the most studied oncogenic lncRNAs, such as HOTAIR, MALAT1, and TUG1, several regulatory axes were identified in ccRCC, demonstrating a number of sites for various miRNAs. Interestingly, the LINC00973/miR-7109/Siglec-15 axis represents a novel agent that can suppress the immune response in patients with ccRCC, serving as a valuable target in addition to the PD1/PD-L1 pathway. Other mechanisms of action of lncRNAs in ccRCC, involving direct binding with proteins, mRNAs, and genes/DNA, are also considered. Our review briefly highlights methods by which various mechanisms of action of lncRNAs were verified. We pay special attention to protein targets and signaling pathways with which lncRNAs are associated in ccRCC. Thus, these new data on the different mechanisms of lncRNA functioning provide a novel basis for understanding the pathogenesis of ccRCC and the identification of new prognostic markers and targets for therapy.
Collapse
|
24
|
Smolarz B, Durczyński A, Romanowicz H, Hogendorf P. The Role of microRNA in Pancreatic Cancer. Biomedicines 2021; 9:biomedicines9101322. [PMID: 34680441 PMCID: PMC8533140 DOI: 10.3390/biomedicines9101322] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/18/2021] [Accepted: 09/22/2021] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs) are small ribonucleic acid molecules that play a key role in regulating gene expression. The increasing number of studies undertaken on the functioning of microRNAs in the tumor formation clearly indicates their important potential in oncological therapy. Pancreatic cancer is one of the deadliest cancers. The expression of miRNAs released into the bloodstream appears to be a good indicator of progression and evaluation of the aggressiveness of pancreatic cancer, as indicated by studies. The work reviewed the latest literature on the importance of miRNAs for pancreatic cancer development.
Collapse
Affiliation(s)
- Beata Smolarz
- Laboratory of Cancer Genetics, Department of Pathology, Polish Mother’s Memorial Hospital Research Institute, 93-338 Lodz, Poland;
- Correspondence: ; Tel.: +48-42-271-1290
| | - Adam Durczyński
- Department of General and Transplant Surgery, N. Barlicki Memorial Clinical Hospital, Medical University of Lodz, 90-153 Lodz, Poland; (A.D.); (P.H.)
| | - Hanna Romanowicz
- Laboratory of Cancer Genetics, Department of Pathology, Polish Mother’s Memorial Hospital Research Institute, 93-338 Lodz, Poland;
| | - Piotr Hogendorf
- Department of General and Transplant Surgery, N. Barlicki Memorial Clinical Hospital, Medical University of Lodz, 90-153 Lodz, Poland; (A.D.); (P.H.)
| |
Collapse
|
25
|
LncRNA SNHG17 promotes tumor progression and predicts poor survival in human renal cell carcinoma via sponging miR-328-3p. Aging (Albany NY) 2021; 13:21232-21250. [PMID: 34497156 PMCID: PMC8457601 DOI: 10.18632/aging.203440] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 08/02/2021] [Indexed: 12/12/2022]
Abstract
Accumulating data shows that dysregulation of long non-coding RNAs (lncRNAs) are involved in human tumors' occurrence and progression. Small nucleolar RNA host genes (SNHGs) are recently revealed to play a carcinogenic role in various human neoplasms. However, the functions and underlying mechanisms of lncRNA SNHG17 in renal cell carcinoma (RCC) are still elusive. We analyzed the relationship between SNHG17 expression levels and clinicopathologic characteristics and prognosis in patients with RCC according to TCGA RNA-sequencing data and our cohort data. Loss-of-function and gain-of-function experiments were conducted to examine the biological behaviors of SNHG17 on RCC cell proliferation, migration, invasion, apoptosis, and tumor growth in vivo. The interaction between SNHG17, miR-328-3p, and Histone’sH2Avariant (H2AX) was verified by bioinformatics, dual-luciferase reporter gene, and RNA immunoprecipitation (RIP). Highly expressed SNHG17 was evident in RCC tissue samples and cell lines, and SNHG17 overexpression was related to advanced TNM stage and reduced relapse-free and overall survival of patients with RCC. Knockdown of SNHG17 prohibited malignant phenotypes, whereas ectopic SNHG17 expression showed the opposite effects. More importantly, SNHG17 could upregulate the expression of H2AX by acting as a miR-328-3p sponge. In vivo experiments confirmed that SNHG17 promoted the growth of RCC tumors. SNHG17/miR-328-3p/H2AXaxis might be involved in RCC progression, which provided a potential therapeutic target for RCC.
Collapse
|
26
|
Bi Y, Ji J, Zhou Y. LncRNA-PVT1 indicates a poor prognosis and promotes angiogenesis via activating the HNF1B/EMT axis in glioma. J Cancer 2021; 12:5732-5744. [PMID: 34475987 PMCID: PMC8408127 DOI: 10.7150/jca.60257] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 07/12/2021] [Indexed: 12/29/2022] Open
Abstract
Recent studies identified that long non-coding RNAs (lncRNAs) exhibited critical roles in tumor migration and invasion. However, the roles of lncRNAs in glioma remain unclear. The aim of this study was to uncover the underlying mechanisms of glioma progression and provide potential therapeutic targets for its treatment in clinic. Our microarray study showed that lncRNA-PVT1 was significantly upregulated in glioma tissues and played an important role in cell proliferation, migration, invasion and angiogenesis. Our data showed that the expression of lncRNA-PVT1 was increased obviously and associated with advanced tumor stage, metastasis, invasion ability, and poor prognosis in glioma patients. Up-regulation of lncRNA-PVT1 was observed to promote glioma cells proliferation, and invasion abilities in vitro as well as tumor growth in vivo by regulating miR-1207-3p expression. Online software (TargetScan, miRDB and miR TarBase) were used to predict the regulating mechanisms of lncRNA-PVT1, miR-1207-3p and HNF1B, which were validated by dual-luciferase reporter gene system. In vivo tumor-bearing mice models were established to validate the cellular results. Therefore, we suggested that lncRNA-PVT1/miR-1207-3p/HNF1B axis might play critical roles in glioma progression, indicating that lncRNA-PVT1/miR-1207-3p/HNF1B signaling axis may serve as novel molecular targets for glioma prevention and treatment.
Collapse
Affiliation(s)
- Yongyan Bi
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Department of Neurosurgery, Minhang Hospital, Fudan University, Minhang, Shanghai, China
| | - Jie Ji
- Department of Rehabilitation Medicine, Minhang Hospital, Fudan University, Minhang, Shanghai, China
| | - Youxin Zhou
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
27
|
Yuan C, Su Z, Liao S, Li D, Zhou Z, Wang Y, Quan M, Zeng L, Lv C, Shen C, Gong W, Wu J, Chen X, Hu W, Lv X, Si W, Yu X. miR-198 inhibits the progression of renal cell carcinoma by targeting BIRC5. Cancer Cell Int 2021; 21:390. [PMID: 34289837 PMCID: PMC8296723 DOI: 10.1186/s12935-021-02092-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 07/14/2021] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND miR-198 is involved in the formation, migration, invasion, and metastasis of various malignant cancers. However, the function and mechanism of action of miR-198 in the tumorigenesis of renal cell carcinoma (RCC) remain elusive. Here, we aimed to explore the role of miR198 in RCC. METHODS Immunohistochemistry was performed to estimate the level of survivin in RCC sections. Quantitative real-time polymerase chain reaction was performed to determine the expression level of miR-198 in fresh RCC tissues. Furthermore, the target relationship between miR-198 and BIRC5 was predicted using the TargetScanHuman 7.2 database and verified via dual-luciferase reporter assay and western blotting. The effects of miR-198 on the viability, apoptosis, invasion, and migration of A498 and ACHN cells were studied using Cell Counting Kit-8, flow cytometry, transwell migration assay, and wound healing assay, respectively. Additionally, a xenograft nude mouse model was established to evaluate the effect of miR-198 on RCC tumorigenesis. RESULTS The expression levels of BIRC5 and miR-198 were respectively higher and lower in RCC tissues than those in normal adjacent tissues. Furthermore, miR-198 could inhibit luciferase activity and reduce the protein level of survivin without affecting the BIRC5 mRNA levels. miR-198 inhibited cell viability, migration, and invasion and promoted cell apoptosis; co-transfection with BIRC5 could rescue these effects. Moreover, miR-198 could repress tumor growth in the xenograft nude mouse model of RCC. CONCLUSIONS Our study demonstrates that miR-198 suppresses RCC progression by targeting BIRC5.
Collapse
Affiliation(s)
- Chao Yuan
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University School of Medicine, Xialu District guilin north, road no. 16, Huangshi, 435003, Hubei, China
| | - Zhenhong Su
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University School of Medicine, Xialu District guilin north, road no. 16, Huangshi, 435003, Hubei, China
| | - Shengjie Liao
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University School of Medicine, Xialu District guilin north, road no. 16, Huangshi, 435003, Hubei, China
| | - Duanzhuo Li
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University School of Medicine, Xialu District guilin north, road no. 16, Huangshi, 435003, Hubei, China
| | - Zhiwen Zhou
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University School of Medicine, Xialu District guilin north, road no. 16, Huangshi, 435003, Hubei, China
| | - Yawen Wang
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University School of Medicine, Xialu District guilin north, road no. 16, Huangshi, 435003, Hubei, China
| | - Mingchun Quan
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University School of Medicine, Xialu District guilin north, road no. 16, Huangshi, 435003, Hubei, China
| | - Lingling Zeng
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University School of Medicine, Xialu District guilin north, road no. 16, Huangshi, 435003, Hubei, China
| | - Cai Lv
- Department of Urology, Haikou Municipal Hospital, Haikou, 570208, Hainan, China
| | - Chenyi Shen
- Yixing Cancer Hospital, Dongshan Dong Lu No. 45, Yixing, 214200, Jiangsu, China
| | - Weida Gong
- Yixing Cancer Hospital, Dongshan Dong Lu No. 45, Yixing, 214200, Jiangsu, China
| | - Jianfeng Wu
- Yixing Cancer Hospital, Dongshan Dong Lu No. 45, Yixing, 214200, Jiangsu, China
| | - Xiaogang Chen
- Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, 435000, Hubei, China
| | - Wenbing Hu
- Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, 435000, Hubei, China
| | - Xu Lv
- Yixing Cancer Hospital, Dongshan Dong Lu No. 45, Yixing, 214200, Jiangsu, China.
| | - Wenxia Si
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University School of Medicine, Xialu District guilin north, road no. 16, Huangshi, 435003, Hubei, China.
| | - Xin Yu
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University School of Medicine, Xialu District guilin north, road no. 16, Huangshi, 435003, Hubei, China. .,Zhaoqing Medical College, Zhaoqing, 526020, Guangdong, China.
| |
Collapse
|
28
|
Jin C, Shi L, Li K, Liu W, Qiu Y, Zhao Y, Zhao B, Li Z, Li Y, Zhu Q. Mechanism of tumor‑derived extracellular vesicles in regulating renal cell carcinoma progression by the delivery of MALAT1. Oncol Rep 2021; 46:187. [PMID: 34278501 PMCID: PMC8298989 DOI: 10.3892/or.2021.8138] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 04/28/2021] [Indexed: 02/06/2023] Open
Abstract
Renal cell carcinoma (RCC) is a major healthcare burden globally. Tumor-derived extracellular vesicles (EVs) contribute to the formation of a pro-metastatic microenvironment. In the present study, we explored the role and mechanism of RCC cell 786-O-derived EVs (786-O-EVs) in RCC. First, 786-O-EVs were extracted and identified, and EV internalization of RCC cells was observed. RCC cell malignant behaviors and long noncoding RNA (lncRNA) metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) expression patterns were detected before and after 786-O-EV treatment. MALAT1 was intervened to evaluate RCC cell behaviors. The downstream mechanism involving MALAT1 was predicted. In addition, the relationship among MALAT1, transcription factor CP2 like 1 (TFCP2L1) and ETS proto-oncogene 1, transcription factor (ETS1) was analyzed. TFCP2L1 expression patterns were measured after 786-O-EV exposure. Tumor xenograft formation assay and lung metastasis model were adopted to verify the role of 786-O-EVs in vivo in RCC. It was found that 786-O-EVs could be internalized by RCC cells. 786-O-EVs promoted RCC cell malignant behaviors, accompanied by elevated MALAT1 expression levels. The 786-O-EVs with MALAT1 knockdown attenuated the promotive effect of sole 786-O-EVs on RCC cells. MALAT1 located ETS1 in the TFCP2L1 promoter and negatively regulated TFCP2L1, and ETS1 protein could specifically bind to MALAT1. 786-O-EVs enhanced the binding of ETS1 and the TFCP2L1 promoter and decreased TFCP2L1 expression. In vivo, 786-O-EVs promoted tumor growth and RCC lung metastasis, which was suppressed following inhibition of MALAT1. Our findings indicated that 786-O-EVs promoted RCC invasion and metastasis by transporting MALAT1 to promote the binding of transcription factor ETS1 and TFCP2L1 promoter.
Collapse
Affiliation(s)
- Chengluo Jin
- Department of Urology, The Second Affiliated Hospital, Harbin Medical University, Nangang, Harbin, Heilongjiang 150086, P.R. China
| | - Linmei Shi
- School of Health Management, Harbin Medical University, Nangang, Harbin, Heilongjiang 150086, P.R. China
| | - Kunlun Li
- Department of Urology, The Second Affiliated Hospital, Harbin Medical University, Nangang, Harbin, Heilongjiang 150086, P.R. China
| | - Wei Liu
- Department of Urology, The Second Affiliated Hospital, Harbin Medical University, Nangang, Harbin, Heilongjiang 150086, P.R. China
| | - Yu Qiu
- Department of Urology, The Second Affiliated Hospital, Harbin Medical University, Nangang, Harbin, Heilongjiang 150086, P.R. China
| | - Yakun Zhao
- Department of Urology, The Second Affiliated Hospital, Harbin Medical University, Nangang, Harbin, Heilongjiang 150086, P.R. China
| | - Bai Zhao
- Department of Urology, The Second Affiliated Hospital, Harbin Medical University, Nangang, Harbin, Heilongjiang 150086, P.R. China
| | - Zhexun Li
- Department of Urology, The Second Affiliated Hospital, Harbin Medical University, Nangang, Harbin, Heilongjiang 150086, P.R. China
| | - Yifei Li
- Department of Urology, The Second Affiliated Hospital, Harbin Medical University, Nangang, Harbin, Heilongjiang 150086, P.R. China
| | - Qingguo Zhu
- Department of Urology, The Second Affiliated Hospital, Harbin Medical University, Nangang, Harbin, Heilongjiang 150086, P.R. China
| |
Collapse
|
29
|
Lu HY, Wang GY, Zhao JW, Jiang HT. Knockdown of lncRNA MALAT1 ameliorates acute kidney injury by mediating the miR-204/APOL1 pathway. J Clin Lab Anal 2021; 35:e23881. [PMID: 34240756 PMCID: PMC8373329 DOI: 10.1002/jcla.23881] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/09/2021] [Accepted: 05/28/2021] [Indexed: 12/11/2022] Open
Abstract
Background Acute kidney injury (AKI) was characterized by loss of renal function, associated with chronic kidney disease, end‐stage renal disease, and length of hospital stay. Long non‐coding RNAs (lncRNAs) participated in AKI development and progression. Here, we aimed to investigate the roles and mechanisms of lncRNA MALAT1 in AKI. Methods AKI serum samples were obtained from 129 AKI patients. ROC analysis was conducted to confirm the diagnostic value of MALAT1 in differentiating AKI from healthy volunteers. After hypoxic treatment on HK‐2 cells, the expressions of inflammatory cytokines, MALAT1, miR‐204, APOL1, p65, and p‐p65, were measured by RT‐qPCR and Western blot assays. The targeted relationship between miR‐204 and MALAT1 or miR‐204 and APOL1 was determined by luciferase reporter assay and RNA pull‐down analysis. After transfection, CCK‐8, flow cytometry, and TUNEL staining assays were performed to evaluate the effects of MALAT1 and miR‐204 on AKI progression. Results From the results, lncRNA MALAT1 was strongly elevated in serum samples from AKI patients, with the high sensitivity and specificity concerning differentiating AKI patients from healthy controls. In vitro, we established the AKI cell model after hypoxic treatment. After experiencing hypoxia, we found significantly increased MALAT1, IL‐1β, IL‐6, and TNF‐α expressions along with decreased miR‐204 level. Moreover, the targeted relationship between MALAT1 and miR‐204 was confirmed. Silencing of MALAT1 could reverse hypoxia‐triggered promotion of HK‐2 cell apoptosis. Meanwhile, the increase of IL‐1β, IL‐6, and TNF‐α after hypoxia treatment could be repressed by MALAT1 knockdown as well. After co‐transfection with MALAT1 silencing and miR‐204 inhibition, we found that miR‐204 could counteract the effects of MALAT1 on HK‐2 cell progression and inflammation after under hypoxic conditions. Finally, NF‐κB signaling was inactivated while APOL1 expression was increased in HK‐2 cells after hypoxia treatment, and lncRNA MALAT1 inhibition reactivated NF‐κB signaling while suppressed APOL1 expression by sponging miR‐204. Conclusions Collectively, these results illustrated that knockdown of lncRNA MALAT1 could ameliorate AKI progression and inflammation by targeting miR‐204 through APOL1/NF‐κB signaling.
Collapse
Affiliation(s)
- Hai-Yuan Lu
- Department of Nephrology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Guo-Yi Wang
- Department of Nephrology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Jin-Wen Zhao
- Department of Nephrology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Hai-Tao Jiang
- Department of Orthopedics, Huai'an First People's Hospital, Huai'an, China
| |
Collapse
|
30
|
BiGAN: LncRNA-disease association prediction based on bidirectional generative adversarial network. BMC Bioinformatics 2021; 22:357. [PMID: 34193046 PMCID: PMC8247109 DOI: 10.1186/s12859-021-04273-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 06/15/2021] [Indexed: 12/11/2022] Open
Abstract
Background An increasing number of studies have shown that lncRNAs are crucial for the control of hormones and the regulation of various physiological processes in the human body, and deletion mutations in RNA are related to many human diseases. LncRNA- disease association prediction is very useful for understanding pathogenesis, diagnosis, and prevention of diseases, and is helpful for labelling relevant biological information. Results In this manuscript, we propose a computational model named bidirectional generative adversarial network (BiGAN), which consists of an encoder, a generator, and a discriminator to predict new lncRNA-disease associations. We construct features between lncRNA and disease pairs by utilizing the disease semantic similarity, lncRNA sequence similarity, and Gaussian interaction profile kernel similarities of lncRNAs and diseases. The BiGAN maps the latent features of similarity features to predict unverified association between lncRNAs and diseases. The computational results have proved that the BiGAN performs significantly better than other state-of-the-art approaches in cross-validation. We employed the proposed model to predict candidate lncRNAs for renal cancer and colon cancer. The results are promising. Case studies show that almost 70% of lncRNAs in the top 10 prediction lists are verified by recent biological research. Conclusion The experimental results indicated that our proposed model had an accurate predictive ability for the association of lncRNA-disease pairs.
Collapse
|
31
|
Liao S, He J, Liu C, Zhang Z, Liao H, Liao Z, Yu C, Guan J, Mo H, Yuan Z, Liang T, Lu Z, Xu G, Wang Z, Chen J, Jiang J, Zhan X. Construction of autophagy prognostic signature and analysis of prospective molecular mechanisms in skin cutaneous melanoma patients. Medicine (Baltimore) 2021; 100:e26219. [PMID: 34087900 PMCID: PMC8183723 DOI: 10.1097/md.0000000000026219] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 05/17/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Autophagy is closely related to skin cutaneous melanoma (SKCM), but the mechanism involved is unclear. Therefore, exploration of the role of autophagy-related genes (ARGs) in SKCM is necessary. MATERIALS AND METHODS Differential expression autophagy-related genes (DEARGs) were first analysed. Univariate and multivariate Cox regression analyses were used to evaluate the expression of DEARGs and prognosis of SKCM. Further, the expression levels of prognosis-related DEARGs were verified by immunohistochemical (IHC) staining. Finally, gene set enrichment analysis (GSEA) was used to explore the underlying molecular mechanisms of SKCM. RESULTS Five ARGs (APOL1, BIRC5, EGFR, TP63, and SPNS1) were positively correlated with the prognosis of SKCM. IHC verified the results of the differential expression of these 5 ARGs in the bioinformatics analysis. According to the receiver operating characteristic curve, the signature had a good performance at predicting overall survival in SKCM. The signature could classify SKCM patients into high-risk or low-risk groups according to distinct overall survival. The nomogram confirmed that the risk score has a particularly large impact on the prognosis of SKCM. Calibration plot displayed excellent agreement between nomogram predictions and actual observations. Principal component analysis indicated that patients in the high-risk group could be distinguished from those in low-risk group. Results of GSEA indicated that the low-risk group is enriched with aggressiveness-related pathways such as phosphatidylinositol-3-kinase/protein kinase B and mitogen-activated protein kinase signalling pathways. CONCLUSION Our study identified a 5-gene signature. It revealed the mechanisms of autophagy that lead to the progression of SKCM and established a prognostic nomogram that can predict overall survival of patients with SKCM. The findings of this study provide novel insights into the relationship between ARGs and prognosis of SKCM.
Collapse
Affiliation(s)
- Shian Liao
- Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi
| | - Juliang He
- Department of Bone and Soft Tissue Surgery, Guangxi Medical University Cancer Hospital
| | - Chong Liu
- Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi
| | - Zide Zhang
- Guangxi Medical University, Nanning, Guangxi
| | - Hongyu Liao
- Southern Medical University, Guangzhou, Guangdong
| | - Zuowei Liao
- Department of General Surgery, The Ninth Affiliated Hospital of Guangxi Medical University, Beihai, Guangxi, China
| | - Chaojie Yu
- Guangxi Medical University, Nanning, Guangxi
| | - Jian Guan
- Department of Bone and Soft Tissue Surgery, Guangxi Medical University Cancer Hospital
| | - Hao Mo
- Department of Bone and Soft Tissue Surgery, Guangxi Medical University Cancer Hospital
| | - Zhenchao Yuan
- Department of Bone and Soft Tissue Surgery, Guangxi Medical University Cancer Hospital
| | - Tuo Liang
- Guangxi Medical University, Nanning, Guangxi
| | - Zhaojun Lu
- Guangxi Medical University, Nanning, Guangxi
| | - Guoyong Xu
- Guangxi Medical University, Nanning, Guangxi
| | - Zequn Wang
- Guangxi Medical University, Nanning, Guangxi
| | - Jiarui Chen
- Guangxi Medical University, Nanning, Guangxi
| | - Jie Jiang
- Guangxi Medical University, Nanning, Guangxi
| | - Xinli Zhan
- Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi
| |
Collapse
|
32
|
Non-Coding RNAs in Kidney Diseases: The Long and Short of Them. Int J Mol Sci 2021; 22:ijms22116077. [PMID: 34199920 PMCID: PMC8200121 DOI: 10.3390/ijms22116077] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 02/07/2023] Open
Abstract
Recent progress in genomic research has highlighted the genome to be much more transcribed than expected. The formerly so-called junk DNA encodes a miscellaneous group of largely unknown RNA transcripts, which contain the long non-coding RNAs (lncRNAs) family. lncRNAs are instrumental in gene regulation. Moreover, understanding their biological roles in the physiopathology of many diseases, including renal, is a new challenge. lncRNAs regulate the effects of microRNAs (miRNA) on mRNA expression. Understanding the complex crosstalk between lncRNA–miRNA–mRNA is one of the main challenges of modern molecular biology. This review aims to summarize the role of lncRNA on kidney diseases, the molecular mechanisms involved, and their function as emerging prognostic biomarkers for both acute and chronic kidney diseases. Finally, we will also outline new therapeutic opportunities to diminish renal injury by targeting lncRNA with antisense oligonucleotides.
Collapse
|
33
|
Zhang L, Li Y, Wang X, Ping Y, Wang D, Cao Y, Dai Y, Liu W, Tao Z. Five-gene signature associating with Gleason score serve as novel biomarkers for identifying early recurring events and contributing to early diagnosis for Prostate Adenocarcinoma. J Cancer 2021; 12:3626-3647. [PMID: 33995639 PMCID: PMC8120165 DOI: 10.7150/jca.52170] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 04/12/2021] [Indexed: 12/14/2022] Open
Abstract
Background: Compared to non-recurrent type, recurrent prostate adenocarcinoma (PCa) is highly fatal, and significantly shortens the survival time of affected patients. Early and accurate laboratory diagnosis is particularly important in identifying patients at high risk of recurrence, necessary for additional systemic intervention. We aimed to develop efficient and accurate diagnostic and prognostic biomarkers for new PCa following radical therapy. Methods: We identified differentially expressed genes (DEGs) and clinicopathological data of PCa patients from Gene Expression Omnibus (GEO) datasets and The Cancer Genome Atlas (TCGA) repositories. We then uncovered the most relevant clinical traits and genes modules associated with PCa prognosis using the Weighted gene correlation network analysis (WGCNA). Univariate Cox regression analysis and multivariate Cox proportional hazards (Cox-PH) models were performed to identify candidate gene signatures related to Disease-Free Interval (DFI). Data for internal and external cohorts were utilized to test and validate the accuracy and clinical utility of the prognostic models. Results: We constructed and validated an accurate and reliable model for predicting the prognosis of PCa using 5 Gleason score-associated gene signatures (ZNF695, CENPA, TROAP, BIRC5 and KIF20A). The ROC and Kaplan-Meier analysis revealed the model was highly accurate in diagnosing and predicting the recurrence and metastases of PCa. The accuracy of the model was validated using the calibration curves based on internal TCGA cohort and external GEO cohort. Using the model, patients could be prognostically stratified in to various groups including TNM classification and Gleason score. Multivariate analysis revealed the model could independently predict the prognosis of PCa patients and its utility was superior to that of clinicopathological characteristics. In addition, we fund the expression of the 5 gene signatures strongly and positively correlated with tumor purity but negatively correlated with infiltration CD8+ T cells to the tumor microenvironment. Conclusions: A 5 gene signatures can accurately be used in the diagnosis and prediction of PCa prognosis. Thus this can guide the treatment and management prostate adenocarcinoma.
Collapse
Affiliation(s)
- Lingyu Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yu Li
- Department of Biochemistry and Molecular Biology, Bengbu Medical College, Anhui 233030, China
| | - Xuchu Wang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Ying Ping
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Danhua Wang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Ying Cao
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yibei Dai
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Weiwei Liu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Zhihua Tao
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| |
Collapse
|
34
|
Zhang M, Yan X, Wen P, Bai W, Zhang Q. CircANKRD52 Promotes the Tumorigenesis of Hepatocellular Carcinoma by Sponging miR-497-5p and Upregulating BIRC5 Expression. Cell Transplant 2021; 30:9636897211008874. [PMID: 33845641 PMCID: PMC8058805 DOI: 10.1177/09636897211008874] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
CircRNAs participate in the pathogenesis of a variety of cancers. Previous studies showed that baculoviral IAP repeat containing 5 (BIRC5) can promote tumor progression. But, the mechanisms by which circRNAs regulate BIRC5 expression in hepatocellular carcinoma (HCC) remain unknown. The clinical prognosis of BIRC5 or miR-497-5p expression in patients with HCC was assessed by TCGA RNA-seq dataset. hsa_circ_0026939 (circANKRD52) or BIRC5 was identified to bind with miR-497-5p by luciferase gene report, RIP and circRIP assays. MTT, colony formation, Transwell assays and a xenograft tumor model were used to estimate the role of miR-497-5p or circANKRD52 in HCC cells. As a result, we found that elevated expression of BIRC5 or decreased expression of miR-497-5p was linked to poor survival in HCC. Restored expression of miR-497-5p repressed cell proliferation, colony formation and invasiveness by targeting BIRC5, but its inhibitor showed the opposite results. Furthermore, circANKRD52 possessed a tumor-promoting effect by acting as a sponge of miR-497-5p and thereby upregulated BIRC5 in HCC cells. In conclusion, our findings demonstrated that circANKRD52 enhances the tumorigenesis of HCC by sponging miR-497-5p and upregulating BIRC5 expression.
Collapse
Affiliation(s)
- Mingzhi Zhang
- Department of Gastroenterology, Tianjin Medical University General Hospital, Tianjin, China.,Department of Gastroenterology, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Xinxin Yan
- Department of Gastroenterology, Tianjin Medical University General Hospital, Tianjin, China.,Geriatric Department, Aerospace Central Hospital, Beijing, China
| | - Peihao Wen
- Department of Liver Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenkun Bai
- Department of Ultrasonography, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Qingyu Zhang
- Department of Gastroenterology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
35
|
Yang Y, Yu Q, Li B, Guan R, Huang C, Yang X. BBOX1-AS1 Accelerates Gastric Cancer Proliferation by Sponging miR-3940-3p to Upregulate BIRC5 Expression. Dig Dis Sci 2021; 66:1054-1062. [PMID: 32394331 DOI: 10.1007/s10620-020-06308-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Accepted: 02/25/2020] [Indexed: 01/28/2023]
Abstract
BACKGROUND Gastric cancer (GC) is one type of the most general malignancies in the globe. Research increasingly suggests long non-coding RNAs (lncRNAs) exert crucial roles in GC. However, the function of BBOX1-AS1 in GC has not been reported yet, it needs more explorations. AIMS The aim of the study is to figure out the role and related regulation mechanism of BBOX1-AS1 in GC. METHODS RT-qPCR assay was applied to detect genes expression. The role of BBOX1-AS1 in GC was investigated by cell counting kit-8, colony formation, tunel detection, and western blot assays. The binding ability between miR-3940-3p and BBOX1-AS1 (or BIRC5) by RIP, RNA pull-down and luciferase reporter assays. RESULTS The expression of BBOX1-AS1 presented significantly upregulation in GC tissues and cells. Moreover, upregulation of BBOX1-AS1 promoted GC cell proliferation, and inhibited GC cell apoptosis. However, downregulation of BBOX1-AS1 led to opposite results. Furtherly, we discovered that BBOX1-AS1 bound with miR-3940-3p and also negatively regulated miR-3940-3p. Besides, it proved that miR-3940-3p interplayed with BIRC5 and negatively regulated BIRC5. Through rescue experiments, we proved that BIRC5 reversed miR-3940-3p-mediated cell proliferation or apoptosis in BBOX1-AS1-dysregulated GC cells. CONCLUSIONS BBOX1-AS1 accelerates GC proliferation by sponging miR-3940-3p to upregulate BIRC5 expression, which may guide a new direction into the therapeutic strategies of GC.
Collapse
Affiliation(s)
- Yan Yang
- Department of Gastroenterology, Tengzhou Central People's Hospital, No. 181, Xingtan Road, Tengzhou, 277500, Shandong, China
| | - Qiong Yu
- Department of Pathology, Zaozhuang Mining Group Central Hospital, Zaozhuang, 277000, Shandong, China
| | - Bing Li
- Department of Gastroenterology, Tengzhou Central People's Hospital, No. 181, Xingtan Road, Tengzhou, 277500, Shandong, China
| | - Renzhen Guan
- Department of Gastroenterology, Tengzhou Central People's Hospital, No. 181, Xingtan Road, Tengzhou, 277500, Shandong, China
| | - ChangYong Huang
- Department of Gastroenterology, Tengzhou Central People's Hospital, No. 181, Xingtan Road, Tengzhou, 277500, Shandong, China
| | - XiuCheng Yang
- Department of Gastroenterology, Tengzhou Central People's Hospital, No. 181, Xingtan Road, Tengzhou, 277500, Shandong, China.
| |
Collapse
|
36
|
LncRNA SNHG12 contributes proliferation, invasion and epithelial-mesenchymal transition of pancreatic cancer cells by absorbing miRNA-320b. Biosci Rep 2021; 40:224896. [PMID: 32432698 PMCID: PMC7276652 DOI: 10.1042/bsr20200805] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/14/2020] [Accepted: 05/18/2020] [Indexed: 01/01/2023] Open
Abstract
Pancreatic cancer is a kind of malignant carcinoma with high mortality, which is devoid of early diagnostic biomarker and effective therapeutic methods. Recently, long non-coding RNAs (lncRNAs) have been reported as a crucial role in regulating the development of various kinds of tumors. Here, we found lncRNA small nuclear RNA host gene 12 (SNHG12) is highly expressed in pancreatic cancer tissues and cell lines through qRT-PCR, which suggested that SNHG12 possibly accelerates the progression of pancreatic cancer. Further study revealed that SNHG12 promoted cancer cells growth and invasion via absorbing miR-320b. Flow cytometry and transwell chamber assay were utilized to verify the promoting effects on proliferation and invasion that SNHG12 acts in pancreatic cancer cells. Evidence that SNHG12 increased cell invasive ability through up-regulated EMT process was lately obtained by Western blotting assay. Consequently, we extrapolated that SNHG12/miR-320b could be invoked as a promising early diagnostic hallmark and therapeutic strategy for pancreatic cancer.
Collapse
|
37
|
LINC00511 exacerbated T-cell acute lymphoblastic leukemia via miR-195-5p/LRRK1 axis. Biosci Rep 2021; 40:222566. [PMID: 32242897 PMCID: PMC7953487 DOI: 10.1042/bsr20193631] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 02/10/2020] [Accepted: 02/17/2020] [Indexed: 01/08/2023] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is a malignant disease arising from the abnormal proliferation of T lymphocyte in marrow. Long non-coding RNAs (lncRNAs) are one kind of non-coding RNAs (ncRNAs), which were reported to modulate the initiation or progression of diverse cancers. However, the role of LINC00511 in T-ALL was unknown. To figure out the function and mechanism of LINC00511 in T-ALL, a series of experiments were carried out. Based on the experimental results, we discovered that LINC00511 boosted cell proliferation and invasion, but hindered cell apoptosis in T-ALL cells. Besides, based on bio-informatics tool, miR-195-5p was selected for further exploration. Then, miR-195-5p was validated to bind with LINC00511. Hereafter, LRRK1 was testified to serve as a target gene of miR-195-5p. At last, rescue assays suggested that LRRK1 overexpression restored sh-LINC00511#1-mediated effects on cell proliferation and apoptosis. All in all, LINC00511 exacerbated T-ALL progression via miR-195-5p/LRRK1 axis, implying a potential therapeutic clue for the patients with T-ALL.
Collapse
|
38
|
Yao J, Wang Z, Cheng Y, Ma C, Zhong Y, Xiao Y, Gao X, Li Z. M2 macrophage-derived exosomal microRNAs inhibit cell migration and invasion in gliomas through PI3K/AKT/mTOR signaling pathway. J Transl Med 2021; 19:99. [PMID: 33676540 PMCID: PMC7937290 DOI: 10.1186/s12967-021-02766-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 02/22/2021] [Indexed: 12/14/2022] Open
Abstract
Background Glioma, the most common primary brain tumor, account Preparing figures for 30 to 40% of all intracranial tumors. Herein, we aimed to study the effects of M2 macrophage-derived exosomal microRNAs (miRNAs) on glioma cells. Methods First, we identified seven differentially expressed miRNAs in infiltrating macrophages and detected the expression of these seven miRNAs in M2 macrophages. We then selected hsa-miR-15a-5p (miR-15a) and hsa-miR-92a-3p (miR-92a) for follow-up studies, and confirmed that miR-15a and miR-92a were under-expressed in M2 macrophage exosomes. Subsequently, we demonstrated that M2 macrophage-derived exosomes promoted migration and invasion of glioma cells, while exosomal miR-15a and miR-92a had the opposite effects on glioma cells. Next, we performed the target gene prediction in four databases and conducted target gene validation by qRT-PCR, western blot and dual luciferase reporter gene assays. Results The results revealed that miR-15a and miR-92a were bound to CCND1 and RAP1B, respectively. Western blot assays demonstrated that interference with the expression of CCND1 or RAP1B reduced the phosphorylation level of AKT and mTOR, indicating that both CCND1 and RAP1B can activate the PI3K/AKT/mTOR signaling pathway. Conclusion Collectively, these findings indicate that M2 macrophage-derived exosomal miR-15a and miR-92a inhibit cell migration and invasion of glioma cells through PI3K/AKT/mTOR signaling pathway. Supplementary information The online version contains supplementary material available at 10.1186/s12967-021-02766-w.
Collapse
Affiliation(s)
- Jie Yao
- Human Genetic Resources Conservation Center of Hubei Province, Wuhan, 430071, China.,Tumor Precision Diagnosis and Treatment Technology and Translation Medicine, Hubei Engineering Research Center, Wuhan, 430071, China
| | - Zefen Wang
- Department of Physiology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Yong Cheng
- Department of Neurology, Hankou Hospital, General Hospital of Central Theater Command of Chinese People's Liberation Army, Wuhan, 430014, China
| | - Chao Ma
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No 169 Donghu Road, Wuhan, 430071, Hubei, China
| | - Yahua Zhong
- Department of Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yilei Xiao
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, 252000, China
| | - Xu Gao
- Department of Neurosurgery, General Hospital of Northern Theater Command of People's Liberation Army, Shenyang, 110000, China
| | - Zhiqiang Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No 169 Donghu Road, Wuhan, 430071, Hubei, China.
| |
Collapse
|
39
|
Liao H, Chen Q, Xiao J. Reflections on the Role of Malat1 in Gynecological Cancer. Cancer Manag Res 2020; 12:13489-13500. [PMID: 33408521 PMCID: PMC7779295 DOI: 10.2147/cmar.s286804] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 12/01/2020] [Indexed: 12/26/2022] Open
Abstract
Non-coding RNAs (ncRNAs) have received significant attention over the last few years. Malat1, as one of the most extensively studied ncRNAs, is believed to be not only a potential biomarker for disease diagnosis and prognosis, but also a candidate drug target for gynecological cancers. This potential is supported by a growing body of experimental evidence demonstrating that Malat1 participates in the occurrence, progression, and metastasis of tumors. Research has also shown that Malat1 can influence patient survival by regulating a range of target genes and signaling pathways. However, previous review articles have generally failed to consider the role of Malat1 in gynecological cancer in detail. In the present review, we summarize recent progress in research relating to the clinical relevance of Malat1 and the molecular mechanisms underlying the action of this ncRNA. Besides, we put forward some action points for further research after taking into consideration the sub-location and other essential properties of Malat1, which might enable us to have a better understanding of the potential of this molecule regarding clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Huiyan Liao
- The 2nd Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province 510006, People's Republic of China
| | - Qi Chen
- The 6th Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province 510006, People's Republic of China
| | - Jing Xiao
- Department of Gynecology, the University Town Branch, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province 510120, People's Republic of China
| |
Collapse
|
40
|
Cai Q, Gao ML, Huang LS, Chen HS, Pan LH. MALAT1/miRNA-203/Wnt5a: A potential mechanism for regulating coronary artery disease. Int J Cardiol 2020; 329:48. [PMID: 33358830 DOI: 10.1016/j.ijcard.2020.12.046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/13/2020] [Accepted: 12/15/2020] [Indexed: 12/19/2022]
Affiliation(s)
- Qiang Cai
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, 530021, China
| | - Mei-Ling Gao
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Lin-Sheng Huang
- Department of hepatobiliary surgery, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Hai-Shao Chen
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, 530021, China
| | - Ling-Hui Pan
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, 530021, China.
| |
Collapse
|
41
|
Wu J, Liu T, Sun L, Zhang S, Dong G. Long noncoding RNA SNHG4 promotes renal cell carcinoma tumorigenesis and invasion by acting as ceRNA to sponge miR-204-5p and upregulate RUNX2. Cancer Cell Int 2020; 20:514. [PMID: 33088220 PMCID: PMC7574175 DOI: 10.1186/s12935-020-01606-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 10/12/2020] [Indexed: 12/17/2022] Open
Abstract
Background Long noncoding RNAs (lncRNAs) are involved in the tumorigenesis and progression of human cancers, including renal cell carcinoma (RCC). Small nucleolar RNA host gene 4 (SNHG4) is reported to play an essential role in tumor growth and progression. However, the molecular mechanisms and function of SNHG4 in RCC remain undocumented. Methods Quantitative real-time polymerase chain reaction (qRT-PCR) was performed to examine expression levels of SNHG4 in RCC tissue samples and cell lines. Cell counting kit-8, western blotting, activities of caspase-3, -8, and -9, wound-healing, and transwell invasion assays were performed to explore cell proliferation, apoptosis, migration, and invasion. The interaction among SNHG4, miR-204-5p, and RUNX2 was verified by bioinformatic analysis, a luciferase gene report, qRT-PCR, western blot analysis, and RNA immunoprecipitation assays. Xenograft mouse models were carried out to examine the role of SNHG4 in RCC in vivo. Results SNHG4 was highly expressed in RCC tissue samples and cell lines, and its upregulation was significantly involved in node involvement, distant metastasis, and reduced overall and relapse-free survival of patients with RCC. SNHG4 acted as an oncogenic lncRNA with promoted RCC cell proliferation, migration, invasion, and inhibited apoptosis. SNHG4 boosted tumor growth in xenograft mouse models. Mechanistically, SNHG4 functioned as a competing endogenous RNA (ceRNA) for sponging miR-204-5p, leading to the upregulation of its target RUNX2 to promote RCC cell proliferation and invasion. Conclusion SNHG4 and miR-204-5p might be indicated in RCC progression via RUNX2, suggesting the potential use of SNHG4/miR-204-5p/RUNX2 axis in RCC treatment.
Collapse
Affiliation(s)
- Jie Wu
- Department of Ultrasound Intervention, The First Affiliated Hospital, Zhengzhou University, 1 Jianshe Dong Road, Zhengzhou, 450052 Henan China
| | - Tingting Liu
- Department of Ultrasound Intervention, The First Affiliated Hospital, Zhengzhou University, 1 Jianshe Dong Road, Zhengzhou, 450052 Henan China
| | - Lulu Sun
- Department of Ultrasound Intervention, The First Affiliated Hospital, Zhengzhou University, 1 Jianshe Dong Road, Zhengzhou, 450052 Henan China
| | - Shaojin Zhang
- Department of Urology Surgery, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan China
| | - Gang Dong
- Department of Ultrasound Intervention, The First Affiliated Hospital, Zhengzhou University, 1 Jianshe Dong Road, Zhengzhou, 450052 Henan China
| |
Collapse
|
42
|
Regulation of Glycolysis by Non-coding RNAs in Cancer: Switching on the Warburg Effect. MOLECULAR THERAPY-ONCOLYTICS 2020; 19:218-239. [PMID: 33251334 PMCID: PMC7666327 DOI: 10.1016/j.omto.2020.10.003] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The “Warburg effect” describes the reprogramming of glucose metabolism away from oxidative phosphorylation toward aerobic glycolysis, and it is one of the hallmarks of cancer cells. Several factors can be involved in this process, but in this review, the roles of non-coding RNAs (ncRNAs) are highlighted in several types of human cancer. ncRNAs, including microRNAs, long non-coding RNAs, and circular RNAs, can all affect metabolic enzymes and transcription factors to promote glycolysis and modulate glucose metabolism to enhance the progression of tumors. In particular, the 5′-AMP-activated protein kinase (AMPK) and the phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathways are associated with alterations in ncRNAs. A better understanding of the roles of ncRNAs in the Warburg effect could ultimately lead to new therapeutic approaches for suppressing cancer.
Collapse
|
43
|
Chen F, Zhong Z, Tan HY, Guo W, Zhang C, Cheng C, Wang N, Ren J, Feng Y. Suppression of lncRNA MALAT1 by betulinic acid inhibits hepatocellular carcinoma progression by targeting IAPs via miR-22-3p. Clin Transl Med 2020; 10:e190. [PMID: 33135336 PMCID: PMC7586994 DOI: 10.1002/ctm2.190] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/16/2020] [Accepted: 09/18/2020] [Indexed: 12/13/2022] Open
Abstract
Betulinic acid (BA) is a natural product extracted from a broad range of medicinal and edible herbal plants. Previous studies showed that BA induces cell death in tumors derived from multiple tissues; however, the underlying mechanism remains obscure. The present study aimed to study the effects of BA on autophagy and apoptosis of hepatocellular carcinoma (HCC). Human HCC cell lines and orthotopic HCC implanted mice were employed to examine the BA-induced tumor suppression; RT2 long noncoding RNA (lncRNA) PCR array and database analysis were used to explore the possible mechanisms; validation of pathways was performed using siRNA and miRNA inhibitors. The results indicated that BA regulated autophagy and induced apoptosis in HCC. The degradation of inhibitor of apoptosis proteins (IAPs), the conversion of LC3-I to LC3-II, and p62 accumulation were enhanced by BA, thereby suggesting that the downregulation of IAPs and autophagic cell death are induced by BA. The addition of autophagy and lysosomal inhibitors indicated that BA induced autophagy-independent apoptosis via degradation of IAPs. Moreover, RT2 lncRNA PCR array and database analysis suggested that BA downregulated the levels of lncRNA MALAT1, which is considered to be an oncogene. Further investigations demonstrated that lncRNA MALAT1 functioned as a ceRNA (competing endogenous RNA) to contribute to BA-mediated degradation of IAPs by sponging miR-22-3p. Therefore, BA could be developed as a potential anticancer agent for HCC.
Collapse
Affiliation(s)
- Feiyu Chen
- School of Chinese Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong S.A.R.P. R. China
| | - Zhangfeng Zhong
- School of Chinese Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong S.A.R.P. R. China
| | - Hor Yue Tan
- School of Chinese Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong S.A.R.P. R. China
| | - Wei Guo
- School of Chinese Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong S.A.R.P. R. China
| | - Cheng Zhang
- School of Chinese Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong S.A.R.P. R. China
| | - Chien‐Shan Cheng
- School of Chinese Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong S.A.R.P. R. China
| | - Ning Wang
- School of Chinese Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong S.A.R.P. R. China
| | - Junguo Ren
- Institute of Basic Medical Sciences, Xiyuan HospitalChina Academy of Chinese Medical SciencesBeijingP. R. China
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong S.A.R.P. R. China
| |
Collapse
|
44
|
LncRNA MALAT1 inhibits apoptosis of endometrial stromal cells through miR-126-5p-CREB1 axis by activating PI3K-AKT pathway. Mol Cell Biochem 2020; 475:185-194. [PMID: 32809092 DOI: 10.1007/s11010-020-03871-y] [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: 03/30/2020] [Accepted: 08/01/2020] [Indexed: 12/13/2022]
Abstract
Endometriosis is a common, chronic and painful disease in women, whose pathogenesis remains not entirely clear. Long non-coding RNA (lncRNA) MALAT1 participates in the development of endometriosis. This study further investigated the regulation of MALAT1-miR-126-5p-CREB1 axis in the pathological process of endometriosis. MALAT1, miR-126-5p, and CREB1 levels in human endometrial stromal cells (HESCs) were detected by quantitative reverse transcription polymerase chain reaction (RT-qPCR). Protein levels were determined by Western blotting. Cell viability and apoptosis was assessed by MTT assay and annexin V-FITC staining, respectively. The interactivity between miR-126-5p and MALAT1 (or CREB1) was assessed by dual luciferase reporter system. Knockdown of MALAT1 or CREB1 restrained proliferation and induced apoptosis as confirmed by upregulating cleaved caspase-3 and Bax, and down-regulating Bcl-2 in HESCs, while inhibition of miR-126-5p presented the opposite results. Moreover, silencing of MALAT1 triggered apoptosis of HESCs via targeting miR-126-5p. In addition, miR-126-5p directly regulated CREB1 expression via binding to its 3' non-coding region. Finally, miR-126-5p inhibitor-mediated apoptosis inhibition was restrained by CREB1 silencing via inactivation of PI3K-AKT pathway in HESCs. Taken together, our study firstly demonstrates that MALAT1 regulates apoptosis of HESCs through miR-126-5p/CREB1 axis mediated PI3K/AKT pathway. Our findings explained the pathogenesis of endometriosis and offered promising therapeutic option for endometriosis.
Collapse
|
45
|
Li Z, Feng Y, Zhang Z, Cao X, Lu X. TMPO-AS1 promotes cell proliferation of thyroid cancer via sponging miR-498 to modulate TMPO. Cancer Cell Int 2020; 20:294. [PMID: 32669970 PMCID: PMC7346673 DOI: 10.1186/s12935-020-01334-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 06/09/2020] [Indexed: 01/14/2023] Open
Abstract
Background Thyroid cancer (TC) is the most frequent endocrine malignancy. Long noncoding RNAs (lncRNAs) have been confirmed to act as significant roles in tumor development. The role of lncRNA TMPO-AS1 in TC is still unclear, so it remains to be explored. The aim of the research is to investigate the role and regulatory mechanism of TMPO-AS1 in TC. Methods TMPO-AS1 and TMPO expression in TC tumors and cells was detected by TCGA database and QRT-PCR assay respectively. CCK-8, EDU, TUNEL and western blot assays were conducted to identify the biological functions of TMPO-AS1 in TC. Luciferase reporter and RNA pull down assays were conducted to measure the interaction among TMPO-AS1, TMPO and miR-498. Results TMPO-AS1 was overexpressed in TC tissues and cell lines. Knockdown of TMPO-AS1 suppressed cell growth and accelerated cell apoptosis in TC. Furthermore, downregulation of TMPO-AS1 suppressed TMPO expression in TC. The data suggested that TMPO expression was upregulated in TC tissues and cell lines and was positively correlated with TMPO-AS1 expression in TC. Furthermore, the expression of miR-498 presented low expression in TC cells. And miR-498 expression was negatively regulated by TMPO-AS1, meanwhile, TMPO expression was negatively regulated by miR-498 in TC cells. Besides, it was confirmed that TMPO-AS1 could bind with miR-498 and TMPO in TC cells. In addition, it was validated that TMPO-AS1 elevated the levels of TMPO via sponging miR-498 in TC cells. Conclusions TMPO-AS1 promotes cell proliferation in TC via sponging miR-498 to modulate TMPO.
Collapse
Affiliation(s)
- Zhenyu Li
- Department of Thyroid Surgery, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East Road, Zhongyuan District, Zhengzhou, 450000 Henan China.,Department of Thyroid Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, 471000 Henan China
| | - Yun Feng
- Department of Thyroid Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, 471000 Henan China
| | - Zhen Zhang
- Department of Thyroid Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, 471000 Henan China
| | - Xiaozhong Cao
- Department of Thyroid Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, 471000 Henan China
| | - Xiubo Lu
- Department of Thyroid Surgery, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East Road, Zhongyuan District, Zhengzhou, 450000 Henan China
| |
Collapse
|
46
|
Wu X, Wang Y, Zhong W, Cheng H, Tian Z. The Long Non-Coding RNA MALAT1 Enhances Ovarian Cancer Cell Stemness by Inhibiting YAP Translocation from Nucleus to Cytoplasm. Med Sci Monit 2020; 26:e922012. [PMID: 32433460 PMCID: PMC7254939 DOI: 10.12659/msm.922012] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The purpose of this work was to unearth the effects and underlying mechanism of long non-coding RNA (lncRNA) MALAT1 in ovarian cancer cell stemness. MATERIAL AND METHODS Western blot, quantitative polymerase chain reaction (qPCR) and sphere forming analysis were performed to evaluate the stem-like traits of cells and MALAT1-induced effects on ovarian cancer cell stemness. Cell viability was performed to evaluate MALAT1 role in the chemoresistance of ovarian cancer cells. RNA immunoprecipitation (RIP) and luciferase reporter analysis were constructed to investigate the underlying mechanisms. RESULTS Here, qPCR assay showed that MALAT1 level was remarkably higher in non-adherent spheres formed by adherent ovarian cancer cells, as well as cisplatin-resistant ovarian cancer cells. Additionally, MALAT1 knockdown reduced ovarian cancer cell stemness, characterized as the decrease of sphere forming ability, expression of stemness regulatory masters, and attenuation of cisplatin resistance. Moreover, MALAT1 interacted with yes-associated protein (YAP), inhibited its nuclear-cytoplasm translocation, promoted YAP protein stability and expression and thus increased its activity. Notably, rescuing expression of YAP attenuated the inhibition of MALAT1 knockdown on ovarian cancer cell stemness. CONCLUSIONS In conclusion, these results demonstrate a MALAT1/YAP axis responsible for ovarian cancer cell stemness.
Collapse
Affiliation(s)
- XingMei Wu
- Department of Gynecology, The People's Hospital of Lishui, Lishui, Zhejiang, China (mainland)
| | - YongHui Wang
- Department of Oncology, Lishui Municipal Central Hospital, Lishui, Zhejiang, China (mainland)
| | - WeiJuan Zhong
- Department of Gynecology, The People's Hospital of Lishui, Lishui, Zhejiang, China (mainland)
| | - HuiFei Cheng
- Department of Radiation Oncology, Lishui Municipal Central Hospital, Lishui, Zhejiang, China (mainland)
| | - Zhifeng Tian
- Department of Radiation Oncology, Lishui Municipal Central Hospital, Lishui, Zhejiang, China (mainland)
| |
Collapse
|
47
|
Chen LJ, Yuan MX, Ji CY, Zhang YB, Peng YM, Zhang T, Gao HQ, Sheng XY, Liu ZY, Xie WX, Yin Q. Long Non-Coding RNA CRNDE Regulates Angiogenesis in Hepatoblastoma by Targeting the MiR-203/VEGFA Axis. Pathobiology 2020; 87:161-170. [PMID: 32182608 DOI: 10.1159/000505131] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 11/28/2019] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE MiR-203 has been shown to participate in multiple malignancies, but the role of miR-203 in hepatoblastoma (HB) remains unclear. The aim of our study was to investigate the effects of miR-203 in HB. METHODS A total of 15 pairs of HB tissues and para-tumour normal tissues were collected for the experiments. RT-qPCR and Western blotting were performed to detect the expression of CRNDE, miR-203, and VEGFA at the mRNA and/or protein levels, respectively. A dual luciferase assay verified the target relationship between miR-203 and the 3'UTR of VEGFA as well as miR-203 and CRNDE. In addition, MTT, wound healing, and tube formation assays were performed to assess the effects of miR-203, VEGFA, and CRNDE on cell proliferation, migration, and angiogenesis, respectively. RESULTS Our data revealed that miR-203 expression was decreased in HB tissues, while long non-coding RNA (lncRNA) CRNDE expression was increased. The dysregulation of miR-203 and CRNDE was closely related to tumour size and stage. Moreover, overexpression of miR-203 inhibited angiogenesis. A dual luciferase assay verified that VEGFA is a direct target of miR-203 and that CRNDE binds to miR-203. Furthermore, our results showed that miR-203 suppressed cell viability, migration, and angiogenesis by regulating VEGFA expression. Additionally, it was confirmed that CRNDE promoted angiogenesis by negatively regulating miR-203 expression. CONCLUSION lncRNA CRNDE targets the miR-203/VEGFA axis and promotes angiogenesis in HB. These results provide insight into the underlying mechanisms of HB and indicate that CRNDE and miR-203 might be potential targets for HB therapy.
Collapse
Affiliation(s)
- Li-Jian Chen
- Department of General Surgery, Hunan Children's Hospital, Changsha, China
| | - Miao-Xian Yuan
- Department of General Surgery, Hunan Children's Hospital, Changsha, China
| | - Chun-Yi Ji
- Department of General Surgery, Hunan Children's Hospital, Changsha, China
| | - Yan-Bing Zhang
- Department of General Surgery, Hunan Children's Hospital, Changsha, China
| | - Yu-Ming Peng
- Department of General Surgery, Hunan Children's Hospital, Changsha, China
| | - Tian Zhang
- Department of General Surgery, Hunan Children's Hospital, Changsha, China
| | - Hong-Qiang Gao
- Department of General Surgery, Hunan Children's Hospital, Changsha, China
| | - Xin-Yi Sheng
- Department of General Surgery, Hunan Children's Hospital, Changsha, China
| | - Zhao-Yang Liu
- Department of General Surgery, Hunan Children's Hospital, Changsha, China
| | - Wei-Xin Xie
- Department of General Surgery, Hunan Children's Hospital, Changsha, China
| | - Qiang Yin
- Department of General Surgery, Hunan Children's Hospital, Changsha, China,
| |
Collapse
|
48
|
Liu N, Feng S, Li H, Chen X, Bai S, Liu Y. Long non-coding RNA MALAT1 facilitates the tumorigenesis, invasion and glycolysis of multiple myeloma via miR-1271-5p/SOX13 axis. J Cancer Res Clin Oncol 2020; 146:367-379. [PMID: 31953613 PMCID: PMC6985203 DOI: 10.1007/s00432-020-03127-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 01/06/2020] [Indexed: 12/19/2022]
Abstract
Background Long non-coding RNAs (lncRNAs) play crucial roles in the regulation and treatment of multiple myeloma (MM). The objective of this research was to study the functional mechanism of metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) in MM. Methods MALAT1, microRNA-1271-5p (miR-1271-5p), and SRY-Box 13 (SOX13) levels were examined by quantitative real-time polymerase chain reaction (qRT-PCR). Cell viability, apoptosis, and invasion were respectively assayed using 3-(4, 5-dimethylthiazol-2-y1)-2, 5-diphenyl tetrazolium bromide (MTT), flow cytometry, and transwell assay. Glycolysis was evaluated by glucose consumption, lactate production, ATP/ADP ratio, and the detection of related enzymes. Associated proteins were measured using Western blot. Target relation was verified via dual-luciferase reporter assay. Xenograft tumor assay was implemented to study the influence of MALAT1 on MM in vivo. Results The up-regulation of MALAT1 and the down-regulation of miR-1271-5p were found in MM serums and cells. MALAT1 knockdown suppressed cell viability, invasion, and glycolysis while expedited cell apoptosis in MM cells. MALAT1 directly targeted miR-1271-5p and miR-1271-5p depression reverted the effects of MALAT1 knockdown on MM cells. SOX13 was a target of miR-1271-5p and SOX13 overexpression weakened the effects of miR-1271-5p on MM. MALAT1 indirectly modulated SOX13 expression through targeting miR-1271-5p. MALAT1 down-regulation inhibited MM growth by miR-1271-5p/SOX13 axis in vivo. Conclusion LncRNA MALAT1 expedited MM tumorigenesis, invasion, and glycolysis via miR-1271-5p/SOX13 axis. MALAT1 might contribute to the therapy of MM as a promising indicator.
Collapse
Affiliation(s)
- Na Liu
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Song Feng
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Huanhuan Li
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Xiaoguang Chen
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Songting Bai
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Yufeng Liu
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
49
|
Zhang H, Wei P, Lv W, Han X, Yang J, Qin S. Long noncoding RNA lnc-DILC stabilizes PTEN and suppresses clear cell renal cell carcinoma progression. Cell Biosci 2019; 9:81. [PMID: 31592114 PMCID: PMC6775667 DOI: 10.1186/s13578-019-0345-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 09/25/2019] [Indexed: 02/06/2023] Open
Abstract
Background Increasing evidence has indicated that long noncoding RNAs (lncRNAs) are crucial regulators affecting the progression of human cancers. Recently, lncRNA downregulated in liver cancer stem cells (lnc-DILC) was identified to function as a tumor suppressor inhibiting the tumorigenesis and metastasis in liver cancer and colorectal cancer. However, to date, little is known about the functional roles of lnc-DILC in modulating malignant phenotypes of clear cell renal cell carcinoma (ccRCC) cells. Methods lnc-DILC expression in human ccRCC tissues was detected by qRT-PCR. Overexpression and knockdown experiments were carried out to determine the effects of lnc-DILC on ccRCC cell proliferation, migration and invasion. To reveal the underlying mechanisms of lnc-DILC functions in ccRCC cells. RNA immunoprecipitation, RNA pull-down, in vivo ubiquitination, co-immunoprecipitation and western blot assays were performed. Results Here, we identified that lnc-DILC levels were dramatically downregulated in ccRCC tissues. Loss of lnc-DILC expression was correlated with larger tumor size, advanced tumor grade and lymph node metastasis, and also predicted worse prognosis in patients with ccRCC. Functionally, knockdown and overexpression experiments demonstrated that lnc-DILC inhibited cell proliferation, migration and invasion in ccRCC cells. Mechanistic investigation revealed that lnc-DILC bound to tumor suppressor PTEN and suppressed its degradation. lnc-DILC repressed the PTEN ubiquitination through blocking the interaction between PTEN and E3 ubiquitin ligase WWP2 and recruiting the deubiquitinase USP11 to PTEN. Moreover, we demonstrated that PTEN–AKT signaling was crucial for lnc-DILC-mediated suppressive effects. Conclusions In summary, our research revealed a novel mechanism by which lnc-DILC regulates PTEN stability via WWP2 and USP11, and shed light on potential therapeutic strategies by the restoration of lnc-DILC expression in patients with ccRCC.
Collapse
Affiliation(s)
- Han Zhang
- Urology Department, Luoyang Central Hospital, No. 288, Zhongzhou Road, Luoyang, 471000 Henan China
| | - Pengtao Wei
- Urology Department, Luoyang Central Hospital, No. 288, Zhongzhou Road, Luoyang, 471000 Henan China
| | - Wenwei Lv
- Urology Department, Luoyang Central Hospital, No. 288, Zhongzhou Road, Luoyang, 471000 Henan China
| | - Xingtao Han
- Urology Department, Luoyang Central Hospital, No. 288, Zhongzhou Road, Luoyang, 471000 Henan China
| | - Jinhui Yang
- Urology Department, Luoyang Central Hospital, No. 288, Zhongzhou Road, Luoyang, 471000 Henan China
| | - Shuaifeng Qin
- Urology Department, Luoyang Central Hospital, No. 288, Zhongzhou Road, Luoyang, 471000 Henan China
| |
Collapse
|
50
|
Zhang H, Li W, Gu W, Yan Y, Yao X, Zheng J. MALAT1 accelerates the development and progression of renal cell carcinoma by decreasing the expression of miR-203 and promoting the expression of BIRC5. Cell Prolif 2019; 52:e12640. [PMID: 31250518 PMCID: PMC6797509 DOI: 10.1111/cpr.12640] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 04/30/2019] [Accepted: 05/04/2019] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE We aimed to investigate the roles of the lncRNA MALAT1 in renal cell carcinoma (RCC) progression. METHODS qRT-PCR was used for the assessment of BIRC5, miRNA-203 and MALAT1 expression. Furthermore, the targeted relationships between miR-203 and BIRC5, as well as MALAT1 and miR-203, were predicted by the miRanda/starBase database and verified by dual-luciferase reporter gene assay. The effects of MALAT1, miRNA-203 and BIRC5 on cell proliferation, cell cycle, cell apoptosis, cell invasion and cell migration were studied by using CCK-8, flow cytometry, transwell and wound healing assays, respectively. In addition, the effects of MALAT1 on RCC tumorigenesis were evaluated in vivo by nude mouse tumorigenesis. RESULTS The expression levels of BIRC5 and MALAT1 were higher in RCC tissues and cell lines than in adjacent normal tissues and a normal renal cortex proximal tubule epithelial cell line. In contrast, the expression of miRNA-203 in RCC tissues and cell lines was higher than that in adjacent normal tissues and a normal renal cortex proximal tubule epithelial cell line. BIRC5 and MALAT1 promoted cell proliferation yet decreased the percentage of RCC cells at G0/G1 phase. CONCLUSIONS Our study demonstrated that MALAT1 functions as a miR-203 decoy to increase BIRC5 expression in RCC.
Collapse
Affiliation(s)
- Haimin Zhang
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wei Li
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wenyu Gu
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yang Yan
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xudong Yao
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Junhua Zheng
- Department of Urology, Shanghai General Hospital, The First People's Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|