1
|
Bogaczyk A, Zawlik I, Zuzak T, Kluz M, Potocka N, Kluz T. The Role of miRNAs in the Development, Proliferation, and Progression of Endometrial Cancer. Int J Mol Sci 2023; 24:11489. [PMID: 37511248 PMCID: PMC10380838 DOI: 10.3390/ijms241411489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/30/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Endometrial cancer is one of the most common cancers in developing and developed countries. Although the detection of this cancer is high at the early stages, there is still a lack of markers to monitor the disease, its recurrence, and metastasis. MiRNAs are in charge of the post-transcriptional regulation of genes responsible for the most important biological processes, which is why they are increasingly used as biomarkers in many types of cancer. Many studies have demonstrated the influence of miRNAs on the processes related to carcinogenesis. The characteristics of miRNA expression profiles in endometrial cancer will allow their use as diagnostic and prognostic biomarkers. This paper focuses on the discussion of selected miRNAs based on the literature and their role in the development of endometrial cancer.
Collapse
Affiliation(s)
- Anna Bogaczyk
- Department of Gynecology, Gynecology Oncology and Obstetrics, Fryderyk Chopin University Hospital, F.Szopena 2, 35-055 Rzeszow, Poland; (A.B.); (T.Z.); (T.K.)
| | - Izabela Zawlik
- Laboratory of Molecular Biology, Centre for Innovative Research in Medical and Natural Sciences, Medical College of Rzeszow University, Warzywna 1a, 35-959 Rzeszow, Poland;
- Institute of Medical Sciences, Medical College of Rzeszow University, Kopisto 2a, 35-959 Rzeszow, Poland
| | - Tomasz Zuzak
- Department of Gynecology, Gynecology Oncology and Obstetrics, Fryderyk Chopin University Hospital, F.Szopena 2, 35-055 Rzeszow, Poland; (A.B.); (T.Z.); (T.K.)
| | - Marta Kluz
- Department of Pathology, Fryderyk Chopin University Hospital, F.Szopena 2, 35-055 Rzeszow, Poland;
| | - Natalia Potocka
- Laboratory of Molecular Biology, Centre for Innovative Research in Medical and Natural Sciences, Medical College of Rzeszow University, Warzywna 1a, 35-959 Rzeszow, Poland;
| | - Tomasz Kluz
- Department of Gynecology, Gynecology Oncology and Obstetrics, Fryderyk Chopin University Hospital, F.Szopena 2, 35-055 Rzeszow, Poland; (A.B.); (T.Z.); (T.K.)
- Institute of Medical Sciences, Medical College of Rzeszow University, Kopisto 2a, 35-959 Rzeszow, Poland
| |
Collapse
|
2
|
Shah JA, Miao Y, Chu J, Chen W, Zhao Q, Cai C, Khattak S, Wang F, Jin J. Feedback Modulation between Human INO80 Chromatin Remodeling Complex and miR-372 in HCT116 Cells. Int J Mol Sci 2023; 24:10685. [PMID: 37445863 DOI: 10.3390/ijms241310685] [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: 05/15/2023] [Revised: 06/21/2023] [Accepted: 06/24/2023] [Indexed: 07/15/2023] Open
Abstract
Human INO80 chromatin remodeling complex (INO80 complex) as a transcription cofactor is widely involved in gene transcription regulation and is frequently highly expressed in tumor cells. However, few reports exist on the mutual regulatory mechanism between INO80 complex and non-coding microRNAs. Herein, we showed evidence that the INO80 complex transcriptionally controls microRNA-372 (miR-372) expression through RNA-Seq analysis and a series of biological experiments. Knocking down multiple subunits in the INO80 complex, including the INO80 catalytic subunit, YY1, Ies2, and Arp8, can significantly increase the expression level of miR-372. Interestingly, mimicking miR-372 expression in HCT116 cells, in turn, post-transcriptionally suppressed INO80 and Arp8 expression at both mRNA and protein levels, indicating the existence of a mutual regulatory mechanism between the INO80 complex and miR-372. The target relationship between miR-372 and INO80 complex was verified using luciferase assays in HCT116 colon cancer cells. As expected, miR-372 mimics significantly suppressed the luciferase activity of pMIR-luc/INO80 and pMIR-luc/Arp8 3'-UTR in cells. In contrast, the miR-372 target sites in the 3'-UTRs linked to the luciferase reporter were mutagenized, and both mutant sites lost their response to miR-372. Furthermore, the mutual modulation between the INO80 complex and miR-372 was involved in cell proliferation and the p53/p21 signaling pathway, suggesting the synergistic anti-tumor role of the INO80 complex and miR372. Our results will provide a solid theoretical basis for exploring miR-372 as a biological marker of tumorigenesis.
Collapse
Affiliation(s)
- Junaid Ali Shah
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Yujuan Miao
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Jinmeng Chu
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Wenqi Chen
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Qingzhi Zhao
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Chengyu Cai
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Saadullah Khattak
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Fei Wang
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Jingji Jin
- School of Life Sciences, Jilin University, Changchun 130012, China
| |
Collapse
|
3
|
Tajik F, Alian F, Yousefi M, Azadfallah A, Hoseini A, Mohammadi F, Karimi-Dehkordi M, Alizadeh-Fanalou S. MicroRNA-372 acts as a double-edged sword in human cancers. Heliyon 2023; 9:e15991. [PMID: 37251909 PMCID: PMC10208947 DOI: 10.1016/j.heliyon.2023.e15991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 04/26/2023] [Accepted: 04/28/2023] [Indexed: 05/31/2023] Open
Abstract
MicroRNAs (miRNAs or miRs) are non-coding, single-stranded, endogenous RNAs that regulate various biological processes, most notably the pathophysiology of many human malignancies. It process is accomplished by binding to 3'-UTR mRNAs and controlling gene expression at the post-transcriptional level. As an oncogene, miRNAs can either accelerate cancer progression or slow it down as a tumor suppressor. MicroRNA-372 (miR-372) has been found to have an abnormal expression in numerous human malignancies, implying that the miRNA plays a role in carcinogenesis. It is both increased and downregulated in various cancers, and it serves as both a tumor suppressor and an oncogene. This study examines the functions of miR-372 as well as the LncRNA/CircRNA-miRNA-mRNA signaling pathways in various malignancies and analyses its potential prognostic, diagnostic, and therapeutic implications.
Collapse
Affiliation(s)
- Fatemeh Tajik
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Alian
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Mohammad Yousefi
- Department of Medicine, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Ali Azadfallah
- Department of Medicine, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Aref Hoseini
- Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Forogh Mohammadi
- Department of Veterinary, Agriculture Faculty, Kermanshah Branch, Islamic Azad University, Kermanshah, Iran
| | - Maryam Karimi-Dehkordi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Shahin Alizadeh-Fanalou
- Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
4
|
A current overview of RhoA, RhoB, and RhoC functions in vascular biology and pathology. Biochem Pharmacol 2022; 206:115321. [DOI: 10.1016/j.bcp.2022.115321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/17/2022] [Accepted: 10/18/2022] [Indexed: 11/24/2022]
|
5
|
Li Y, Li F, Feng C, Wu T, Chen Y, Shah JA, Wang F, Cai Y, Wang J, Jin J. MiR-372-3p Functions as a Tumor Suppressor in Colon Cancer by Targeting MAP3K2. Front Genet 2022; 13:836256. [PMID: 35432472 PMCID: PMC9006175 DOI: 10.3389/fgene.2022.836256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/15/2022] [Indexed: 11/13/2022] Open
Abstract
MicroRNAs (miRNAs) as small non-coding RNA transcripts bind their complementary sequences in the 3′-untranslated region (3′-UTR) of target messenger RNAs (mRNAs) to regulate their expression. It is known that miR-372 belongs to the miR-371–373 gene cluster and has been found to be abnormally expressed in a variety of cancers, but its precise mechanism in cancer remains to be discovered. In this study, miR-372-3p expression was assessed in 153 frozen tissue samples, including primary diagnosed colon cancer and matched normal and adjacent tissues, using real time quantitative polymerase chain reaction (qPCR). An analysis of qPCR data revealed a significant reduction in miR-372-3p expression (by >2-fold) in colon cancer tissues in 51.5% (34/66) of patients. Consistent with this, mimicking the increased miR-372-3p levels in SW480 colon cancer cells significantly suppressed cell growth and proliferation. Although no direct correlation was found between the low level of miR-372-3p and certain tumor-related factors, such as p53, HRE-2, PMS2, MLH1, MSH2, MSH6, HDAC4, p21, and Wee1, in colon cancer tissues, an inverse relationship between miR-372-3p and Ki67 (a marker of proliferation) or miR-372-3p and MAP3K2(MEKK2), which plays a critical role in the MAPK signaling pathways, was confirmed using tissue samples. The target relationship between miR-372-3p and MAP3K2 was verified using luciferase assays in SW480 colon cancer cells. As expected, miR-372-3p mimics significantly suppressed the luciferase activity of pMIR-luc/MAP3K2 3′-UTR in cells, suggesting that miR-372-3p modulates the expression of MAP3K2 by directly targeting its 3′-UTR. Overall, the results obtained herein suggest that miR-372-3p may function as a tumor-suppressor miRNA in colon cancer by targeting MAP3K2.
Collapse
Affiliation(s)
- Yana Li
- School of Life Sciences, Jilin University, Changchun, China
- Department of Ophthalmology and Otorhinolaryngology, Changchun Children’s Hospital, Changchun, China
| | - Fuqiang Li
- School of Life Sciences, Jilin University, Changchun, China
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Chang Feng
- School of Life Sciences, Jilin University, Changchun, China
| | - Tingting Wu
- School of Life Sciences, Jilin University, Changchun, China
| | - Yuyang Chen
- School of Life Sciences, Jilin University, Changchun, China
| | | | - Fei Wang
- School of Life Sciences, Jilin University, Changchun, China
| | - Yong Cai
- School of Life Sciences, Jilin University, Changchun, China
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Jianfeng Wang
- Department of Radiotherapy, China-Japan Union Hospital, Jilin University, Changchun, China
- *Correspondence: Jianfeng Wang, ; Jingji Jin,
| | - Jingji Jin
- School of Life Sciences, Jilin University, Changchun, China
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
- *Correspondence: Jianfeng Wang, ; Jingji Jin,
| |
Collapse
|
6
|
Chen Z, Gan J, Wei Z, Zhang M, Du Y, Xu C, Zhao H. The Emerging Role of PRMT6 in Cancer. Front Oncol 2022; 12:841381. [PMID: 35311114 PMCID: PMC8931394 DOI: 10.3389/fonc.2022.841381] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/09/2022] [Indexed: 01/01/2023] Open
Abstract
Protein arginine methyltransferase 6 (PRMT6) is a type I PRMT that is involved in epigenetic regulation of gene expression through methylating histone or non-histone proteins, and other processes such as alternative splicing, DNA repair, cell proliferation and senescence, and cell signaling. In addition, PRMT6 also plays different roles in various cancers via influencing cell growth, migration, invasion, apoptosis, and drug resistant, which make PRMT6 an anti-tumor therapeutic target for a variety of cancers. As a result, many PRMT6 inhibitors are being utilized to explore their efficacy as potential drugs for various cancers. In this review, we summarize the current knowledge on the function and structure of PRMT6. At the same time, we highlight the role of PRMT6 in different cancers, including the differentiation of its promotive or inhibitory effects and the underlying mechanisms. Apart from the above, current research progress and the potential mechanisms of PRMT6 behind them were also summarized.
Collapse
Affiliation(s)
- Zhixian Chen
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai, China
| | - Jianfeng Gan
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai, China
| | - Zhi Wei
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai, China
| | - Mo Zhang
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai, China
| | - Yan Du
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai, China
| | - Congjian Xu
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai, China
- *Correspondence: Hongbo Zhao, ; Congjian Xu,
| | - Hongbo Zhao
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai, China
- *Correspondence: Hongbo Zhao, ; Congjian Xu,
| |
Collapse
|
7
|
Zhang J, Zhang D, Yan X, Jiang F. The expression level and prognostic value of microRNA-15a-5p in endometrial carcinoma. Transl Cancer Res 2021; 10:4838-4844. [PMID: 35116336 PMCID: PMC8798199 DOI: 10.21037/tcr-21-2079] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/29/2021] [Indexed: 11/06/2022]
Abstract
BACKGROUND Recent studies have shown that the microRNA-15a-5p (miR-15a-5p) plays varying roles in different malignancies. However, to date, the role and prognostic value of miR-15a-5p in patients with endometrial cancer has not been explored. This study investigated the expression level of miR-15a-5p in endometrial carcinoma and its prognostic value. METHODS A total of 108 patients with endometrial cancer treated in our hospital from January 2015 to January 2016 were enrolled in this study. The patients were followed up for 5 years. Patients who experienced recurrence or metastasis after surgery were assigned into the recurrence and metastasis group (n=45) and the remaining patients were assigned into the control group (n=63). The expression level of microRNA-15a-5p in endometrial cancer was analyzed. Furthermore, the correlation between the expression of miR-15a-5p and the pathological features and prognosis was examined. RESULTS The expression of miR-15a-5p in endometrial carcinoma was significantly lower than that in adjacent healthy tissues (2.22±0.75 vs. 2.59±0.91, P=0.000). Furthermore, the expression of miR-15a-5p in the endometrial cancer tissues of patients in the recurrence and metastasis group was significantly lower than that observed in patients in the control group (1.91±0.62 vs. 2.45±0.75, P=0.000). The receiver operating characteristic curve was used to analyze the predictive value of miR-15a-5p in endometrial cancer tissue for postoperative recurrence or metastasis in endometrial cancer patients. The area under the curve was 0.690 [95% confidence interval (CI): 0.601 to 0.798, P=0.000], the best cut-off value of diagnosis was 2.325, the sensitivity was 0.619, and the specificity was 0.733. Multivariate logistic regression analysis showed that miR-15a-5p expression <2.325 was a risk factor for postoperative recurrence or metastasis of endometrial cancer [odds ratio (OR) =3.544 (95% CI: 1.489 to 8.436), P=0.004]. Furthermore, the expression of miR-15a-5p in endometrial carcinoma was correlated with lymph node metastasis, TNM stage, and patient mortality. CONCLUSIONS The expression of miR-15a-5p in endometrial carcinoma is related to lymph node metastasis, TNM stage, and mortality. Furthermore, the expression of miR-15a-5p was significantly decreased in endometrial cancer patients with recurrence or metastasis and thus, miR-15a-5p may have certain value in predicting postoperative recurrence or metastasis in such patients.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Gynecology, Baoji Central Hospital, Baoji, China
| | - Dandan Zhang
- Department of Obstetrics and Gynecology, Xichang People’s Hospital, Xichang, China
| | - Xiaofang Yan
- Department of Obstetrics and Gynecology, Yixing People’s Hospital, Yixing, China
| | - Feizhou Jiang
- Department of Obstetrics and Gynecology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, China
| |
Collapse
|
8
|
Lou Y, Jiang Y, Liang Z, Liu B, Li T, Zhang D. Role of RhoC in cancer cell migration. Cancer Cell Int 2021; 21:527. [PMID: 34627249 PMCID: PMC8502390 DOI: 10.1186/s12935-021-02234-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/27/2021] [Indexed: 12/20/2022] Open
Abstract
Migration is one of the five major behaviors of cells. Although RhoC-a classic member of the Rho gene family-was first identified in 1985, functional RhoC data have only been widely reported in recent years. Cell migration involves highly complex signaling mechanisms, in which RhoC plays an essential role. Cell migration regulated by RhoC-of which the most well-known function is its role in cancer metastasis-has been widely reported in breast, gastric, colon, bladder, prostate, lung, pancreatic, liver, and other cancers. Our review describes the role of RhoC in various types of cell migration. The classic two-dimensional cell migration cycle constitutes cell polarization, adhesion regulation, cell contraction and tail retraction, most of which are modulated by RhoC. In the three-dimensional cell migration model, amoeboid migration is the most classic and well-studied model. Here, RhoC modulates the formation of membrane vesicles by regulating myosin II, thereby affecting the rate and persistence of amoeba-like migration. To the best of our knowledge, this review is the first to describe the role of RhoC in all cell migration processes. We believe that understanding the detail of RhoC-regulated migration processes will help us better comprehend the mechanism of cancer metastasis. This will contribute to the study of anti-metastatic treatment approaches, aiding in the identification of new intervention targets for therapeutic or genetic transformational purposes.
Collapse
Affiliation(s)
- Yingyue Lou
- Department of Plastic and Reconstructive Surgery, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yuhan Jiang
- Department of Plastic and Reconstructive Surgery, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Zhen Liang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Bingzhang Liu
- Department of Plastic and Reconstructive Surgery, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Tian Li
- Department of Plastic and Reconstructive Surgery, The First Hospital of Jilin University, Changchun, Jilin, China.
| | - Duo Zhang
- Department of Plastic and Reconstructive Surgery, The First Hospital of Jilin University, Changchun, Jilin, China.
| |
Collapse
|
9
|
Shah JA, Khattak S, Rauf MA, Cai Y, Jin J. Potential Biomarkers of miR-371-373 Gene Cluster in Tumorigenesis. Life (Basel) 2021; 11:life11090984. [PMID: 34575133 PMCID: PMC8465240 DOI: 10.3390/life11090984] [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] [Received: 08/27/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 12/12/2022] Open
Abstract
microRNAs (miRNAs) are small non-coding RNA transcripts (20–24 nucleotides) that bind to their complementary sequences in the 3′-untranslated regions (3′-UTR) of targeted genes to negatively or positively regulate their expression. miRNAs affect the expression of genes in cells, thereby contributing to several important biological processes, including tumorigenesis. Identifying the miRNA cluster as a human embryonic stem cell (hESC)-specific miRNAs initially led to the identification of miR-371, miR-372, miR-373, and miR-373*, which can ultimately be translated into mature miRNAs. Recent evidence suggests that miR-371–373 genes are abnormally expressed in various cancers and act either as oncogenes or tumor suppressors, indicating they may be suitable as molecular biomarkers for cancer diagnosis and prevention. In this article, we summarize recent studies linking miR-371–373 functions to tumorigenesis and speculate on the potential applications of miR-371–373 as biomarkers for cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Junaid Ali Shah
- School of Life Sciences, Jilin University, Changchun 130012, China; (J.A.S.); (Y.C.)
| | - Saadullah Khattak
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China;
| | - Mohd Ahmar Rauf
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; or
| | - Yong Cai
- School of Life Sciences, Jilin University, Changchun 130012, China; (J.A.S.); (Y.C.)
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Jingji Jin
- School of Life Sciences, Jilin University, Changchun 130012, China; (J.A.S.); (Y.C.)
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China
- Correspondence:
| |
Collapse
|
10
|
Klicka K, Grzywa TM, Klinke A, Mielniczuk A, Włodarski PK. The Role of miRNAs in the Regulation of Endometrial Cancer Invasiveness and Metastasis-A Systematic Review. Cancers (Basel) 2021; 13:3393. [PMID: 34298609 PMCID: PMC8304659 DOI: 10.3390/cancers13143393] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/18/2021] [Accepted: 07/02/2021] [Indexed: 12/17/2022] Open
Abstract
Endometrial cancer (EC) is the most common genital cancer in women with increasing death rates. MiRNAs are short non-coding RNAs that regulate gene expression on the post-transcriptional levels. Multiple studies demonstrated a fundamental role of miRNAs in the regulation of carcinogenesis. This systematic review is a comprehensive overview of the role of miRNAs in the regulation of cancer cell invasiveness and metastasis in EC. The literature was searched for studies investigating the role of miRNAs in the regulation of invasiveness and metastasis in EC. We explored PubMed, Embase, and Scopus using the following keywords: miRNA, metastasis, invasiveness, endometrial cancer. Data were collected from 163 articles that described the expression and role of 106 miRNAs in the regulation of EC invasiveness and metastasis out of which 63 were tumor suppressor miRNAs, and 38 were oncomiRNAs. Five miRNAs had a discordant role in different studies. Moreover, we identified 66 miRNAs whose expression in tumor tissue or concentration in serum correlated with at least one clinical parameter. These findings suggest a crucial role of miRNAs in the regulation of EC invasiveness and metastasis and present them as potential prognostic factors for patients with EC.
Collapse
Affiliation(s)
- Klaudia Klicka
- Department of Methodology, Medical University of Warsaw, 02-091 Warsaw, Poland; (K.K.); (T.M.G.); (A.K.); (A.M.)
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Tomasz M. Grzywa
- Department of Methodology, Medical University of Warsaw, 02-091 Warsaw, Poland; (K.K.); (T.M.G.); (A.K.); (A.M.)
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Alicja Klinke
- Department of Methodology, Medical University of Warsaw, 02-091 Warsaw, Poland; (K.K.); (T.M.G.); (A.K.); (A.M.)
| | - Aleksandra Mielniczuk
- Department of Methodology, Medical University of Warsaw, 02-091 Warsaw, Poland; (K.K.); (T.M.G.); (A.K.); (A.M.)
| | - Paweł K. Włodarski
- Department of Methodology, Medical University of Warsaw, 02-091 Warsaw, Poland; (K.K.); (T.M.G.); (A.K.); (A.M.)
| |
Collapse
|
11
|
Piergentili R, Zaami S, Cavaliere AF, Signore F, Scambia G, Mattei A, Marinelli E, Gulia C, Perelli F. Non-Coding RNAs as Prognostic Markers for Endometrial Cancer. Int J Mol Sci 2021; 22:3151. [PMID: 33808791 PMCID: PMC8003471 DOI: 10.3390/ijms22063151] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/12/2021] [Accepted: 03/12/2021] [Indexed: 02/06/2023] Open
Abstract
Endometrial cancer (EC) has been classified over the years, for prognostic and therapeutic purposes. In recent years, classification systems have been emerging not only based on EC clinical and pathological characteristics but also on its genetic and epigenetic features. Noncoding RNAs (ncRNAs) are emerging as promising markers in several cancer types, including EC, for which their prognostic value is currently under investigation and will likely integrate the present prognostic tools based on protein coding genes. This review aims to underline the importance of the genetic and epigenetic events in the EC tumorigenesis, by expounding upon the prognostic role of ncRNAs.
Collapse
Affiliation(s)
- Roberto Piergentili
- Institute of Molecular Biology and Pathology, Italian National Research Council (CNR-IBPM), 00185 Rome, Italy;
| | - Simona Zaami
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, “Sapienza” University of Rome, Viale Regina Elena 336, 00161 Rome, Italy
| | - Anna Franca Cavaliere
- Gynecology and Obstetric Department, Azienda USL Toscana Centro, Santo Stefano Hospital, 59100 Prato, Italy;
| | - Fabrizio Signore
- Obstetrics and Gynecology Department, USL Roma2, Sant’Eugenio Hospital, 00144 Rome, Italy;
| | - Giovanni Scambia
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Gynecologic Oncology Unit, 00168 Rome, Italy;
- Universita’ Cattolica Del Sacro Cuore, 00168 Rome, Italy
| | - Alberto Mattei
- Gynecology and Obstetric Department, Azienda USL Toscana Centro, Santa Maria Annunziata Hospital, 50012 Florence, Italy; (A.M.); (F.P.)
| | - Enrico Marinelli
- Unit of Forensic Toxicology (UoFT), Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University, 00161 Rome, Italy;
| | - Caterina Gulia
- Department of Urology, Misericordia Hospital, 58100 Grosseto, Italy;
| | - Federica Perelli
- Gynecology and Obstetric Department, Azienda USL Toscana Centro, Santa Maria Annunziata Hospital, 50012 Florence, Italy; (A.M.); (F.P.)
| |
Collapse
|
12
|
Wang JS, Ruan F, Guo LZ, Wang FG, Wang FL, An HM. B3GNT3 acts as a carcinogenic factor in endometrial cancer via facilitating cell growth, invasion and migration through regulating RhoA/RAC1 pathway-associated markers. Genes Genomics 2021; 43:447-457. [PMID: 33683574 DOI: 10.1007/s13258-021-01072-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 02/19/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Aberrant expression of beta-1,3-N-acetylglucosaminyltransferase-3 (B3GNT3) has been frequently clarified in various cancers, however, its role in endometrial cancer (EC) has not been assessed in detail. PURPOSE This study aimed to investigate the biological role of B3GNT3 in EC and simply explored the detailed mechanism. METHODS The EC RNA-Seq dataset from TCGA database was applied to evaluate the expression of B3GNT3 and assess its role on prognostic value. HEC-1-A and KLE cell lines of EC were used to perform loss- and gain-of-function B3GNT3 assays respectively. Quantitative real-time PCR (qRT-PCR) and western blot were used to measure the mRNA and protein levels of indicated molecules respectively. Cell counting kit-8, clone formation tests, and Transwell assay served to determine the changes of proliferative, invasive and migratory abilities of EC cells after altering the expression of B3GNT3. RESULTS B3GNT3 was found to be highly expressed in EC tissues compared to normal tissues according to the online public databases, which confirmed by the following qRT-PCR in 3 EC cell lines. Besides, high B3GNT3 expression presented a worse overall survival in EC patients as compared with low B3GNT3 expression group. Furthermore, functional experiments in vitro indicated that B3GNT3 could facilitate the cell growth, invasion and migration. Moreover, we found that downregulation of B3GNT3 significantly reduced the expression level of GTP-RhoA and GTP-RAC1, whereas upregulation of B3GNT3 presented the opposite results. CONCLUSION The results of current study demonstrate that B3GNT3 acts as an oncogene that promotes EC cells growth, invasion and migration possibly through regulating the RhoA/RAC1 signaling pathway-related markers, suggesting that B3GNT3 may be a candidate biomarker for EC therapeutic intervention.
Collapse
Affiliation(s)
- Ji-Shui Wang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Jining Medical University, Jining, 272029, Shandong, China
| | - Fang Ruan
- Department of Obstetrics, Affiliated Hospital of Jining Medical University, No.89 of Guhuai Road, Jining, 272029, Shandong, China
| | - Li-Zhu Guo
- Department of Obstetrics, Affiliated Hospital of Jining Medical University, No.89 of Guhuai Road, Jining, 272029, Shandong, China
| | - Feng-Ge Wang
- Department of Obstetrics, Affiliated Hospital of Jining Medical University, No.89 of Guhuai Road, Jining, 272029, Shandong, China
| | - Fu-Ling Wang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Jining Medical University, Jining, 272029, Shandong, China
| | - Hong-Min An
- Department of Obstetrics, Affiliated Hospital of Jining Medical University, No.89 of Guhuai Road, Jining, 272029, Shandong, China.
| |
Collapse
|
13
|
Joshi H, Vastrad B, Joshi N, Vastrad C, Tengli A, Kotturshetti I. Identification of Key Pathways and Genes in Obesity Using Bioinformatics Analysis and Molecular Docking Studies. Front Endocrinol (Lausanne) 2021; 12:628907. [PMID: 34248836 PMCID: PMC8264660 DOI: 10.3389/fendo.2021.628907] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 05/19/2021] [Indexed: 01/01/2023] Open
Abstract
Obesity is an excess accumulation of body fat. Its progression rate has remained high in recent years. Therefore, the aim of this study was to diagnose important differentially expressed genes (DEGs) associated in its development, which may be used as novel biomarkers or potential therapeutic targets for obesity. The gene expression profile of E-MTAB-6728 was downloaded from the database. After screening DEGs in each ArrayExpress dataset, we further used the robust rank aggregation method to diagnose 876 significant DEGs including 438 up regulated and 438 down regulated genes. Functional enrichment analysis was performed. These DEGs were shown to be significantly enriched in different obesity related pathways and GO functions. Then protein-protein interaction network, target genes - miRNA regulatory network and target genes - TF regulatory network were constructed and analyzed. The module analysis was performed based on the whole PPI network. We finally filtered out STAT3, CORO1C, SERPINH1, MVP, ITGB5, PCM1, SIRT1, EEF1G, PTEN and RPS2 hub genes. Hub genes were validated by ICH analysis, receiver operating curve (ROC) analysis and RT-PCR. Finally a molecular docking study was performed to find small drug molecules. The robust DEGs linked with the development of obesity were screened through the expression profile, and integrated bioinformatics analysis was conducted. Our study provides reliable molecular biomarkers for screening and diagnosis, prognosis as well as novel therapeutic targets for obesity.
Collapse
Affiliation(s)
- Harish Joshi
- Department of Endocrinology, Endocrine and Diabetes Care Center, Hubbali, India
| | - Basavaraj Vastrad
- Department of Biochemistry, Basaveshwar College of Pharmacy, Gadag, India
| | - Nidhi Joshi
- Department of Medicine, Dr. D. Y. Patil Medical College, Kolhapur, India
| | - Chanabasayya Vastrad
- Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad, India
- *Correspondence: Chanabasayya Vastrad,
| | - Anandkumar Tengli
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Mysuru and JSS Academy of Higher Education & Research, Mysuru, India
| | - Iranna Kotturshetti
- Department of Ayurveda, Rajiv Gandhi Education Society`s Ayurvedic Medical College, Ron, India
| |
Collapse
|
14
|
Grzywa TM, Klicka K, Włodarski PK. Regulators at Every Step-How microRNAs Drive Tumor Cell Invasiveness and Metastasis. Cancers (Basel) 2020; 12:E3709. [PMID: 33321819 PMCID: PMC7763175 DOI: 10.3390/cancers12123709] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 02/06/2023] Open
Abstract
Tumor cell invasiveness and metastasis are the main causes of mortality in cancer. Tumor progression is composed of many steps, including primary tumor growth, local invasion, intravasation, survival in the circulation, pre-metastatic niche formation, and metastasis. All these steps are strictly controlled by microRNAs (miRNAs), small non-coding RNA that regulate gene expression at the post-transcriptional level. miRNAs can act as oncomiRs that promote tumor cell invasion and metastasis or as tumor suppressor miRNAs that inhibit tumor progression. These miRNAs regulate the actin cytoskeleton, the expression of extracellular matrix (ECM) receptors including integrins and ECM-remodeling enzymes comprising matrix metalloproteinases (MMPs), and regulate epithelial-mesenchymal transition (EMT), hence modulating cell migration and invasiveness. Moreover, miRNAs regulate angiogenesis, the formation of a pre-metastatic niche, and metastasis. Thus, miRNAs are biomarkers of metastases as well as promising targets of therapy. In this review, we comprehensively describe the role of various miRNAs in tumor cell migration, invasion, and metastasis.
Collapse
Affiliation(s)
- Tomasz M. Grzywa
- Department of Methodology, Medical University of Warsaw, 02-091 Warsaw, Poland; (T.M.G.); (K.K.)
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Klaudia Klicka
- Department of Methodology, Medical University of Warsaw, 02-091 Warsaw, Poland; (T.M.G.); (K.K.)
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Paweł K. Włodarski
- Department of Methodology, Medical University of Warsaw, 02-091 Warsaw, Poland; (T.M.G.); (K.K.)
| |
Collapse
|
15
|
Regulators at Every Step—How microRNAs Drive Tumor Cell Invasiveness and Metastasis. Cancers (Basel) 2020. [DOI: 10.3390/cancers12123709
expr 991289423 + 939431153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Tumor cell invasiveness and metastasis are the main causes of mortality in cancer. Tumor progression is composed of many steps, including primary tumor growth, local invasion, intravasation, survival in the circulation, pre-metastatic niche formation, and metastasis. All these steps are strictly controlled by microRNAs (miRNAs), small non-coding RNA that regulate gene expression at the post-transcriptional level. miRNAs can act as oncomiRs that promote tumor cell invasion and metastasis or as tumor suppressor miRNAs that inhibit tumor progression. These miRNAs regulate the actin cytoskeleton, the expression of extracellular matrix (ECM) receptors including integrins and ECM-remodeling enzymes comprising matrix metalloproteinases (MMPs), and regulate epithelial–mesenchymal transition (EMT), hence modulating cell migration and invasiveness. Moreover, miRNAs regulate angiogenesis, the formation of a pre-metastatic niche, and metastasis. Thus, miRNAs are biomarkers of metastases as well as promising targets of therapy. In this review, we comprehensively describe the role of various miRNAs in tumor cell migration, invasion, and metastasis.
Collapse
|
16
|
Rho GTPases in Gynecologic Cancers: In-Depth Analysis toward the Paradigm Change from Reactive to Predictive, Preventive, and Personalized Medical Approach Benefiting the Patient and Healthcare. Cancers (Basel) 2020; 12:cancers12051292. [PMID: 32443784 PMCID: PMC7281750 DOI: 10.3390/cancers12051292] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/12/2020] [Accepted: 05/13/2020] [Indexed: 12/24/2022] Open
Abstract
Rho guanosine triphospatases (GTPases) resemble a conserved family of GTP-binding proteins regulating actin cytoskeleton dynamics and several signaling pathways central for the cell. Rho GTPases create a so-called Ras-superfamily of GTPases subdivided into subgroups comprising at least 20 members. Rho GTPases play a key regulatory role in gene expression, cell cycle control and proliferation, epithelial cell polarity, cell migration, survival, and apoptosis, among others. They also have tissue-related functions including angiogenesis being involved in inflammatory and wound healing processes. Contextually, any abnormality in the Rho GTPase function may result in severe consequences at molecular, cellular, and tissue levels. Rho GTPases also play a key role in tumorigenesis and metastatic disease. Corresponding mechanisms include a number of targets such as kinases and scaffold/adaptor-like proteins initiating GTPases-related signaling cascades. The accumulated evidence demonstrates the oncogenic relevance of Rho GTPases for several solid malignancies including breast, liver, bladder, melanoma, testicular, lung, central nervous system (CNS), head and neck, cervical, and ovarian cancers. Furthermore, Rho GTPases play a crucial role in the development of radio- and chemoresistance e.g. under cisplatin-based cancer treatment. This article provides an in-depth overview on the role of Rho GTPases in gynecological cancers, highlights relevant signaling pathways and pathomechanisms, and sheds light on their involvement in tumor progression, metastatic spread, and radio/chemo resistance. In addition, insights into a spectrum of novel biomarkers and innovative approaches based on the paradigm shift from reactive to predictive, preventive, and personalized medicine are provided.
Collapse
|
17
|
Jiang N, Li QL, Pan W, Li J, Zhang MF, Cao T, Su SG, Shen H. PRMT6 promotes endometrial cancer via AKT/mTOR signaling and indicates poor prognosis. Int J Biochem Cell Biol 2019; 120:105681. [PMID: 31884111 DOI: 10.1016/j.biocel.2019.105681] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/04/2019] [Accepted: 12/19/2019] [Indexed: 12/24/2022]
Abstract
Arginine methylation plays essential roles in post-transcriptional modification and signal transduction. Dysregulation of protein arginine methyltransferases (PRMTs) has been reported in human cancers, yet the expression and biological function of PRMT6 in endometrial cancer (EMC) remains unclear. Here, we show that PRMT6 is upregulated in EMC and exhibits oncogenic activities via activation of AKT/mTOR pathway. The expression of PRMT6 in EMC is much higher than that in the adjacent nontumorous tissues. Elevated PRMT6 expression is significantly associated with higher histological tumor grade and unfavorable prognosis in two independent cohorts consisting of a total of 564 patients with EMC. In vitro data demonstrate that PRMT6 expression was identified as a downstream target of miR-372-3p. Ectopic expression of miR-372-3p downregulates PRMT6. Overexpression of PRMT6 promotes EMC cell proliferation and migration, whereas knockdown of PRMT6 leads to opposite phenotypes. Mechanistically, PRMT6 induces the phosphorylation of AKT and mTOR in EMC cells. Inhibition of AKT/mTOR signaling by MK2206 or rapamycin attenuates the PRMT6-mediated EMC progression. In clinical samples, high expression of PRMT6 was correlated to low expression of miR-372-3p and high expression of phosphorylated AKT. Collectively, our findings suggest PRMT6 may function as an oncogene to promote tumor progression, and be of prognostic value to predict disease-free survival of patients with EMC. The newly identified miR-372-3p/PRMT6/AKT/mTOR axis represents a new promising target for EMC management.
Collapse
Affiliation(s)
- Nan Jiang
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Qiu-Li Li
- Department of Head and Neck Surgery, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Wenwei Pan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Jinhui Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Mei-Fang Zhang
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Tiefeng Cao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Shu-Guang Su
- Department of Pathology, The Affiliated Hexian Memorial Hospital of Southern Medical University, Guangzhou, China.
| | - Huimin Shen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
18
|
Thomas P, Pranatharthi A, Ross C, Srivastava S. RhoC: a fascinating journey from a cytoskeletal organizer to a Cancer stem cell therapeutic target. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:328. [PMID: 31340863 PMCID: PMC6651989 DOI: 10.1186/s13046-019-1327-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 07/15/2019] [Indexed: 01/05/2023]
Abstract
Tumor heterogeneity results in differential response to therapy due to the existence of plastic tumor cells, called cancer stem cells (CSCs), which exhibit the property of resistance to therapy, invasion and metastasis. These cells have a distinct, signaling network active at every stage of progression. It is difficult to envisage that the CSCs will have a unique set of signaling pathways regulating every stage of disease progression. Rather, it would be easier to believe that a single pivotal pathway having significant contribution at every stage, which can further turn on a battery of signaling mechanisms specific to that stage, would be instrumental in regulating the signaling network, enabling easy transition from one state to another. In this context, we discuss the role of RhoC which has contributed to several phenotypes during tumor progression. RhoC (Ras homolog gene family member C) has been widely reported to regulate actin organization. It has been shown to impact the motility of cancer cells, resultantly affecting invasion and metastasis, and has contributed to carcinoma progression of the breast, pancreas, lung, ovaries and cervix, among several others. The most interesting finding has been its indispensable role in metastasis. Also, it has the ability to modulate various other phenotypes like angiogenesis, motility, invasion, metastasis, and anoikis resistance. These observations suggest that RhoC imparts the plasticity required by tumor cells to exhibit such diverse functions based on microenvironmental cues. This was further confirmed by recent reports which show that it regulates cancer stem cells in breast, ovary and head and neck cancers. Studies also suggest that the inhibition of RhoC results in abolition of advanced tumor phenotypes. Our review throws light on how RhoC, which is capable of modulating various phenotypes may be the apt core signaling candidate regulating disease progression. Additionally, mice studies show that RhoC is not essential for embryogenesis, giving scope for its development as a possible therapeutic target. This review thus stresses on the need to understand the protein and its functioning in greater detail to enable its development as a stem cell marker and a possible therapeutic target.
Collapse
Affiliation(s)
- Pavana Thomas
- Translational and Molecular Biology Laboratory (TMBL), St. John's Research Institute (SJRI), Bangalore, 560034, India.,School of Integrative Health Sciences, The University of Trans-Disciplinary Health Sciences and Technology (TDU), Bangalore, 560064, India
| | - Annapurna Pranatharthi
- Rajiv Gandhi University of Health Sciences (RGUHS), Bangalore, 560041, India.,National Centre for Biological Sciences (NCBS), Bangalore, 560065, India.,Translational and Molecular Biology Laboratory (TMBL), Department of Medicine, St. John's Medical College Hospital (SJMCH), Bangalore, 560034, India
| | - Cecil Ross
- Translational and Molecular Biology Laboratory (TMBL), Department of Medicine, St. John's Medical College Hospital (SJMCH), Bangalore, 560034, India
| | - Sweta Srivastava
- Translational and Molecular Biology Laboratory (TMBL), Department of Transfusion Medicine and Immunohematology, St. John's Medical College Hospital (SJMCH), Bangalore, 560034, India.
| |
Collapse
|
19
|
MEHP promotes the proliferation of oral cancer cells via down regulation of miR-27b-5p and miR-372-5p. Toxicol In Vitro 2019; 58:35-41. [PMID: 30858031 DOI: 10.1016/j.tiv.2019.03.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/24/2019] [Accepted: 03/07/2019] [Indexed: 01/06/2023]
Abstract
Mono-2-ethyhexyl phthalate (MEHP), an environmental xenoestrogen, is widely used in the production of polyvinyl chloride materials and can be easily accumulated into human body. Emerging evidences showed that MEHP can regulate the progression of various cancers. Oral cancer cells could be directly exposed to MEHP during food and water digestion, while the roles of MEHP on the progression of oral cancer were rarely investigated. Our present study found that MEHP can trigger the proliferation of oral squamous cell carcinoma (OSCC) cells and increase the expression of proliferating cell nuclear antigen (PCNA). We checked the expression of various miRNAs which can target the 3'UTR of PCNA. Specifically, MEHP can decrease the expression of miR-27b-5p and miR-372-5p, which can directly bind with the 3'UTR of PCNA to inhibit its expression. Over expression of miR-27b-5p and miR-372-5p can abolish MEHP induced cell proliferation and expression of PCNA in OSCC cells. Further, MEHP can induce the expression of c-Myc, which can suppress the transcription of miR-27b-5p in OSCC cells. In vivo xenograft study on the basis of SCC-4 cells confirmed that MEHP can trigger the growth of OSCC and suppress the expression of miR-27b-5p and miR-372-5p. Collectively, our present study suggested that MEHP can promote the growth and progression of OSCC via down regulation of miR-27b-5p and miR-372-5p.
Collapse
|
20
|
Wang Z, Mao JW, Liu GY, Wang FG, Ju ZS, Zhou D, Wang RY. MicroRNA-372 enhances radiosensitivity while inhibiting cell invasion and metastasis in nasopharyngeal carcinoma through activating the PBK-dependent p53 signaling pathway. Cancer Med 2019; 8:712-728. [PMID: 30656832 PMCID: PMC6382924 DOI: 10.1002/cam4.1924] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 11/23/2018] [Accepted: 11/23/2018] [Indexed: 01/05/2023] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a common cancer found in the nasopharynx, which plagues countless NPC patients. MicroRNA‐372 (miR‐372) has been reported to be involved in various tumors. Here, we explored the important role of miR‐372 in radiosensitivity, invasion, and metastasis of NPC. Microarray analysis was conducted to search the NPC‐related differentially expressed genes (DEGs) and predict the miRs regulating PBK, which suggested that miR‐372 could influence the development of NPC via PBK and the p53 signaling pathway. Importantly, miR‐372 was observed to target PBK, thus down‐regulating its expression. Then, NPC 5‐8F and C666‐1 cells were selected, and treated with ionization radiation and alteration of miR‐372 and PBK expression to explore the functional role of miR‐372 in NPC. The expression of miR‐372, PBK, Bcl‐2, p53, and Bax as well as the extent of Akt phosphorylation were measured. In addition, cell colony formation, cell cycle, proliferation, apoptosis, migration, and invasion were detected. At last, tumor growth and the effect of miR‐372 on radiosensitivity of NPC were evaluated. Besides, over‐expressed miR‐372 down‐regulated Bcl‐2 and PBK expression and the extent of Akt phosphorylation while up‐regulated the expression of p53 and Bax. Additionally, miR‐372 over‐expression and radiotherapy inhibited cell clone formation, proliferation, tumor growth, migration, invasion, and cell cycle entry, but promoted cell apoptosis. However, the restoration of PBK in NPC cells expressing miR‐372 reversed the anti‐tumor effect of miR‐372 and activation of the p53 signaling pathway. In conclusion, the study shows that up‐regulated miR‐372 promotes radiosensitivity by activating the p53 signaling pathway via inhibition of PBK.
Collapse
Affiliation(s)
- Zhe Wang
- Department of Medical Oncology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China.,The Key Laboratory of Biomarker High Throughput Screening and Target Translation of Breast and Gastrointestinal Tumor, Dalian University, Dalian, China
| | - Ji-Wei Mao
- Department of Medical Oncology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China.,The Key Laboratory of Biomarker High Throughput Screening and Target Translation of Breast and Gastrointestinal Tumor, Dalian University, Dalian, China
| | - Guang-Yan Liu
- College of Basic Medical Sciences, Shenyang Medical College, Shenyang, China
| | - Fu-Guang Wang
- Department of Medical Oncology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Zai-Shuang Ju
- Department of Medical Oncology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Dong Zhou
- Department of Medical Oncology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Ruo-Yu Wang
- Department of Medical Oncology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China.,The Key Laboratory of Biomarker High Throughput Screening and Target Translation of Breast and Gastrointestinal Tumor, Dalian University, Dalian, China
| |
Collapse
|
21
|
Ji S, Su X, Zhang H, Han Z, Zhao Y, Liu Q. MicroRNA-372 functions as a tumor suppressor in cell invasion, migration and epithelial-mesenchymal transition by targeting ATAD2 in renal cell carcinoma. Oncol Lett 2018; 17:2400-2408. [PMID: 30719113 PMCID: PMC6350190 DOI: 10.3892/ol.2018.9871] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 12/05/2018] [Indexed: 12/30/2022] Open
Abstract
In recent years, renal cell carcinoma (RCC) has exhibited an increasing incidence and mortality rate worldwide. Accumulating evidence has identified that microRNAs (miRNAs) function as negative or positive regulators of many malignant tumors; however, the roles of miR-372 in RCC remain unclear. The focus of the present study was the functions of miR-372 in RCC metastasis and EMT. Data revealed that miR-372 expression levels were significantly downregulated in RCC tissue samples and cells. Moreover, the decreased expression levels were strongly associated with the poor survival rates and adverse clinical characteristics of RCC patients. Accordingly, miR-372 overexpression markedly inhibited RCC cell invasion, migration and EMT. In terms of the potential mechanisms, ATAD2, the expression of which was inversely correlated with miR-372 expression in RCC, was identified as a direct functional target of miR-372. Notably, ATAD2 silence exerted suppressive functions in RCC cells, being similar to the effects of miR-372 overexpression. In conclusion, findings of this study indicate that miR-372 repressed RCC EMT and metastasis via targeting ATAD2, suggesting that the miR-372/ATAD2 axis may be therapeutic biomarkers for RCC.
Collapse
Affiliation(s)
- Shiqi Ji
- Department of Urology, Beijing Ditan Hospital Capital Medical University, Capital Medical University, Beijing 100015, P.R. China
| | - Xiaolin Su
- Department of Emergency, Beijing First Hospital of Integrated Chinese and Western Medicine, Beijing 100021, P.R. China
| | - Haijian Zhang
- Department of Urology, Beijing Ditan Hospital Capital Medical University, Capital Medical University, Beijing 100015, P.R. China
| | - Zhixing Han
- Department of Urology, Beijing Ditan Hospital Capital Medical University, Capital Medical University, Beijing 100015, P.R. China
| | - Yuqian Zhao
- Department of Urology, Beijing Ditan Hospital Capital Medical University, Capital Medical University, Beijing 100015, P.R. China
| | - Qingjun Liu
- Department of Urology, Beijing Ditan Hospital Capital Medical University, Capital Medical University, Beijing 100015, P.R. China
| |
Collapse
|
22
|
Ding M, Lu X, Wang C, Zhao Q, Ge J, Xia Q, Wang J, Zen K, Zhang CY, Zhang C. The E2F1-miR-520/372/373-SPOP Axis Modulates Progression of Renal Carcinoma. Cancer Res 2018; 78:6771-6784. [PMID: 30348808 DOI: 10.1158/0008-5472.can-18-1662] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 09/11/2018] [Accepted: 10/15/2018] [Indexed: 11/16/2022]
Abstract
: Although renal cell carcinoma (RCC) is the most malignant urologic cancer, its pathogenesis remains unclear, and effective treatments for advanced RCC are still lacking. Here, we report that a novel E2F1-miR-520/372/373-SPOP axis controls RCC carcinogenesis. Speckle-type POZ protein (SPOP) was upregulated in over 90% of RCC tissues, whereas the miR-520/372/373 family was downregulated and correlated inversely with SPOP protein levels in RCC tissues. The miR-520/372/373 family targeted the SPOP 3'-UTR and suppressed SPOP protein expression, leading to elevation of PTEN and DUSP7 levels and, consequently, decreased proliferation, invasion/migration, and metastasis of RCC cells in vitro and in vivo. Tail-vein delivery of therapeutic miR-520/372/373 family significantly decreased both tumor size and lung metastasis ratio in mice bearing orthotopic xenograft tumors. Decreased expression of miR-520/372/373 family was mediated by transcription factor E2F1. In conclusion, our results demonstrate that the E2F1-miR-520/372/373-SPOP axis functions as a key signaling pathway in RCC progression and metastasis and represents a promising opportunity for targeted therapies. SIGNIFICANCE: These findings show that the E2F1-miR-520/372/373 family-SPOP axis promotes RCC progression, thereby contributing to our understanding of RCC pathogenesis and unveiling new avenues for more effective targeted therapies.
Collapse
Affiliation(s)
- Meng Ding
- Department of Clinical Laboratory, Jinling Hospital, State Key Laboratory of Analytical Chemistry for Life Science, NJU Advanced Institute for Life Sciences (NAILS), School of Life Sciences, Nanjing University, Nanjing, China.,State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute for Life Sciences (NAILS), School of Life Science, Nanjing University, Nanjing, China
| | - Xiaolan Lu
- Department of Clinical Laboratory, Jinling Hospital, State Key Laboratory of Analytical Chemistry for Life Science, NJU Advanced Institute for Life Sciences (NAILS), School of Life Sciences, Nanjing University, Nanjing, China.,State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute for Life Sciences (NAILS), School of Life Science, Nanjing University, Nanjing, China
| | - Cheng Wang
- Department of Clinical Laboratory, Jinling Hospital, State Key Laboratory of Analytical Chemistry for Life Science, NJU Advanced Institute for Life Sciences (NAILS), School of Life Sciences, Nanjing University, Nanjing, China.,State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute for Life Sciences (NAILS), School of Life Science, Nanjing University, Nanjing, China
| | - Quan Zhao
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute for Life Sciences (NAILS), School of Life Science, Nanjing University, Nanjing, China
| | - Jingping Ge
- Department of Urology, Jinling Hospital, Nanjing University School of Medicine, Nanjing University, Nanjing, China
| | - Qiuyuan Xia
- Department of Pathology, Jinling Hospital, Nanjing University School of Medicine, Nanjing University, Nanjing, China
| | - Junjun Wang
- Department of Clinical Laboratory, Jinling Hospital, State Key Laboratory of Analytical Chemistry for Life Science, NJU Advanced Institute for Life Sciences (NAILS), School of Life Sciences, Nanjing University, Nanjing, China
| | - Ke Zen
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute for Life Sciences (NAILS), School of Life Science, Nanjing University, Nanjing, China
| | - Chen-Yu Zhang
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute for Life Sciences (NAILS), School of Life Science, Nanjing University, Nanjing, China.
| | - Chunni Zhang
- Department of Clinical Laboratory, Jinling Hospital, State Key Laboratory of Analytical Chemistry for Life Science, NJU Advanced Institute for Life Sciences (NAILS), School of Life Sciences, Nanjing University, Nanjing, China. .,State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute for Life Sciences (NAILS), School of Life Science, Nanjing University, Nanjing, China
| |
Collapse
|
23
|
Verdelli C, Forno I, Morotti A, Creo P, Guarnieri V, Scillitani A, Cetani F, Vicentini L, Balza G, Beretta E, Ferrero S, Vaira V, Corbetta S. The aberrantly expressed miR-372 partly impairs sensitivity to apoptosis in parathyroid tumor cells. Endocr Relat Cancer 2018; 25:761-771. [PMID: 29724878 DOI: 10.1530/erc-17-0204] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 05/01/2018] [Indexed: 01/08/2023]
Abstract
Parathyroid tumors deregulate microRNAs belonging to the two clusters on the chromosome 19, the C19MC and miR-371-373 clusters. Here, we report that the embryonic miR-372 is aberrantly expressed in half of parathyroid adenomas (PAds) in most of atypical adenomas and carcinomas (n = 15). Through in situ hybridization, we identified that miR-372-positive parathyroid tumor cells were scattered throughout the tumor parenchyma. In PAd-derived cells, ectopic miR-372 inhibited the expression of its targets CDKN1A/p21 and LATS2 at both mRNA and protein levels. Although the viability of parathyroid cells was not affected by miR-372 overexpression, the miRNA blunted camptothecin-induced apoptosis in primary PAd-derived cultures. miR-372 overexpression in parathyroid tumor cells increased parathormone (PTH) mRNA levels, and it positively correlated in vivo with circulating PTH levels. Conversely, the parathyroid-specific genes TBX1 and GCM2 were not affected by miR-372 mimic transfection. Finally, miR-372 dampened the Wnt pathway in parathyroid tumor cells through DKK1 upregulation. In conclusion, miR-372 is a novel mechanism exploited by a subset of parathyroid tumor cells to partially decrease sensitivity to apoptosis, to increase PTH synthesis and to deregulate Wnt signaling.
Collapse
Affiliation(s)
- Chiara Verdelli
- Laboratory of Experimental EndocrinologyIRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Irene Forno
- Department of Pathophysiology and TransplantationUniversity of Milan, Milan, Italy
- Division of PathologyFondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Annamaria Morotti
- Department of Pathophysiology and TransplantationUniversity of Milan, Milan, Italy
- Division of PathologyFondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Pasquale Creo
- Laboratory of Stem Cells for Tissue EngineeringIRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - Vito Guarnieri
- Medical GeneticsIRCCS Hospital Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Alfredo Scillitani
- Endocrine UnitIRCCS Hospital Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Filomena Cetani
- Department of Endocrinology and MetabolismUniversity of Pisa, Pisa, Italy
| | - Leonardo Vicentini
- Endocrine SurgeryIRCCS Fondazione Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Gianni Balza
- Endocrinology UnitOspedale Manzoni, Lecco, Italy
| | | | - Stefano Ferrero
- Division of PathologyFondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of BiomedicalSurgical and Dental Sciences, University of Milan, Milan, Italy
| | - Valentina Vaira
- Department of Pathophysiology and TransplantationUniversity of Milan, Milan, Italy
- Division of PathologyFondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Sabrina Corbetta
- Endocrinology UnitDepartment of Biomedical Sciences for Health, University of Milan, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| |
Collapse
|
24
|
Nomikou E, Livitsanou M, Stournaras C, Kardassis D. Transcriptional and post-transcriptional regulation of the genes encoding the small GTPases RhoA, RhoB, and RhoC: implications for the pathogenesis of human diseases. Cell Mol Life Sci 2018; 75:2111-2124. [PMID: 29500478 PMCID: PMC11105751 DOI: 10.1007/s00018-018-2787-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 01/25/2018] [Accepted: 02/26/2018] [Indexed: 12/15/2022]
Abstract
Rho GTPases are highly conserved proteins that play critical roles in many cellular processes including actin dynamics, vesicular trafficking, gene transcription, cell-cycle progression, and cell adhesion. The main mode of regulation of Rho GTPases is through guanine nucleotide binding (cycling between an active GTP-bound form and an inactive GDP-bound form), but transcriptional, post-transcriptional, and post-translational modes of Rho regulation have also been described. In the present review, we summarize recent progress on the mechanisms that control the expression of the three members of the Rho-like subfamily (RhoA, RhoB, and RhoC) at the level of gene transcription as well as their post-transcriptional regulation by microRNAs. We also discuss the progress made in deciphering the mechanisms of cross-talk between Rho proteins and the transforming growth factor β signaling pathway and their implications for the pathogenesis of human diseases such as cancer metastasis and fibrosis.
Collapse
Affiliation(s)
- Eirini Nomikou
- Laboratory of Biochemistry, Department of Medicine, University of Crete, 71003, Heraklion, Greece
| | - Melina Livitsanou
- Laboratory of Biochemistry, Department of Medicine, University of Crete, 71003, Heraklion, Greece
| | - Christos Stournaras
- Laboratory of Biochemistry, Department of Medicine, University of Crete, 71003, Heraklion, Greece
| | - Dimitris Kardassis
- Laboratory of Biochemistry, Department of Medicine, University of Crete, 71003, Heraklion, Greece.
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, 71110, Heraklion, Greece.
| |
Collapse
|
25
|
An J, Xu J, Li J, Jia S, Li X, Lu Y, Yang Y, Lin Z, Xin X, Wu M, Zheng Q, Pu H, Gui X, Li T, Lu D. HistoneH3 demethylase JMJD2A promotes growth of liver cancer cells through up-regulating miR372. Oncotarget 2018; 8:49093-49109. [PMID: 28467776 PMCID: PMC5564752 DOI: 10.18632/oncotarget.17095] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 04/01/2017] [Indexed: 11/25/2022] Open
Abstract
Changes in histone lysine methylation status have been observed during cancer formation. JMJD2A protein is a demethylase that is overexpressed in several tumors. Herein, our results demonstrate that JMJD2A accelerates malignant progression of liver cancer cells in vitro and in vivo. Mechanistically, JMJD2A promoted the expression and mature of pre-miR372 epigenetically. Notably, miR372 blocks the editing of 13th exon-introns-14th exon and forms a novel transcript(JMJD2AΔ) of JMJD2A. In particular, JMJD2A inhibited P21(WAF1/Cip1) expression by decreasing H3K9me3 dependent on JMJD2AΔ. Thereby, JMJD2A could enhance Pim1 transcription by suppressing P21(WAF1/Cip1). Furthermore, through increasing the expression of Pim1, JMJD2A could facilitate the interaction among pRB, CDK2 and CyclinE which prompts the transcription and translation of oncogenic C-myc. Strikingly, JMJD2A may trigger the demethylation of Pim1. On the other hand, Pim1 knockdown and P21(WAF1/Cip1) overexpression fully abrogated the oncogenic function of JMJD2A. Our observations suggest that JMJD2A promotes liver cancer cell cycle progress through JMJD2A-miR372-JMJD2AΔ-P21WAF1/Cip1-Pim1-pRB-CDK2-CyclinE-C-myc axis. This study elucidates a novel mechanism for JMJD2A in liver cancer cells and suggests that JMJD2A can be used as a novel therapeutic targets of liver cancer.
Collapse
Affiliation(s)
- Jiahui An
- School of Life Science and Technology, Tongji University, Shanghai, 20092, China
| | - Jie Xu
- School of Life Science and Technology, Tongji University, Shanghai, 20092, China
| | - Jiao Li
- School of Medicine, Tongji University, Shanghai, 200092, China
| | - Song Jia
- School of Medicine, Tongji University, Shanghai, 200092, China
| | - Xiaonan Li
- School of Life Science and Technology, Tongji University, Shanghai, 20092, China
| | - Yanan Lu
- School of Life Science and Technology, Tongji University, Shanghai, 20092, China
| | - Yuxin Yang
- School of Life Science and Technology, Tongji University, Shanghai, 20092, China
| | - Zhuojia Lin
- School of Life Science and Technology, Tongji University, Shanghai, 20092, China
| | - Xiaoru Xin
- School of Life Science and Technology, Tongji University, Shanghai, 20092, China
| | - Mengying Wu
- School of Life Science and Technology, Tongji University, Shanghai, 20092, China
| | - Qidi Zheng
- School of Life Science and Technology, Tongji University, Shanghai, 20092, China
| | - Hu Pu
- School of Life Science and Technology, Tongji University, Shanghai, 20092, China
| | - Xin Gui
- School of Life Science and Technology, Tongji University, Shanghai, 20092, China
| | - Tianming Li
- School of Life Science and Technology, Tongji University, Shanghai, 20092, China
| | - Dongdong Lu
- School of Life Science and Technology, Tongji University, Shanghai, 20092, China
| |
Collapse
|
26
|
Lin Z, Lu Y, Meng Q, Wang C, Li X, Yang Y, Xin X, Zheng Q, Xu J, Gui X, Li T, Pu H, Xiong W, Li J, Jia S, Lu D. miR372 Promotes Progression of Liver Cancer Cells by Upregulating erbB-2 through Enhancement of YB-1. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 11:494-507. [PMID: 29858084 PMCID: PMC5992473 DOI: 10.1016/j.omtn.2018.04.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 03/30/2018] [Accepted: 04/04/2018] [Indexed: 12/16/2022]
Abstract
MicroRNAs are known to be involved in carcinogenesis. Recently, microRNA-372 (miR372) has been proven to play a substantial role in several human cancers, but its functions in liver cancer remain unclear. Herein, our results demonstrate that miR372 accelerates growth of liver cancer cells in vitro and in vivo. Mechanistically, miR372 enhances expression of Y-box-binding protein 1 (YB-1) by targeting for phosphatase and tensin homolog (PTEN) directly and consequently promotes phosphorylation of YB-1 via HULC looping dependent on ERK1/2 and PTEN. In particular, HULC knockdown or PTEN overexpression abrogated this miR372 action. Moreover, miR372 inhibits the degradation of β-catenin dependent on phosphorylation of YB-1 and then enhances the expression and activity of pyruvate kinase M2 isoform (PKM2) by β-catenin-LEF/TCF4 pathway. Furthermore, the loading of LEF/TCF4 on PKM2 promoter region was significantly increased in miR372 overexpressing Hep3B, and thus, glycolytic proton efflux rate (glycoPER) was significantly increased in rLV-miR372 group compared to the rLV group. Moreover, β-catenin knockdown abrogates this function of miR372. Ultimately, miR372 promotes the expression of erbB-2 through PKM2-pH3T11-acetylation on histone H3 lysine 9 (H3K9Ac) pathway. Of significance, both YB-1 knockdown and erbB-2 knockdown abrogate oncogenic action of miR372. Our observations suggest that miR372 promotes liver cancer cell cycle progress by activating cyclin-dependent kinase 2 (CDK2)-cyclin E-P21/Cip1 complex through miR372-YB-1-β-catenin-LEF/TCF4-PKM2-erbB-2 axis. This study elucidates a novel mechanism for miR372 in liver cancer cells and suggests that miR372 can be used as a novel therapeutic target of liver cancer.
Collapse
Affiliation(s)
- Zhuojia Lin
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Yanan Lu
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Qiuyu Meng
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Chen Wang
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Xiaonan Li
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Yuxin Yang
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Xiaoru Xin
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Qidi Zheng
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Jie Xu
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Xin Gui
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Tianming Li
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Hu Pu
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Wujun Xiong
- Department of Hepatology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Jiao Li
- School of Medicine, Tongji University, Shanghai 200092, China
| | - Song Jia
- School of Medicine, Tongji University, Shanghai 200092, China
| | - Dongdong Lu
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, Shanghai 200092, China.
| |
Collapse
|
27
|
Li L, Ma L. Upregulation of miR-582-5p regulates cell proliferation and apoptosis by targeting AKT3 in human endometrial carcinoma. Saudi J Biol Sci 2018; 25:965-970. [PMID: 30108448 PMCID: PMC6088104 DOI: 10.1016/j.sjbs.2018.03.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 03/07/2018] [Accepted: 03/08/2018] [Indexed: 02/07/2023] Open
Abstract
The human endometrial carcinoma is one of the most common female malignancies, and there is an urgent requirement to explore new therapeutic strategies. There is accumulating evidence that microRNAs (miRNAs) can serve as potential diagnostic and prognostic biomarkers for various types of cancer, but the significance of miR-582-5p still remains largely unknown in the endometrial carcinoma. The aims of this study were to understand and identify the influence of miR-582-5p on the proliferation and apoptosis of human endometrial carcinoma and its relevant mechanism. First, quantitative real-time PCR (qRT-PCR) was used to detect miR-582-5p and AKT3 expression in human tissue samples and cells. Then, CyQuant assay and 2D colony assay were employed to evaluate cell proliferation. Western blotting was used to determine protein expression. Subsequently, the luciferase reporter assay was used to identify the target of miR-582-5p. Finally, Annexin V assay was used to detect cell apoptosis. We found that miR-582-5p expression was significantly decreased in human endometrial carcinoma tissues, and miR-582-5p upregulation in human endometrial carcinoma cells inhibit cell proliferation and promote apoptosis. Moreover, AKT3 was validated as a target of miR-582-5p and AKT3 expression was inversely correlated with miR-582-5p expression. Besides, AKT3 upregulation efficiently abrogates the effect of miR-582-5p on the cells. These results demonstrated that miR-582-5p regulates cell proliferation and apoptosis in human endometrial carcinoma via AKT3. Thus, miR-582-5p represents a potential therapeutic target in human endometrial carcinoma meriting further investigation.
Collapse
Affiliation(s)
- Lingling Li
- Department of Gynecology of Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang 471000, PR China
| | - Li Ma
- Department of Gynecology of Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang 471000, PR China
| |
Collapse
|
28
|
Cheng X, Chen J, Huang Z. miR-372 promotes breast cancer cell proliferation by directly targeting LATS2. Exp Ther Med 2018; 15:2812-2817. [PMID: 29456685 PMCID: PMC5795589 DOI: 10.3892/etm.2018.5761] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 01/05/2018] [Indexed: 12/14/2022] Open
Abstract
MicroRNAs (miRs) have previously been demonstrated to be important in the tumorigenesis and progression of breast cancer. miR-372 was previously revealed to be involved in various types of human cancer, however its function in breast cancer remains largely unknown. The present study demonstrated that miR-372 is frequently overexpressed in breast cancer cell lines and tissues. The downregulation of miR-372 markedly inhibited cell proliferation, arrested the cell cycle in the G1/S phase, and increased the apoptosis of breast cancer cells. Consistently, an in vivo xenograft study also demonstrated the suppressive effects of miR-372 knockdown on tumor growth. Further studies revealed that miR-372 modulated the expression of large tumor suppressor kinase 2 (LATS2) by directly targeting its 3′-untranslated region in breast cancer cells. Furthermore, silencing of LATS2 was able to rescue the effect of the miR-372 inhibitor. Overall, the results suggest that miR-372 functions as an oncogenic miRNA in breast cancer by targeting LATS2.
Collapse
Affiliation(s)
- Xueyuan Cheng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China.,Department of General Surgery, Beihai People's Hospital, Beihai, Guangxi 536000, P.R. China
| | - Junqiang Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Zhong Huang
- Department of General Surgery, Beihai People's Hospital, Beihai, Guangxi 536000, P.R. China
| |
Collapse
|
29
|
Srivastava SK, Ahmad A, Zubair H, Miree O, Singh S, Rocconi RP, Scalici J, Singh AP. MicroRNAs in gynecological cancers: Small molecules with big implications. Cancer Lett 2017; 407:123-138. [PMID: 28549791 PMCID: PMC5601032 DOI: 10.1016/j.canlet.2017.05.011] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 05/10/2017] [Accepted: 05/15/2017] [Indexed: 12/14/2022]
Abstract
Gynecological cancers (GCs) are often diagnosed at advanced stages, limiting the efficacy of available therapeutic options. Thus, there remains an urgent and unmet need for innovative research for the efficient clinical management of GC patients. Research over past several years has revealed the enormous promise of miRNAs. These small non-coding RNAs can aid in the diagnosis, prognosis and therapy of all major GCs, viz., ovarian cancers, cervical cancers and endometrial cancers. Mechanistic details of the miRNAs-mediated regulation of multiple biological functions are under constant investigation, and a number of miRNAs are now believed to influence growth, proliferation, invasion, metastasis, chemoresistance and the relapse of different GCs. Modulation of tumor microenvironment by miRNAs can possibly explain some of their reported biological effects. miRNA signatures have been proposed as biomarkers for the early detection of GCs, even the various subtypes of individual GCs. miRNA signatures are also being pursued as predictors of response to therapies. This review catalogs the knowledge gained from collective studies, so as to assess the progress made so far. It is time to ponder over the knowledge gained, so that more meaningful pre-clinical and translational studies can be designed to better realize the potential that miRNAs have to offer.
Collapse
Affiliation(s)
- Sanjeev K Srivastava
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA.
| | - Aamir Ahmad
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | - Haseeb Zubair
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | - Orlandric Miree
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | - Seema Singh
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA; Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA
| | - Rodney P Rocconi
- Division of Gynecologic Oncology, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | - Jennifer Scalici
- Division of Gynecologic Oncology, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | - Ajay P Singh
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA; Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA.
| |
Collapse
|
30
|
Zhao YX, Liu HC, Ying WY, Wang CY, Yu YJ, Sun WJ, Liu JF. microRNA‑372 inhibits proliferation and induces apoptosis in human breast cancer cells by directly targeting E2F1. Mol Med Rep 2017; 16:8069-8075. [PMID: 28944922 PMCID: PMC5779890 DOI: 10.3892/mmr.2017.7591] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 04/19/2017] [Indexed: 12/23/2022] Open
Abstract
Breast cancer is the most prevalent cancer and the leading cause of cancer-associated mortalities among women worldwide today. Accumulating evidence suggested that miR-372 may serve important roles in the initiation and development of various human cancers. However, the role of miR-372 in breast cancer remains unknown. The present study demonstrated that the expression level of miR-372 in human breast cancer tissues and cell lines is significantly reduced compared with normal breast tissues cell lines. Furthermore, results of functional assays indicated that miR-372 inhibits cell proliferation and induces apoptosis in the MCF-7 human breast cancer cell line. E2F1 was identified as a direct functional target of miR-372 in breast cancer. In conclusion, the findings revealed that miR-372 may have the potential to act as a novel molecule for the diagnosis and therapy of patients with breast cancer.
Collapse
Affiliation(s)
- Ya-Xin Zhao
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Hua-Cheng Liu
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Wei-Yang Ying
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Cheng-Yu Wang
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Yao-Jun Yu
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Wei-Jian Sun
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Jie-Fan Liu
- Department of General Practice, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| |
Collapse
|
31
|
Abstract
Rho GTPases are regulators of many cellular functions and are often dysregulated in cancer. However, the precise role of Rho proteins for tumor development is not well understood. In breast cancer, overexpression of RhoC is linked with poor prognosis. Here, we aim to compare the function of RhoC and its homolog family member RhoA in breast cancer progression. We established stable breast epithelial cell lines with inducible expression of RhoA and RhoC, respectively. Moreover, we made use of Rho-activating bacterial toxins (Cytotoxic Necrotizing Factors) to stimulate the endogenous pool of Rho GTPases in benign breast epithelial cells and simultaneously knocked down specific Rho proteins. Whereas activation of Rho GTPases was sufficient to induce an invasive phenotype in three-dimensional culture systems, overexpression of RhoA or RhoC were not. However, RhoC but not RhoA was required for invasion, whereas RhoA and RhoC equally regulated proliferation. We further identified downstream target genes of RhoC involved in invasion and identified PTGS2 (COX-2) being preferentially upregulated by RhoC. Consistently, the COX-2 inhibitor Celecoxib blocked the invasive phenotype induced by the Rho-activating toxins.
Collapse
|
32
|
Chen H, Zhang Z, Lu Y, Song K, Liu X, Xia F, Sun W. Downregulation of ULK1 by microRNA-372 inhibits the survival of human pancreatic adenocarcinoma cells. Cancer Sci 2017; 108:1811-1819. [PMID: 28677209 PMCID: PMC5581518 DOI: 10.1111/cas.13315] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 06/28/2017] [Accepted: 07/02/2017] [Indexed: 12/12/2022] Open
Abstract
Dysregulation of microRNA (miRNA) expression in various cancers and their role in cancer progression is well documented. The purpose of this study was to investigate the biological role of miR‐372 in human pancreatic adenocarcinoma (HPAC). We collected 20 pairs of HPAC tissues and adjacent non‐cancerous tissues to detect miR‐372 expression levels. We transfected BXPC‐3 and PANC‐1 cells with miR‐372 inhibitor/mimics to study their effect on cell proliferation, apoptosis, invasion, migration and autophagy. In addition, miR‐372 mimics and a tumor protein UNC51‐like kinase 1 (ULK1) siRNA were co‐transfected into BXPC‐3 and PANC‐1 cells to explore the mechanism of miR‐372 and ULK1 on HPAC tumorigenesis. We found that the expression of miR‐372 was markedly downregulated in HPAC cells compared to adjacent normal tissues. Furthermore, functional assays showed that miR‐372 inhibited cell proliferation, invasion, migration and autophagy in BXPC‐3 and PANC‐1 cells. An inverse correlation between miR‐372 expression and ULK1 expression was observed in HPAC tissues. Downregulation of ULK1 inhibited the overexpression effects of miR‐372, and upregulation of ULK1 reversed the effects of overexpressed miR‐372. Finally, we found that silencing ULK1 or inhibiting autophagy partly rescued the effects of miR‐372 knockdown in HPAC cells, which may explain the influence of miR‐372/ULK1 in HPAC development. Taken together, these results revealed a significant role of the miR‐372/ULK1 axis in suppressing HPAC cell proliferation, migration, invasion and autophagy.
Collapse
Affiliation(s)
- Hongxi Chen
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Zhipeng Zhang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Yebin Lu
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Kun Song
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Xiwu Liu
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Fada Xia
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Weijia Sun
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
33
|
The role of miR-372 in ovarian carcinoma cell proliferation. Gene 2017; 624:14-20. [DOI: 10.1016/j.gene.2017.04.043] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Accepted: 04/25/2017] [Indexed: 01/22/2023]
|
34
|
Wang Q, Liu S, Zhao X, Wang Y, Tian D, Jiang W. MiR-372-3p promotes cell growth and metastasis by targeting FGF9 in lung squamous cell carcinoma. Cancer Med 2017; 6:1323-1330. [PMID: 28440022 PMCID: PMC5463061 DOI: 10.1002/cam4.1026] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 12/26/2016] [Accepted: 01/06/2017] [Indexed: 01/04/2023] Open
Abstract
The aim of this study was to study the role of miR‐372‐3p in lung squamous cell carcinoma (LSCC) cell proliferation and invasion by suppressing FGF9. RT‐PCR was used to determine miR‐372‐3p and FGF9 mRNA expression in tissues and cells. Western blot was used to determine FGF9 expression in tissues and NCI‐H520 cell line. Dual luciferase reporter gene assay was conducted to confirm that FGF9 can be directly targeted by miR‐372‐3p. MTT, colony formation assays were conducted to investigate the effects of ectopic miR‐372‐3p and FGF9 expression on NCI‐H520 cell growth. Flow cytometry was used to analyze the influence of miR‐372‐3p and FGF9 expression on cell cycle distribution and apoptosis. Transwell assay was also conducted to see the effects of miR‐372‐3p and FGF9 expression on NCI‐H520 cell invasiveness. MiR‐372‐3p was found significantly overexpressed in both LSCC tissues and cell lines, whereas FGF9 mRNA was found underexpressed in LSCC tissues. MiR‐372‐3p directly bound to wild‐type FGF9 mRNA 3′UTR, therefore led to the reduction in FGF9 expression. The upregulation of FGF9 or the downregulation of miR‐372‐3p substantially retarded LSCC cell growth, mitosis, and invasion. MiR‐372‐3p enhanced LSCC cell proliferation and invasion through inhibiting FGF9.
Collapse
Affiliation(s)
- Qing Wang
- Department of Thoracic Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110032, China
| | - Siyang Liu
- Department of Thoracic Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110032, China
| | - Xitong Zhao
- Department of Thoracic Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110032, China
| | - Yuan Wang
- Department of Thoracic Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110032, China
| | - Dali Tian
- Department of Thoracic Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110032, China
| | - Wenjun Jiang
- Department of Thoracic Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110032, China
| |
Collapse
|
35
|
Stope MB, Koensgen D, Weimer J, Paditz M, Burchardt M, Bauerschlag D, Mustea A. The future therapy of endometrial cancer: microRNA's functionality, capability, and putative clinical application. Arch Gynecol Obstet 2016; 294:889-895. [PMID: 27637583 DOI: 10.1007/s00404-016-4194-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 09/05/2016] [Indexed: 01/27/2023]
Abstract
PURPOSE Endometrial cancer (EC) therapy is characterized by the heterogeneity of EC subtypes resulting in unclear clinical behavior as well as in unsatisfactory treatment options. The available biomarkers, such as cellular tumor antigen p53 (TP53), phosphatidylinositol 3,4,5-trisphosphate 3-phosphatase and dual-specificity protein phosphatase (PTEN), and phosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) genes alone might not be sufficient, and thus, new predictive and prognostic biomarkers are urgently required. The biomolecule class of microRNA represents a group of endogenously expressed regulatory factors primarily involved in control of pivotal cancer-related mechanisms including cell cycle, proliferation, apoptosis, and metastasis. Here, we review the current state of science regarding microRNA functionality in EC progression.
Collapse
Affiliation(s)
- Matthias B Stope
- Cancer Laboratory, Department of Urology, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475, Greifswald, Germany.
| | - Dominique Koensgen
- Department of Gynaecology and Obstetrics, University Medicine Greifswald, Greifswald, Germany
| | - Jörg Weimer
- Department of Gynecology and Obstetrics, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Madeleine Paditz
- Department of Gynaecology and Obstetrics, University Medicine Greifswald, Greifswald, Germany
| | - Martin Burchardt
- Cancer Laboratory, Department of Urology, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475, Greifswald, Germany
| | - Dirk Bauerschlag
- Department of Gynecology and Obstetrics, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Alexander Mustea
- Department of Gynaecology and Obstetrics, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|