1
|
Yu J, Guo K, Xia M, Ma X. EEE1F1 could serve as a prognostic biomarker for patients with endometrial cancer: A comprehensive analysis by integrating bulk and single-cell sequencing. Asian J Surg 2024:S1015-9584(24)02668-X. [PMID: 39668036 DOI: 10.1016/j.asjsur.2024.11.065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 11/07/2024] [Indexed: 12/14/2024] Open
Affiliation(s)
- Jiayuan Yu
- Department of Gynecology and Obstetrics, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, 264000, Yantai, China
| | - Kui Guo
- Department of Gynecology and Obstetrics, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, 264000, Yantai, China
| | - Min Xia
- Department of Gynecology and Obstetrics, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, 264000, Yantai, China.
| | - Xiaohong Ma
- Department of Gynecology and Obstetrics, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, 264000, Yantai, China.
| |
Collapse
|
2
|
Wang L, Cao J, Tao J, Liang Y. STMN1 promotes cell malignancy and bortezomib resistance of multiple myeloma cell lines via PI3K/AKT signaling. Expert Opin Drug Saf 2024; 23:277-286. [PMID: 37642368 DOI: 10.1080/14740338.2023.2251384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/31/2023] [Accepted: 08/09/2023] [Indexed: 08/31/2023]
Abstract
BACKGROUND This study investigates the biological functions of Stathmin1 (STMN1) involving drug resistance and cell proliferation in multiple myeloma (MM) and its related mechanisms. METHODS Bone marrow aspirates were collected from 20 MM patients, and the bone marrow mononuclear cells (BMMCs) were separated by Ficoll-Hypaque density gradient centrifugation. Blood samples of 20 patients with monoclonal gammopathy of undetermined significance (MGUS) and 20 healthy donors were collected. Normal plasma cells sorted from the peripheral blood of MGUS patients and healthy subject as controls. Two bortezomib (BTZ)-resistant MM cell lines were established, namely NCI-H929/BTZ and KM3/BTZ cells, and then transfected with lentiviruses packaging sh-STMN1 to knock down STMN1 level in BTZ-resistant cells. Expression of STMN1 was assessed by RT-qPCR and western blotting. CCK-8 assays were performed to assess 50% growth inhibition (IC50) values. Green fluorescent protein in BTZ-resistant cells infected with lentiviruses was observed by fluorescence microscopy. Cell viability, proliferation, cell cycle, and apoptosis were evaluated through MTT assays, colony formation assays, flow cytometry analyses, and TUNEL staining. RESULTS STMN1 was upregulated in MM cells and bone marrow aspirates of MM patients. Additionally, STMN1 depletion attenuated BTZ resistance in MM cells. Moreover, downregulation of STMN1 limited the malignant phenotypes of BTZ-resistant cells. Mechanistically, the PI3K/Akt signaling was inactivated by STMN1 downregulation in BTZ-resistant cells. CONCLUSION STMN1 silencing inhibits cell proliferation and BTZ resistance in MM by inactivating the PI3K/Akt signaling.
Collapse
Affiliation(s)
- Ling Wang
- Department of Hematology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Jie Cao
- Department of Pathology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Jian Tao
- Department of Hematology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Yan Liang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medicine University, Nanjing, China
| |
Collapse
|
3
|
Yousif A, Ebeid A, Kacsoh B, Bazzaro M, Chefetz I. The Ovary-Brain Connection. Cells 2024; 13:94. [PMID: 38201298 PMCID: PMC10778337 DOI: 10.3390/cells13010094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 12/26/2023] [Indexed: 01/12/2024] Open
Abstract
The brain and the ovaries are in a state of continuous communication [...].
Collapse
Affiliation(s)
- Abdelrahman Yousif
- Department of Obstetrics and Gynecology, Texas Tech University Health Sciences Center, El Paso, TX 79905, USA
| | - Ahmed Ebeid
- Department of Obstetrics and Gynecology, The George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA
| | - Balint Kacsoh
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA 31207, USA
| | - Martina Bazzaro
- Masonic Cancer Center and Department of Obstetrics, Gynecology and Women’s Health, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Biomedical and Clinical Science, Linköping University, SE-581 85 Linköping, Sweden
| | - Ilana Chefetz
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA 31207, USA
| |
Collapse
|
4
|
Yang Z, Liu T, Ren X, Yang M, Tu C, Li Z. Mir-34a: a regulatory hub with versatile functions that controls osteosarcoma networks. Cell Cycle 2022; 21:2121-2131. [PMID: 35699451 DOI: 10.1080/15384101.2022.2087755] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Osteosarcoma (OS) is one of the most prevalent and highly aggressive bone malignancies. The treatment strategies of OS is under standard regimens, including surgical resection, chemotherapy, and other adjuvant therapy. However, the 5-year survival rate is still unsatisfactory. Previous studies have demonstrated that the expression of miR-34a decreases in osteosarcoma, which is involved in regulating numerous genes directly or indirectly at the post-transcriptional level and other pathways. Thus, miR-34a plays an important role in mediating OS cell proliferation, differentiation, migration, and apoptosis, and might be a pivotal biomarker for OS with diagnostic and therapeutic potentials. In this review, we aim to summarize the relationship between miR-34a and OS, with an emphasis on the specific mechanisms in OS development referring to miR-34a. Moreover, the potential role of miR-34a as a diagnostic, prognostic, and therapeutic candidate for OS would be presented in detail. However, the molecular mechanisms related to miR-34a and OS remain elusive, and more investigations are needed to reach a comprehensive understanding.
Collapse
Affiliation(s)
- Zhimin Yang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Hunan, China.,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Hunan, China
| | - Tang Liu
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Hunan, China.,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Hunan, China
| | - Xiaolei Ren
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Hunan, China.,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Hunan, China
| | - Mei Yang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Hunan, China.,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Hunan, China
| | - Chao Tu
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Hunan, China.,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Hunan, China
| | - Zhihong Li
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Hunan, China.,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Hunan, China
| |
Collapse
|
5
|
Overexpression of Stathmin 1 Predicts Poor Prognosis and Promotes Cancer Cell Proliferation and Migration in Ovarian Cancer. DISEASE MARKERS 2022; 2022:3554100. [PMID: 35186166 PMCID: PMC8849943 DOI: 10.1155/2022/3554100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 01/25/2022] [Accepted: 01/28/2022] [Indexed: 12/12/2022]
Abstract
Purpose The aim of this study was to investigate the expression of stathmin 1 (STMN1) in ovarian cancer and its effect on prognosis. The effect and mechanism of STMN1 on the proliferation and migration of ovarian cancer cells were also investigated. Methods Expression of STMN1 was measured by immunohistochemical staining in ovarian cancer tissues. The effects of STMN1 on the proliferation and migration capacity of ovarian cancer were evaluated using Cell Counting Kit-8 (CCK-8) assays, colony formation assays, immunofluorescence staining, wound healing assays, and Transwell assays. Transcription factors were predicted by bioinformatic analysis of TCGA database. Results STMN1 was upregulated in ovarian cancer tissues as compared to paracancerous tissues and associated with shorter overall survival. STMN1 expression significantly correlated with FIGO staging and tumor differentiation (P < 0.05). Furthermore, STMN1 promoted proliferation and migration in ovarian cancer cell lines. Bioinformatic analysis revealed that STMN1 was potentially regulated by E2F transcription factors. Then, we found that E2F1 regulated the expression of STMN1 and affected proliferation. Conclusion STMN1 is overexpressed in ovarian cancer, and its high expression suggests a poor prognosis. STMN1 promotes the proliferation and migration of ovarian cancer and is regulated by E2F1. Thus, STMN1 may serve as a negative prognostic factor and possible target for the treatment of ovarian cancer patients.
Collapse
|
6
|
Han R, Feng P, Pang J, Zou D, Li X, Geng C, Li L, Min J, Shi J. A Novel HCC Prognosis Predictor EEF1E1 Is Related to Immune Infiltration and May Be Involved in EEF1E1/ATM/p53 Signaling. Front Oncol 2021; 11:700972. [PMID: 34282404 PMCID: PMC8285289 DOI: 10.3389/fonc.2021.700972] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 06/18/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND EEF1E1 has been reported to play a role in ovarian cancer, breast cancer, non-small cell lung cancer and other cancers, but its role and mechanism in hepatocellular carcinoma (HCC) are still unknown. METHODS EEF1E1 expression in human HCC was analyzed via the GTEx and TCGA database. Logistic regression analysis was used to analyze the clinicopathological correlation of EEF1E1 expression. The correlation between EEF1E1 expression and patients' prognosis was analyzed in HCC, shown by forest plots, nomogram and Kaplan-Meier curves. Hazard ratio (HR) with 95% confidence intervals and log-rank p-value were calculated via multivariate/univariate survival analyses. Moreover, the correlation between EEF1E1 and tumor immune infiltration was analyzed using the gsva package with the ssgsea algorithm. Pearson correlation was used to investigate the correlation between EEF1E1 expression and p53 pathway genes expression. Two third-party databases were used to validate the content of EEF1E1 protein and mRNA expression patterns and prognosis analysis. The immunohistochemistry and multiplex immunohistochemistry was used to verify the bio-informatics results. RESULTS EEF1E1 mRNA and protein expression in tumor was statistically higher than normal (EEF1E1 mRNA: p < 0.001; EEF1E1 protein: p < 0.01). Results from paired t-test (cancer and adjacent tissues) exhibited consistent trend (t = 7.572, p < 0.001). Immunohistochemistry showed that EEF1E1 is highly expressed in cancer. The expression of EEF1E1 was positively correlated with body weight, BMI, tumor status, vascular invasion, AFP, logistic grade, T stage and pathological stage. The univariate Cox model revealed that high EEF1E1 expression was strongly associated with worse OS (HR=2.581; 95% CI: 1.782-3.739; p < 0.001), as was T stage, pathologic stage, Histologic grade. High EEF1E1 expression was the only independent prognostic factor associated with OS (HR=2.57; 95% CI: 1.715-3.851; p < 0.001) in the multivariate analysis. EEF1E1 was significantly correlated with various immune cells, including cytotoxic cells, DC, macrophages, neutrophils, NK cd56bright, TFH, Tgd, Th17, Th2, Treg. Multiplex immunohistochemistry showed that the EEF1E1 protein level is positively correlated to the CD3, CD4, PD1 and is negatively correlated to the CD8. The expression level of EEF1E1 in HCC was significantly correlated with the key genes involved in the p53 pathway. The expression of EEF1E1, ATM, p53 and CASPASE3 in HCC tissues was significantly higher than that in adjacent tissues. CONCLUSION EEF1E1 is highly expressed in cancer tissues in HCC. EEF1E1's high expression is significantly correlated with worse prognosis and immune cell infiltration of HCC. EEF1E1 may be participating in EEF1E1/ATM/p53 signaling pathway in HCC.
Collapse
Affiliation(s)
- Ruiqin Han
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Penghui Feng
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Junyi Pang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dingfeng Zou
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaolu Li
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chao Geng
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lili Li
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie Min
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Jing Shi
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
7
|
Kazmierczak D, Jopek K, Sterzynska K, Ginter-Matuszewska B, Nowicki M, Rucinski M, Januchowski R. The Significance of MicroRNAs Expression in Regulation of Extracellular Matrix and Other Drug Resistant Genes in Drug Resistant Ovarian Cancer Cell Lines. Int J Mol Sci 2020; 21:ijms21072619. [PMID: 32283808 PMCID: PMC7177408 DOI: 10.3390/ijms21072619] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/02/2020] [Accepted: 04/07/2020] [Indexed: 12/11/2022] Open
Abstract
Ovarian cancer rates the highest mortality among all gynecological malignancies. The main reason for high mortality is the development of drug resistance. It can be related to increased expression of drug transporters and increased expression of extracellular matrix (ECM) proteins. Our foremost aim was to exhibit alterations in the miRNA expression levels in cisplatin (CIS), paclitaxel (PAC), doxorubicin (DOX), and topotecan (TOP)-resistant variants of the W1 sensitive ovarian cancer cell line-using miRNA microarray. The second goal was to identify miRNAs responsible for the regulation of drug-resistant genes. According to our observation, alterations in the expression of 40 miRNAs were present. We could observe that, in at least one drug-resistant cell line, the expression of 21 miRNAs was upregulated and that of 19 miRNAs was downregulated. We identified target genes for 22 miRNAs. Target analysis showed that miRNA regulates key genes responsible for drug resistance. Among others, we observed regulation of the ATP-binding cassette subfamily B member 1 gene (ABCB1) in the paclitaxel-resistant cell line by miR-363 and regulation of the collagen type III alpha 1 chain gene (COL3A1) in the topotekan-resistant cell line by miR-29a.
Collapse
|
8
|
Huang Z, Wu X, Li J. miR-101 suppresses colon cancer cell migration through regulation of EZH2. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2020; 113:255-260. [DOI: 10.17235/reed.2020.6800/2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
9
|
Rajan A, Zhao C. Deciphering the biology of thymic epithelial tumors. MEDIASTINUM (HONG KONG, CHINA) 2019; 3:36. [PMID: 31608319 PMCID: PMC6788633 DOI: 10.21037/med.2019.08.03] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 08/11/2019] [Indexed: 12/25/2022]
Abstract
Thymic cancers arise from epithelial cells of the thymus and have a predilection for intrathoracic spread. Clinical behavior varies from relatively indolent to highly aggressive with a capacity to metastasize widely and adversely affect survival. Paraneoplastic autoimmune disorders are frequently observed in association with thymoma and have a significant impact on quality of life. Underlying immune deficits associated with thymic epithelial tumors (TETs) increase the risk for development of opportunistic infections and emergence of extrathymic malignancies. Advances in the molecular characterization of thymic tumors have revealed the lowest tumor mutation burden among all adult cancers and the occurrence of distinct molecular subtypes of these diseases. Mutations in general transcription factor IIi (GTF2I) are unique to TETs and are rarely observed in other malignancies. The infrequency of actionable mutations has created obstacles for the development of biologic therapies and has spurred research to uncover druggable genomic targets. Persistence of autoreactive T cells due to altered thymic function increases the risk for development of severe immune-related toxicity and limits opportunities for use of immune-based therapies, especially in patients with thymoma. In this paper we review emerging data on the molecular characterization and immunobiology of thymic tumors and highlight clinical implications of these discoveries.
Collapse
Affiliation(s)
- Arun Rajan
- Thoracic and Gastrointestinal Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Chen Zhao
- Thoracic and Gastrointestinal Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
10
|
MicroRNAs as Potential Biomarkers for Chemoresistance in Adenocarcinomas of the Esophagogastric Junction. JOURNAL OF ONCOLOGY 2019; 2019:4903152. [PMID: 31467538 PMCID: PMC6701342 DOI: 10.1155/2019/4903152] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 07/15/2019] [Indexed: 12/11/2022]
Abstract
Concerning adenocarcinomas of the esophagogastric junction, neoadjuvant chemotherapy is regularly implemented, but patients' response varies greatly, with some cases showing no therapeutic effect, being deemed as chemoresistant. Small, noncoding RNAs (miRNAs) have evolved as key players in biological processes, including malignant diseases, often promoting tumor growth and expansion. In addition, specific miRNAs have been implicated in the development of chemoresistance through evasion of apoptosis, cell cycle alterations, and drug target modification. We performed a retrospective study of 33 patients receiving neoadjuvant chemotherapy by measuring their miRNA expression profiles. Histologic tumor regression was evaluated using resection specimens, while miRNA profiles were prepared using preoperative biopsies without prior therapy. A preselected panel of 96 miRNAs, known to be of importance in various malignancies, was used to test for significant differences between responsive (chemosensitive) and nonresponsive (chemoresistant) cases. The cohort consisted of 12 nonresponsive and 21 responsive cases with the following 4 miRNAs differentially expressed between both the groups: hsa-let-7f-5p, hsa-miRNA-221-3p, hsa-miRNA-31-5p, and hsa-miRNA-191-5p. The former 3 showed upregulation in chemoresistant cases, while the latter showed upregulation in chemosensitive cases. In addition, significant correlation between high expression of hsa-miRNA-194-5p and prolonged survival could be demonstrated (p value <0.0001). In conclusion, we identified a panel of 3 miRNAs predicting chemoresistance and a single miRNA contributing to chemosensitivity. These miRNAs might function as prognostic biomarkers and enable clinicians to better predict the effect of one or more reliably select patients benefitting from (neoadjuvant) chemotherapy.
Collapse
|
11
|
Tan Y, Bi X, Wang Q, Li Y, Zhang N, Lao J, Liu X. Dexmedetomidine protects PC12 cells from lidocaine-induced cytotoxicity via downregulation of Stathmin 1. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:2067-2079. [PMID: 31308624 PMCID: PMC6618032 DOI: 10.2147/dddt.s199572] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 04/04/2019] [Indexed: 12/26/2022]
Abstract
Background: Understanding of lidocaine-induced neurotoxicity is not complete, resulting in the unsuccessful treatment in some clinical settings. Dexmedetomidine (DEX) has been shown to alleviate lidocaine-induced neurotoxicity in our previous cell model. However, the rationale for DEX combined with lidocaine to reduce lidocaine-induced neurotoxicity in the clinical setting remains to be further clarified in the detailed molecular mechanism. Methods: In this study, we established a cellular injury model by lidocaine preconditioning. Cell Counting Kit-8 (CCK-8) and 5-ethynyl-2'-deoxyuridine (EdU) proliferation assay kit were used to analyze cell proliferation. Cell apoptosis was measured by flow cytometry and Hoechst 33342 staining. Cell cycle progression was detected by flow cytometry. The protein expression levels were detected by Western blotting and immunofluorescence staining. Results: Our results showed that DEX dose-dependently restored impaired proliferation of PC12 cells induced by lidocaine,as reflected by the increased cell viability and EdU positive cells, which were consistent with the decreased expression of tumor suppressor protein p21 and increased expression of cell cycle-related cyclin D1 and CDK1. In addition, DEX dose-dependently reduced apoptotic PC12 cells induced by lidocaine,as reflected by the decreased expression of apoptosis-related Bax, caspase-3 and caspase-9 and increased expression of anti-apoptotic Bcl-2 compared to the cells only treated with lidocaine. Mechanistically, with gain-or-loss-of-function of STMN1, we showed that DEX-mediated neuroprotection by lidocaine-induced damage is associated with downregulation of STMN1 which might be an upstream molecule involved in regulation of mitochondria death pathway. Conclusion: Our results reveal that DEX is likely to be an effective adjunct to alleviate chronic neurotoxicity induced by lidocaine.
Collapse
Affiliation(s)
- Yonghong Tan
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong 510623, People's Republic of China
| | - Xiaobao Bi
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong 510623, People's Republic of China
| | - Qiong Wang
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong 510623, People's Republic of China
| | - Yu Li
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong 510623, People's Republic of China
| | - Na Zhang
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong 510623, People's Republic of China
| | - Jianxin Lao
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong 510623, People's Republic of China
| | - Xiaoping Liu
- Department of Hematology, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong 510623, People's Republic of China
| |
Collapse
|
12
|
Peng H, Wang L, Su Q, Yi K, Du J, Wang Z. MiR-31-5p promotes the cell growth, migration and invasion of colorectal cancer cells by targeting NUMB. Biomed Pharmacother 2019; 109:208-216. [PMID: 30396078 DOI: 10.1016/j.biopha.2018.10.048] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 10/08/2018] [Accepted: 10/09/2018] [Indexed: 02/07/2023] Open
Abstract
The aim of this study was to investigate the role and specific molecular mechanism of miR-31-5 in colorectal cancer. The relative expression of miR-31-5p and NUMB in colorectal cancer tissues was analyzed by qRT-PCR. To knock down the expression of miR-31-5p, the transfection of miR-31-5p inhibitor was performed. The transfection with miR-31-5p mimic was used for miR-31-5p overexpression and pcDNA3.0-NUMB plasmid was used for NUMB overexpression. CCK-8 assay was used to analyze the cell proliferation. Flow cytometry was used to evaluate the cell apoptosis and cell cycle. Matrigel invasion assay was performed to assess the invasion potency and migration assay was performed to assess the migration potency. Hoechst 33258 staining assay was performed to analyze the cell apoptosis of HT29 cells after the indicated transfection. Luciferase activity assays were performed to confirmed the potential binding site for miR-31-5p in 3'-UTR region of NUMB. MiR-31-5p is highly expressed in colorectal cancer and is critical for the cell proliferation, cell cycle, migration, invasion and apoptosis. NUMB is target of miR-31-5p and NUMB overexpression inhibited the cell proliferation, migration, invasion and induced cell cycle arrest and apoptosis in HT29 colorectal cancer cells. In conclusion, miR-31-5p promoted the cell growth, migration and invasion by targeting NUMB in colorectal cancer cells.
Collapse
Affiliation(s)
- Hong Peng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China; Department of Anorectal Surgery, Nanchong Central Hospital, Nanchong, Sichuan 637000, PR China
| | - Longfei Wang
- Department of Pharmacy, Nanchong Central Hospital, Nanchong, Sichuan 637000, PR China
| | - Qiang Su
- Department of Clinical Pharmacy, Nanchong Central Hospital, Nanchong, Sichuan 637000, PR China
| | - Kun Yi
- Department of Image Diagnoses, Nanchong Central Hospital, Nanchong, Sichuan 637000, PR China
| | - Jingwei Du
- Department of Otolaryngology, Nanchong Central Hospital, Nanchong, Sichuan 637000, PR China
| | - Ziwei Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|
13
|
Clinical importance of the EMSY gene expression and polymorphisms in ovarian cancer. Oncotarget 2018; 9:17735-17755. [PMID: 29707144 PMCID: PMC5915152 DOI: 10.18632/oncotarget.24878] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 02/28/2018] [Indexed: 11/25/2022] Open
Abstract
EMSY, a BRCA2–associated protein, is amplified and overexpressed in various sporadic cancers. This is the first study assessing the clinical impact of its expression and polymorphisms on ovarian cancer (OvCa) outcome in the context of the chemotherapy regimen used. In 134 frozen OvCa samples, we assessed EMSY mRNA expression with Reverse Transcription-quantitative PCR, and also investigated the EMSY gene sequence using SSCP and/or PCR-sequencing. Clinical relevance of changes in EMSY mRNA expression and DNA sequence was evaluated in two subgroups treated with either taxane/platinum (TP, n=102) or platinum/cyclophosphamide (PC, n=32). High EMSY expression negatively affected overall survival (OS), disease-free survival (DFS) and sensitivity to treatment (PS) in the TP-treated subgroup (p-values: 0.001, 0.002 and 0.010, respectively). Accordingly, our OvCa cell line studies showed that the EMSY gene knockdown sensitized A2780 and IGROV1 cells to paclitaxel. Interestingly, EMSY mRNA expression in surviving cells was similar as in the control cells. Additionally, we identified 24 sequence alterations in the EMSY gene, including the previously undescribed: c.720G>C, p.(Lys240Asn); c.1860G>A, p.(Lys620Lys); c.246-76A>G; c.421+68A>C. In the PC-treated subgroup, a heterozygous genotype comprising five SNPs (rs4300410, rs3814711, rs4245443, rs2508740, rs2513523) negatively correlated with OS (p-value=0.009). The same SNPs exhibited adverse borderline associations with PS in the TP-treated subgroup. This is the first study providing evidence that high EMSY mRNA expression is a negative prognostic and predictive factor in OvCa patients treated with TP, and that the clinical outcome may hinge on certain SNPs in the EMSY gene as well.
Collapse
|
14
|
Current updates on microRNAs as regulators of chemoresistance. Biomed Pharmacother 2017; 95:1000-1012. [DOI: 10.1016/j.biopha.2017.08.084] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 08/11/2017] [Accepted: 08/23/2017] [Indexed: 12/28/2022] Open
|
15
|
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: 9.5] [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
|
16
|
Chakravarthi BVSK, Chandrashekar DS, Agarwal S, Balasubramanya SAH, Pathi SS, Goswami MT, Jing X, Wang R, Mehra R, Asangani IA, Chinnaiyan AM, Manne U, Sonpavde G, Netto GJ, Gordetsky J, Varambally S. miR-34a Regulates Expression of the Stathmin-1 Oncoprotein and Prostate Cancer Progression. Mol Cancer Res 2017; 16:1125-1137. [PMID: 29025958 DOI: 10.1158/1541-7786.mcr-17-0230] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 08/24/2017] [Accepted: 10/09/2017] [Indexed: 12/27/2022]
Abstract
In aggressive prostate cancers, the oncoprotein STMN1 (also known as stathmin 1 and oncoprotein 18) is often overexpressed. STMN1 is involved in various cellular processes, including cell proliferation, motility, and tumor metastasis. Here, it was found that the expression of STMN1 RNA and protein is elevated in metastatic prostate cancers. Knockdown of STMN1 resulted in reduced proliferation and invasion of cells and tumor growth and metastasis in vivo Furthermore, miR-34a downregulated STMN1 by directly binding to its 3'-UTR. Overexpression of miR-34a in prostate cancer cells reduced proliferation and colony formation, suggesting that it is a tumor suppressor. The transcriptional corepressor C-terminal binding protein 1 (CtBP1) negatively regulated expression of miR-34a. Furthermore, gene expression profiling of STMN1-modulated prostate cancer cells revealed molecular alterations, including elevated expression of growth differentiation factor 15 (GDF15), which is involved in cancer progression and potentially in STMN1-mediated oncogenesis. Thus, in prostate cancer, CtBP1-regulated miR-34a modulates STMN1 expression and is involved in cancer progression through the CtBP1\miR-34a\STMN1\GDF15 axis.Implications: The CtBP1\miR-34a\STMN1\GDF15 axis is a potential therapeutic target for treatment of aggressive prostate cancer. Mol Cancer Res; 16(7); 1125-37. ©2017 AACR.
Collapse
Affiliation(s)
- Balabhadrapatruni V S K Chakravarthi
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama.,Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
| | | | - Sumit Agarwal
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | | | - Satya S Pathi
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
| | - Moloy T Goswami
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
| | - Xiaojun Jing
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan.,Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Rui Wang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
| | - Rohit Mehra
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan.,Department of Pathology, University of Michigan, Ann Arbor, Michigan.,Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Irfan A Asangani
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
| | - Arul M Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan.,Department of Pathology, University of Michigan, Ann Arbor, Michigan.,Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan.,Department of Urology, University of Michigan, Ann Arbor, Michigan.,Howard Hughes Medical Institute, University of Michigan, Ann Arbor, Michigan
| | - Upender Manne
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama.,Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Guru Sonpavde
- Department of Medical Oncology, GU section, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - George J Netto
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jennifer Gordetsky
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Sooryanarayana Varambally
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama. .,Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama.,Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
17
|
Vetter NS, Kolb EA, Mills CC, Sampson VB. The Microtubule Network and Cell Death Are Regulated by an miR-34a/Stathmin 1/βIII-Tubulin Axis. Mol Cancer Res 2017; 15:953-964. [PMID: 28275089 PMCID: PMC5500423 DOI: 10.1158/1541-7786.mcr-16-0372] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 12/06/2016] [Accepted: 03/02/2017] [Indexed: 01/05/2023]
Abstract
MicroRNA-34a (miR-34a) is a master regulator of signaling networks that maintains normal physiology and disease and is currently in development as a miRNA-based therapy for cancer. Prior studies have reported low miR-34a expression in osteosarcoma; however, the molecular mechanisms underlying miR-34a activity in osteosarcoma are not well-defined. Therefore, this study evaluated the role of miR-34a in regulating signal transduction pathways that influence cell death in osteosarcoma. Levels of miR-34a were attenuated in human osteosarcoma cells and xenografts of the Pediatric Preclinical Testing Consortium (PPTC). Bioinformatics predictions identified stathmin 1 (STMN1) as a potential miR-34a target. Biotin pull-down assay and luciferase reporter analysis confirmed miR-34a target interactions within the STMN1 mRNA 3'-untranslated region. Overexpression of miR-34a in osteosarcoma cells suppressed STMN1 expression and reduced cell growth in vitro Restoration of miR-34a led to microtubule destabilization and increased βIII-tubulin expression, with corresponding G1-G2 phase cell-cycle arrest and apoptosis. Knockdown of the Sp1 transcription factor, by siRNA silencing, also upregulated βIII-tubulin expression in osteosarcoma cells, suggesting that miR-34a indirectly affects Sp1. Validating the coordinating role of miR-34a in microtubule destabilization, when miR-34a was combined with either microtubule inhibitors or chemotherapy, STMN1 phosphorylation was suppressed and there was greater cytotoxicity in osteosarcoma cells. These results demonstrate that miR-34a directly represses STMN1 gene and protein expression and upregulates βIII-tubulin, leading to disruption of the microtubule network and cell death.Implications: The miR-34a/STMN1/βIII-tubulin axis maintains the microtubule cytoskeleton in osteosarcoma, and combining miR-34a with microtubule inhibitors can be investigated as a novel therapeutic strategy. Mol Cancer Res; 15(7); 953-64. ©2017 AACR.
Collapse
Affiliation(s)
- Nancy S Vetter
- Nemours Center for Cancer and Blood Disorders, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware
| | - E A Kolb
- Nemours Center for Cancer and Blood Disorders, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware
| | | | - Valerie B Sampson
- Nemours Center for Cancer and Blood Disorders, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware.
| |
Collapse
|
18
|
Ayers D, Vandesompele J. Influence of microRNAs and Long Non-Coding RNAs in Cancer Chemoresistance. Genes (Basel) 2017; 8:genes8030095. [PMID: 28273813 PMCID: PMC5368699 DOI: 10.3390/genes8030095] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 02/15/2017] [Accepted: 02/24/2017] [Indexed: 12/16/2022] Open
Abstract
Innate and acquired chemoresistance exhibited by most tumours exposed to conventional chemotherapeutic agents account for the majority of relapse cases in cancer patients. Such chemoresistance phenotypes are of a multi-factorial nature from multiple key molecular players. The discovery of the RNA interference pathway in 1998 and the widespread gene regulatory influences exerted by microRNAs (miRNAs) and other non-coding RNAs have certainly expanded the level of intricacy present for the development of any single physiological phenotype, including cancer chemoresistance. This review article focuses on the latest research efforts in identifying and validating specific key molecular players from the two main families of non-coding RNAs, namely miRNAs and long non-coding RNAs (lncRNAs), having direct or indirect influences in the development of cancer drug resistance properties and how such knowledge can be utilised for novel theranostics in oncology.
Collapse
Affiliation(s)
- Duncan Ayers
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida MSD2080, Malta.
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M1 7DN, UK.
| | - Jo Vandesompele
- Center for Medical Genetics Ghent, Ghent University, Ghent 9000, Belgium.
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent 9000, Belgium.
| |
Collapse
|
19
|
Abstract
Based on the histological features and outcome, the current WHO classification separates thymomas into A, AB, B1, B2 and B3 subtypes. It is hypothesized that the type A thymomas are derived from the thymic medulla while the type B thymomas are derived from the cortex. Due to occasional histological overlap between the tumor subtypes creating difficulties in their separation, the aim of this study was to provide their proteomic characterization and identify potential immunohistochemical markers aiding in tissue diagnosis. Pair-wise comparison of neoplastic and normal thymus by liquid chromatography tandem mass spectrometry (LC-MS/MS) of formalin fixed paraffin embedded tissue revealed 61 proteins differentially expressed in thymomas compared to normal tissue. Hierarchical clustering showed distinct segregation of subtypes AB, B1 and B2 from that of A and B3. Most notably, desmoyokin, a protein that is encoded by the AHNAK gene, was associated with type A thymomas and medulla of normal thymus, by LC-MS/MS and immunohistochemistry. In this global proteomic characterization of the thymoma, several proteins unique to different thymic compartments and thymoma subtypes were identified. Among differentially expressed proteins, desmoyokin is a marker specific for thymic medulla and is potentially promising immunohistochemical marker in separation of type A and B3 thymomas.
Collapse
|
20
|
Biaoxue R, Xiguang C, Hua L, Shuanying Y. Stathmin-dependent molecular targeting therapy for malignant tumor: the latest 5 years' discoveries and developments. J Transl Med 2016; 14:279. [PMID: 27670291 PMCID: PMC5037901 DOI: 10.1186/s12967-016-1000-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 08/03/2016] [Indexed: 12/20/2022] Open
Abstract
Knowledge of the molecular mechanisms on malignant tumors is very critical for the development of new treatment strategies like molecularly targeted therapies. In last 5 years, many investigations suggest that stathmin is over-expressed in a variety of human malignant tumors, and potentially promotes the occurrence and development of tumors. Rather, down-regulation of stathmin can reduce cell proliferation, motility and metastasis and induce apoptosis of malignant tumors. Thus, a stathmin antagonist, such as a specific inhibitor (antibody, small molecule compound, peptide, or siRNA), may be a novel strategy of molecular targeted therapy. This review summarizes the research progress of recent 5 years on the role of stathmin in tumorigenesis, the molecular mechanisms and development of anti-stathmin treatment, which suggest that continued investigations into the function of stathmin in the tumorigenesis could lead to more rationally designed therapeutics targeting stathmin for treating human malignant tumors.
Collapse
Affiliation(s)
- Rong Biaoxue
- Department of Respiratory Medicine, First Affiliated Hospital, Xi'an Medical University, Xi'an, China.
| | - Cai Xiguang
- Department of Respiratory Medicine, Gansu Provincial Hospital, Lanzhou, China
| | - Liu Hua
- Department of Respiratory Medicine, Gansu Provincial Hospital, Lanzhou, China
| | - Yang Shuanying
- Department of Respiratory Medicine, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
21
|
Nymoen DA, Slipicevic A, Holth A, Emilsen E, Hetland Falkenthal TE, Tropé CG, Reich R, Flørenes VA, Davidson B. MiR-29a is a candidate biomarker of better survival in metastatic high-grade serous carcinoma. Hum Pathol 2016; 54:74-81. [DOI: 10.1016/j.humpath.2016.03.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Revised: 03/02/2016] [Accepted: 03/05/2016] [Indexed: 12/27/2022]
|
22
|
STEPICHEVA NADEZDAA, SONG JIAL. Function and regulation of microRNA-31 in development and disease. Mol Reprod Dev 2016; 83:654-74. [PMID: 27405090 PMCID: PMC6040227 DOI: 10.1002/mrd.22678] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Accepted: 06/29/2016] [Indexed: 12/13/2022]
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs that orchestrate numerous cellular processes both under normal physiological conditions as well as in diseases. This review summarizes the functional roles and transcriptional regulation of the highly evolutionarily conserved miRNA, microRNA-31 (miR-31). miR-31 is an important regulator of embryonic implantation, development, bone and muscle homeostasis, and immune system function. Its own regulation is disrupted during the onset and progression of cancer and autoimmune disorders such as psoriasis and systemic lupus erythematosus. Limited studies suggest that miR-31 is transcriptionally regulated by epigenetics, such as methylation and acetylation, as well as by a number of transcription factors. Overall, miR-31 regulates diverse cellular and developmental processes by targeting genes involved in cell proliferation, apoptosis, cell differentiation, and cell motility. Mol. Reprod. Dev. 83: 654-674, 2016 © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - JIA L. SONG
- Department of Biological Sciences, University of Delaware, Newark, Delaware
| |
Collapse
|
23
|
Lin X, Yu T, Zhang L, Chen S, Chen X, Liao Y, Long D, Shen F. Silencing Op18/stathmin by RNA Interference Promotes the Sensitivity of Nasopharyngeal Carcinoma Cells to Taxol and High-Grade Differentiation of Xenografted Tumours in Nude Mice. Basic Clin Pharmacol Toxicol 2016; 119:611-620. [DOI: 10.1111/bcpt.12633] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Accepted: 06/01/2016] [Indexed: 01/01/2023]
Affiliation(s)
- Xuechi Lin
- Department of Medical Laboratory; Changsha Medical University; Changsha China
- Department of Anatomy, Histology and Embryology; Institute of Neuroscience; Changsha Medical University; Changsha China
| | - Ting Yu
- Department of Medical Laboratory; Changsha Medical University; Changsha China
| | - Lingxi Zhang
- Department of Medical Laboratory; Changsha Medical University; Changsha China
| | - Sangyan Chen
- Department of Medical Laboratory; Changsha Medical University; Changsha China
| | - Xian Chen
- Department of Medical Laboratory; Changsha Medical University; Changsha China
| | - Ying Liao
- Department of Medical Laboratory; Changsha Medical University; Changsha China
| | - Dan Long
- Department of Medical Laboratory; Changsha Medical University; Changsha China
| | - Fang Shen
- Department of Medical Laboratory; Changsha Medical University; Changsha China
- Department of Clinical Laboratory; the First Affiliated Hospital of Hunan Normal University; Changsha Hunan China
| |
Collapse
|
24
|
Prahm KP, Novotny GW, Høgdall C, Høgdall E. Current status on microRNAs as biomarkers for ovarian cancer. APMIS 2016; 124:337-55. [PMID: 26809719 DOI: 10.1111/apm.12514] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 12/10/2015] [Indexed: 01/05/2023]
Abstract
Ovarian cancer (OC) is the most lethal gynecological malignancy in the Western world, and has a very poor prognosis, often due to late diagnosis and emergence of chemotherapy resistance. Therefore, there is an essential need for new diagnostic and prognostic markers that can improve and initiate more personalized treatment, eventually improving survival of the patients. MicroRNAs are small, non-coding RNA molecules, that post-transcriptionally regulate gene expression. Several studies have within the last decade shown that microRNAs are deregulated in OC and have potential as diagnostic and prognostic biomarkers for OC. Recently studies have also focused on microRNAs as predictors of chemotherapy responses and their potential as therapeutic targets. However, many of the published studies are difficult to interpret as a whole due to various methods of analysis. Future focus should be aimed at developing a general standardized analytical method, which can limit differences between studies thus allowing easier comparison across them. In addition, validation of studies in independent series that ideally should be histotype-specific is essential to determine the clinical role of microRNAs in different types of OC. In this review we summarize the current knowledge of microRNAs as potential biomarkers for OC, with focus on their clinical relevance.
Collapse
Affiliation(s)
- Kira Philipsen Prahm
- Molecular Unit, Department of Pathology, Danish Cancer Biobank, Herlev Hospital, University of Copenhagen, Herlev, Denmark.,Department of Gynaecology, Juliane Marie Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Guy Wayne Novotny
- Molecular Unit, Department of Pathology, Danish Cancer Biobank, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | - Claus Høgdall
- Department of Gynaecology, Juliane Marie Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Estrid Høgdall
- Molecular Unit, Department of Pathology, Danish Cancer Biobank, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| |
Collapse
|