1
|
Chauhan P, Pramodh S, Hussain A, Elsori D, Lakhanpal S, Kumar R, Alsaweed M, Iqbal D, Pandey P, Al Othaim A, Khan F. Understanding the role of miRNAs in cervical cancer pathogenesis and therapeutic responses. Front Cell Dev Biol 2024; 12:1397945. [PMID: 39263322 PMCID: PMC11387185 DOI: 10.3389/fcell.2024.1397945] [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: 03/08/2024] [Accepted: 08/08/2024] [Indexed: 09/13/2024] Open
Abstract
Cervical cancer (CC) is the most common cancer in women and poses a serious threat to health. Despite familiarity with the factors affecting its etiology, initiation, progression, treatment strategies, and even resistance to therapy, it is considered a significant problem for women. However, several factors have greatly affected the previous aspects of CC progression and treatment in recent decades. miRNAs are short non-coding RNA sequences that regulate gene expression by inhibiting translation of the target mRNA. miRNAs play a crucial role in CC pathogenesis by promoting cancer stem cell (CSC) proliferation, postponing apoptosis, continuing the cell cycle, and promoting invasion, angiogenesis, and metastasis. Similarly, miRNAs influence important CC-related molecular pathways, such as the PI3K/AKT/mTOR signaling pathway, Wnt/β-catenin system, JAK/STAT signaling pathway, and MAPK signaling pathway. Moreover, miRNAs affect the response of CC patients to chemotherapy and radiotherapy. Consequently, this review aims to provide an acquainted summary of onco miRNAs and tumor suppressor (TS) miRNAs and their potential role in CC pathogenesis and therapy responses by focusing on the molecular pathways that drive them.
Collapse
Affiliation(s)
| | - Sreepoorna Pramodh
- Department of Biomedical Sciences, University of Birmingham Dubai, Dubai, United Arab Emirates
| | - Arif Hussain
- School of Life Sciences, Manipal Academy of Higher Education, Dubai, United Arab Emirates
| | - Deena Elsori
- Faculty of Resilience, Rabdan Academy, Abu Dhabi, United Arab Emirates
| | - Sorabh Lakhanpal
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Rahul Kumar
- Chitkara Centre for Research and Development, Chitkara University, Baddi, Himachal Pradesh, India
| | - Mohammed Alsaweed
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al-Majmaah, Saudi Arabia
| | - Danish Iqbal
- Department of Health Information Management, College of Applied Medical Sciences, Buraydah Private Colleges, Buraydah, Saudi Arabia
| | - Pratibha Pandey
- Centre for Research Impact and Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, India
- Centre for Research Impact and Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Ayoub Al Othaim
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al-Majmaah, Saudi Arabia
| | - Fahad Khan
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
| |
Collapse
|
2
|
Ebrahimi N, Manavi MS, Faghihkhorasani F, Fakhr SS, Baei FJ, Khorasani FF, Zare MM, Far NP, Rezaei-Tazangi F, Ren J, Reiter RJ, Nabavi N, Aref AR, Chen C, Ertas YN, Lu Q. Harnessing function of EMT in cancer drug resistance: a metastasis regulator determines chemotherapy response. Cancer Metastasis Rev 2024; 43:457-479. [PMID: 38227149 DOI: 10.1007/s10555-023-10162-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 12/08/2023] [Indexed: 01/17/2024]
Abstract
Epithelial-mesenchymal transition (EMT) is a complicated molecular process that governs cellular shape and function changes throughout tissue development and embryogenesis. In addition, EMT contributes to the development and spread of tumors. Expanding and degrading the surrounding microenvironment, cells undergoing EMT move away from the main location. On the basis of the expression of fibroblast-specific protein-1 (FSP1), fibroblast growth factor (FGF), collagen, and smooth muscle actin (-SMA), the mesenchymal phenotype exhibited in fibroblasts is crucial for promoting EMT. While EMT is not entirely reliant on its regulators like ZEB1/2, Twist, and Snail proteins, investigation of upstream signaling (like EGF, TGF-β, Wnt) is required to get a more thorough understanding of tumor EMT. Throughout numerous cancers, connections between tumor epithelial and fibroblast cells that influence tumor growth have been found. The significance of cellular crosstalk stems from the fact that these events affect therapeutic response and disease prognosis. This study examines how classical EMT signals emanating from various cancer cells interfere to tumor metastasis, treatment resistance, and tumor recurrence.
Collapse
Affiliation(s)
- Nasim Ebrahimi
- Genetics Division, Department of Cell and Molecular Biology and Microbiology, Faculty of Science and Technology, University of Isfahan, Isfahan, Iran
| | | | | | - Siavash Seifollahy Fakhr
- Department of Biotechnology, Faculty of Applied Ecology, Agricultural Science and Biotechnology, Campus Hamar, Inland Norway University of Applied Sciences, Hamar, Norway
| | | | | | - Mohammad Mehdi Zare
- Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Nazanin Pazhouhesh Far
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Science, Islamic Azad University, Tehran, Iran
| | - Fatemeh Rezaei-Tazangi
- Department of Anatomy, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Jun Ren
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Russel J Reiter
- Department of Cellular and Structural Biology, UT Health Science Center, San Antonio, TX, 77030, USA
| | - Noushin Nabavi
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H3Z6, Canada
| | - Amir Reza Aref
- Translational Medicine Group, Xsphera Biosciences, 6 Tide Street, Boston, MA, 02210, USA.
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | - Chu Chen
- Department of Cardiology, Affiliated Hospital of Nantong University, Jiangsu, 226001, China
| | - Yavuz Nuri Ertas
- ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, 38039, Türkiye.
- Department of Biomedical Engineering, Erciyes University, Kayseri, 38039, Türkiye.
| | - Qi Lu
- Department of Cardiology, Affiliated Hospital of Nantong University, Jiangsu, 226001, China.
| |
Collapse
|
3
|
Le Compte M, De La Hoz EC, Peeters S, Fortes FR, Hermans C, Domen A, Smits E, Lardon F, Vandamme T, Lin A, Vanlanduit S, Roeyen G, Van Laere S, Prenen H, Peeters M, Deben C. Single-organoid analysis reveals clinically relevant treatment-resistant and invasive subclones in pancreatic cancer. NPJ Precis Oncol 2023; 7:128. [PMID: 38066116 PMCID: PMC10709344 DOI: 10.1038/s41698-023-00480-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 11/09/2023] [Indexed: 06/28/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal diseases, characterized by a treatment-resistant and invasive nature. In line with these inherent aggressive characteristics, only a subset of patients shows a clinical response to the standard of care therapies, thereby highlighting the need for a more personalized treatment approach. In this study, we comprehensively unraveled the intra-patient response heterogeneity and intrinsic aggressive nature of PDAC on bulk and single-organoid resolution. We leveraged a fully characterized PDAC organoid panel (N = 8) and matched our artificial intelligence-driven, live-cell organoid image analysis with retrospective clinical patient response. In line with the clinical outcomes, we identified patient-specific sensitivities to the standard of care therapies (gemcitabine-paclitaxel and FOLFIRINOX) using a growth rate-based and normalized drug response metric. Moreover, the single-organoid analysis was able to detect resistant as well as invasive PDAC organoid clones, which was orchestrates on a patient, therapy, drug, concentration and time-specific level. Furthermore, our in vitro organoid analysis indicated a correlation with the matched patient progression-free survival (PFS) compared to the current, conventional drug response readouts. This work not only provides valuable insights on the response complexity in PDAC, but it also highlights the potential applications (extendable to other tumor types) and clinical translatability of our approach in drug discovery and the emerging era of personalized medicine.
Collapse
Affiliation(s)
- Maxim Le Compte
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
| | | | - Sofía Peeters
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
| | - Felicia Rodrigues Fortes
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
| | - Christophe Hermans
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
| | - Andreas Domen
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
- Department of Oncology, Multidisciplinary Oncological Center Antwerp, Antwerp University Hospital, Antwerp, Belgium
| | - Evelien Smits
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium
| | - Filip Lardon
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
| | - Timon Vandamme
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
- Department of Oncology, Multidisciplinary Oncological Center Antwerp, Antwerp University Hospital, Antwerp, Belgium
| | - Abraham Lin
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
- Plasma Lab for Applications in Sustainability and Medicine ANTwerp (PLASMANT), University of Antwerp, Antwerp, Belgium
| | | | - Geert Roeyen
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
- Department of Hepatobiliary Transplantation and Endocrine Surgery, University Hospital Antwerp (UZA), Antwerp, Belgium
| | - Steven Van Laere
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
| | - Hans Prenen
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
- Department of Oncology, Multidisciplinary Oncological Center Antwerp, Antwerp University Hospital, Antwerp, Belgium
| | - Marc Peeters
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
- Department of Oncology, Multidisciplinary Oncological Center Antwerp, Antwerp University Hospital, Antwerp, Belgium
| | - Christophe Deben
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
4
|
Du S, Qu H, Zhang Y, Zhu S, Wang Y, Zhang S, Wang Z, Yang Q, Fu S, Dong K. MiR-375 promotes cisplatin sensitivity of lung adenocarcinoma. Pathol Res Pract 2023; 249:154765. [PMID: 37625279 DOI: 10.1016/j.prp.2023.154765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 07/30/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023]
Abstract
BACKGROUND Cisplatin-based chemotherapy has been widely used in the treatment of lung adenocarcinoma (LUAD). However, the development of cisplatin resistance becomes a major obstacle impeding the curative effect. It remains necessary to uncover the molecular mechanism of cisplatin resistance. METHODS Based on the CCLE database, lung cancer cell lines were divided into cisplatin-resistant and cisplatin-sensitive groups. The differentially expressed miRNAs were filtered and further identified by survival prognosis analysis. After transfection with miR-375 inhibitor or mimic, cell cytotoxicity assay, flow cytometry and western blot were conducted to validate the role of miR-375. The transcription factor (TF)-miRNA network was constructed based on TransmiR. The target genes of miR-375 were predicted by Starbase and further verified by RT-qPCR and immunohistochemistry results in the Human Protein Atlas. Functional enrichment analysis was performed with GO terms and KEGG. RESULTS In this study, miR-375 showed the ability to promote cisplatin sensitivity and apoptosis of LUAD. Genes correlated with miR-375 in LUAD were analyzed and ABCC8 showed the strongest positive correlation. Moreover, transcription factors that regulate miR-375 expression were predicted. MBNL1, PTPN3, PRKD1 and RPN1 were identified as the target genes of miR-375. Enrichment analysis demonstrated that miR-375-related genes associated with promoting cell proliferation and anti-apoptosis were involved in the MAPK signaling pathway. CONCLUSION Overall, this study provides new insights into the role of miR-375 in the cisplatin sensitivity of LUAD. Our present findings may serve as a theoretical basis for new therapeutic strategies and predictive models of cisplatin resistance in LUAD.
Collapse
Affiliation(s)
- Shuomeng Du
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin 150081, China; Laboratory of Medical Genetics, Harbin Medical University, Harbin 150081, China
| | - Han Qu
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin 150081, China; Laboratory of Medical Genetics, Harbin Medical University, Harbin 150081, China
| | - Ying Zhang
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin 150081, China; Laboratory of Medical Genetics, Harbin Medical University, Harbin 150081, China
| | - Shihao Zhu
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin 150081, China; Laboratory of Medical Genetics, Harbin Medical University, Harbin 150081, China
| | - Yang Wang
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin 150081, China; Laboratory of Medical Genetics, Harbin Medical University, Harbin 150081, China
| | - Shuopeng Zhang
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin 150081, China; Laboratory of Medical Genetics, Harbin Medical University, Harbin 150081, China
| | - Zhao Wang
- Department of Gynecological Radiotherapy, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Qian Yang
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin 150081, China; Laboratory of Medical Genetics, Harbin Medical University, Harbin 150081, China
| | - Songbin Fu
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin 150081, China; Laboratory of Medical Genetics, Harbin Medical University, Harbin 150081, China
| | - Kexian Dong
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin 150081, China; Laboratory of Medical Genetics, Harbin Medical University, Harbin 150081, China.
| |
Collapse
|
5
|
Matulić M, Gršković P, Petrović A, Begić V, Harabajsa S, Korać P. miRNA in Molecular Diagnostics. Bioengineering (Basel) 2022; 9:bioengineering9090459. [PMID: 36135005 PMCID: PMC9495386 DOI: 10.3390/bioengineering9090459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 08/05/2022] [Accepted: 09/07/2022] [Indexed: 11/17/2022] Open
Abstract
MicroRNAs are a class of small non-coding RNA molecules that regulate gene expression on post-transcriptional level. Their biogenesis consists of a complex series of sequential processes, and they regulate expression of many genes involved in all cellular processes. Their function is essential for maintaining the homeostasis of a single cell; therefore, their aberrant expression contributes to development and progression of many diseases, especially malignant tumors and viral infections. Moreover, they can be associated with certain states of a specific disease, obtained in the least invasive manner for patients and analyzed with basic molecular methods used in clinical laboratories. Because of this, they have a promising potential to become very useful biomarkers and potential tools in personalized medicine approaches. In this review, miRNAs biogenesis, significance in cancer and infectious diseases, and current available test and methods for their detection are summarized.
Collapse
Affiliation(s)
- Maja Matulić
- Division of Molecular Biology, Department of Biology, Faculty of Science, University of Zagreb, 10000 Zagreb, Croatia
| | - Paula Gršković
- Division of Molecular Biology, Department of Biology, Faculty of Science, University of Zagreb, 10000 Zagreb, Croatia
| | - Andreja Petrović
- Division of Molecular Biology, Department of Biology, Faculty of Science, University of Zagreb, 10000 Zagreb, Croatia
- Institute of Clinical Pathology and Cytology, Merkur University Hospital, 10000 Zagreb, Croatia
| | - Valerija Begić
- Division of Molecular Biology, Department of Biology, Faculty of Science, University of Zagreb, 10000 Zagreb, Croatia
- Primary School “Sesvetski Kraljevec”, 10361 Sesvetski Kraljevec, Croatia
| | - Suzana Harabajsa
- Division of Molecular Biology, Department of Biology, Faculty of Science, University of Zagreb, 10000 Zagreb, Croatia
- Department of Pathology and Cytology, Division of Pulmonary Cytology Jordanovac, University Hospital Centre Zagreb, 10000 Zagreb, Croatia
| | - Petra Korać
- Division of Molecular Biology, Department of Biology, Faculty of Science, University of Zagreb, 10000 Zagreb, Croatia
- Correspondence: ; Tel.: +385-1-4606-278
| |
Collapse
|
6
|
Matuszyk J. MALAT1-miRNAs network regulate thymidylate synthase and affect 5FU-based chemotherapy. Mol Med 2022; 28:89. [PMID: 35922756 PMCID: PMC9351108 DOI: 10.1186/s10020-022-00516-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/22/2022] [Indexed: 12/12/2022] Open
Abstract
Background The active metabolite of 5-Fluorouracil (5FU), used in the treatment of several types of cancer, acts by inhibiting the thymidylate synthase encoded by the TYMS gene, which catalyzes the rate-limiting step in DNA replication. The major failure of 5FU-based cancer therapy is the development of drug resistance. High levels of TYMS-encoded protein in cancerous tissues are predictive of poor response to 5FU treatment. Expression of TYMS is regulated by various mechanisms, including involving non-coding RNAs, both miRNAs and long non-coding RNAs (lncRNAs). Aim To delineate the miRNAs and lncRNAs network regulating the level of TYMS-encoded protein. Main body Several miRNAs targeting TYMS mRNA have been identified in colon cancers, the levels of which can be regulated to varying degrees by lncRNAs. Due to their regulation by the MALAT1 lncRNA, these miRNAs can be divided into three groups: (1) miR-197-3p, miR-203a-3p, miR-375-3p which are downregulated by MALAT1 as confirmed experimentally and the levels of these miRNAs are actually reduced in colon and gastric cancers; (2) miR-140-3p, miR-330-3p that could potentially interact with MALAT1, but not yet supported by experimental results; (3) miR-192-5p, miR-215-5p whose seed sequences do not recognize complementary response elements within MALAT1. Considering the putative MALAT1-miRNAs interaction network, attention is drawn to the potential positive feedback loop causing increased expression of MALAT1 in colon cancer and hepatocellular carcinoma, where YAP1 acts as a transcriptional co-factor which, by binding to the TCF4 transcription factor/ β-catenin complex, may increase the activation of the MALAT1 gene whereas the MALAT1 lncRNA can inhibit miR-375-3p which in turn targets YAP1 mRNA. Conclusion The network of non-coding RNAs may reduce the sensitivity of cancer cells to 5FU treatment by upregulating the level of thymidylate synthase.
Collapse
Affiliation(s)
- Janusz Matuszyk
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 12 R. Weigla Street, 53-114, Wroclaw, Poland.
| |
Collapse
|
7
|
Wu CZ, Chu YC, Lai SW, Hsieh MS, Yadav VK, Fong IH, Deng L, Huang CC, Tzeng YM, Yeh CT, Chen JS. Urokinase plasminogen activator induces epithelial-mesenchymal and metastasis of pancreatic cancer through plasmin/MMP14/TGF-β axis, which is inhibited by 4-acetyl-antroquinonol B treatment. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 100:154062. [PMID: 35366491 DOI: 10.1016/j.phymed.2022.154062] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 03/09/2022] [Accepted: 03/14/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND The current standard therapy for metastatic pancreatic cancer is ineffective, necessitating a new treatment approach for prognosis improvement. The urokinase-plasmin activator (uPA) is a critical factor in epithelial-mesenchymal transition (EMT) and cancer metastasis, but its underlying mechanisms in pancreatic cancer remains elusive. METHODS We investigated uPA expression in our pancreatic cancer cohort. A bioinformatics approach was used to further determine the role of uPA in pancreatic cancer. We employed MiaPaCa-2 and PANC-1 cell lines to investigate how uPA regulates EMT and metastasis in pancreatic cancer and present a novel approach aimed at inhibiting uPA in pancreatic cancer. RESULTS We observed that higher uPA mRNA expression was significantly associated with overall-poor survival and progression-free survival in pancreatic cancer. uPA was highly expressed in tumor tissue. Gene set enrichment analysis revealed a positive association between uPA mRNA expression and EMT and transforming growth factor β (TGF-β) signaling pathways. Moreover, shRNA-mediated uPA gene knockdown reduced plasmin, MMP14, and TGF-β activation, leading to the inhibition of PANC-1 cells' EMT marker expression, migration, invasion, and cell viability. Notably, 4-acetyl-antroquinonol B (4-AAQB) treatment suppressed MiaPaCa-2 and PANC-1 cell migratory and invasive abilities by inhibiting the uPA/MMP14/TGF-β axis through upregulation of miR-181d-5p. In the xenograft mouse model of orthotropic pancreatic cancer, 4-AAQB treatment has reduced tumor growth and metastasis rate by deactivating uPA and improving the survival of the mice model. CONCLUSION Accordingly, to extent of our knowledge and previous studies, we demonstrated that 4-AAQB is an anti Pan-Cancer drug, and may inhibit pancreatic cancer EMT and metastasis and serve as a new therapeutic approach for patients with late-stage pancreatic cancer.
Collapse
Affiliation(s)
- Chung-Ze Wu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei City, Taiwan, ROC; Division of Endocrinology and Metabolism, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan, ROC
| | - Yi Cheng Chu
- Department of Medicine, School of Medicine, St. George's University, St. George SW17 0RE, Grenada
| | - Shiue-Wei Lai
- Division of Hematology-Oncology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC; Department of Internal Medicine, Tri-Service General Hospital Penghu Branch, Penghu, Taiwan, ROC
| | - Ming-Shou Hsieh
- Department of Medical Research & Education, Taipei Medical University - Shuang Ho Hospital, New Taipei City, 235, Taiwan, ROC
| | - Vijesh Kumar Yadav
- Department of Medical Research & Education, Taipei Medical University - Shuang Ho Hospital, New Taipei City, 235, Taiwan, ROC
| | - Iat-Hang Fong
- Department of Medical Research & Education, Taipei Medical University - Shuang Ho Hospital, New Taipei City, 235, Taiwan, ROC
| | - Li Deng
- Beijing Bioprocess Key Laboratory, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; Amoy-BUCT Industrial Bio-Technovation Institute, Amoy 361022, China
| | - Chun-Chih Huang
- Center for General Education, National Taitung University, Taitung, Taiwan, ROC
| | - Yew-Min Tzeng
- Center for General Education, National Taitung University, Taitung, Taiwan, ROC
| | - Chi-Tai Yeh
- Department of Medical Research & Education, Taipei Medical University - Shuang Ho Hospital, New Taipei City, 235, Taiwan, ROC; Department of Medical Laboratory Science and Biotechnology, Yuanpei University of Medical Technology, Hsinchu 300, Taiwan, ROC.
| | - Jin-Shuen Chen
- Administration Department and Division of Nephrology, Kaohsiung Veterans General Hospital, Kaohsiung City, Taiwan, ROC; Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC.
| |
Collapse
|
8
|
Ren S, Tan X, Fu MZ, Ren S, Wu X, Chen T, Latham PS, Lin P, Man YG, Fu SW. Downregulation of miR-375 contributes to ERBB2-mediated VEGFA overexpression in esophageal cancer. J Cancer 2021; 12:7138-7146. [PMID: 34729115 PMCID: PMC8558641 DOI: 10.7150/jca.63836] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 09/28/2021] [Indexed: 12/21/2022] Open
Abstract
Esophageal cancer (EC) is a lethal cancer with an extremely aggressive nature and poor survival rate. However, the molecular mechanisms driving the occurrence and progression of EC are not well understood. MicroRNAs (miRNAs) are small RNA molecules that regulate the expression of protein-coding genes. miRNA-mediated gene regulation plays an important role in EC. By cross-referencing studies from NCBI, we found that microRNA-375 (miR-375) is one of the most frequently downregulated miRNAs in EC. We assessed expression of miR-375 in EC cell lines and primary EC tissues and their matched normal tissues. We found significant downregulation of miR-375 in both cell lines and EC tissues. Forced expression of miR-375 attenuated EC cell proliferation and invasion. Human epidermal growth factor receptor 2 (HER2, ERBB2), a known proto-oncogene, was identified here as one of the potential target genes of miR-375. Ectopic expression of miR-375 significantly suppressed the expression of ERBB2 and subsequently downregulated one of its target genes, vascular endothelial growth factor A (VEGFA), which is related to cancer invasion and metastasis. These findings suggest that miR-375 acts as a tumor suppressor by blocking the ERBB2/VEGFA pathway with the potential to modulate the occurrence and/ or progression of EC.
Collapse
Affiliation(s)
- Shuchang Ren
- Department of Medicine, Division of Genomic Medicine, and Department of Microbiology, Immunology and Tropical Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Xiaohui Tan
- Department of Medicine, Division of Genomic Medicine, and Department of Microbiology, Immunology and Tropical Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Melinda Z Fu
- Department of Medicine, Division of Genomic Medicine, and Department of Microbiology, Immunology and Tropical Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Shuyang Ren
- Department of Medicine, Division of Genomic Medicine, and Department of Microbiology, Immunology and Tropical Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Xiaoling Wu
- Department of Medicine, Chengdu Military General Hospital, Chengdu, Sichuan, China
| | - Tao Chen
- Department of Medicine, Chengdu Military General Hospital, Chengdu, Sichuan, China
| | - Patricia S Latham
- Department of Pathology, The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Paul Lin
- Department of Surgery, The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Yan-Gao Man
- Department of Pathology, Hackensack Meridian Health-Hackensack, University Medical Center, Hackensack, NJ; the International Union for Difficult to treat Diseases (IUDD), Silver Spring, MD
| | - Sidney W Fu
- Department of Medicine, Division of Genomic Medicine, and Department of Microbiology, Immunology and Tropical Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC
| |
Collapse
|
9
|
MicroRNA-375: potential cancer suppressor and therapeutic drug. Biosci Rep 2021; 41:229736. [PMID: 34494089 PMCID: PMC8458691 DOI: 10.1042/bsr20211494] [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: 06/23/2021] [Revised: 08/31/2021] [Accepted: 09/06/2021] [Indexed: 02/06/2023] Open
Abstract
MiR-375 is a conserved noncoding RNA that is known to be involved in tumor cell proliferation, migration, and drug resistance. Previous studies have shown that miR-375 affects the epithelial-mesenchymal transition (EMT) of human tumor cells via some key transcription factors, such as Yes-associated protein 1 (YAP1), Specificity protein 1 (SP1) and signaling pathways (Wnt signaling pathway, nuclear factor κB (NF-κB) pathway and transforming growth factor β (TGF-β) signaling pathway) and is vital for the development of cancer. Additionally, recent studies have identified microRNA (miRNA) delivery system carriers for improved in vivo transportation of miR-375 to specific sites. Here, we discussed the role of miR-375 in different types of cancers, as well as molecular mechanisms, and analyzed the potential of miR-375 as a molecular biomarker and therapeutic target to improve the efficiency of clinical diagnosis of cancer.
Collapse
|
10
|
Wang S, Xu L, Zhang Z, Wang P, Zhang R, He H, Chen L. Overexpressed miR-375-Loaded Restrains Development of Cervical Cancer Through Down-Regulation of Frizzled Class Receptor 4 (FZD4) with Liposome Nanoparticle as a Carrier. J Biomed Nanotechnol 2021; 17:1882-1889. [PMID: 34688334 DOI: 10.1166/jbn.2021.3145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Dysregulation expression of miR-375 is noted to correlate with progression of cervical cancer. This study attempted to investigate the impact of overexpressed miR-375-loaded liposome nanoparticles on proliferation of cervical cancer (CC), to provide an insight on pathogenesis of CC disorder. CC cells were co-cultured with pure liposome nanoparticles (empty vector group), miR-375 agonist-loaded liposome nanoparticles, or transfected with miR-375 antagonist. Besides, some cells were exposed to TGF-β/Smads signaling pathway inhibitor or activator whilst cell proliferation was assessed by MTT assay, and expressions of FZD4 and miR-375 were determined. Western blot analysis was carried out to detect the expression of TGF-β pathway factors (TGF-β, Smad2, Smad7, p-Smad2) and its downstream Smads pathway. The interaction between miR-375 and FZD4 was evaluated by dual-luciferase reporter gene assay. Overexpression of miR-375 induced arrest at the G0/G1 phase of cell cycle and elevation of Smad2 protein expression (P <0.05), with lower expressions of TGF-β, Smad7, p-Smad2, and FZD4, while transfection with miR-375 inhibitor exhibited opposite activity. Presence of miR-375 agonist-loaded liposome nanoparticles induced decreased cell proliferation. There was a targeting relationship between miR-375 and FZD4, and administration with TGF-β/Smads agonist resulted in increased miR-375 and Smad2 expressions, as well as decreased TGF-β, Smad7, p-Smad2, FZD4 protein expression, and the number of S phase and G2/M phase cells (P < 0.05). The signaling inhibitor oppositely suppressed cell proliferation decreasing miR-375 expression. miR-375-loaded liposome nanoparticles activated TGF-β/Smads signaling pathway to restrain cell cycle and suppress cell division, and proliferation through targeting FZD4 in CC. Its molecular mechanism is related to activation of TGF-β/Smads signaling pathway.
Collapse
Affiliation(s)
- Suqin Wang
- The First Department of Gynecology of Shanxi Provincial People's Hospital, Taiyuan, 030012, Shanxi, China
| | - Lina Xu
- The First Department of Gynecology of Shanxi Provincial People's Hospital, Taiyuan, 030012, Shanxi, China
| | - Zhiqiang Zhang
- The First Department of Gynecology of Shanxi Provincial People's Hospital, Taiyuan, 030012, Shanxi, China
| | - Ping Wang
- Department of Gynecology, Shanxi Tumour Hospital, Taiyuan, 030013, Shanxi, China
| | - Rong Zhang
- The Second Department of Gynecology of Shanxi Provincial People's Hospital, Taiyuan, 030012, Shanxi, China
| | - Hui He
- The First Department of Gynecology of Shanxi Provincial People's Hospital, Taiyuan, 030012, Shanxi, China
| | - Ling Chen
- The First Department of Gynecology of Shanxi Provincial People's Hospital, Taiyuan, 030012, Shanxi, China
| |
Collapse
|
11
|
Mitra T, Elangovan S. Cervical cancer development, chemoresistance, and therapy: a snapshot of involvement of microRNA. Mol Cell Biochem 2021; 476:4363-4385. [PMID: 34453645 DOI: 10.1007/s11010-021-04249-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 08/17/2021] [Indexed: 12/24/2022]
Abstract
Cervical cancer (CC) is one of the leading causes of death in women due to cancer and a major concern in the developing world. Persistent human papilloma virus (HPV) infection is the major causative agent for CC. Besides HPV infection, genetic and epigenetic factors including microRNA (miRNA) also contribute to the malignant transformation. Earlier studies have revealed that miRNAs participate in cell proliferation, invasion and metastasis, angiogenesis, and chemoresistance processes by binding and inversely regulating the target oncogenes or tumor suppressor genes. Based on functions and mechanistic insights, miRNAs have been identified as cellular modulators that have an enormous role in diagnosis, prognosis, and cancer therapy. Signatures of miRNA could be used as diagnostic markers which are necessary for early diagnosis and management of CC. The therapeutic potential of miRNAs has been shown in CC; however, more comprehensive clinical trials are required for the clinical translation of miRNA-based diagnostics and therapeutics. Understanding the molecular mechanism of miRNAs and their target genes has been useful to develop miRNA-based therapeutic strategies for CC and overcome chemoresistance. In this review, we summarize the role of miRNAs in the development, progression, and metastasis of CC as well as chemoresistance. Further, we discuss the diagnostic and therapeutic potential of miRNAs to overcome chemoresistance and treatment of CC.
Collapse
Affiliation(s)
- Tandrima Mitra
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Deemed To Be University, Bhubaneswar, Odisha, 751024, India
| | - Selvakumar Elangovan
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Deemed To Be University, Bhubaneswar, Odisha, 751024, India.
| |
Collapse
|
12
|
Sasaki M, Ishikawa T, Ishiguro M, Okazaki S, Yamauchi S, Kikuchi A, Matsuyama T, Kawada K, Tokunaga M, Uetake H, Kinugasa Y. The effectiveness of plasma miR-33a-5p as a predictive biomarker for the efficacy of colorectal cancer chemotherapy. Oncol Lett 2021; 21:489. [PMID: 33968205 PMCID: PMC8100963 DOI: 10.3892/ol.2021.12749] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 03/09/2021] [Indexed: 02/06/2023] Open
Abstract
Several chemotherapeutic options are available for patients with metastatic colorectal cancer (mCRC), making it important to individualize treatment regimens. Individualization requires the clinical application of biomarkers for regimen selection, which is presently insufficient. miRNAs serve an important role in the control of biological processes in several types of cancer, acting as plasma biomarkers. The current study aimed to evaluate novel plasma microRNAs for predicting chemo-resistance in chemotherapy for patients with colorectal cancer (CRC) by employing a Toray 3D-Gene microRNA array-based approach, which compared plasma content before and during treatment. Specific miRNAs that acted as biomarkers of the fluoropyrimidine (FP) + oxaliplatin (OX) + bevacizumab (BEV) regime, a common first-line treatment for mCRC, were searched. The plasma samples of 110 patients with mCRC who had received the FP+OX+BEV regimen were subjected to microarray analyses using the 3D-Gene miRNA microarray platform, after which miRNAs levels were quantified via reverse transcription- quantitative PCR. Patients exhibiting complete response, partial response (PR) and reduced stable disease (SD) were defined as responders. Patients with extended SD and progression disease (PD) were defined as non-responders. Following microarray analysis, miR-33a-5p was selected as the candidate miRNA as it was upregulated in non-responder plasma samples. The expression of miR-33a-5p was upregulated in the non-responders (n=15) compared with the responders (n=95) (P=0.032). The high expression group demonstrated significantly poor progression-free survival (P<0.01). To evaluate whether miR-33a-5p can serve as a marker of chemo-resistance, miR-33a-5p expression levels were assessed at the following three time-points: Pre-point (before chemotherapy); PR-point (3-months after chemotherapy began); and PD-point (the time at which recurrence or progression was recorded). The results revealed that expression levels were significantly increased at the PD-point when compared with that at the pre-point (P=0.024). The current study determined that the miR-33a-5p expression level in the plasma may serve as a predictive marker of efficacy and as a biomarker of chemo-resistance.
Collapse
Affiliation(s)
- Megumi Sasaki
- Department of Gastrointestinal Surgery, Graduate School, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Toshiaki Ishikawa
- Department of Specialized Surgeries, Graduate School, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Megumi Ishiguro
- Department of Translational Oncology, Graduate School, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Satoshi Okazaki
- Department of Specialized Surgeries, Graduate School, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Shinichi Yamauchi
- Department of Gastrointestinal Surgery, Graduate School, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Akifumi Kikuchi
- Department of Gastrointestinal Surgery, Graduate School, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Takatoshi Matsuyama
- Department of Gastrointestinal Surgery, Graduate School, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Kenro Kawada
- Department of Gastrointestinal Surgery, Graduate School, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Masanori Tokunaga
- Department of Gastrointestinal Surgery, Graduate School, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Hiroyuki Uetake
- Department of Specialized Surgeries, Graduate School, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Yusuke Kinugasa
- Department of Gastrointestinal Surgery, Graduate School, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8519, Japan
| |
Collapse
|
13
|
Dong B, Li S, Zhu S, Yi M, Luo S, Wu K. MiRNA-mediated EMT and CSCs in cancer chemoresistance. Exp Hematol Oncol 2021; 10:12. [PMID: 33579377 PMCID: PMC7881653 DOI: 10.1186/s40164-021-00206-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 01/30/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer stem cells (CSCs) are a small group of cancer cells, which contribute to tumorigenesis and cancer progression. Cancer cells undergoing epithelial-to-mesenchymal transition (EMT) acquire the chemoresistant ability, which is regarded as an important feature of CSCs. Thus, there emerges an opinion that the generation of CSCs is considered to be driven by EMT. In this complex process, microRNAs (miRNAs) are found to play a key role. In order to overcome the drug resistance, inhibiting EMT as well as CSCs phenotype seem feasible. Thereinto, regulating the EMT- or CSCs-associated miRNAs is a crucial approach. Herein, we conduct this review to elaborate on the complicated interplay between EMT and CSCs in cancer chemoresistance, which is modulated by miRNAs. In addition, we elucidate the therapeutic strategy to overcome drug resistance through targeting EMT and CSCs.
Collapse
Affiliation(s)
- Bing Dong
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008 China
| | - Shiyu Li
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Shuangli Zhu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Ming Yi
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Suxia Luo
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008 China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008 China
| |
Collapse
|
14
|
Subramaniam Y, Subban K, Chelliah J. A novel synergistic anticancer effect of fungal cholestanol glucoside and paclitaxel: Apoptosis induced by an intrinsic pathway through ROS generation in cervical cancer cell line (HeLa). Toxicol In Vitro 2021; 72:105079. [PMID: 33422634 DOI: 10.1016/j.tiv.2021.105079] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 01/02/2021] [Accepted: 01/05/2021] [Indexed: 01/18/2023]
Abstract
In the search for efficient therapeutics with economically viable for cancer treatment, combination therapy has developed as a keystone in the pursuit of novel approaches for drug discovery. In this regard, we confirmed the presence of cholestanol glucoside (CG) in Lasiodiplodia theobromae culture filtrate and its production was estimated to be 20.01 mg/l. The purified fungal CG was obtained with a molecular mass of 550.18 m/z. The combination of CG and paclitaxel (PTX) was found to have potent cytotoxicity against HeLa cells. We revealed that the synergistic effect of CG and PTX induced apoptosis through the formation of nuclear fragments, DNA fragmentation and sub G1 cell cycle arrest. Further, it was proven that apoptosis took place by loss of the mitochondrial membrane potential (MMP) through reactive oxygen species (ROS) production and caspase 3/7 activity. Moreover, the data suggests that the synergistic effect of CG and PTX played a role in a mitochondrial intrinsic pathway through the apoptotic gene expression of Bax, caspase-9 and caspase-3. In addition, the down-regulation of Bcl-2 strongly described the induced apoptosis through an intrinsic pathway using the Western blot analysis. The conclusion of this study is that a combination of CG and PTX has synergistic apoptotic effects in HeLa cells, which provides a possible therapeutic strategy for cancer therapy in the future.
Collapse
Affiliation(s)
| | - Kamalraj Subban
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| | | |
Collapse
|
15
|
Singh S, Raza W, Parveen S, Meena A, Luqman S. Flavonoid display ability to target microRNAs in cancer pathogenesis. Biochem Pharmacol 2021; 189:114409. [PMID: 33428895 DOI: 10.1016/j.bcp.2021.114409] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/01/2021] [Accepted: 01/05/2021] [Indexed: 12/11/2022]
Abstract
MicroRNAs (miRNAs) are non-coding, conserved, single-stranded nucleotide sequences involved in physiological and developmental processes. Recent evidence suggests an association between miRNAs' deregulation with initiation, promotion, progression, and drug resistance in cancer cells. Besides, miRNAs are known to regulate the epithelial-mesenchymal transition, angiogenesis, autophagy, and senescence in different cancer types. Previous reports proposed that apart from the antioxidant potential, flavonoids play an essential role in miRNAs modulation associated with changes in cancer-related proteins, tumor suppressor genes, and oncogenes. Thus, flavonoids can suppress proliferation, help in the development of drug sensitivity, suppress metastasis and angiogenesis by modulating miRNAs expression. In the present review, we summarize the role of miRNAs in cancer, drug resistance, and the chemopreventive potential of flavonoids mediated by miRNAs. The potential of flavonoids to modulate miRNAs expression in different cancer types demonstrate their selectivity and importance as regulators of carcinogenesis. Flavonoids as chemopreventive agents targeting miRNAs are extensively studied in vitro, in vivo, and pre-clinical studies, but their efficiency in targeting miRNAs in clinical studies is less investigated. The evidence presented in this review highlights the potential of flavonoids in cancer prevention/treatment by regulating miRNAs, although further investigations are required to validate and establish their clinical usefulness.
Collapse
Affiliation(s)
- Shilpi Singh
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Waseem Raza
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, Uttar Pradesh, India; Jawahar Lal Nehru University, New Delhi 110067, India
| | - Shahnaz Parveen
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Abha Meena
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Suaib Luqman
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India.
| |
Collapse
|
16
|
Zhao X, Sun W, Ren Y, Lu Z. Therapeutic potential of p53 reactivation in cervical cancer. Crit Rev Oncol Hematol 2020; 157:103182. [PMID: 33276182 DOI: 10.1016/j.critrevonc.2020.103182] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 10/23/2020] [Accepted: 11/11/2020] [Indexed: 12/19/2022] Open
Abstract
Cervical cancer (CC) is one of most common malignancies affecting women worldwide. To date, surgical resection is the only effective radical remedy for CC at its early stages, while the prognosis of metastatic or recurrent CC is very poor. Dysfunction of the tumor suppressor p53 due to aberrant expression, post-translational modification, mutations, SNPs, and LOH as well as sequestration by viral antigens and MDM2/HDM2-mediated degradation is closely associated with the therapeutic insensitivity and relapse of many malignancies, including CC. Accumulating studies have demonstrated that restoration of p53 activity can induce cell cycle arrest and apoptosis, eliminate radio- and chemotherapy resistance, and inhibit tumor growth in CC cells. Therefore, activation of wild-type p53 as well as restoration of p53 function seems appealing as a therapeutic strategy. In this review, we focus on the potential roles of p53 reactivation in CC treatment and their underlying molecular mechanisms towards the development of novel therapies.
Collapse
Affiliation(s)
- Xiangxuan Zhao
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, 110004, LN, China.
| | - Wei Sun
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, 110004, LN, China
| | - Ying Ren
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, 110004, LN, China
| | - Zaiming Lu
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, 110004, LN, China
| |
Collapse
|
17
|
Sen P, Ghosal S, Hazra R, Mohanty R, Arega S, Sahu B, Ganguly N. CRISPR-mediated knockdown of miR-214 modulates cell fate in response to anti-cancer drugs in HPV-negative and HPV-positive cervical cancer cells. J Biosci 2020. [DOI: 10.1007/s12038-020-00054-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
18
|
Miao J, Regenstein JM, Xu D, Zhou D, Li H, Zhang H, Li C, Qiu J, Chen X. The roles of microRNA in human cervical cancer. Arch Biochem Biophys 2020; 690:108480. [PMID: 32681832 DOI: 10.1016/j.abb.2020.108480] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 06/19/2020] [Accepted: 06/20/2020] [Indexed: 12/13/2022]
Abstract
Although a potentially preventable disease, cervical cancer (CC) is the second most commonly diagnosed gynaecological cancer with at least 530,000 new cases annually, and the prognosis with CC is still poor. Studies suggest that aberrant expression of microRNA (miRNA) contributes to the progression of CC. As a group of small non-coding RNA with 18-25 nucleotides, miRNA regulate about one-third of all human genes. They function by repressing translation or inducing mRNA cleavage or degradation, including genes involved in diverse and important cellular processes, including cell cycling, proliferation, differentiation, and apoptosis. Results showed that misexpression of miRNA is closely related to the onset and progression of CC. This review will provide an overview of the function of miRNA in CC and the mechanisms involved in cervical carcinogenesis.
Collapse
Affiliation(s)
- Jingnan Miao
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Haikou, Hainan, 570100, China; School of Pharmacy, Hainan Medical University, Haikou, Hainan, 570100, China
| | - Joe M Regenstein
- Department of Food Science, Cornell University, Ithaca, NY, 14853-7201, USA
| | - Dan Xu
- School of Pharmacy, Hainan Medical University, Haikou, Hainan, 570100, China
| | - Dan Zhou
- School of Pharmacy, Hainan Medical University, Haikou, Hainan, 570100, China
| | - Haixia Li
- School of Pharmacy, Hainan Medical University, Haikou, Hainan, 570100, China
| | - Hua Zhang
- Department of Food Science, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang, 150010, China
| | - Chunfeng Li
- Gastrointestinal Surgical Ward, Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Junqiang Qiu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Haikou, Hainan, 570100, China; School of Pharmacy, Hainan Medical University, Haikou, Hainan, 570100, China; Hainan Provincial Key Laboratory of R & D on Tropical Herbs, Haikou, Hainan, 570100, China.
| | - Xun Chen
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Haikou, Hainan, 570100, China; School of Pharmacy, Hainan Medical University, Haikou, Hainan, 570100, China; Hainan Provincial Key Laboratory of R & D on Tropical Herbs, Haikou, Hainan, 570100, China
| |
Collapse
|
19
|
Guan Y, Bhandari A, Xia E, Kong L, Zhang X, Wang O. Downregulating integrin subunit alpha 7 (ITGA7) promotes proliferation, invasion, and migration of papillary thyroid carcinoma cells through regulating epithelial-to-mesenchymal transition. Acta Biochim Biophys Sin (Shanghai) 2020; 52:116-124. [PMID: 31942970 DOI: 10.1093/abbs/gmz144] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 07/19/2019] [Accepted: 11/18/2019] [Indexed: 12/14/2022] Open
Abstract
Thyroid cancer is one of the common malignancies of the endocrine system and the number of thyroid cancer cases is increasing constantly. Significant work has focused on the molecular mechanisms of thyroid cancer, but many mechanisms remain undiscovered. In this study, we employed a comprehensive analysis of whole-transcriptome resequencing derived from paired papillary thyroid cancer (PTC) and normal thyroid tissues. We performed a massive parallel whole-transcriptome resequencing of matched PTC and normal thyroid tissues in 19 patients and found that integrin subunit alpha 7 (ITGA7) was downregulated in thyroid tumor tissues, but the function of ITGA7 in this cancer is still unclear. We also discovered that ITGA7 gene in thyroid cancer tissues was downregulated compared to paired adjacent non-tumor tissues by real-time quantitative polymerase chain reaction. After transfection with small interfering RNA to knock down ITGA7, the abilities of colony formation, proliferation, migration, and invasion were enhanced in PTC cell lines (TPC1 and KTC-1). Meanwhile, ITGA7 knockdown decreased apoptotic cell death in thyroid cells but promoted the expressions of N-cadherin and vimentin and decreased E-cadherin expression by epithelial-to-mesenchymal transition, which may induce invasion and migration. In conclusion, these results indicated that ITGA7 is involved in the progress of PTC and might act as a tumor suppressor gene.
Collapse
Affiliation(s)
- Yaoyao Guan
- Department of Thyroid & Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Adheesh Bhandari
- Department of Thyroid & Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Erjie Xia
- Department of Thyroid & Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Lingguo Kong
- Department of Thyroid & Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Xiaohua Zhang
- Department of Thyroid & Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Ouchen Wang
- Department of Thyroid & Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| |
Collapse
|
20
|
Condrat CE, Thompson DC, Barbu MG, Bugnar OL, Boboc A, Cretoiu D, Suciu N, Cretoiu SM, Voinea SC. miRNAs as Biomarkers in Disease: Latest Findings Regarding Their Role in Diagnosis and Prognosis. Cells 2020; 9:E276. [PMID: 31979244 PMCID: PMC7072450 DOI: 10.3390/cells9020276] [Citation(s) in RCA: 740] [Impact Index Per Article: 148.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/14/2020] [Accepted: 01/21/2020] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (miRNAs) represent a class of small, non-coding RNAs with the main roles of regulating mRNA through its degradation and adjusting protein levels. In recent years, extraordinary progress has been made in terms of identifying the origin and exact functions of miRNA, focusing on their potential use in both the research and the clinical field. This review aims at improving the current understanding of these molecules and their applicability in the medical field. A thorough analysis of the literature consulting resources available in online databases such as NCBI, PubMed, Medline, ScienceDirect, and UpToDate was performed. There is promising evidence that in spite of the lack of standardized protocols regarding the use of miRNAs in current clinical practice, they constitute a reliable tool for future use. These molecules meet most of the required criteria for being an ideal biomarker, such as accessibility, high specificity, and sensitivity. Despite present limitations, miRNAs as biomarkers for various conditions remain an impressive research field. As current techniques evolve, we anticipate that miRNAs will become a routine approach in the development of personalized patient profiles, thus permitting more specific therapeutic interventions.
Collapse
Affiliation(s)
- Carmen Elena Condrat
- Alessandrescu-Rusescu National Institute for Mother and Child Health, Fetal Medicine Excellence Research Center, 020395 Bucharest, Romania; (C.E.C.); (D.C.T.); (M.G.B.); (O.L.B.); (A.B.); (D.C.); (N.S.)
| | - Dana Claudia Thompson
- Alessandrescu-Rusescu National Institute for Mother and Child Health, Fetal Medicine Excellence Research Center, 020395 Bucharest, Romania; (C.E.C.); (D.C.T.); (M.G.B.); (O.L.B.); (A.B.); (D.C.); (N.S.)
| | - Madalina Gabriela Barbu
- Alessandrescu-Rusescu National Institute for Mother and Child Health, Fetal Medicine Excellence Research Center, 020395 Bucharest, Romania; (C.E.C.); (D.C.T.); (M.G.B.); (O.L.B.); (A.B.); (D.C.); (N.S.)
| | - Oana Larisa Bugnar
- Alessandrescu-Rusescu National Institute for Mother and Child Health, Fetal Medicine Excellence Research Center, 020395 Bucharest, Romania; (C.E.C.); (D.C.T.); (M.G.B.); (O.L.B.); (A.B.); (D.C.); (N.S.)
| | - Andreea Boboc
- Alessandrescu-Rusescu National Institute for Mother and Child Health, Fetal Medicine Excellence Research Center, 020395 Bucharest, Romania; (C.E.C.); (D.C.T.); (M.G.B.); (O.L.B.); (A.B.); (D.C.); (N.S.)
| | - Dragos Cretoiu
- Alessandrescu-Rusescu National Institute for Mother and Child Health, Fetal Medicine Excellence Research Center, 020395 Bucharest, Romania; (C.E.C.); (D.C.T.); (M.G.B.); (O.L.B.); (A.B.); (D.C.); (N.S.)
- Department of Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd., 050474 Bucharest, Romania
| | - Nicolae Suciu
- Alessandrescu-Rusescu National Institute for Mother and Child Health, Fetal Medicine Excellence Research Center, 020395 Bucharest, Romania; (C.E.C.); (D.C.T.); (M.G.B.); (O.L.B.); (A.B.); (D.C.); (N.S.)
- Division of Obstetrics, Gynecology and Neonatology, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd., 050474 Bucharest, Romania
- Department of Obstetrics and Gynecology, Polizu Clinical Hospital, Alessandrescu-Rusescu National Institute for Mother and Child Health, 020395 Bucharest, Romania
| | - Sanda Maria Cretoiu
- Department of Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd., 050474 Bucharest, Romania
| | - Silviu Cristian Voinea
- Department of Surgical Oncology, Prof. Dr. Alexandru Trestioreanu Oncology Institute, Carol Davila University of Medicine and Pharmacy, 252 Fundeni Rd., 022328 Bucharest, Romania;
| |
Collapse
|
21
|
Chemotherapeutic Stress Influences Epithelial-Mesenchymal Transition and Stemness in Cancer Stem Cells of Triple-Negative Breast Cancer. Int J Mol Sci 2020; 21:ijms21020404. [PMID: 31936348 PMCID: PMC7014166 DOI: 10.3390/ijms21020404] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 12/10/2019] [Accepted: 12/31/2019] [Indexed: 12/13/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer characterized by the absence of estrogen and progesterone receptors (ER, PR) and lacking an overexpression of human epidermal growth factor receptor 2 (HER2). Apart from this lack of therapeutic targets, TNBC also shows an increased capacity for early metastasis and therapy resistance. Currently, many TNBC patients receive neoadjuvant chemotherapy (NACT) upon detection of the disease. With TNBC likely being driven at least in part by a cancer stem-like cell type, we wanted to evaluate the response of primary cancer stem cells (CSCs) to standard chemotherapeutics. Therefore, we set up a survival model using primary CSCs to mimic tumor cells in patients under chemotherapy. Breast cancer stem cells (BCSCs) were exposed to chemotherapeutics with a sublethal dose for six days. Surviving cells were allowed to recover in culture medium without chemotherapeutics. Surviving and recovered cells were examined in regard to proliferation, migratory capacity, sphere forming capacity, epithelial–mesenchymal transition (EMT) factor expression at the mRNA level, and cancer-related microRNA (miRNA) profile. Our results indicate that chemotherapeutic stress enhanced sphere forming capacity of BCSCs, and changed cell morphology and EMT-related gene expression at the mRNA level, whereas the migratory capacity was unaffected. Six miRNAs were identified as potential regulators in this process.
Collapse
|
22
|
Huang L, Gan X, He L, Wang L, Yu J. Silencing of long non-coding RNA NCK1-AS1 inhibits cell proliferation and migration via inhibition of microRNA-134 in cervical cancer. Exp Ther Med 2019; 18:2314-2322. [PMID: 31410182 DOI: 10.3892/etm.2019.7799] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Accepted: 04/26/2019] [Indexed: 12/19/2022] Open
Abstract
Long non-coding RNA NCK1-antisense 1 (AS1) has recently been demonstrated to promote cell proliferation and induce cell cycle progression through the crosstalk NCK1-AS1/microRNA (miR)-6857/cyclin dependent kinase 1 pathway in cervical cancer. However, the regulatory mechanism of NCK1-AS1 in cervical cancer growth and metastasis remains largely unclear. In the present study, it was identified that NCK1-AS1 was significantly upregulated in cervical cancer tissues compared with the levels in adjacent non-tumour tissues. High expression levels of NCK1-AS1 were associated with tumour progression and poor prognosis in patients with cervical cancer. Silencing of NCK1-AS1 expression significantly decreased the levels of proliferation and migration of cervical cancer cells, and decreased the protein expression levels of matrix metalloproteinase (MMP)-2 and MMP-9. The results of the luciferase reporter gene assay indicated that there was an miR-134 binding site within the NCK1-AS1 gene in cervical cancer cells. miR-134 was significantly downregulated in cervical cancer tissues compared with the miR-134 levels in adjacent non-tumour tissues, and the expression level of miR-134 was inversely correlated with the NCK1-AS1 expression levels in cervical cancer tissues. Knockdown of miR-134 attenuated the inhibitory effects of NCK1-AS1 downregulation on the proliferation and migration of cervical cancer cells. Therefore, the data from the present study suggested that NCK1-AS1 serves a promotive role in cervical cancer cell proliferation and migration by functioning as a molecular sponge for miR-134. NCK1-AS1 may become a novel therapeutic target for cervical cancer.
Collapse
Affiliation(s)
- Lu Huang
- Department of Obstetrics and Gynaecology, Chengdu Women and Children's Central Hospital, Chengdu, Sichuan 610031, P.R. China
| | - Xiaoqin Gan
- Department of Obstetrics and Gynaecology, Chengdu Women and Children's Central Hospital, Chengdu, Sichuan 610031, P.R. China
| | - Li He
- Department of Obstetrics and Gynaecology, Chengdu Women and Children's Central Hospital, Chengdu, Sichuan 610031, P.R. China
| | - Luying Wang
- Department of Obstetrics and Gynaecology, Chengdu Women and Children's Central Hospital, Chengdu, Sichuan 610031, P.R. China
| | - Jie Yu
- Department of Obstetrics and Gynaecology, Chengdu Women and Children's Central Hospital, Chengdu, Sichuan 610031, P.R. China
| |
Collapse
|
23
|
Nahand JS, Taghizadeh-Boroujeni S, Karimzadeh M, Borran S, Pourhanifeh MH, Moghoofei M, Bokharaei-Salim F, Karampoor S, Jafari A, Asemi Z, Tbibzadeh A, Namdar A, Mirzaei H. microRNAs: New prognostic, diagnostic, and therapeutic biomarkers in cervical cancer. J Cell Physiol 2019; 234:17064-17099. [PMID: 30891784 DOI: 10.1002/jcp.28457] [Citation(s) in RCA: 143] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/24/2019] [Accepted: 01/28/2019] [Indexed: 12/11/2022]
Abstract
Cervical cancer is as a kind of cancer beginning from the cervix. Given that cervical cancer could be observed in women who infected with papillomavirus, regular oral contraceptives, and multiple pregnancies. Early detection of cervical cancer is one of the most important aspects of the therapy of this malignancy. Despite several efforts, finding and developing new biomarkers for cervical cancer diagnosis are required. Among various prognostic, diagnostic, and therapeutic biomarkers, miRNA have been emerged as powerful biomarkers for detection, treatment, and monitoring of response to therapy in cervical cancer. Here, we summarized various miRNAs as an employable platform for prognostic, diagnostic, and therapeutic biomarkers in the treatment of cervical cancer.
Collapse
Affiliation(s)
- Javid Sadri Nahand
- Department of Virology, Student Research Committee, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sima Taghizadeh-Boroujeni
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Brujen, Iran
| | - Mohammad Karimzadeh
- Department of Virology, Student Research Committee, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sarina Borran
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Pourhanifeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohsen Moghoofei
- Department of Microbiology, Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Farah Bokharaei-Salim
- Department of Virology, Student Research Committee, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sajad Karampoor
- Department of Virology, Student Research Committee, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amir Jafari
- Department of Medical Nanotechnology, Faculty of Advanced Technology in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Alireza Tbibzadeh
- Department of Virology, Student Research Committee, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Afshin Namdar
- Department of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
24
|
Liu L, Yu TT, Ren CC, Yang L, Cui SH, Zhang XA. CP-31398 inhibits the progression of cervical cancer through reversing the epithelial mesenchymal transition via the downregulation of PAX2s. J Cell Physiol 2019; 234:2929-2942. [PMID: 30132866 DOI: 10.1002/jcp.27109] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 06/28/2018] [Indexed: 01/18/2023]
Abstract
CP-31398, a styrylquinazoline, emerges from a screen for therapeutic agents that restore the wild-type DNA-binding conformation of mutant p53 to suppress tumors in vivo, but its effects on cervical cancer (CC) remain unknown. Hence, this study aimed to explore the effects CP-31398 has on the CC cells and to investigate whether it is associated with paired box 2 (PAX2) expression. CC cells were treated with different concentrations of CP-31398 (1, 2, 4, 6, 8, and 10 μg/ml) to determine the optimum concentration using fluorometric microculture cytotoxicity assay. After constructing the sh-PAX2 vector, CC cells were transfected with sh-PAX2 or treated with CP-31398. The effects of CP-31398 or PAX2 silencing on CC cell proliferation, apoptosis, invasion, and migration were evaluated. Epithelial mesenchymal transition (EMT)-related genes such as E-cadherin, vimentin, N-cadherin, snail, and twist in CC cells were detected. Tumor formation experiment in nude mice was performed to observe tumor growth. The optimum concentration of CP-31398 was 2 μg/ml. PAX2 was overexpressed in CC cells. CC cells treated with CP-31398 or treated with sh-PAX2 inhibited proliferation, invasion, and migration but promoted apoptosis with decreased PAX2 expression. The EMT process in CC cells was also reversed after treatment with CP-31398 or sh-PAX2. Moreover, the tumor formation experiment in nude mice revealed the inhibitory activity of CP-31398 in CC tumor in nude mice by suppressing PAX2. Our results provide evidence that CP-31398 could inhibit EMT and promote apoptosis of CC cells to curb CC tumor growth by downregulating PAX2.
Collapse
Affiliation(s)
- Ling Liu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tan-Tan Yu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chen-Chen Ren
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Li Yang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shi-Hong Cui
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiao-An Zhang
- Department of Imaging, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
25
|
Joyce MH, Lu C, James ER, Hegab R, Allen SC, Suggs LJ, Brock A. Phenotypic Basis for Matrix Stiffness-Dependent Chemoresistance of Breast Cancer Cells to Doxorubicin. Front Oncol 2018; 8:337. [PMID: 30234012 PMCID: PMC6134055 DOI: 10.3389/fonc.2018.00337] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 08/03/2018] [Indexed: 12/12/2022] Open
Abstract
The persistence of drug resistant cell populations following chemotherapeutic treatment is a significant challenge in the clinical management of cancer. Resistant subpopulations arise via both cell intrinsic and extrinsic mechanisms. Extrinsic factors in the microenvironment, including neighboring cells, glycosaminoglycans, and fibrous proteins impact therapy response. Elevated levels of extracellular fibrous proteins are associated with tumor progression and cause the surrounding tissue to stiffen through changes in structure and composition of the extracellular matrix (ECM). We sought to determine how this progressively stiffening microenvironment affects the sensitivity of breast cancer cells to chemotherapeutic treatment. MDA-MB-231 triple negative breast carcinoma cells cultured in a 3D alginate-based hydrogel system displayed a stiffness-dependent response to the chemotherapeutic doxorubicin. MCF7 breast carcinoma cells cultured in the same conditions did not exhibit this stiffness-dependent resistance to the drug. This differential therapeutic response was coordinated with nuclear translocation of YAP, a marker of mesenchymal differentiation. The stiffness-dependent response was lost when cells were transferred from 3D to monolayer cultures, suggesting that endpoint ECM conditions largely govern the response to doxorubicin. To further examine this response, we utilized a platform capable of dynamic ECM stiffness modulation to allow for a change in matrix stiffness over time. We found that MDA-MB-231 cells have a stiffness-dependent resistance to doxorubicin and that duration of exposure to ECM stiffness is sufficient to modulate this response. These results indicate the need for additional tools to integrate mechanical stiffness with therapeutic response and inform decisions for more effective use of chemotherapeutics in the clinic.
Collapse
Affiliation(s)
- M. Hunter Joyce
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, United States
| | - Carolyne Lu
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, United States
| | - Emily R. James
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, United States
| | - Rachel Hegab
- Department of Biomedical Engineering, Louisiana Tech University, Ruston, LA, United States
| | - Shane C. Allen
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, United States
| | - Laura J. Suggs
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, United States
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX, United States
| | - Amy Brock
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, United States
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
26
|
Li RM, Nai MM, Duan SJ, Li SX, Yin BN, An F, Zhai YQ, Liu J, Chu YR, Yu Y, Song WY. Down-expression of GOLM1 enhances the chemo-sensitivity of cervical cancer to methotrexate through modulation of the MMP13/EMT axis. Am J Cancer Res 2018; 8:964-980. [PMID: 30034935 PMCID: PMC6048392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 03/29/2018] [Indexed: 06/08/2023] Open
Abstract
The highly refractory nature of cervical cancer to chemotherapeutic drugs and its epithelial-to-mesenchymal transition (EMT) are the key reasons contributing to the poor prognosis of this disease. Golgi Membrane Protein 1 (GOLM1), a protein involved in the trafficking of proteins through the Golgi apparatus, has been shown to be oncogenic in a variety of human cancers. Herein, we found GOLM1 was markedly up-regulated in cervical cancer and GOLM1 down-expression enhanced the anti-tumor effect of methotrexate. By performing mechanistic studies using both in vitro and in vivo models, we found that GOLM1 could target matrix metallopeptidase 13 (MMP13), a member of the MMPs, and regulate the EMT process. Moreover, altered EMT progression compromised the chemotherapy-enhancing effects of GOLM1 knock-down. Finally, we found significantly higher levels of GOLM1 and MMP13 in cervical cancer tissues compared with adjacent noncancerous tissues, and this was also associated with poor cervical cancer patients' prognosis. Taken together, our results suggest that the GOLM1/MMP13/EMT axis is an important factor involved in regulating methotrexate in cervical cancer, and highlights the potential of novel GOLM1-based clinical modalities as a therapeutic approach in cervical cancer patients.
Collapse
Affiliation(s)
- Rui Min Li
- Department of Gynecology, Jiaozuo Maternal and Child Care Service CentreJiaozuo, Henan Province, China
| | - Man Man Nai
- Department of Gynecology, The Third Affiliated Hospital of Zhengzhou UniversityZhengzhou, Henan Province, China
| | - She Jiao Duan
- Department of Gynecology, Jiaozuo Maternal and Child Care Service CentreJiaozuo, Henan Province, China
| | - Shu Xing Li
- Department of Gynecology, Jiaozuo Maternal and Child Care Service CentreJiaozuo, Henan Province, China
| | - Bao Na Yin
- Department of Gynecology, Jiaozuo Maternal and Child Care Service CentreJiaozuo, Henan Province, China
| | - Fang An
- Department of Gynecology, Jiaozuo Maternal and Child Care Service CentreJiaozuo, Henan Province, China
| | - Yao Qing Zhai
- Department of Gynecology, Jiaozuo Maternal and Child Care Service CentreJiaozuo, Henan Province, China
| | - Jie Liu
- Department of Gynecology, Jiaozuo Maternal and Child Care Service CentreJiaozuo, Henan Province, China
| | - Yan Rong Chu
- Department of Gynecology, Jiaozuo Maternal and Child Care Service CentreJiaozuo, Henan Province, China
| | - Yang Yu
- Department of Endocrinology, Jiaozuo People’s HospitalJiaozuo, Henan Province, China
| | - Wen Yue Song
- Department of Gynecology, Jiaozuo Maternal and Child Care Service CentreJiaozuo, Henan Province, China
| |
Collapse
|
27
|
Goto M, Osada S, Imagawa M, Nishizuka M. FAD104, a regulator of adipogenesis, is a novel suppressor of TGF-β-mediated EMT in cervical cancer cells. Sci Rep 2017; 7:16365. [PMID: 29180690 PMCID: PMC5703855 DOI: 10.1038/s41598-017-16555-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 11/15/2017] [Indexed: 01/05/2023] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is a biological process in which epithelial cells translate into a mesenchymal phenotype with invasive capacities, contributing to tumour progression, metastasis, and the acquisition of chemotherapy resistance. To identify new therapeutic targets for cancers, it is important to clarify the molecular mechanism of induction of EMT. We have previously reported that fad104, a positive regulator of adipocyte differentiation, suppressed the invasion and metastasis of melanoma and breast cancer cells. In this study, we showed that FAD104 functions as a novel suppressor of transforming growth factor-β (TGF-β)–mediated EMT in cervical cancer cells. Expression of FAD104 is upregulated during TGF-β–mediated EMT in human cervical cancer HeLa cells. Reduction of fad104 expression enhanced TGF-β–mediated EMT and migration in HeLa cells. Conversely, overexpression of FAD104 suppressed TGF-β–induced EMT. In addition, we showed that FAD104 negatively regulated phosphorylation of Smad2 and Smad3 but positively regulated phosphorylation of Smad1/5/8 via treatment with TGF-β. These findings demonstrate that FAD104 is a novel suppressor of TGF-β signalling and represses TGF-β–mediated EMT in cervical cancer cells.
Collapse
Affiliation(s)
- Motoharu Goto
- Department of Molecular Biology, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi, 467-8603, Japan
| | - Shigehiro Osada
- Department of Molecular Biology, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi, 467-8603, Japan
| | - Masayoshi Imagawa
- Department of Molecular Biology, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi, 467-8603, Japan
| | - Makoto Nishizuka
- Department of Molecular Biology, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi, 467-8603, Japan.
| |
Collapse
|
28
|
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
|
29
|
Liu Q, Xue Y, Chen Q, Chen H, Zhang X, Wang L, Han C, Que S, Lou M, Lan J. PomGnT1 enhances temozolomide resistance by activating epithelial-mesenchymal transition signaling in glioblastoma. Oncol Rep 2017; 38:2911-2918. [PMID: 29048655 DOI: 10.3892/or.2017.5964] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 07/28/2017] [Indexed: 11/06/2022] Open
Abstract
Temozolomide (TMZ) is commonly used in glioblastoma (GBM) chemotherapy. However, a great challenge for TMZ treatment is the rapid development of resistance and subsequent tumor recurrence and poor outcome. In the present study we established TMZ-resistant GBM cells (U87-TR and U251-TR) and found that the expression of PomGnT1 was significantly upregulated in TMZ-resistant GBM cells compared with the TMZ-sensitive counterparts. Furthermore, overexpression of PomGnT1 in U87-MG and U251-MG cells led to increased IC50 values for TMZ and reduced apoptosis of cells. Knockdown of PomGnT1 in both U87-TR and U251-TR cells led to decreased IC50 values for TMZ and enhanced apoptosis. Biochemical analysis revealed that PomGnT1 regulates the expression of factors in epithelial-mesenchymal transition signaling including TCF8, vimentin, β-catenin and Slug in GBM cells. These findings demonstrate that PomGnT1 might be a new focus of GBM research for treatment of recurrent TMZ-resistant GBM.
Collapse
Affiliation(s)
- Qi Liu
- Department of Neurosurgery, Jingdezhen Second Hospital, Jingdezhen, Jiangxi, P.R. China
| | - Yajun Xue
- Department of Neurosurgery, General Hospital, Shanghai, P.R. China
| | - Qingshan Chen
- Department of Neurosurgery, The Second People's Hospital of Liaocheng City, Liaocheng, Shandong, P.R. China
| | - Huairui Chen
- Department of Neurosurgery, General Hospital, Shanghai, P.R. China
| | - Xiaofei Zhang
- Department of Neurosurgery, General Hospital, Shanghai, P.R. China
| | - Leiping Wang
- Department of Neurosurgery, General Hospital, Shanghai, P.R. China
| | - Cong Han
- Department of Neurosurgery, General Hospital, Shanghai, P.R. China
| | - Shuanglin Que
- Department of Neurosurgery, Longyan First Hospital, Fujian Medical University, Longyan, Fujian, P.R. China
| | - Meiqing Lou
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Jin Lan
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| |
Collapse
|
30
|
Zou Q, Yi W, Huang J, Fu F, Chen G, Zhong D. MicroRNA-375 targets PAX6 and inhibits the viability, migration and invasion of human breast cancer MCF-7 cells. Exp Ther Med 2017; 14:1198-1204. [PMID: 28810579 DOI: 10.3892/etm.2017.4593] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 02/10/2017] [Indexed: 12/13/2022] Open
Abstract
MicroRNAs (miRs) are a type of small non-coding RNA that serve crucial roles in the development and progression of breast cancer. However, the exact role and underlying molecular mechanism of miR-375 in mediating the growth and metastasis of breast cancer remains unknown. In the present study, reverse transcription-quantitative polymerase chain reaction and western blot analysis were conducted to examine RNA and protein expression. A luciferase reporter assay was performed to determine the association between miR-375 and paired box 6 (PAX6). The results of the current study indicate that the expression of miR-375 was reduced in breast cancer tissues compared with matched adjacent normal tissues. Transfection with miR-375 mimics led to a significant increase in levels of miR-375 in human breast cancer Michigan Cancer Foundation (MCF)-7 cells (P<0.05). The increase in miR-375 expression caused a significant decrease in the viability, migration and invasion of MCF-7 cells (P<0.05), accompanied by a reduced expression of matrix metalloproteinase (MMP) 2 and MMP9 proteins. Luciferase reporter assay identified PAX6 as a novel target of miR-375 and miR-375 in turn, negatively regulated the protein expression of PAX6 in MCF-7 cells. By contrast, overexpression of PAX6 led to a significant increase in MCF-7 cell viability (P<0.01) but did not affect the migration and invasion of MCF-7 cells, suggesting that the inhibitory effect of miR-375 on MCF-7 cell viability may be occurring, in part, via the direct targeting of PAX6.
Collapse
Affiliation(s)
- Qiongyan Zou
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Wenjun Yi
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Jianghai Huang
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Fenfen Fu
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Gannong Chen
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Dewu Zhong
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
31
|
Shen CJ, Cheng YM, Wang CL. LncRNA PVT1 epigenetically silences miR-195 and modulates EMT and chemoresistance in cervical cancer cells. J Drug Target 2017; 25:637-644. [PMID: 28296507 DOI: 10.1080/1061186x.2017.1307379] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The plasmacytoma variant translocation 1 gene (PVT1) is an oncogenic lncRNA with regulative effect on chemosensitivity in cervical cancer. However, the underlying mechanisms were not fully understood. In this study, HPV16 positive CaSki and SiHa cells were used as in vitro cell model. Knockdown of HPV16 E7 significantly inhibited PVT1 and restored miR-195 expression. PVT1 directly interacts with EZH2 and the complex anchors in the promoter region of miR-195. PVT1 overexpression resulted in increased H3K27me3 levels in the miR-195 promoter region, while PVT1 knockdown decreased H3K27me3 levels in the promoter region. In addition, PVT1 could competitively bind with miR-195. MiR-195 overexpression suppressed PVT1 expression in the cancer cells. Both PVT1 and miR-195 could inhibit paclitaxel (PTX) induced epithelial-to-mesenchymal transition (EMT) and also sensitize CaSki cells to PTX. Based on these findings, we infer that PVT1 could decrease miR-195 expression via enhancing histone H3K27me3 in the miR-195 promoter region and also via direct sponging of miR-195. In addition, the PVT1/miR-195 axis can modulate responses of the cancer cells to PTX via regulating EMT.
Collapse
Affiliation(s)
- Ching-Ju Shen
- a Department of Gynecology and Obstetrics , Kaohsiung Medical University Hospital, Kaohsiung Medical University , Kaohsiung , Taiwan
| | - Ya-Min Cheng
- b Department of Obstetrics and Gynecology , Institute of Clinical Medicine, College of Medicine, National Cheng Kung University , Tainan , Taiwan
| | - Chiu-Lin Wang
- c Department of Obstetrics and Gynecology , Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung Medical University , Kaohsiung , Taiwan
| |
Collapse
|
32
|
Downregulation of microRNA-145 promotes epithelial-mesenchymal transition via regulating Snail in osteosarcoma. Cancer Gene Ther 2017; 24:83-88. [PMID: 28186090 DOI: 10.1038/cgt.2017.1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 11/21/2016] [Accepted: 01/05/2017] [Indexed: 12/12/2022]
Abstract
Metastasis is the principal cause of cancer death and occurs through multiple, complex processes. Epithelial to mesenchymal transition (EMT) is an important process during embryonic development and has also been hypothesized to exhibit a significant role in cancer cell invasion and metastasis. MicroRNAs (miRNAs) are a class of widespread noncoding RNAs. In recent years, many studies have shown that miRNAs could influence the signaling pathways and downstream events that define EMT on a molecular level. However, the exact role and mechanisms of miR-145 in EMT of osteosarcoma (OS) was unknown. In the present study, miR-145 was downregulated in OS tissues and cell lines and it was shown that miR-145 expression was closely correlated with advanced tumor progression in patients of OS. In addition, miR-145 upregulation by miR-145 agomir significantly inhibited MG63 cells invasion and migration ability. MiR-145 was reported to be able to inhibit EMT in cancers. Following the examination of changes in cell epithelial and mesenchymal markers, it was found that upregulation of miR-145 strongly reversed EMT in MG63 cells. Meanwhile, the expression of Snail, a strong E-cadherin transcription repressor was also attenuated by miR-145 agomir. Furthermore, the decreased EMT and invasion and metastasis caused by miR-145 agomir could be restored by Snail siRNA. In conclusion, the results demonstrated that miR-145 could mediate EMT by targeting Snail and miR-145 might be a novel EMT regulating transcription factor that involved in the progression of OS. The specific drugs targeting miR-145-mediated EMT process might be new promising cancer therapies.
Collapse
|
33
|
Juang V, Lee HP, Lin AMY, Lo YL. Cationic PEGylated liposomes incorporating an antimicrobial peptide tilapia hepcidin 2-3: an adjuvant of epirubicin to overcome multidrug resistance in cervical cancer cells. Int J Nanomedicine 2016; 11:6047-6064. [PMID: 27895479 PMCID: PMC5117904 DOI: 10.2147/ijn.s117618] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Antimicrobial peptides (AMPs) have been recently evaluated as a new generation of adjuvants in cancer chemotherapy. In this study, we designed PEGylated liposomes encapsulating epirubicin as an antineoplastic agent and tilapia hepcidin 2–3, an AMP, as a multidrug resistance (MDR) transporter suppressor and an apoptosis/autophagy modulator in human cervical cancer HeLa cells. Cotreatment of HeLa cells with PEGylated liposomal formulation of epirubicin and hepcidin 2–3 significantly increased the cytotoxicity of epirubicin. The liposomal formulations of epirubicin and/or hepcidin 2–3 were found to noticeably escalate the intracellular H2O2 and O2− levels of cancer cells. Furthermore, these treatments considerably reduced the mRNA expressions of MDR protein 1, MDR-associated protein (MRP) 1, and MRP2. The addition of hepcidin 2–3 in liposomes was shown to markedly enhance the intracellular epirubicin uptake and mainly localized into the nucleus. Moreover, this formulation was also found to trigger apoptosis and autophagy in HeLa cells, as validated by significant increases in the expressions of cleaved poly ADP ribose polymerase, caspase-3, caspase-9, and light chain 3 (LC3)-II, as well as a decrease in mitochondrial membrane potential. The apoptosis induction was also confirmed by the rise in sub-G1 phase of cell cycle assay and apoptosis percentage of annexin V/propidium iodide assay. We found that liposomal epirubicin and hepcidin 2–3 augmented the accumulation of GFP-LC3 puncta as amplified by chloroquine, implying the involvement of autophagy. Interestingly, the partial inhibition of necroptosis and the epithelial–mesenchymal transition by this combination was also verified. Altogether, our results provide evidence that coincubation with PEGylated liposomes of hepcidin 2–3 and epirubicin caused programmed cell death in cervical cancer cells through modulation of multiple signaling pathways, including MDR transporters, apoptosis, autophagy, and/or necroptosis. Thus, this formulation may provide a new platform for the combined treatment of traditional chemotherapy and hepcidin 2–3 as a new adjuvant for effective MDR reversal.
Collapse
Affiliation(s)
- Vivian Juang
- Department and Institute of Pharmacology, National Yang-Ming University
| | - Hsin-Pin Lee
- Department of Biological Sciences and Technology, National University of Tainan
| | - Anya Maan-Yuh Lin
- Department and Institute of Pharmacology, National Yang-Ming University; Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
| | - Yu-Li Lo
- Department and Institute of Pharmacology, National Yang-Ming University
| |
Collapse
|
34
|
Wang Y, Lieberman R, Pan J, Zhang Q, Du M, Zhang P, Nevalainen M, Kohli M, Shenoy NK, Meng H, You M, Wang L. miR-375 induces docetaxel resistance in prostate cancer by targeting SEC23A and YAP1. Mol Cancer 2016; 15:70. [PMID: 27832783 PMCID: PMC5105253 DOI: 10.1186/s12943-016-0556-9] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 11/02/2016] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Treatment options for metastatic castrate-resistant prostate cancer (mCRPC) are limited and typically are centered on docetaxel-based chemotherapy. We previously reported that elevated miR-375 levels were significantly associated with poor overall survival of mCRPC patients. In this study, we evaluated if miR-375 induced chemo-resistance to docetaxel through regulating target genes associated with drug resistance. METHODS We first compared miR-375 expression level between prostate cancer tissues and normal prostate tissues using data from The Cancer Genome Atlas (TCGA). To examine the role of miR-375 in docetaxel resistance, we transfected miR-375 using a pre-miRNA lentiviral vector and examined the effects of exogenously overexpressed miR-375 on cell growth in two prostate cancer cell lines, DU145 and PC-3. To determine the effect of overexpressed miR-375 on tumor growth and chemo-resistance in vivo, we injected prostate cancer cells overexpressing miR-375 into nude mice subcutaneously and evaluated tumor growth rate during docetaxel treatment. Lastly, we utilized qRT-PCR and Western blot assay to examine two miR-375 target genes, SEC23A and YAP1, for their expression changes after miR-375 transfection. RESULTS By examining 495 tumor tissues and 52 normal tissues from TCGA data, we found that compared to normal prostate, miR-375 was significantly overexpressed in prostate cancer tissues (8.45-fold increase, p value = 1.98E-23). Docetaxel treatment induced higher expression of miR-375 with 5.83- and 3.02-fold increases in DU145 and PC-3 cells, respectively. Interestingly, miR-375 appeared to play a dual role in prostate cancer proliferation. While miR-375 overexpression caused cell growth inhibition and cell apoptosis, elevated miR-375 also significantly reduced cell sensitivity to docetaxel treatment in vitro, as evidenced by decreased apoptotic cells. In vivo xenograft mouse study showed that tumors with increased miR-375 expression were more tolerant to docetaxel treatment, demonstrated by greater tumor weight and less apoptotic cells in miR-375 transfected group when compared to empty vector control group. In addition, we examined expression levels of the two miR-375 target genes (SEC23A and YAP1) and observed significant reduction in the expression at both protein and mRNA levels in miR-375 transfected prostate cancer cell lines. TCGA dataset analysis further confirmed the negative correlations between miR-375 and the two target genes (r = -0.62 and -0.56 for SEC23A and YAP1, respectively; p < 0.0001). CONCLUSIONS miR-375 is involved in development of chemo-resistance to docetaxel through regulating SEC23A and YAP1 expression. Our results suggest that miR-375 or its target genes, SEC23A or YAP1, might serve as potential predictive biomarkers to docetaxel-based chemotherapy and/or therapeutic targets to overcome chemo-resistance in mCRPC stage.
Collapse
Affiliation(s)
- Yuan Wang
- Key Laboratory of Hydrobiology in Liaoning Province's Universities, Dalian Ocean University, Dalian, 116021, China.,Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Rachel Lieberman
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Jing Pan
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Qi Zhang
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Meijun Du
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Peng Zhang
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Marja Nevalainen
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.,Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Manish Kohli
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Niraj K Shenoy
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Hui Meng
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Ming You
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Liang Wang
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
| |
Collapse
|
35
|
Peng F, Xiong L, Tang H, Peng C, Chen J. Regulation of epithelial-mesenchymal transition through microRNAs: clinical and biological significance of microRNAs in breast cancer. Tumour Biol 2016; 37:14463-14477. [DOI: 10.1007/s13277-016-5334-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Accepted: 09/06/2016] [Indexed: 12/16/2022] Open
|
36
|
Biersack B. Current state of phenolic and terpenoidal dietary factors and natural products as non-coding RNA/microRNA modulators for improved cancer therapy and prevention. Noncoding RNA Res 2016; 1:12-34. [PMID: 30159408 PMCID: PMC6096431 DOI: 10.1016/j.ncrna.2016.07.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 07/20/2016] [Accepted: 07/20/2016] [Indexed: 02/06/2023] Open
Abstract
The epigenetic regulation of cancer cells by small non-coding RNA molecules, the microRNAs (miRNAs), has raised particular interest in the field of oncology. These miRNAs play crucial roles concerning pathogenic properties of cancer cells and the sensitivity of cancer cells towards anticancer drugs. Certain miRNAs are responsible for an enhanced activity of drugs, while others lead to the formation of tumor resistance. In addition, miRNAs regulate survival and proliferation of cancer cells, in particular of cancer stem-like cells (CSCs), that are especially drug-resistant and, thus, cause tumor relapse in many cases. Various small molecule compounds were discovered that target miRNAs that are known to modulate tumor aggressiveness and drug resistance. This review comprises the effects of naturally occurring small molecules (phenolic compounds and terpenoids) on miRNAs involved in cancer diseases.
Collapse
Key Words
- 1,25-D, 1,25-dihydroxyvitamin D3
- 18-AGA, 18α-glycyrrhetinic acid
- 3,6-DHF, 3,6-dihydroxyflavone
- AKBA, 3-acetyl-11-keto-β-boswellic acid
- Anticancer drugs
- CAPE, caffeic acid phenethyl ester
- CDODA-Me, methyl 2-cyano-3,11-dioxo-18β-olean-1,12-dien-30-oate
- Dox, doxorubicin
- EGCG, (−)-epigallocatechin-3-O-gallate
- MicroRNA
- PEG, polyethylene glycol
- PPAP, polycyclic polyprenylated acylphloroglucinol
- Polyphenols
- RA, retinoic acid
- ROS, reactive oxygen species
- TQ, thymoquinone
- Terpenes
Collapse
Affiliation(s)
- Bernhard Biersack
- Organic Chemistry Laboratory, University of Bayreuth, Universitätsstraße 30, 95447 Bayreuth, Germany
| |
Collapse
|
37
|
Hao M, Zhao W, Zhang L, Wang H, Yang X. Low folate levels are associated with methylation-mediated transcriptional repression of miR-203 and miR-375 during cervical carcinogenesis. Oncol Lett 2016; 11:3863-3869. [PMID: 27313708 DOI: 10.3892/ol.2016.4449] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 01/22/2016] [Indexed: 12/14/2022] Open
Abstract
The aim of the present study was to investigate the correlation between a lack of folic acid and the abnormal expression of microRNA (miR)-203 and miR-375 in cervical cancer. In total, 60 tissue samples of cervical intraepithelial neoplasia (CIN) or stage IA-IIA cervical cancer (study group), and 30 samples without soluble interleukin or malignancy (control group) were examined. The expression of miR-203 and miR-375 was detected using reverse transcription-quantitative polymerase chain reaction (RT-qPCR), and the difference in expression levels was quantified using the 2-ΔΔCq method. In addition, CaSki cervical cancer cells were cultured in vitro and treated with various concentrations of folic acid. The DNA methylation states of miR-203 and miR-375 were subsequently detected by methylation-specific PCR, and the expression levels were evaluated using RT-PCR. miR-203 and miR-375 were significantly downregulated in CIN and cervical cancer tissues, compared with the control group. There was a marked difference in terms of the expression levels of miR-375 between the two groups (P<0.05). In CaSki cells, as the concentration of folic acid increased, the positive rate of DNA methylation of miR-203 and miR-375 decreased, while the expression levels of miR-203 and miR-375 demonstrated a gradual increase, which indicated that the latter two parameters were negatively correlated (P<0.05). Compared with normal cervical tissue, the expression levels of miR-203 and miR-375 were downregulated in CIN and cervical cancer. Methylation of these two miRs was apparent in CaSki cells, and was associated with a lack of folic acid. Therefore, reduced levels of folic acid, leading to increased methylation of miR-203 and miR-375, may be significant events during cervical carcinogenesis.
Collapse
Affiliation(s)
- Min Hao
- Department of Obstetrics and Gynecology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Weihong Zhao
- Department of Obstetrics and Gynecology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Lili Zhang
- Department of Obstetrics and Gynecology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Honghong Wang
- Department of Obstetrics and Gynecology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Xin Yang
- Department of Obstetrics and Gynecology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| |
Collapse
|
38
|
Zhao W, Wang H, Han X, Ma J, Zhou Y, Chen Z, Zhou H, Xu H, Sun Z, Kong B, Fang H. ΔNp63α attenuates tumor aggressiveness by suppressing miR-205/ZEB1-mediated epithelial-mesenchymal transition in cervical squamous cell carcinoma. Tumour Biol 2016; 37:10621-32. [PMID: 26864590 DOI: 10.1007/s13277-016-4921-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 01/27/2016] [Indexed: 12/31/2022] Open
Abstract
Cervical cancer is one of the most common female cancers worldwide. Although the therapeutic outcomes of patients with early-stage cervical cancer have been significantly improved in the past decades, tumor metastasis and recurrence remain the major causes of cervical cancer-related deaths. In cervical squamous cell carcinoma (SCC), the aberrant activation of epithelial-mesenchymal transition (EMT), a crucial process in invasion and metastasis of epithelial cancer, could promote lymph nodal metastasis and recurrence, and predicts poor prognosis. In this study, we show that the expression levels of EMT markers, β-catenin and Vimentin, are associated with the p63 isoform ΔNp63α in SCC by using immunohistochemistry staining and analysis. Compared to the control SiHa cells (SiHa-NC), the expression of E-cadherin and β-catenin are upregulated, while Vimentin and ZEB1 are downregulated in the constructed SiHa cell line with stable ΔNp63α overexpression (SiHa-ΔNp63α). Besides, the migration and invasion abilities are also suppressed in SiHa-ΔNp63α cells with a typical epithelial morphology with cobblestone-like shape, suggesting that ΔNp63α is a vital EMT repressor in SCC cells. In addition, the involvement of miR-205/ZEB1 axis in the inhibition effect of ΔNp63α on EMT program is revealed by a miRNA array and confirmed by the subsequent transfection of the miR-205 mimic and antagomir. Moreover, SCC patients with low ΔNp63α expression and high EMT level show more frequent metastasis and recurrence as well as reduced overall survival. Therefore, EMT program and its vital repressor ΔNp63α could be used as biomarkers for tumor metastasis and recurrence in cervical cancer.
Collapse
Affiliation(s)
- Weidong Zhao
- Department of Obstetrics and Gynecology, Anhui Provincial Hospital affiliated to Anhui Medical University, Hefei, China. .,Department of Gynecologic Oncology, Anhui Provincial Cancer Hospital, Hefei, China. .,Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, China.
| | - Huiyan Wang
- Department of Gynecologic Oncology, Anhui Provincial Cancer Hospital, Hefei, China
| | - Xiaohui Han
- Department of Obstetrics and Gynecology, Anhui Provincial Hospital affiliated to Anhui Medical University, Hefei, China
| | - Jie Ma
- Department of Gynecologic Oncology, Anhui Provincial Cancer Hospital, Hefei, China
| | - Yuanyuan Zhou
- Department of Gynecologic Oncology, Anhui Provincial Cancer Hospital, Hefei, China
| | - Zhengzheng Chen
- Department of Obstetrics and Gynecology, Anhui Provincial Hospital affiliated to Anhui Medical University, Hefei, China
| | - Hu Zhou
- Department of Gynecologic Oncology, Anhui Provincial Cancer Hospital, Hefei, China
| | - Hanjie Xu
- Department of Obstetrics and Gynecology, Anhui Provincial Hospital affiliated to Anhui Medical University, Hefei, China
| | - Zhengwei Sun
- Department of Obstetrics and Gynecology, Anhui Provincial Hospital affiliated to Anhui Medical University, Hefei, China
| | - Beihua Kong
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, China
| | - Huiying Fang
- Department of Nursing, Anhui Vocational Institute of Population, Chizhou, China
| |
Collapse
|
39
|
Salony, Solé X, Alves CP, Dey-Guha I, Ritsma L, Boukhali M, Lee JH, Chowdhury J, Ross KN, Haas W, Vasudevan S, Ramaswamy S. AKT Inhibition Promotes Nonautonomous Cancer Cell Survival. Mol Cancer Ther 2015; 15:142-53. [PMID: 26637368 DOI: 10.1158/1535-7163.mct-15-0414] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 10/21/2015] [Indexed: 12/20/2022]
Abstract
Small molecule inhibitors of AKT (v-akt murine thymoma viral oncogene homolog) signaling are being evaluated in patients with various cancer types, but have so far proven therapeutically disappointing for reasons that remain unclear. Here, we treat cancer cells with subtherapeutic doses of Akti-1/2, an allosteric small molecule AKT inhibitor, in order to experimentally model pharmacologic inhibition of AKT signaling in vitro. We then apply a combined RNA, protein, and metabolite profiling approach to develop an integrated, multiscale, molecular snapshot of this "AKT(low)" cancer cell state. We find that AKT-inhibited cancer cells suppress thousands of mRNA transcripts, and proteins related to the cell cycle, ribosome, and protein translation. Surprisingly, however, these AKT-inhibited cells simultaneously upregulate a host of other proteins and metabolites posttranscriptionally, reflecting activation of their endo-vesiculo-membrane system, secretion of inflammatory proteins, and elaboration of extracellular microvesicles. Importantly, these microvesicles enable rapidly proliferating cancer cells of various types to better withstand different stress conditions, including serum deprivation, hypoxia, or cytotoxic chemotherapy in vitro and xenografting in vivo. These findings suggest a model whereby cancer cells experiencing a partial inhibition of AKT signaling may actually promote the survival of neighbors through non-cell autonomous communication.
Collapse
Affiliation(s)
- Salony
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts
| | - Xavier Solé
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts
| | - Cleidson P Alves
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts
| | - Ipsita Dey-Guha
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts
| | - Laila Ritsma
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts
| | - Myriam Boukhali
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts
| | - Ju H Lee
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts
| | - Joeeta Chowdhury
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts
| | - Kenneth N Ross
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts
| | - Wilhelm Haas
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts
| | - Shobha Vasudevan
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts
| | - Sridhar Ramaswamy
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts. Broad Institute of Harvard & MIT, Cambridge, Massachusetts. Harvard Stem Cell Institute, Cambridge, Massachusetts. Harvard-Ludwig Center for Cancer Research, Boston, Massachusetts.
| |
Collapse
|
40
|
PAK4 confers the malignance of cervical cancers and contributes to the cisplatin-resistance in cervical cancer cells via PI3K/AKT pathway. Diagn Pathol 2015; 10:177. [PMID: 26411419 PMCID: PMC4584462 DOI: 10.1186/s13000-015-0404-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 08/28/2015] [Indexed: 01/16/2023] Open
Abstract
Background Multiple protein or microRNA markers have been recognized to contribute to the progression and recurrence of cervical cancers. Particular those, which are associated with the chemo- or radio-resistance of cervical cancers, have been proposed to be promising and to facilitate the definition for cervical cancer treatment options. Methods This study was designed to explore the potential prognosis value of p21-activated kinase (PAK)-4 in cervical cancer, via the Kaplan–Meier analysis, log-rank test and Cox regression analysis, and then to investigate the regulatory role of PAK4 in the cisplatin resistance in cervical cancer cells, via the strategies of both PAK4 overexpression and PAK4 knockout. Results It was demonstrated that PAK4 was upregulated in cervical cancer tissues, in an association with the cancer’s malignance variables such as FIGO stage, lymph node or distant metastasis and the poor histological grade. The high PAK4 expression was also independently associated with poor prognosis to cervical cancer patients. Moreover, PAK4 confers cisplatin resistance in cervical cancer Hela or Caski cells. In addition, the PI3K/Akt pathway has been implicated in the PAK4-confered cisplatin resistance. And the PI3K/Akt inhibitor, LY294002, markedly deteriorated the cisplatin-mediated viability reduction of Hela or Caski cells, indicating the involvement of PI3K/Akt pathway in the cisplatin resistance in cervical cancer cells. Conclusion This study has confirmed the significant prognostic role of PAK4 level in cervical cancer patients and has recognized the regulatory role in cervical cancer progression. Moreover, our study has indicated that PAK4 also confers the chemoresistance of cervical cancer cells in a PI3K/Akt-dependent way. Thus, our study indicates PAK4 as a promising marker for cervical cancer treatment.
Collapse
|
41
|
Song L, Liu S, Zeng S, Zhang L, Li X. miR-375 Modulates Radiosensitivity of HR-HPV-Positive Cervical Cancer Cells by Targeting UBE3A through the p53 Pathway. Med Sci Monit 2015. [PMID: 26224081 PMCID: PMC4524565 DOI: 10.12659/msm.893859] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Background Prediction of radioresistance of HR-HPV-positive (+) cervical cancer, especially before the course of radiotherapy, is quite beneficial to develop an optimal treatment strategy for individual patients. Unfortunately, the mechanisms responsible for radioresistance of cervical cancer are still largely unexplored. HR-HPV infection leads to a series of changes to normal biophysical process, including miRNAs expression. In this study, we explored the association between miR-375 and radioresistance in HR-HPV (+) cervical cancer. Material/Methods qRT-PCR analysis was performed to determine miR-375 expression in HR-HPV-positive (+) cervical cancer patients and in HPV-16-positive SiHa and HPV-18-positive HeLa cervical cancer cell lines. The influence of miR-375 on radiosensitivity and the downstream regulative network were further explored in the cell line models. Results The results verified a putative binding site between miR-375 and UBE3A. miR-375 overexpression could significantly reduce UBE3A expression. UBE3A knockdown led to significantly reduced cell survival under radiation treatment. miR-375 promoted radiosensitivity of HR-HPV (+) cancer through decreasing p53 degradation and thereby increasing radiation-induced apoptosis. Conclusions The miR-375-UBE3A axis is important in modulating radiosensitivity of HR-HPV (+) cervical cancer.
Collapse
Affiliation(s)
- Lili Song
- Department of Gynaecology and Obstetrics, Cangzhou Central Hospital, Cangzhou, Hebei, China (mainland)
| | - Shikai Liu
- Department of Gynaecology and Obstetrics, Cangzhou Central Hospital, Cangzhou, Hebei, China (mainland)
| | - Saitian Zeng
- Department of Gynaecology and Obstetrics, Cangzhou Central Hospital, Cangzhou, Hebei, China (mainland)
| | - Liang Zhang
- Department of Gynaecology and Obstetrics, Cangzhou Central Hospital, Cangzhou, Hebei, China (mainland)
| | - Xia Li
- Department of Gynaecology and Obstetrics, Cangzhou Central Hospital, Cangzhou, Hebei, China (mainland)
| |
Collapse
|
42
|
Naidu S, Magee P, Garofalo M. MiRNA-based therapeutic intervention of cancer. J Hematol Oncol 2015; 8:68. [PMID: 26062952 PMCID: PMC4465004 DOI: 10.1186/s13045-015-0162-0] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 05/28/2015] [Indexed: 12/27/2022] Open
Abstract
MicroRNAs (miRNAs) are important modulators of eukaryotic gene expression. By targeting protein coding transcripts, miRNAs influence the cellular transcriptome and proteome, thus helping to determine cell fate. MiRNAs have emerged as crucial molecules in cancer research, in which recent studies have linked erratic expression of miRNAs to carcinogenesis and have provided solid evidence for their potential in cancer therapy. This review briefly summarises the recent knowledge on the involvement of miRNAs in tumourigenesis and reviews current studies on the therapeutic strategies and advances in the delivery of miRNAs.
Collapse
Affiliation(s)
- Srivatsava Naidu
- Transcriptional Networks in Lung Cancer Group, Cancer Research UK Manchester Institute, University of Manchester, Wilmslow Road, Manchester, M20 4BX, UK.
| | - Peter Magee
- Transcriptional Networks in Lung Cancer Group, Cancer Research UK Manchester Institute, University of Manchester, Wilmslow Road, Manchester, M20 4BX, UK.
| | - Michela Garofalo
- Transcriptional Networks in Lung Cancer Group, Cancer Research UK Manchester Institute, University of Manchester, Wilmslow Road, Manchester, M20 4BX, UK.
| |
Collapse
|
43
|
MicroRNA-375 functions as a tumor suppressor in osteosarcoma by targeting PIK3CA. Tumour Biol 2015; 36:8579-84. [PMID: 26036761 DOI: 10.1007/s13277-015-3614-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 05/26/2015] [Indexed: 01/06/2023] Open
Abstract
Osteosarcoma has become one of the most common primary malignant bone tumors in childhood and adult. Numerous studies have demonstrated that aberrant microRNA (miRNA) expression is involved in human disease including cancer. To date, the potential miRNAs regulating osteosarcoma growth and progression are not fully identified yet. Herein, we showed that miR-375 was frequently downregulated in osteosarcoma tissue and cell lines compared to normal human colon tissues. Overexpression of miR-375 resulted in decreased expression of PIK3CA (phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit alpha) at both mRNA and protein levels. We found that miR-375 overexpression markedly suppressed cell proliferation in vitro. And inhibition of miR-375 promotes osteosarcoma growth. Mechanistic studies showed that PIK3CA was a potential target of miR-375 and it mediated reduction of PIK3CA resulted in suppression of PI3K/Akt pathway. Taken together, our results demonstrate that miR-375 functions as a growth-suppressive miRNA and plays an important role in inhibiting the tumorigenesis through targeting PIK3CA in osteosarcoma.
Collapse
|
44
|
Krakowsky RHE, Tollefsbol TO. Impact of Nutrition on Non-Coding RNA Epigenetics in Breast and Gynecological Cancer. Front Nutr 2015; 2:16. [PMID: 26075205 PMCID: PMC4445322 DOI: 10.3389/fnut.2015.00016] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Accepted: 05/02/2015] [Indexed: 12/21/2022] Open
Abstract
Cancer is the second leading cause of death in females. According to the American Cancer Society, there are 327,660 new cases in breast and gynecological cancers estimated in 2014, placing emphasis on the need for cancer prevention and new cancer treatment strategies. One important approach to cancer prevention involves phytochemicals, biologically active compounds derived from plants. A variety of studies on the impact of dietary compounds found in cruciferous vegetables, green tea, and spices like curry and black pepper have revealed epigenetic changes in female cancers. Thus, an important emerging topic comprises epigenetic changes due to the modulation of non-coding RNA levels. Since it has been shown that non-coding RNAs such as microRNAs and long non-coding RNAs are aberrantly expressed in cancer, and furthermore are linked to distinct cancer phenotypes, understanding the effects of dietary compounds and supplements on the epigenetic modulator non-coding RNA is of great interest. This article reviews the current findings on nutrition-induced changes in breast and gynecological cancers at the non-coding RNA level.
Collapse
Affiliation(s)
- Rosanna H E Krakowsky
- Department of Biology, University of Alabama at Birmingham , Birmingham, AL , USA ; Department of Biochemistry, University of Leipzig , Leipzig , Germany
| | - Trygve O Tollefsbol
- Department of Biology, University of Alabama at Birmingham , Birmingham, AL , USA ; Comprehensive Center for Healthy Ageing, University of Alabama at Birmingham , Birmingham, AL , USA ; Comprehensive Cancer Center, University of Alabama at Birmingham , Birmingham, AL , USA ; Nutrition Obesity Research Center, University of Alabama at Birmingham , Birmingham, AL , USA ; Comprehensive Diabetes Center, University of Alabama at Birmingham , Birmingham, AL , USA
| |
Collapse
|