1
|
Gujrati H, Ha S, Wang BD. Deregulated microRNAs Involved in Prostate Cancer Aggressiveness and Treatment Resistance Mechanisms. Cancers (Basel) 2023; 15:3140. [PMID: 37370750 PMCID: PMC10296615 DOI: 10.3390/cancers15123140] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/06/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Prostate cancer (PCa) is the most frequently diagnosed cancer and the second leading cause of cancer deaths among American men. Complex genetic and epigenetic mechanisms are involved in the development and progression of PCa. MicroRNAs (miRNAs) are short noncoding RNAs that regulate protein expression at the post-transcriptional level by targeting mRNAs for degradation or inhibiting protein translation. In the past two decades, the field of miRNA research has rapidly expanded, and emerging evidence has revealed miRNA dysfunction to be an important epigenetic mechanism underlying a wide range of diseases, including cancers. This review article focuses on understanding the functional roles and molecular mechanisms of deregulated miRNAs in PCa aggressiveness and drug resistance based on the existing literature. Specifically, the miRNAs differentially expressed (upregulated or downregulated) in PCa vs. normal tissues, advanced vs. low-grade PCa, and treatment-responsive vs. non-responsive PCa are discussed. In particular, the oncogenic and tumor-suppressive miRNAs involved in the regulation of (1) the synthesis of the androgen receptor (AR) and its AR-V7 splice variant, (2) PTEN expression and PTEN-mediated signaling, (3) RNA splicing mechanisms, (4) chemo- and hormone-therapy resistance, and (5) racial disparities in PCa are discussed and summarized. We further provide an overview of the current advances and challenges of miRNA-based biomarkers and therapeutics in clinical practice for PCa diagnosis/prognosis and treatment.
Collapse
Affiliation(s)
- Himali Gujrati
- Department of Pharmaceutical Sciences, University of Maryland Eastern Shore School of Pharmacy, Princess Anne, MD 21853, USA
| | - Siyoung Ha
- Department of Pharmaceutical Sciences, University of Maryland Eastern Shore School of Pharmacy, Princess Anne, MD 21853, USA
| | - Bi-Dar Wang
- Department of Pharmaceutical Sciences, University of Maryland Eastern Shore School of Pharmacy, Princess Anne, MD 21853, USA
- Hormone Related Cancers Program, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
| |
Collapse
|
2
|
Saleh AA, El-Hefnawy SM, Kasemy ZA, Alhagaa AA, Nooh MZ, Arafat ES. Mi-RNA-93 and Mi-RNA-152 in the Diagnosis of Type 2 Diabetes and Diabetic Retinopathy. Br J Biomed Sci 2022; 79:10192. [PMID: 35996507 PMCID: PMC8915732 DOI: 10.3389/bjbs.2021.10192] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/23/2021] [Indexed: 12/03/2022]
Abstract
Background and Aim: Diabetes mellitus (DM) is a chronic disorder with diabetic retinopathy (DR) as one of its main microvascular outcomes, being a prime cause of vision loss. Dysregulation of microRNAs (miRNAs) has been associated with some diabetic microvascular complications such as diabetic retinopathy. This hypothesised changes in the serum of miR-93 and miR-152 in diabetes and diabetic retinopathy. Methods: The study cohort consisted of 80 healthy volunteers, 80 type 2 diabetic patients, and 80 diabetic retinopathy patients, of whom 40 had proliferative (PDR) and 40 non-proliferative retinopathy (NPDR). Serum fasting and 2-hour postprandial glucose (2hPP), glycated haemoglobin (HbA1c), fasting insulin, and HOMA-IR were evaluated by routine methods, miR-93 and miR-152 expression by quantitative real-time PCR. Results: FBG, 2hPP, fasting insulin, HOMA-IR, and miR-152 showed an increasing trend across groups while miR-93 showed a decreasing trend (all p < 0.001). Binary logistic regression analysis for prediction of DR found that the most significant were miR-152 (OR 1.37, 95% CI: 1.18-1.58, <0.001), BMI (1.13, [1.07-1.31], p = 0.004), duration of disease (1.29 [1.04-1.6] p = 0.018), and miR-152 (0.01, [0.0-0.47] p = 0.019). The most significant predictors of PDR were miR-152 (OR = 1.47, 95% CI: 1.12-1.92, p = 0.005), HOMA-IR (2.66 [1.30-5.45] p = 0.007), and miR-93 (0.25 [0.07-0.86] p = 0.028). Conclusion: MiR-93 and miR-152 can differentiate patients with diabetes and those with DR. Both miRNAs might be potential biomarkers for diabetes and diabetic retinopathy, and specifically for proliferative diabetic retinopathy.
Collapse
Affiliation(s)
- A. A. Saleh
- Department of Medical Biochemistry and Molecular Biology , Faculty of Medicine, Menoufia University, Shebein Alkom, Egypt
| | - S. M. El-Hefnawy
- Department of Medical Biochemistry and Molecular Biology , Faculty of Medicine, Menoufia University, Shebein Alkom, Egypt
| | - Z. A. Kasemy
- Department of Public Health and Community Medicine, Faculty of Medicine, Menoufia University, Shebein Alkom, Egypt
| | - A. A. Alhagaa
- Department of Ophthalmology, Faculty of Medicine, Menoufia University, Shebein Alkom, Egypt
| | - M. Z. Nooh
- Department of Internal Medicine, Faculty of Medicine, Menoufia University, Shebein Alkom, Egypt
| | - E. S. Arafat
- Department of Medical Biochemistry and Molecular Biology , Faculty of Medicine, Menoufia University, Shebein Alkom, Egypt
| |
Collapse
|
3
|
Gurbuz V, Sozen S, Bilen CY, Konac E. miR-148a, miR-152 and miR-200b promote prostate cancer metastasis by targeting DNMT1 and PTEN expression. Oncol Lett 2021; 22:805. [PMID: 34630712 PMCID: PMC8488332 DOI: 10.3892/ol.2021.13066] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 09/07/2021] [Indexed: 12/14/2022] Open
Abstract
MicroRNAs (miRs) modulate the expression of target genes in the signal pathway on transcriptome level. The present study investigated the ‘epigenetic-based miRNA (epi-miRNA)-mRNA’ regulatory network of miR-34b, miR-34c, miR-148a, miR-152, miR-200a and miR-200b epi-miRNAs and their target genes, DNA methyltransferase (DNMT1, 3a and 3b), phosphate and tensin homolog (PTEN) and NK3 Homeobox 1 (NKX3.1), in prostate cancer (PCa) using reverse transcription-quantitative PCR. The expression level of NKX3.1 were not significantly different between the PCa, Met-PCa and control groups. However, in the PCa and Met-PCa groups, the expression level of DNMT1 was upregulated, while DNMT3a, DNMT3b and PTEN were downregulated. Overexpression of DNMT1 (~5 and ~6-fold increase in the PCa and Met-PCa groups respectively) was accompanied by a decreased expression in PTEN, indicating a potential negative association. Both groups indicated that a high level of DNMT1 is associated with the aggressiveness of cancer, and there is a a directly proportional relationship between this gene and PSA, GS and TNM staging. A significant ~2 to ~5-fold decrease in the expression levels of DNMT3a and DNMT3b was found in both groups. In the PCa group, significant associations were identified between miR-34b and DNMT1/DNMT3b; between miR-34c/miR-148a and all target genes; between miR-152 and DNMT1/DNMT3b and PTEN; and between miR-200a/b and DNMT1. In the Met-PCa group, miR-148a, miR-152 and miR-200b exhibited a significant association with all target genes. A significant negative association was identified between PTEN and DNMT1 in the Met-PCa group. It was also revealed that that miR-148a, miR-152 and miR-200b increased the expression of DNMT1 and suppressed PTEN. Furthermore, the ‘epi-miRNA-mRNA’ bidirectional feedback loop was emphasised and the methylation pattern in PCa anti-cancer therapeutics was highlighted.
Collapse
Affiliation(s)
- Venhar Gurbuz
- Department of Medical Biology and Genetics, Faculty of Medicine, Gazi University, Ankara 06510, Turkey
| | - Sinan Sozen
- Department of Urology, Faculty of Medicine, Gazi University, Ankara 06510, Turkey
| | - Cenk Y Bilen
- Department of Urology, Faculty of Medicine, Hacettepe University, Ankara 06100, Turkey
| | - Ece Konac
- Department of Medical Biology and Genetics, Faculty of Medicine, Gazi University, Ankara 06510, Turkey
| |
Collapse
|
4
|
Carron J, Torricelli C, Silva JK, Queiroz GSR, Ortega MM, Lima CSP, Lourenço GJ. microRNAs deregulation in head and neck squamous cell carcinoma. Head Neck 2020; 43:645-667. [PMID: 33159410 DOI: 10.1002/hed.26533] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 09/30/2020] [Accepted: 10/23/2020] [Indexed: 12/24/2022] Open
Abstract
Head and neck (HN) squamous cell carcinoma (SCC) is the eighth most common human cancer worldwide. Besides tobacco and alcohol consumption, genetic and epigenetic alterations play an important role in HNSCC occurrence and progression. microRNAs (miRNAs) are small noncoding RNAs that regulate cell cycle, proliferation, development, differentiation, and apoptosis by interfering in gene expression. Expression profiling of miRNAs showed that some miRNAs are upregulated or downregulated in tumor cells when compared with the normal cells. The present review focuses on the role of miRNAs deregulations in HNSCC, enrolled in risk, development, outcome, and therapy sensitivity. Moreover, the influence of single nucleotide variants in miRNAs target sites, miRNAs seed sites, and miRNAs-processing genes in HNSCC was also revised. Due to its potential for cancer diagnosis, progression, and as a therapeutic target, miRNAs may bring new perspectives in HNSCC understanding and therapy, especially for those patients with no or insufficient treatment options.
Collapse
Affiliation(s)
- Juliana Carron
- Laboratory of Cancer Genetics, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Caroline Torricelli
- Laboratory of Cancer Genetics, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Janet K Silva
- Laboratory of Cancer Genetics, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Gabriela S R Queiroz
- Laboratory of Cancer Genetics, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Manoela M Ortega
- Laboratory of Cell and Molecular Tumor Biology and Bioactive Compounds, São Francisco University, Bragança Paulista, Brazil
| | - Carmen S P Lima
- Laboratory of Cancer Genetics, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Gustavo J Lourenço
- Laboratory of Cancer Genetics, School of Medical Sciences, University of Campinas, Campinas, Brazil
| |
Collapse
|
5
|
Bryant J, White L, Coen N, Shields L, McClean B, Meade AD, Lyng FM, Howe O. MicroRNA Analysis of ATM-Deficient Cells Indicate PTEN and CCDN1 as Potential Biomarkers of Radiation Response. Radiat Res 2020; 193:520-530. [PMID: 32216710 DOI: 10.1667/rr15462.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 03/06/2020] [Indexed: 11/03/2022]
Abstract
Genetic and epigenetic profile changes associated with individual radiation sensitivity are well documented and have led to enhanced understanding of the mechanisms of the radiation-induced DNA damage response. However, the search continues to identify reliable biomarkers of individual radiation sensitivity. Herein, we report on a multi-biomarker approach using traditional cytogenetic biomarkers, DNA damage biomarkers and transcriptional microRNA (miR) biomarkers coupled with their potential gene targets to identify radiosensitivity in ataxia-telangiectasia mutated (ATM)-deficient lymphoblastoid cell lines (LCL); ATM-proficient cell lines were used as controls. Cells were 0.05 and 0.5 Gy irradiated, using a linear accelerator, with sham-irradiated cells as controls. At 1 h postirradiation, cells were fixed for γ-H2AX analysis as a measurement of DNA damage, and cytogenetic analysis using the G2 chromosomal sensitivity assay, G-banding and FISH techniques. RNA was also isolated for genetic profiling by microRNA (miR) and RT-PCR analysis. A panel of 752 miR were analyzed, and potential target genes, phosphatase and tensin homolog (PTEN) and cyclin D1 (CCND1), were measured. The cytogenetic assays revealed that although the control cell line had functional cell cycle checkpoints, the radiosensitivity of the control and AT cell lines were similar. Analysis of DNA damage in all cell lines, including an additional control cell line, showed elevated γ-H2AX levels for only one AT cell line. Of the 752 miR analyzed, eight miR were upregulated, and six miR were downregulated in the AT cells compared to the control. Upregulated miR-152-3p, miR-24-5p and miR-92-15p and all downregulated miR were indicated as modulators of PTEN and CCDN1. Further measurement of both genes validated their potential role as radiation-response biomarkers. The multi-biomarker approach not only revealed potential candidates for radiation response, but provided additional mechanistic insights into the response in AT-deficient cells.
Collapse
Affiliation(s)
- Jane Bryant
- Radiation and Environmental Science Centre (RESC), FOCAS Research Institute
| | - Lisa White
- Radiation and Environmental Science Centre (RESC), FOCAS Research Institute.,School of Biological and Health Sciences, Technological University Dublin, City Campus, Dublin 8, Ireland
| | - Natasha Coen
- Department of Clinical Genetics, Division of Cytogenetics, Our Lady's Children's Hospital, Crumlin, Dublin 12, Ireland
| | - Laura Shields
- Medical Physics Department, St Luke's Radiation Oncology Centre, Rathgar, Dublin 6, Ireland
| | - Brendan McClean
- Medical Physics Department, St Luke's Radiation Oncology Centre, Rathgar, Dublin 6, Ireland
| | - Aidan D Meade
- Radiation and Environmental Science Centre (RESC), FOCAS Research Institute.,School of Physics & Clinical & Optometric Sciences, Technological University Dublin, City Campus, Dublin 8, Ireland
| | - Fiona M Lyng
- Radiation and Environmental Science Centre (RESC), FOCAS Research Institute.,School of Physics & Clinical & Optometric Sciences, Technological University Dublin, City Campus, Dublin 8, Ireland
| | - Orla Howe
- Radiation and Environmental Science Centre (RESC), FOCAS Research Institute.,School of Biological and Health Sciences, Technological University Dublin, City Campus, Dublin 8, Ireland
| |
Collapse
|
6
|
Zhang T, Lei F, Jiang T, Xie L, Huang P, Li P, Huang Y, Tang X, Gong J, Lin Y, Cheng A, Huang W. H19/miR-675-5p Targeting SFN Enhances the Invasion and Metastasis of Nasalpharyngeal Cancer Cells. Curr Mol Pharmacol 2020; 12:324-333. [PMID: 31677258 DOI: 10.2174/1874467212666190719120446] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 05/27/2019] [Accepted: 06/18/2019] [Indexed: 12/25/2022]
Abstract
AIMS The aim is to study the role of miR-675-5p coded by long non-coding RNA H19 in the development of Nasopharyngeal Cancer (NPC) and whether miR-675-5p regulates the invasion and metastasis of NPC through targeting SFN (14-3-3σ). The study further validated the relationship between H19, miR-675-5p and SFN in NPC and their relationship with the invasion and metastasis of NPC. METHODS Western blot was used to detect the expression of 14-3-3σ protein in immortalized normal nasopharyngeal epithelial cells NP69 and different metastatic potential NPC cells, 6-10B and 5-8F. At the same time, to find out the relationship between 14-3-3σ protein and the expression of H19 and miR-675-5p, the expression of H19 and miR-675-5p in normal nasopharynx epithelial cells NP69 and varied nasopharyngeal carcinoma cells 6-10B and 5-8F were quantified by real-time PCR. MiR-675-5p mimic and inhibitor were transfected into NPC 6-10B to over-express and down-express miR-675-5p; miR-675-5p mimic negative control and inhibitor negative control were transfected into NPC 6-10B as control groups. The effect of over-expression and down-expression by miR-675-5p on the expression of 14-3-3σ protein was detected by Western blotting. The 3'-UTR segments of SFN, containing miR-675-5p binding sites were amplified by PCR and the luciferase activity in the transfected cells was assayed to detect whether SFN is the direct target of miR-675-5p. Transwell and scratch assays were used to verify the changes in NPC invasion and metastasis ability of mimics and inhibitors transfected with miR-675-5p. RESULTS The expression of 14-3-3σ protein in normal nasopharynx epithelial cells NP69 is significantly higher than in varied nasopharyngeal carcinoma cells, 6-10B and 5-8F (P<0.05), and the 14-3-3σ protein levels in low-metastatic nasopharyngeal carcinoma cell 6-10B is higher than in high-metastatic nasopharyngeal carcinoma cell 5-8F. The expression of H19 and miR-675-5p are significantly higher in NPC cells than in NP69 cell (P<0.05). The expression of H19 and miR-675-5p in high-Metastatic nasopharyngeal carcinoma cell 5-8F was higher than in low-Metastatic nasopharyngeal carcinoma cell 6-10B. The expression of 14-3-3σ protein in miR-675-5p mimic cells was significantly lower than in mimic NC (negative control) group and blank control group. However, compared with the blank control group, mimic NC showed no significant difference in 14-3-3σ protein between the two groups. The miR-675-5p inhibitor group was significantly higher than the inhibitor NC group and the blank control group (p<0.05), but there was no significant difference in the expression of 14-3-3σ protein in the inhibitor NC group and the blank control group (p>0.05). Dual-luciferase reporter assay system shows the 3'-UTR segments of SFN containing miR-675-5p binding sites. SFN was the target gene of miR-675-5p. CONCLUSION 14-3-3σ is downregulated in NPC and is involved in the development of NPC. H19 and miR- 675-5p are upregulated in NPC, which is related to the development of NPC. The over-expression of miR- 675-5p inhibits the expression of 14-3-3σ protein. SFN is the target gene of miR-675-5p. MiR-675-5p targets SFN, downregulates its protein expression and promotes the invasion and metastasis of NPC.
Collapse
Affiliation(s)
- Ting Zhang
- Cancer Research Institute, Hengyang Medical College of University of South China, Hengyang 421001, Hunan Province, China
| | - Fanghong Lei
- Cancer Research Institute, Hengyang Medical College of University of South China, Hengyang 421001, Hunan Province, China
| | - Tao Jiang
- Cancer Research Institute, Hengyang Medical College of University of South China, Hengyang 421001, Hunan Province, China
| | - Lisha Xie
- Cancer Research Institute, Hengyang Medical College of University of South China, Hengyang 421001, Hunan Province, China
| | - Pin Huang
- Cancer Research Institute, Hengyang Medical College of University of South China, Hengyang 421001, Hunan Province, China
| | - Pei Li
- Cancer Research Institute, Hengyang Medical College of University of South China, Hengyang 421001, Hunan Province, China
| | - Yun Huang
- Cancer Research Institute, Hengyang Medical College of University of South China, Hengyang 421001, Hunan Province, China
| | - Xia Tang
- Hengyang Medical College of University of South China, Hengyang 421001, Hunan Province, China
| | - Jie Gong
- Hengyang Medical College of University of South China, Hengyang 421001, Hunan Province, China
| | - Yunpeng Lin
- Hengyang Medical College of University of South China, Hengyang 421001, Hunan Province, China
| | - Ailan Cheng
- Cancer Research Institute, Hengyang Medical College of University of South China, Hengyang 421001, Hunan Province, China.,Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology (2016TP1015), Hengyang 421001, Hunan Province, China
| | - Weiguo Huang
- Cancer Research Institute, Hengyang Medical College of University of South China, Hengyang 421001, Hunan Province, China.,Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology (2016TP1015), Hengyang 421001, Hunan Province, China
| |
Collapse
|
7
|
Bekric D, Neureiter D, Ritter M, Jakab M, Gaisberger M, Pichler M, Kiesslich T, Mayr C. Long Non-Coding RNAs in Biliary Tract Cancer-An Up-to-Date Review. J Clin Med 2020; 9:jcm9041200. [PMID: 32331331 PMCID: PMC7231154 DOI: 10.3390/jcm9041200] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 03/31/2020] [Accepted: 04/07/2020] [Indexed: 02/07/2023] Open
Abstract
The term long non-coding RNA (lncRNA) describes non protein-coding transcripts with a length greater than 200 base pairs. The ongoing discovery, characterization and functional categorization of lncRNAs has led to a better understanding of the involvement of lncRNAs in diverse biological and pathological processes including cancer. Aberrant expression of specific lncRNA species was demonstrated in various cancer types and associated with unfavorable clinical characteristics. Recent studies suggest that lncRNAs are also involved in the development and progression of biliary tract cancer, a rare disease with high mortality and limited therapeutic options. In this review, we summarize current findings regarding the manifold roles of lncRNAs in biliary tract cancer and give an overview of the clinical and molecular consequences of aberrant lncRNA expression as well as of underlying regulatory functions of selected lncRNA species in the context of biliary tract cancer.
Collapse
Affiliation(s)
- Dino Bekric
- Institute of Physiology and Pathophysiology, Paracelsus Medical University, 5020 Salzburg, Austria; (D.B.); (M.R.); (M.J.); (M.G.); (T.K.)
| | - Daniel Neureiter
- Institute of Pathology, Paracelsus Medical University/Salzburger Landeskliniken (SALK), 5020 Salzburg, Austria;
- Cancer Cluster Salzburg, 5020 Salzburg, Austria
| | - Markus Ritter
- Institute of Physiology and Pathophysiology, Paracelsus Medical University, 5020 Salzburg, Austria; (D.B.); (M.R.); (M.J.); (M.G.); (T.K.)
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Paracelsus Medical University, 5020 Salzburg, Austria
- Gastein Research Institute, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Martin Jakab
- Institute of Physiology and Pathophysiology, Paracelsus Medical University, 5020 Salzburg, Austria; (D.B.); (M.R.); (M.J.); (M.G.); (T.K.)
| | - Martin Gaisberger
- Institute of Physiology and Pathophysiology, Paracelsus Medical University, 5020 Salzburg, Austria; (D.B.); (M.R.); (M.J.); (M.G.); (T.K.)
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Paracelsus Medical University, 5020 Salzburg, Austria
- Gastein Research Institute, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Martin Pichler
- Research Unit of Non-Coding RNAs and Genome Editing, Division of Clinical Oncology, Department of Medicine, Comprehensive Cancer Center Graz, Medical University of Graz, 8036 Graz, Austria;
| | - Tobias Kiesslich
- Institute of Physiology and Pathophysiology, Paracelsus Medical University, 5020 Salzburg, Austria; (D.B.); (M.R.); (M.J.); (M.G.); (T.K.)
- Department of Internal Medicine I, Paracelsus Medical University/Salzburger Landeskliniken (SALK), 5020 Salzburg, Austria
| | - Christian Mayr
- Institute of Physiology and Pathophysiology, Paracelsus Medical University, 5020 Salzburg, Austria; (D.B.); (M.R.); (M.J.); (M.G.); (T.K.)
- Department of Internal Medicine I, Paracelsus Medical University/Salzburger Landeskliniken (SALK), 5020 Salzburg, Austria
- Correspondence:
| |
Collapse
|
8
|
Effect of adenovirus-mediated overexpression of PTEN on brain oxidative damage and neuroinflammation in a rat kindling model of epilepsy. Chin Med J (Engl) 2020; 132:2628-2635. [PMID: 31658159 PMCID: PMC6846256 DOI: 10.1097/cm9.0000000000000496] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background: Epilepsy is a chronic and severe neurological disorder. Phosphatase and tensin homolog deleted on chromosome ten (PTEN)-deficient mice exhibit learning and memory deficits and spontaneous epilepsy. The aim of this study was to investigate the role of PTEN in brain oxidative damage and neuroinflammation in a rat model of epilepsy. Methods: An adenovirus (Ad)-PTEN vector was constructed, and status epilepticus (SE) was induced in 41 model rats using lithium chloride-pilocarpine. Thirty-six SE rats were then allocated into the Ad-PTEN, Ad-LacZ, and SE groups, those were administered intracerebroventricular injections of Ad-PTEN, Ad-enhanced green fluorescent protein, and phosphate buffer saline, respectively. The normal group was comprised of healthy Sprague-Dawley rats. Nissl staining was conducted to evaluate neuronal damage, and immunohistochemistry was conducted to observe the morphology of cells in the hippocampal CA1 region and the distribution of ionized calcium-binding adaptor molecule 1 (Iba1) and ED1 (rat homologue of human CD68). Levels of apoptosis-related proteins, inflammatory-related factors, and oxidative stress-related markers (reactive oxygen species [ROS], glutathione [GSH], superoxide dismutase [SOD], and malondialdehyde [MDA]) were measured. Comparisons between multiple groups were conducted using one-way analysis of variance (ANOVA), and pairwise comparisons after ANOVA were conducted using the Tukey multiple comparisons test. Results: After SE induction, PTEN expression in the rat brain exhibited a four-fold decrease (P = 0.000) and the expression of both Iba1 and ED1 increased. Furthermore, significant neuronal loss, oxidative damage, and neuroinflammation were observed in the SE rat brain. After intracerebroventricular injection of Ad-PTEN, PTEN expression exhibited a three-fold increase (P = 0.003), and the expression of both Iba1 and ED1 decreased. Additionally, neurons were restored and neuronal apoptosis was inhibited. Furthermore, ROS and MDA levels decreased, GSH level and SOD activity increased, and neuroinflammation was reduced. Conclusion: Our study demonstrated that brain oxidative damage and neuroinflammation in SE rats were ameliorated by intracerebroventricular injection of Ad-PTEN.
Collapse
|
9
|
Li Y, Huang S, Wei Z, Yang B. A putative competing endogenous RNA network in cisplatin-resistant lung adenocarcinoma cells identifying potentially rewarding research targets. Oncol Lett 2020; 19:4040-4052. [PMID: 32382346 PMCID: PMC7202328 DOI: 10.3892/ol.2020.11483] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 02/28/2020] [Indexed: 12/12/2022] Open
Abstract
Lung adenocarcinoma (LUAD) is the most common type of non-small cell lung cancer and has a poor 5 year survival rate (<10%). Cisplatin is one of the most effective chemotherapeutic treatments for LUAD, even though it is of limited overall utility due to acquired drug resistance. To identify possible genetic targets for the mitigation of cisplatin resistance, gene expression data from cisplatin-resistant cell lines were integrated with patient information. Expression data for cisplatin-resistant and cisplatin-sensitive A549 cell lines were obtained from the Gene Expression Omnibus database, while LUAD patient data was obtained from The Cancer Genome Atlas (TCGA) database. Differentially expressed mRNAs (DEmRNAs), microRNAs (DEmiRNAs) and long non-coding RNAs (DElncRNAs) were identified between the cisplatin-sensitive and cisplatin-resistant cells. Using the TCGA patient data, 33 DEmRNAs associated with survival were identified. A total of 74 DElncRNAs co-expressed with the survival-associated DEmRNAs, and 11 DEmiRNAs that regulated the survival-associated DEmRNAs, were also identified. A competing endogenous RNA (ceRNA) network was constructed based on the aforementioned results, which included 17 survival-associated DEmRNAs, 9 DEmiRNAs and 16 DElncRNAs. This network revealed 8 ceRNA pathway axes possibly associated with cisplatin resistance in A549 cells. Specifically, the network suggested that the lncRNAs HOXD-AS2, LINC01123 and FIRRE may act as ceRNAs to increase cisplatin resistance in human LUAD cells. Therefore, it was speculated that these lncRNAs represent potentially rewarding research targets.
Collapse
Affiliation(s)
- Yepeng Li
- Department of Oncology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Shiqing Huang
- Department of Oncology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Zhongheng Wei
- Department of Oncology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Bo Yang
- Key Laboratory of Guangxi College and Universities, Biomedical Research Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| |
Collapse
|
10
|
MicroRNA-152-3p protects neurons from oxygen-glucose-deprivation/reoxygenation-induced injury through upregulation of Nrf2/ARE antioxidant signaling by targeting PSD-93. Biochem Biophys Res Commun 2019; 517:69-76. [DOI: 10.1016/j.bbrc.2019.07.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 07/04/2019] [Indexed: 11/21/2022]
|
11
|
Wang Z, Zhu Z, Lin Z, Luo Y, Liang Z, Zhang C, Chen J, Peng P. miR-429 suppresses cell proliferation, migration and invasion in nasopharyngeal carcinoma by downregulation of TLN1. Cancer Cell Int 2019; 19:115. [PMID: 31068760 PMCID: PMC6492405 DOI: 10.1186/s12935-019-0831-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 04/20/2019] [Indexed: 02/06/2023] Open
Abstract
Background miR-429 and TLN1 have been shown to affect the biological behaviours of many carcinomas. However, their effects in nasopharyngeal carcinoma (NPC) are not yet clear. Here, we investigated their regulatory relationships and effects on NPC cells. Methods TargetScan was used to predict the regulatory relationships of miR-429 and TLN1 in NPC cells. Then, Western blotting and quantitative real-time PCR (qPCR) were performed to examine TLN1 levels, and qPCR was used to determine miR-429 levels in NPC cell lines with different metastatic characteristics (5-8F, CNE-2, CNE-1, 6-10B and NP69), to investigate whether TLN1 and miR-429 are correlated with the metastatic characteristics of these cells. Next, we upregulated or downregulated miR-429 in 5-8F and 6-10B cells, which have different tumourigenicity and transferability, and examined TLN1 expression by western blotting and qPCR after transfection. QPCR was also performed to confirm successful transfection of miR-429 mimic into 5-8F and 6-10B cells. Dual luciferase reporter gene assay was performed to investigate whether miR-429 regulates TLN1 by binding to its 3′UTR. After transfection, Cell Counting Kit-8 (CCK8) and IncuCyte were used to examine the proliferation of these cells, and wound-healing assay, Transwell migration assay, and invasion assays were performed to investigate the changes in migration and invasion after transfection. Results Western blotting and qPCR analyses showed that the protein level of TLN1 was negatively correlated with miR-429 in NPC cell lines (P < 0.05), while the mRNA level showed no relation with miR429 expression (P > 0.05). In addition, cells with high transferability showed high TLN1 expression at the protein level, while miR429 expression showed the opposite trend (P < 0.05), but there were no differences at the mRNA level between the different cell lines. Overexpression of miR429 in 5-8F and 6-10B cells was accompanied by downregulation of TLN1 at the protein level (P < 0.05), while there were no significant differences at the mRNA level (P > 0.05). In addition, transferability, proliferation, and invasion were downregulated by miR429 overexpression (P < 0.05). However, dual-luciferase reporter gene assay indicated that TLN1 was not a direct target of miR-429. Conclusion This study showed that miR-429 functions as a tumour suppressor in NPC by downregulation of TLN1, although the relationship is not direct.
Collapse
Affiliation(s)
- Zhihui Wang
- 1Department of Thoracic Oncology, The Cancer Center of The Fifth Affiliated Hospital of Sun Yat-sen University, 52 Meihua East Road, Zhuhai, Guangdong China
| | - Zhiquan Zhu
- 1Department of Thoracic Oncology, The Cancer Center of The Fifth Affiliated Hospital of Sun Yat-sen University, 52 Meihua East Road, Zhuhai, Guangdong China
| | - Zhong Lin
- 1Department of Thoracic Oncology, The Cancer Center of The Fifth Affiliated Hospital of Sun Yat-sen University, 52 Meihua East Road, Zhuhai, Guangdong China
| | - Youli Luo
- 1Department of Thoracic Oncology, The Cancer Center of The Fifth Affiliated Hospital of Sun Yat-sen University, 52 Meihua East Road, Zhuhai, Guangdong China
| | - Zibin Liang
- 1Department of Thoracic Oncology, The Cancer Center of The Fifth Affiliated Hospital of Sun Yat-sen University, 52 Meihua East Road, Zhuhai, Guangdong China
| | - Caibin Zhang
- 2Department of Neurology, The Fifth Affiliated Hospital of Sun Yat-sen University, 52 Meihua East Road, Zhuhai, Guangdong China
| | - Jianxu Chen
- 3Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, 52 Meihua East Road, Zhuhai, Guangdong China
| | - Peijian Peng
- 1Department of Thoracic Oncology, The Cancer Center of The Fifth Affiliated Hospital of Sun Yat-sen University, 52 Meihua East Road, Zhuhai, Guangdong China
| |
Collapse
|
12
|
Yang D, Zhang Q. miR-152 may function as an early diagnostic and prognostic biomarker in patients with cervical intraepithelial neoplasia and patients with cervical cancer. Oncol Lett 2019; 17:5693-5698. [PMID: 31186794 DOI: 10.3892/ol.2019.10233] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 02/15/2019] [Indexed: 12/11/2022] Open
Abstract
Previous studies have demonstrated that circulating miRNAs are effective biomarkers of various types of cancer. It has also been indicated that miR-152 is upregulated in cervical cancer. However, whether miR-152 may be used as an early detection method for patients with cervical cancer is yet to be elucidated. The results of the current study demonstrated that miR-152 levels were the lowest in healthy controls, high in patients with cervical intraepithelial neoplasia (CIN), and the highest in patients with cervical cancer. Furthermore, miR-152 levels in peripheral blood were higher in patients with high-grade CIN compared with those with low-grade CIN. It was also demonstrated that miR-152 levels increased as the clinical stage of cervical cancer advanced. Compared with healthy controls, squamous cell carcinoma antigen (SSC-Ag) levels were significantly higher in patients with cervical cancer. However, no significant differences were identified in patients with CIN, indicating that SCC-Ag could not be used for the early detection of CIN. In contrast, miR-152 was elevated along with SCC-Ag in patients with CIN and cervical cancer. Receiver operating characteristic (ROC) analysis demonstrated that miR-152 preferentially distinguished patients with CIN (95% confidence interval, 0.688-0.973; P<0.001) and patients with cervical cancer (95% confidence interval, 0.817-0.996; P<0.001) from healthy controls. Additionally, miR-152 levels were markedly reduced in patients with cervical cancer who received chemotherapy (28 patients) or chemotherapy and radiation therapy (22 patients). In conclusion, the level of miR-153 in peripheral blood may be utilized as an effective biomarker for the early detection of cervical cancer, thus decreasing the requirement for invasive cervical biopsies. Furthermore, it may be utilized to predict the most effective form of treatment for patients with cervical cancer.
Collapse
Affiliation(s)
- Dongmei Yang
- Department Two of Gynecology, Linyi People's Hospital, Linyi, Shandong 276003, P.R. China
| | - Qiumei Zhang
- Outpatient Department of Obstetrics and Gynecology, Linyi People's Hospital, Linyi, Shandong 276003, P.R. China
| |
Collapse
|
13
|
Chen L, Qian H, Xue J, Zhang J, Chen H. MicroRNA‑152 regulates insulin secretion and pancreatic β cell proliferation by targeting PI3Kα. Mol Med Rep 2018; 18:4113-4121. [PMID: 30106118 DOI: 10.3892/mmr.2018.9359] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 11/02/2017] [Indexed: 11/06/2022] Open
Abstract
An increasing number of microRNAs (miRNAs/miRs) are reported to have important roles in diabetes. Glucose‑stimulated insulin secretion and pancreatic β cell proliferation are essential in the control of metabolic disorder, however, the underlying molecular mechanisms remain unclear. The present study investigated the function of miR‑152 in diabetes. The results of reverse transcription‑quantitative polymerase chain reaction demonstrated that miR‑152 levels in the blood were markedly reduced in patients with diabetes compared with nondiabetic controls. In addition, a high blood glucose concentration was significantly associated with reduced miR‑152 expression. Furthermore, overexpression of miR‑152 using miR‑152 mimics promoted the proliferation of INS‑1 and MIN6 cells, as determined by an MTT assay, in addition to insulin secretion, while knockdown of miR‑152 using an inhibitor led to the opposite effects. Phosphatidylinositol 3‑kinase (PI3K) signaling has been reported to inhibit insulin secretion, however, the regulation of PI3K in the pancreatic β cell is poorly understood. The present study identified that PI3K catalytic subunit α (PI3Kα) was a direct target gene of miR‑152 using a luciferase reporter assay, and miR‑152 inhibited the expression of PI3Kα at the protein level, which was determined by western blotting. Therefore, the regulation of insulin secretion and pancreatic β cell proliferation may occur via the miR‑152/PI3Kα axis. The overexpression of PI3Kα in INS‑1 and MIN6 cells partially reduced the effects of miR‑152 overexpression on insulin secretion. Consistently, PI3Kα levels were reduced in murine pancreatic islets following treatment with 20 mM glucose, and increased in blood samples from patients with diabetes compared with healthy individuals. In conclusion, the results of the present study demonstrate that miR‑152 may have an important role in pancreatic β cell function, and established an association between miR‑152 and the PI3Kα axis. Therefore, targeting PI3Kα may be a potential therapeutic option for diabetes.
Collapse
Affiliation(s)
- Li Chen
- Department of Endocrinology, Jingzhou Central Hospital, The Second Clinical Medical College, Yangtze University, Jingzhou, Hubei 434020, P.R. China
| | - Haiyun Qian
- Department of Cardiothoracic Surgery, Jingzhou Central Hospital, The Second Clinical Medical College, Yangtze University, Jingzhou, Hubei 434020, P.R. China
| | - Junli Xue
- Department of Endocrinology, Jingzhou Central Hospital, The Second Clinical Medical College, Yangtze University, Jingzhou, Hubei 434020, P.R. China
| | - Juan Zhang
- Department of Endocrinology, Jingzhou Central Hospital, The Second Clinical Medical College, Yangtze University, Jingzhou, Hubei 434020, P.R. China
| | - Hao Chen
- Department of Cardiothoracic Surgery, Jingzhou Central Hospital, The Second Clinical Medical College, Yangtze University, Jingzhou, Hubei 434020, P.R. China
| |
Collapse
|
14
|
Nogueira I, Dias F, Teixeira AL, Medeiros R. miRNAs as potential regulators of mTOR pathway in renal cell carcinoma. Pharmacogenomics 2018; 19:249-261. [PMID: 29334302 DOI: 10.2217/pgs-2017-0160] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Renal cell carcinoma (RCC) is the most commonly occurring solid cancer of the adult kidney with the majority of RCC cases being detected accidentally. The most aggressive subtype is clear cell RCC (ccRCC). miRNAs, a family of small noncoding RNAs regulating gene expression have been identified as key biological modulators. The von Hippel-Lindau pathway is one of the signaling pathways involved in the pathophysiology of ccRCC. Another oncogenic mechanism involves the activation of PI3K/AKT/mTOR signaling and serves as a central regulator of cell metabolism, proliferation and survival. Several studies have described the involvement of miRNA dysregulation in the pathogenesis and progression of ccRCC. These molecules can be considered as potential diagnostic and prognostic biomarkers, allowing response to therapy to be monitored.
Collapse
Affiliation(s)
- Inês Nogueira
- Molecular Oncology and Viral Pathology Group, IPO-Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal.,FMUP, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Francisca Dias
- Molecular Oncology and Viral Pathology Group, IPO-Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal.,ICBAS, Abel Salazar Institute for the Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal
| | - Ana Luísa Teixeira
- Molecular Oncology and Viral Pathology Group, IPO-Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, IPO-Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal.,FMUP, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal.,Research Department, LPCC-Portuguese League, Against Cancer (NRNorte), 4200-172 Porto, Portugal.,CEBIMED, Faculty of Health Sciences, Fernando Pessoa University, 4200-150 Porto, Portugal
| |
Collapse
|
15
|
Zhang S, Xiao J, Chai Y, Du YY, Liu Z, Huang K, Zhou X, Zhou W. LncRNA-CCAT1 Promotes Migration, Invasion, and EMT in Intrahepatic Cholangiocarcinoma Through Suppressing miR-152. Dig Dis Sci 2017; 62:3050-3058. [PMID: 28921383 DOI: 10.1007/s10620-017-4759-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 09/08/2017] [Indexed: 01/17/2023]
Abstract
BACKGROUND Increasing evidence has suggested that lncRNA CCAT1 is upregulated and functions as a potential tumor promoter in many cancers. However, the potential biological roles and regulatory mechanisms of CCAT1 in intrahepatic cholangiocarcinoma (ICC) remain unclear. METHODS We used real-time PCR to measure CCAT1 expression in ICC tissues and the adjacent normal tissues. The statistical analyses were applied to evaluate the prognostic value and associations of CCAT1 expression with clinical parameters. The CCAT1 was silenced with siRNA in ICC cells. The migration and invasion of ICC cells were detected with Transwell assay. The expressions of epithelial-mesenchymal transition (EMT)-related proteins were evaluated to discover whether the process of EMT was involved. RESULTS We found that CCAT1 expression was elevated in ICC tissues compared to the adjacent normal tissues. We also found that high CCAT1 expression is closely correlated with tumor progression in ICC patients. Furthermore, our results show that knockdown of CCAT1 significantly suppressed the migration and invasion of ICC cells. Additionally, CCAT1 silencing remarkably reverses the EMT phenotype of ICC cells. Moreover, bioinformatics analysis and luciferase reporter assay revealed that CCAT1 directly bound to the miR-152, which has been reported to serve as a tumor suppressor in variety cancers. Further investigation demonstrated that CCAT1 led to the metastasis and EMT activation of ICC cells through inhibiting miR-152. CONCLUSIONS Our results suggested that CCAT1 functions as an oncogenic lncRNA in ICC, which could serve as a potential diagnostic and therapeutic target for ICC patients.
Collapse
Affiliation(s)
- Shouhua Zhang
- Department of General Surgery, Jiangxi Children's Hospital, Nanchang, 330006, Jiangxi Province, China
| | - Juhua Xiao
- Department of Ultrasound, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, 330006, Jiangxi Province, China
| | - Yong Chai
- Department of General Surgery, Jiangxi Children's Hospital, Nanchang, 330006, Jiangxi Province, China
| | - Yun Yan Du
- Department of Otolaryngology, Jiangxi Provincial People's Hospital, Nanchang, 330006, Jiangxi Province, China
| | - Zhiqiang Liu
- Department of General Surgery, Jiangxi Children's Hospital, Nanchang, 330006, Jiangxi Province, China
| | - Kai Huang
- Department of Gastrointestinal Surgery, Jiangxi Provincial Cancer Hospital, No. 519, Beijing East Road, Nanchang, 330029, Jiangxi Province, China
| | - Xin Zhou
- Department of Ultrasound, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, 330006, Jiangxi Province, China
| | - Wei Zhou
- Department of Gastrointestinal Surgery, Jiangxi Provincial Cancer Hospital, No. 519, Beijing East Road, Nanchang, 330029, Jiangxi Province, China.
| |
Collapse
|
16
|
Low concentration of formononetin promotes proliferation of estrogen receptor-positive cells through an ERα-miR-375-PTEN-ERK1/2-bcl-2 pathway. Oncotarget 2017; 8:100045-100055. [PMID: 29245959 PMCID: PMC5725001 DOI: 10.18632/oncotarget.21923] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 08/23/2017] [Indexed: 11/25/2022] Open
Abstract
A low dose of formononetin accelerates the proliferation of nasopharyngeal carcinoma cells in vitro; however, the underlying mechanism remains unknown. Here, we investigated the molecular mechanism of formononetin in CNE2 cell proliferation. CNE2 cells were treated with 0 to 1 μM formononetin. To inhibit mitogen activated protein kinase / extracellular regulate kinase (MAPK/ERK) kinase (MEK) and microRNA (miR)-375, cells were pretreated with either PD98059 or a miR-375 inhibitor, respectively, followed by co-treatment with formononetin (0.3 μM) plus an inhibitor. Female rats were ovariectomized (OVX), and some OVX rats received formononetin or estrogen (E2) injections. Sham operated animals were used as controls. Compared to control, 0.3 μM formononetin accelerated proliferation and decreased late apoptosis of CNE2 cells. However, formononetin-induced pro-growth and anti-apoptosis activity was abolished by PD98059 and the miR-375 inhibitor. In addition, 0.1 and 0.3 μM formononetin significantly increased estrogen receptor-α (ERα) and bcl-2, but decreased protein-phosphatase and tensin homologue (PTEN) protein expression, all of which was reversed by the miR-375 inhibitor. Additionally, formononetin treatment resulted in a transient upregulation of phosphorylated (p)-ERK1/2. In vivo studies indicated that formononetin significantly increased endometrium thickness and down-regulated ERα expression in OVX rats. Taken together, our study demonstrates that a low concentration of formononetin can promote growth of CNE2 cells and uterine tissues, possibly through regulating the ERα-miR-375-PTEN-ERK1/2-bcl-2 signaling pathway.
Collapse
|
17
|
Yuan W, Wang D, Liu Y, Tian D, Wang Y, Zhang R, Yin L, Deng Z. miR‑494 inhibits cell proliferation and metastasis via targeting of CDK6 in osteosarcoma. Mol Med Rep 2017; 16:8627-8634. [PMID: 28990071 PMCID: PMC5779916 DOI: 10.3892/mmr.2017.7709] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 06/01/2017] [Indexed: 12/25/2022] Open
Abstract
Tumorigenesis is a multistep process involving various cell growth-associated factors. Accumulated evidence indicates that the disordered regulation of microRNAs (miRNAs) contributes to tumorigenesis. However, the detailed mechanism underlying the involvement of miRNAs in oncogenesis remains to be fully elucidated. In the present study, the repressed expression of microRNA (miR)-494 was identified in 18 patients with osteosarcoma (OS) and OS cell lines, compared with corresponding controls. To determine whether deregulated miR-494 exerts tumor-suppressive effects in the development of OS, the effects of miR-494 on cell proliferation and metastasis were evaluated. It was found that the restoration of miR-494 in MG-63 and U2OS cells led to inhibited cell proliferation and attenuated migratory propensity in vitro, determined through analysis using MTT, colony formation and Transwell assays. In addition, overexpression of miR-494 markedly suppressed the tumor volume and weight in vivo. In accordance, the ectopic expression of miR-494 induced cell cycle arrest at the G1/S phase in OS cells. Bioinformatics analysis and luciferase reporter assays were performed to investigate the potential regulatory role of miR-494, the results of which indicated that miR-494 directly targeted cyclin-dependent kinase 6 (CDK6). Of note, the data obtained through reverse transcription-quantitative polymerase chain reaction and western blot analyses suggested that the elevated expression of miR-494 resulted in reduced mRNA and protein expression levels of CDK6. Taken together, these findings indicated that the miR-494/CDK6 axis has a significant tumor-suppressive effect on OS, and maybe a diagnostic and therapeutic target for the treatment of OS.
Collapse
Affiliation(s)
- Wei Yuan
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Du Wang
- Department of Orthopedics, Wuhan Hospital No. 3 and Tongren Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yang Liu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Dongdong Tian
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Yang Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Ranxi Zhang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Liangjun Yin
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Zhongliang Deng
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| |
Collapse
|
18
|
Ma C, Han J, Dong D, Wang N. MicroRNA-152 Suppresses Human Osteosarcoma Cell Proliferation and Invasion by Targeting E2F Transcription Factor 3. Oncol Res 2017; 26:765-773. [PMID: 28810933 PMCID: PMC7844728 DOI: 10.3727/096504017x15021536183535] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
MicroRNA-152 (miR-152) expression has been reported to be downregulated in osteosarcoma (OS). However, the role of miR-152 in OS is not well documented. In the present study, we aimed to explore the function and underlying mechanism of miR-152 in OS. We found that miR-152 was underexpressed in OS tissues and cell lines. Decreased miR-152 was inversely correlated with lymph node metastasis and advanced clinical stage. Overexpression of miR-152 significantly inhibited cell proliferation, colony formation, migration, and invasion of OS cells. Bioinformatics analyses showed that miR-152 directly targeted E2F transcription factor 3 (E2F3), as further confirmed by a dual-luciferase reporter assay. E2F3 expression was upregulated and inversely correlated with miR-152 expression level in human OS tissues. Moreover, the inhibitory effects of miR-152 on OS growth and invasion were attenuated by E2F3 overexpression. Taken together, our findings indicated that miR-152 reduced OS growth and invasion by targeting E2F3 and provided new evidence of miR-152 as a potential therapeutic target for OS.
Collapse
Affiliation(s)
- Chao Ma
- The First Hospital of Jilin University, Changchun, P.R. China
| | - Jinfeng Han
- The First Hospital of Jilin University, Changchun, P.R. China
| | - Dong Dong
- The First Hospital of Jilin University, Changchun, P.R. China
| | - Nanya Wang
- The First Hospital of Jilin University, Changchun, P.R. China
| |
Collapse
|
19
|
Fang Y, Zhang L, Li Z, Li Y, Huang C, Lu X. MicroRNAs in DNA Damage Response, Carcinogenesis, and Chemoresistance. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 333:1-49. [DOI: 10.1016/bs.ircmb.2017.03.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|