1
|
Liao M, Qin M, Liu L, Huang H, Chen N, Du H, Huang D, Wang P, Zhou H, Tong G. Exosomal microRNA profiling revealed enhanced autophagy suppression and anti-tumor effects of a combination of compound Phyllanthus urinaria and lenvatinib in hepatocellular carcinoma. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 122:155091. [PMID: 37844378 DOI: 10.1016/j.phymed.2023.155091] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 08/23/2023] [Accepted: 09/12/2023] [Indexed: 10/18/2023]
Abstract
BACKGROUND Compound Phyllanthus urinaria (CP), a traditional Chinese herbal remedy, possesses strong anti-cancer effects and is extensively employed in the clinical management of hepatocellular carcinoma (HCC). While lenvatinib and other oral tyrosine kinase inhibitors have been authorized as initial treatments for advanced unresectable HCC, the survival of patients is ultimately restricted due to the gradual development of drug resistance. Fortunately, the co-administration of CP and lenvatinib holds promise for anti-cancer applications. PURPOSE Our objective was to understand the molecular-level mechanisms of bioactive phytocompounds in CP, in order to explore the anti-HCC effects of combining CP and lenvatinib treatment and reveal the underlying mechanisms. Furthermore, we discovered new miRNAs associated with autophagy that are common to both HepG2-derived exosomes and HepG2 cells. These miRNAs play a role in the advancement of HCC and were identified through the utilization of CP and lenvatinib. METHODS To assess the anti-HCC effects of CP in combination with lenvatinib, both an in vitro CCK-8 assay and an in vivo xenograft model assay were performed. TEM, NTA, and nano-flow cytometry were employed for the identification of isolated exosomes. To ascertain the miRNA expression patterns in HepG2 cells and HepG2-derived exosomes, miRNA-sequencing analysis was conducted. Further investigation involved the use of real-time PCR, examination of the fusion protein GFP-mRFP-LC3, TEM analysis, and western blotting. RESULTS In vitro and in vivo, the combination of CP and lenvatinib showed a stronger and more powerful impact on HCC compared to either CP or lenvatinib alone. The combination of CP and lenvatinib had a significant impact on autophagy-related miRNAs in HepG2-derived exosomes and HepG2 cells, as demonstrated by cellular and exosomal miRNA sequencing. Additional tests indicated that the increased inhibition of autophagy in HepG2 cells subjected to CP treatment, as well as the combination of CP and lenvatinib, was accomplished through the regulation of Beclin-1, LC3-II, and P62 expression. CONCLUSION In conclusion, our results indicate that the combination of CP and lenvatinib can effectively inhibit HCC by promoting the exosome-mediated suppression of autophagy. This novel therapeutic option is highly efficient and durable, making it a promising treatment for HCC. Moreover, the miRNAs that are differentially expressed and associated with exosome-mediated autophagy, which have been discovered in this study, could potentially be targeted for clinical treatment of HCC.
Collapse
Affiliation(s)
- Mianmian Liao
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No.1 Fuhua Road, Futian District, Shenzhen, Guangdong 518033, China
| | - Meirong Qin
- Shenzhen Institute for Drug Control, No. 28, The second Gaoxin Road, Nanshan District, Shenzhen, Guangdong 518000, China
| | - Linhua Liu
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No.1 Fuhua Road, Futian District, Shenzhen, Guangdong 518033, China
| | - Houshuang Huang
- Shenzhen Institute for Drug Control, No. 28, The second Gaoxin Road, Nanshan District, Shenzhen, Guangdong 518000, China
| | - Ning Chen
- Shenzhen Institute for Drug Control, No. 28, The second Gaoxin Road, Nanshan District, Shenzhen, Guangdong 518000, China
| | - Haiyan Du
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No.1 Fuhua Road, Futian District, Shenzhen, Guangdong 518033, China
| | - Danping Huang
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No.1 Fuhua Road, Futian District, Shenzhen, Guangdong 518033, China; Department of Integrated Traditional Chinese and Western Medicine, School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510006, China
| | - Ping Wang
- Shenzhen Institute for Drug Control, No. 28, The second Gaoxin Road, Nanshan District, Shenzhen, Guangdong 518000, China.
| | - Hua Zhou
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune, Disease Research, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China.
| | - Guangdong Tong
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No.1 Fuhua Road, Futian District, Shenzhen, Guangdong 518033, China.
| |
Collapse
|
2
|
Baker AR, Miliotis C, Ramírez-Moya J, Marc T, Vlachos IS, Santisteban P, Slack FJ. Transcriptome profiling of ADAR1 targets in triple-negative breast cancer cells reveals mechanisms for regulating growth and invasion. Mol Cancer Res 2022; 20:960-971. [PMID: 35247916 DOI: 10.1158/1541-7786.mcr-21-0604] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 12/23/2021] [Accepted: 02/28/2022] [Indexed: 11/16/2022]
Abstract
ADARs catalyze Adenosine-to-Inosine (A-to-I) editing of double-stranded RNA and regulate global gene expression output through interactions with RNA and other proteins. ADARs play important roles in development and disease, and previous work has shown that ADAR1 is oncogenic in a growing list of cancer types. Here we show that ADAR1 is a critical gene for triple-negative breast cancer cells, as ADAR1 loss results in reduced growth (viability and cell cycle progression), invasion, and mammosphere formation. Whole transcriptome sequencing analyses demonstrate that ADAR1 regulates both coding and non-coding targets by altering gene expression level, A-to-I editing, and splicing. We determine that a recoding edit in filamin B (FLNB chr3:58156064) reduces the tumor suppressive activities of the protein to promote growth and invasion. We also show that several tumor suppressor microRNAs are upregulated upon ADAR1 loss and suppress cell cycle progression and invasion. Implications: This work describes several novel mechanisms of ADAR1-mediated oncogenesis in triple-negative breast cancer, providing support to strategies targeting ADAR1 in this aggressive cancer type that has few treatment options.
Collapse
Affiliation(s)
| | | | - Julia Ramírez-Moya
- Boston Children's Hospital / Harvard Medical School, Boston, MA, United States
| | | | | | - Pilar Santisteban
- Instituto de Investigaciones Biomedicas CSIC/UAM, Madrid, Madrid, Spain
| | | |
Collapse
|
3
|
Slabáková E, Kahounová Z, Procházková J, Souček K. Regulation of Neuroendocrine-like Differentiation in Prostate Cancer by Non-Coding RNAs. Noncoding RNA 2021; 7:ncrna7040075. [PMID: 34940756 PMCID: PMC8704250 DOI: 10.3390/ncrna7040075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/18/2021] [Accepted: 11/29/2021] [Indexed: 12/21/2022] Open
Abstract
Neuroendocrine prostate cancer (NEPC) represents a variant of prostate cancer that occurs in response to treatment resistance or, to a much lesser extent, de novo. Unravelling the molecular mechanisms behind transdifferentiation of cancer cells to neuroendocrine-like cancer cells is essential for development of new treatment opportunities. This review focuses on summarizing the role of small molecules, predominantly microRNAs, in this phenomenon. A published literature search was performed to identify microRNAs, which are reported and experimentally validated to modulate neuroendocrine markers and/or regulators and to affect the complex neuroendocrine phenotype. Next, available patients’ expression datasets were surveyed to identify deregulated microRNAs, and their effect on NEPC and prostate cancer progression is summarized. Finally, possibilities of miRNA detection and quantification in body fluids of prostate cancer patients and their possible use as liquid biopsy in prostate cancer monitoring are discussed. All the addressed clinical and experimental contexts point to an association of NEPC with upregulation of miR-375 and downregulation of miR-34a and miR-19b-3p. Together, this review provides an overview of different roles of non-coding RNAs in the emergence of neuroendocrine prostate cancer.
Collapse
|
4
|
Wambecke A, Ahmad M, Morice PM, Lambert B, Weiswald LB, Vernon M, Vigneron N, Abeilard E, Brotin E, Figeac M, Gauduchon P, Poulain L, Denoyelle C, Meryet-Figuiere M. The lncRNA 'UCA1' modulates the response to chemotherapy of ovarian cancer through direct binding to miR-27a-5p and control of UBE2N levels. Mol Oncol 2021; 15:3659-3678. [PMID: 34160887 PMCID: PMC8637575 DOI: 10.1002/1878-0261.13045] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 03/24/2021] [Accepted: 06/22/2021] [Indexed: 01/28/2023] Open
Abstract
Ovarian cancer (OC) is the leading cause of death in patients with gynecologic cancers. Due to late diagnosis and resistance to chemotherapy, the 5‐year survival rate in patients with OC is below 40%. We observed that UCA1, a lncRNA previously reported to play an oncogenic role in several malignancies, is overexpressed in the chemoresistant OC cell line OAW42‐R compared to their chemotherapy‐sensitive counterpart OAW42. Additionally, UCA1 overexpression was related to poor prognosis in two independent patient cohorts. Currently, the molecular mechanisms through which UCA1 acts in OC are poorly understood. We demonstrated that downregulation of the short isoform of UCA1 sensitized OC cells to cisplatin and that UCA1 acted as competing endogenous RNA to miR‐27a‐5p. Upon UCA1 downregulation, miR‐27a‐5p downregulated its direct target UBE2N leading to the upregulation of BIM, a proapoptotic protein of the Bcl2 family. The upregulation of BIM is the event responsible for the sensitization of OC cells to cisplatin. In order to model response to therapy in patients with OC, we used several patient‐derived organoid cultures, a model faithfully mimicking patient’s response to therapy. Inhibition of UBE2N sensitized patient‐derived organoids to platinum salts. In conclusion, response to treatment in patients with OC is regulated by the UCA1/miR‐27a‐5p/UBE2N axis, where UBE2N inhibition could potentially represent a novel therapeutic strategy to counter chemoresistance in OC.
Collapse
Affiliation(s)
- Anaïs Wambecke
- UNICAEN, Inserm U1086 ANTICIPE (Interdisciplinary Research Unit for Cancer Prevention and Treatment), Normandie Univ, Caen, France.,Cancer Centre François Baclesse, UNICANCER, Caen, France
| | - Mohammad Ahmad
- UNICAEN, Inserm U1086 ANTICIPE (Interdisciplinary Research Unit for Cancer Prevention and Treatment), Normandie Univ, Caen, France.,Cancer Centre François Baclesse, UNICANCER, Caen, France
| | - Pierre-Marie Morice
- UNICAEN, Inserm U1086 ANTICIPE (Interdisciplinary Research Unit for Cancer Prevention and Treatment), Normandie Univ, Caen, France.,Cancer Centre François Baclesse, UNICANCER, Caen, France
| | - Bernard Lambert
- UNICAEN, Inserm U1086 ANTICIPE (Interdisciplinary Research Unit for Cancer Prevention and Treatment), Normandie Univ, Caen, France.,Cancer Centre François Baclesse, UNICANCER, Caen, France.,CNRS, Normandy Regional Delegation, Caen, France
| | - Louis-Bastien Weiswald
- UNICAEN, Inserm U1086 ANTICIPE (Interdisciplinary Research Unit for Cancer Prevention and Treatment), Normandie Univ, Caen, France.,Cancer Centre François Baclesse, UNICANCER, Caen, France
| | - Mégane Vernon
- UNICAEN, Inserm U1086 ANTICIPE (Interdisciplinary Research Unit for Cancer Prevention and Treatment), Normandie Univ, Caen, France.,Cancer Centre François Baclesse, UNICANCER, Caen, France
| | - Nicolas Vigneron
- UNICAEN, Inserm U1086 ANTICIPE (Interdisciplinary Research Unit for Cancer Prevention and Treatment), Normandie Univ, Caen, France.,Cancer Centre François Baclesse, UNICANCER, Caen, France
| | - Edwige Abeilard
- UNICAEN, Inserm U1086 ANTICIPE (Interdisciplinary Research Unit for Cancer Prevention and Treatment), Normandie Univ, Caen, France.,Cancer Centre François Baclesse, UNICANCER, Caen, France
| | - Emilie Brotin
- UNICAEN, Inserm U1086 ANTICIPE (Interdisciplinary Research Unit for Cancer Prevention and Treatment), Normandie Univ, Caen, France.,Cancer Centre François Baclesse, UNICANCER, Caen, France.,ImpedanCELL Core Facility, Federative Structure 4206 ICORE, UNICAEN, Caen, France
| | - Martin Figeac
- Functional and structural genomics platform, Institute for Cancer Research, Lille Univ, France
| | - Pascal Gauduchon
- UNICAEN, Inserm U1086 ANTICIPE (Interdisciplinary Research Unit for Cancer Prevention and Treatment), Normandie Univ, Caen, France.,Cancer Centre François Baclesse, UNICANCER, Caen, France
| | - Laurent Poulain
- UNICAEN, Inserm U1086 ANTICIPE (Interdisciplinary Research Unit for Cancer Prevention and Treatment), Normandie Univ, Caen, France.,Cancer Centre François Baclesse, UNICANCER, Caen, France
| | - Christophe Denoyelle
- UNICAEN, Inserm U1086 ANTICIPE (Interdisciplinary Research Unit for Cancer Prevention and Treatment), Normandie Univ, Caen, France.,Cancer Centre François Baclesse, UNICANCER, Caen, France.,ImpedanCELL Core Facility, Federative Structure 4206 ICORE, UNICAEN, Caen, France
| | - Matthieu Meryet-Figuiere
- UNICAEN, Inserm U1086 ANTICIPE (Interdisciplinary Research Unit for Cancer Prevention and Treatment), Normandie Univ, Caen, France.,Cancer Centre François Baclesse, UNICANCER, Caen, France
| |
Collapse
|
5
|
Baghbani E, Noorolyai S, Duijf PHG, Silvestris N, Kolahian S, Hashemzadeh S, Baghbanzadeh Kojabad A, FallahVazirabad A, Baradaran B. The impact of microRNAs on myeloid-derived suppressor cells in cancer. Hum Immunol 2021; 82:668-678. [PMID: 34020831 DOI: 10.1016/j.humimm.2021.04.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 04/28/2021] [Accepted: 04/28/2021] [Indexed: 02/08/2023]
Abstract
Inflammation promotes cancer development. To a large extent, this can be attributed to the recruitment of myeloid-derived suppressor cells (MDSCs) to tumors. These cells are known for establishing an immunosuppressive tumor microenvironment by suppressing T cell activities. However, MDSCs also promote metastasis and angiogenesis. Critically, as small non-coding RNAs that regulate gene expression, microRNAs (miRNAs) control MDSC activities. In this review, we discuss how miRNA networks regulate key MDSC signaling pathways, how they shape MDSC development, differentiation and activation, and how this impacts tumor development. By targeting the expression of miRNAs in MDSCs, we can alter their main signaling pathways. In turn, this can compromise their ability to promote multiple hallmarks of cancer. Therefore, this may represent a new powerful strategy for cancer immunotherapy.
Collapse
Affiliation(s)
- Elham Baghbani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Noorolyai
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pascal H G Duijf
- Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Australia; University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Nicola Silvestris
- IRCCS Bari, Italy. Medical Oncology Unit-IRCCS Istituto Tumori "Giovanni Paolo II" of Bari, Bari, Italy, Department of Biomedical Sciences and Human Oncology DIMO-University of Bari, Bari, Italy
| | - Saeed Kolahian
- Department of Experimental and Clinical Pharmacology and Pharmacogenomics, Division of Pharmacogenomics, University of Tübingen, Tübingen, Germany; Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Philipps University of Marburg, Marburg, Germany; Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Marburg, Germany
| | - Shahryar Hashemzadeh
- General and Vascular Surgery Department, Imam Reza Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
6
|
Chen Y, Maniakas A, Tan L, Cui M, Le X, Niedzielski JS, Michel KA, Harlan CJ, Lu W, Henderson YC, Mohamed ASR, Lorenzi PL, Putluri N, Bankson JA, Sandulache VC, Lai SY. Development of a rational strategy for integration of lactate dehydrogenase A suppression into therapeutic algorithms for head and neck cancer. Br J Cancer 2021; 124:1670-1679. [PMID: 33742144 PMCID: PMC8110762 DOI: 10.1038/s41416-021-01297-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 01/09/2021] [Accepted: 01/27/2021] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Lactate dehydrogenase (LDH) is a critical metabolic enzyme. LDH A (LDHA) overexpression is a hallmark of aggressive malignancies and has been linked to tumour initiation, reprogramming and progression in multiple tumour types. However, successful LDHA inhibition strategies have not materialised in the translational and clinical space. We sought to develop a rational strategy for LDHA suppression in the context of solid tumour treatment. METHODS We utilised a doxycycline-inducible short hairpin RNA (shRNA) system to generate LDHA suppression. Lactate and LDH activity levels were measured biochemically and kinetically using hyperpolarised 13C-pyruvate nuclear magnetic resonance spectroscopy. We evaluated effects of LDHA suppression on cellular proliferation and clonogenic survival, as well as on tumour growth, in orthotopic models of anaplastic thyroid carcinoma (ATC) and head and neck squamous cell carcinoma (HNSCC), alone or in combination with radiation. RESULTS shRNA suppression of LDHA generated a time-dependent decrease in LDH activity with transient shifts in intracellular lactate levels, a decrease in carbon flux from pyruvate into lactate and compensatory shifts in metabolic flux in glycolysis and the Krebs cycle. LDHA suppression decreased cellular proliferation and temporarily stunted tumour growth in ATC and HNSCC xenografts but did not by itself result in tumour cure, owing to the maintenance of residual viable cells. Only when chronic LDHA suppression was combined with radiation was a functional cure achieved. CONCLUSIONS Successful targeting of LDHA requires exquisite dose and temporal control without significant concomitant off-target toxicity. Combinatorial strategies with conventional radiation are feasible as long as the suppression is targeted, prolonged and non-toxic.
Collapse
Affiliation(s)
- Yunyun Chen
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anastasios Maniakas
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Hôpital Maisonneuve-Rosemont, University of Montreal, Montreal, QC, Canada
| | - Lin Tan
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Meng Cui
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Head Neck and Thyroid, Henan Cancer Hospital affiliated to Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Xiangdong Le
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joshua S Niedzielski
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Keith A Michel
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Collin J Harlan
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wuhao Lu
- Department of Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, TX, USA
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ying C Henderson
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Abdallah S R Mohamed
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Philip L Lorenzi
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nagireddy Putluri
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - James A Bankson
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vlad C Sandulache
- Department of Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, TX, USA.
| | - Stephen Y Lai
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
7
|
Salmani T, Ghaderian SMH, Hajiesmaeili M, Rezaeimirghaed O, Hoseini MS, Rakhshan A, Nasiri MJ, Ghaedi H, Akbarzadeh R. Hsa-miR-27a-3p and epidermal growth factor receptor expression analysis in glioblastoma FFPE samples. Asia Pac J Clin Oncol 2020; 17:e185-e190. [PMID: 33029912 DOI: 10.1111/ajco.13399] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 05/23/2020] [Indexed: 12/27/2022]
Abstract
AIM Glioblastoma multiforme (GBM) is the most invasive type of glial tumors. MicroRNAs as the small, noncoding RNAs have been revealed that play an important role in tumorigenic processes. So, they may be used as potential biomarkers for detection and prognosis of cancers at the early stages. In addition, they can be applied as therapeutic targets. In the present study, the expression levels of hsa-miR-27a-3p and EGFR were investigated in GBM. METHODS Real-time RT-PCR was applied to evaluate hsa-miR-27a-3p and EGFR expressions in the formalin-fixed paraffin-embedded (FFPE) tissue samples obtained from 50 GBM and 50 normal people. RESULTS The expression level of hsa-miR-27a-3p and EGFR was significantly different between cases and controls. Positive association was found between gene expressions and immunohistochemistry markers, such as Ki67 and glial fibrillary acidic protein, except for IDH1 status. CONCLUSION We showed the association of hsa-miR-27a-3p and EGFR with GBM and it can be concluded that they have a promising potential to use as primary cancer biomarkers.
Collapse
Affiliation(s)
- Tayyebali Salmani
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sayyed Mohammad Hossein Ghaderian
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Hajiesmaeili
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Omidvar Rezaeimirghaed
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Azadeh Rakhshan
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Javad Nasiri
- Department of Microbiology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Ghaedi
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Akbarzadeh
- Institute of Anatomy, University of Lübeck, Lübeck, Germany.,Skin Research Centre, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Chari NS, Ivan C, Le X, Li J, Mijiti A, Patel AA, Osman AA, Peterson CB, Williams MD, Pickering CR, Caulin C, Myers JN, Calin GA, Lai SY. Disruption of TP63-miR-27a* Feedback Loop by Mutant TP53 in Head and Neck Cancer. J Natl Cancer Inst 2020; 112:266-277. [PMID: 31124563 DOI: 10.1093/jnci/djz097] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 04/03/2019] [Accepted: 05/22/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Alterations in the epidermal growth factor receptor and PI3K pathways in head and neck squamous cell carcinomas (HNSCCs) are frequent events that promote tumor progression. Ectopic expression of the epidermal growth factor receptor-targeting microRNA (miR), miR-27a* (miR-27a-5p), inhibits tumor growth. We sought to identify mechanisms mediating repression of miR-27a* in HNSCC, which have not been previously identified. METHODS We quantified miR-27a* in 47 oral cavity squamous cell carcinoma patient samples along with analysis of miR-27a* in 73 oropharyngeal and 66 human papillomavirus-positive (HPV+) samples from The Cancer Genome Atlas. In vivo and in vitro TP53 models engineered to express mutant TP53, along with promoter analysis using chromatin immunoprecipitation and luciferase assays, were used to identify the role of TP53 and TP63 in miR-27a* transcription. An HNSCC cell line engineered to conditionally express miR-27a* was used in vitro to determine effects of miR-27a* on target genes and tumor cells. RESULTS miR-27a* expression was repressed in 47 oral cavity tumor samples vs matched normal tissue (mean log2 difference = -0.023, 95% confidence interval = -0.044 to -0.002; two-sided paired t test, P = .03), and low miR-27a* levels were associated with poor survival in HPV+ and oropharyngeal HNSCC samples. Binding of ΔNp63α to the promoter led to an upregulation of miR-27a*. In vitro and in vivo findings showed that mutant TP53 represses the miR-27a* promoter, downregulating miR-27a* levels. ΔNp63α and nucleoporin 62, a protein involved in ΔNP63α transport, were validated as novel targets of miR-27a*. CONCLUSION Our results characterize a negative feedback loop between TP63 and miR-27a*. Genetic alterations in TP53, a frequent event in HNSCC, disrupt this regulatory loop by repressing miR-27a* expression, promoting tumor survival.
Collapse
Affiliation(s)
- Nikhil S Chari
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Cristina Ivan
- Department of Experimental Therapeutics and The Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Xiandong Le
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jinzhong Li
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX.,Department of Oral and Maxillofacial-Head and Neck Oncology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - Ainiwaer Mijiti
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX.,Department of Stomatology, Shenzen Luohu People's Hospital, Shenzen, Guandong, China
| | - Ameeta A Patel
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Abdullah A Osman
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Christine B Peterson
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Michelle D Williams
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Curtis R Pickering
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Carlos Caulin
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX.,Department of Otolaryngology-Head and Neck Surgery, The University of Arizona and University of Arizona Cancer Center, Tucson, AZ
| | - Jeffrey N Myers
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - George A Calin
- Department of Experimental Therapeutics and The Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Stephen Y Lai
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX.,Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX.,Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
9
|
Harsha C, Banik K, Ang HL, Girisa S, Vikkurthi R, Parama D, Rana V, Shabnam B, Khatoon E, Kumar AP, Kunnumakkara AB. Targeting AKT/mTOR in Oral Cancer: Mechanisms and Advances in Clinical Trials. Int J Mol Sci 2020; 21:ijms21093285. [PMID: 32384682 PMCID: PMC7246494 DOI: 10.3390/ijms21093285] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/02/2020] [Accepted: 05/03/2020] [Indexed: 12/18/2022] Open
Abstract
Oral cancer (OC) is a devastating disease that takes the lives of lots of people globally every year. The current spectrum of treatment modalities does not meet the needs of the patients. The disease heterogeneity demands personalized medicine or targeted therapies. Therefore, there is an urgent need to identify potential targets for the treatment of OC. Abundant evidence has suggested that the components of the protein kinase B (AKT)/ mammalian target of rapamycin (mTOR) pathway are intrinsic factors for carcinogenesis. The AKT protein is central to the proliferation and survival of normal and cancer cells, and its downstream protein, mTOR, also plays an indispensable role in the cellular processes. The wide involvement of the AKT/mTOR pathway has been noted in oral squamous cell carcinoma (OSCC). This axis significantly regulates the various hallmarks of cancer, like proliferation, survival, angiogenesis, invasion, metastasis, autophagy, and epithelial-to-mesenchymal transition (EMT). Activated AKT/mTOR signaling is also associated with circadian signaling, chemoresistance and radio-resistance in OC cells. Several miRNAs, circRNAs and lncRNAs also modulate this pathway. The association of this axis with the process of tumorigenesis has culminated in the identification of its specific inhibitors for the prevention and treatment of OC. In this review, we discussed the significance of AKT/mTOR signaling in OC and its potential as a therapeutic target for the management of OC. This article also provided an update on several AKT/mTOR inhibitors that emerged as promising candidates for therapeutic interventions against OC/head and neck cancer (HNC) in clinical studies.
Collapse
Affiliation(s)
- Choudhary Harsha
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India; (C.H.); (K.B.); (S.G.); (R.V.); (D.P.); (V.R.); (B.S.); (E.K.)
| | - Kishore Banik
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India; (C.H.); (K.B.); (S.G.); (R.V.); (D.P.); (V.R.); (B.S.); (E.K.)
| | - Hui Li Ang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore;
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Sosmitha Girisa
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India; (C.H.); (K.B.); (S.G.); (R.V.); (D.P.); (V.R.); (B.S.); (E.K.)
| | - Rajesh Vikkurthi
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India; (C.H.); (K.B.); (S.G.); (R.V.); (D.P.); (V.R.); (B.S.); (E.K.)
| | - Dey Parama
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India; (C.H.); (K.B.); (S.G.); (R.V.); (D.P.); (V.R.); (B.S.); (E.K.)
| | - Varsha Rana
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India; (C.H.); (K.B.); (S.G.); (R.V.); (D.P.); (V.R.); (B.S.); (E.K.)
| | - Bano Shabnam
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India; (C.H.); (K.B.); (S.G.); (R.V.); (D.P.); (V.R.); (B.S.); (E.K.)
| | - Elina Khatoon
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India; (C.H.); (K.B.); (S.G.); (R.V.); (D.P.); (V.R.); (B.S.); (E.K.)
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore;
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
- Correspondence: (A.P.K.); (A.B.K.); Tel.: +65-6516-5456 (A.P.K.); +91-361-258-2231 (A.B.K.); Fax: +65-6873-9664 (A.P.K.); +91-361-258-2249 (A.B.K.)
| | - Ajaikumar B. Kunnumakkara
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India; (C.H.); (K.B.); (S.G.); (R.V.); (D.P.); (V.R.); (B.S.); (E.K.)
- Correspondence: (A.P.K.); (A.B.K.); Tel.: +65-6516-5456 (A.P.K.); +91-361-258-2231 (A.B.K.); Fax: +65-6873-9664 (A.P.K.); +91-361-258-2249 (A.B.K.)
| |
Collapse
|
10
|
Maximov VV, Akkawi R, Khawaled S, Salah Z, Jaber L, Barhoum A, Or O, Galasso M, Kurek KC, Yavin E, Aqeilan RI. MiR-16-1-3p and miR-16-2-3p possess strong tumor suppressive and antimetastatic properties in osteosarcoma. Int J Cancer 2019; 145:3052-3063. [PMID: 31018244 DOI: 10.1002/ijc.32368] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 03/18/2019] [Accepted: 04/10/2019] [Indexed: 01/07/2023]
Abstract
Osteosarcoma (OS) is an aggressive malignancy affecting mostly children and adolescents. MicroRNAs (miRNAs) play important roles in OS development and progression. Here we found that miR-16-1-3p and miR-16-2-3p "passenger" strands, as well as the "lead" miR-16-5p strand, are frequently downregulated and possess strong tumor suppressive functions in human OS. Furthermore, we report different although strongly overlapping functions for miR-16-1-3p and miR-16-2-3p in OS cells. Ectopic expression of these miRNAs affected primary tumor growth, metastasis seeding and chemoresistance and invasiveness of human OS cells. Loss-of-function experiments verified tumor suppressive functions of these miRNAs at endogenous levels of expression. Using RNA immunoprecipitation (RIP) assays, we identify direct targets of miR-16-1-3p and miR-16-2-3p in OS cells. Moreover, validation experiments identified FGFR2 as a direct target for miR-16-1-3p and miR-16-2-3p. Overall, our findings underscore the importance of passenger strand miRNAs, at least some, in osteosarcomagenesis.
Collapse
Affiliation(s)
- Vadim V Maximov
- The Lautenberg Center for General and Tumor Immunology, Department of Immunology and Cancer Research-IMRIC, The Hebrew University-Hadassah Medical School at Ein-Kerem, Jerusalem, Israel
| | - Rania Akkawi
- The Lautenberg Center for General and Tumor Immunology, Department of Immunology and Cancer Research-IMRIC, The Hebrew University-Hadassah Medical School at Ein-Kerem, Jerusalem, Israel
| | - Saleh Khawaled
- The Lautenberg Center for General and Tumor Immunology, Department of Immunology and Cancer Research-IMRIC, The Hebrew University-Hadassah Medical School at Ein-Kerem, Jerusalem, Israel
| | - Zaidoun Salah
- Al Quds-Bard College, Al-Quds University, Al-Bireh, East Jerusalem, Palestine
| | - Lina Jaber
- The Lautenberg Center for General and Tumor Immunology, Department of Immunology and Cancer Research-IMRIC, The Hebrew University-Hadassah Medical School at Ein-Kerem, Jerusalem, Israel
| | - Ahlam Barhoum
- The Lautenberg Center for General and Tumor Immunology, Department of Immunology and Cancer Research-IMRIC, The Hebrew University-Hadassah Medical School at Ein-Kerem, Jerusalem, Israel
| | - Omer Or
- Department of Orthopedics Surgery, Hebrew University Hadassah Medical Center, Jerusalem, Israel
| | - Marco Galasso
- Biosystems Analysis, LTTA, Department of Morphology, Surgery and Experimental Medicine, Università degli Studi, Ferrara, Italy
| | - Kyle C Kurek
- Department of Pathology and Medical Genetics Cumming School of Medicine, University of Calgary, Alberta Children's Hospital & Research Institute, Calgary, AB, Canada
| | - Eylon Yavin
- The Institute for Drug Research, The School of Pharmacy, The Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Rami I Aqeilan
- The Lautenberg Center for General and Tumor Immunology, Department of Immunology and Cancer Research-IMRIC, The Hebrew University-Hadassah Medical School at Ein-Kerem, Jerusalem, Israel.,Department of Cancer Biology & Genetics, The Ohio State University Wexner Medical Center, Columbus, OH
| |
Collapse
|
11
|
Tang H, Xu X, Xiao W, Liao Y, Xiao X, Li L, Li K, Jia X, Feng H. Silencing of microRNA-27a facilitates autophagy and apoptosis of melanoma cells through the activation of the SYK-dependent mTOR signaling pathway. J Cell Biochem 2019; 120:13262-13274. [PMID: 30994959 DOI: 10.1002/jcb.28600] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 02/11/2019] [Indexed: 12/19/2022]
Abstract
Melanoma is considered as an aggressive neoplastic transformation and featured with high metastatic potential. Although some studies have provided targets for novel therapeutic interventions, clinical development of targeted drugs for melanoma still remains obscure. Therefore, this study aims to identify the role of microRNA-27a (miR-27a) in autophagy and apoptosis of melanoma cells in regulating spleen tyrosine kinase (SYK)-mediated the mammalian target of rapamycin (mTOR) signaling pathway. A microarray-based analysis was made to screen differentially expressed genes and predict target miRNA. Melanoma specimens were collected with pigmented nevus as a control. Melanoma cell line Mel-RM was treated with miR-27a inhibitor or pcDNA-SYK to prove their effects on autophagy and apoptosis of melanoma cells. The volume change and tumor mass of nude mice in each group were detected by the tumorigenesis assay. Microarray-based analysis results showed that SYK was lowly expressed in melanoma cells and may be regulated by miR-27a. Besides, miR-27a expression was increased whereas SYK expression was decreased in melanoma tissues. Meanwhile, miR-27a was positively correlated with tumor stage and lymph node metastasis of melanoma tissues. Furthermore, miR-27a targeted SYK and silencing of miR-27a or overexpression of SYK cells promoted autophagy and apoptosis of melanoma cells and reduced their tumorigenic ability in vivo. In conclusion, this study proves that silencing of miR-27a facilitates autophagy and apoptosis of melanoma cells by upregulating SYK expression and activating the mTOR signaling pathway. The finding offers new ideas for the clinical development of melanoma.
Collapse
Affiliation(s)
- Hua Tang
- Department of Dermatology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, P.R. China
| | - Xiaopeng Xu
- Department of Dermatology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, P.R. China
| | - Weirong Xiao
- Department of Dermatology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, P.R. China
| | - Yangying Liao
- Department of Dermatology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, P.R. China
| | - Xiao Xiao
- Department of Dermatology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, P.R. China
| | - Lan Li
- Department of Dermatology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, P.R. China
| | - Ke Li
- Department of Dermatology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, P.R. China
| | - Xiaomin Jia
- Department of Pathology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, P.R. China
| | - Hao Feng
- Department of Dermatology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, P.R. China
| |
Collapse
|
12
|
Wang XH, Yao DX, Luan XS, Wang Y, Liu HX, Liu B, Liu Y, Zhao L, Ji XM, Wang TL. MicroRNA expression in the hippocampal CA1 region under deep hypothermic circulatory arrest. Neural Regen Res 2019; 14:2003-2010. [PMID: 31290459 PMCID: PMC6676878 DOI: 10.4103/1673-5374.253174] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Using deep hypothermic circulatory arrest, thoracic aorta diseases and complex heart diseases can be subjected to corrective procedures. However, mechanisms underlying brain protection during deep hypothermic circulatory arrest are unclear. After piglet models underwent 60 minutes of deep hypothermic circulatory arrest at 14°C, expression of microRNAs (miRNAs) was analyzed in the hippocampus by microarray. Subsequently, TargetScan 6.2, RNA22 v2.0, miRWalk 2.0, and miRanda were used to predict potential targets, and gene ontology enrichment analysis was carried out to identify functional pathways involved. Quantitative reverse transcription-polymerase chain reaction was conducted to verify miRNA changes. Deep hypothermic circulatory arrest altered the expression of 35 miRNAs. Twenty-two miRNAs were significantly downregulated and thirteen miRNAs were significantly upregulated in the hippocampus after deep hypothermic circulatory arrest. Six out of eight targets among the differentially expressed miRNAs were enriched for neuronal projection (cyclin dependent kinase, CDK16 and SLC1A2), central nervous system development (FOXO3, TYRO3, and SLC1A2), ion transmembrane transporter activity (ATP2B2 and SLC1A2), and interleukin-6 receptor binding (IL6R) – these are the key functional pathways involved in cerebral protection during deep hypothermic circulatory arrest. Quantitative reverse transcription-polymerase chain reaction confirmed the results of microarray analysis. Our experimental results illustrate a new role for transcriptional regulation in deep hypothermic circulatory arrest, and provide significant insight for the development of miRNAs to treat brain injuries. All procedures were approved by the Animal Care Committee of Xuanwu Hospital, Capital Medical University, China on March 1, 2017 (approval No. XW-INI-AD2017-0112).
Collapse
Affiliation(s)
- Xiao-Hua Wang
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University; Institute of Geriatrics; National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Dong-Xu Yao
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University; Institute of Geriatrics; National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Xiu-Shu Luan
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University; Institute of Geriatrics; National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Yu Wang
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University; Institute of Geriatrics; National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Hai-Xia Liu
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University; Institute of Geriatrics; National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Bei Liu
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University; Institute of Geriatrics; National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Yang Liu
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University; Institute of Geriatrics; National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Lei Zhao
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University; Institute of Geriatrics; National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Xun-Ming Ji
- Department of Neurosurgery; Cerebrovascular Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Tian-Long Wang
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University; Institute of Geriatrics; National Clinical Research Center for Geriatric Disorders, Beijing, China
| |
Collapse
|
13
|
Álvarez-Díaz DA, Gutiérrez-Díaz AA, Orozco-García E, Puerta-González A, Bermúdez-Santana CI, Gallego-Gómez JC. Dengue virus potentially promotes migratory responses on endothelial cells by enhancing pro-migratory soluble factors and miRNAs. Virus Res 2018; 259:68-76. [PMID: 30367889 DOI: 10.1016/j.virusres.2018.10.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 10/21/2018] [Accepted: 10/23/2018] [Indexed: 12/31/2022]
Abstract
The most life-threatening effect of the Dengue virus (DENV) infection is an acute destabilization of the microvascular endothelial cell (MEC) barrier leading to plasma leakage, hypovolemic shock and haemorrhage. However, the underlying cellular mechanisms responsible for the dysfunction of MECs are not well understood. To identify potential cellular processes altered during DENV infection of MECs, expression profiles of cytokines/growth factors and microRNAs were measured by Luminex assay and next generation sequencing, respectively. Synchronously DENV2-infected MECs increase the secretion of IL-6, IL-8, FGF-2, GM-CSF, G-CSF, TGF-α, GRO, RANTES, MCP-1 and MCP-3. Conditioned media of infected MECs increased the migration of non-infected MECs. Furthermore, six miRNAs deregulated at 24 hpi were predicted to regulate host genes involved in cell migration and vascular developmental processes such as angiogenesis. These in silico analyses provide insights that support that DENV promotes an acute migratory phenotype in MECs that contributes to the vascular destabilization observed in severe dengue cases.
Collapse
Affiliation(s)
- Diego Alejandro Álvarez-Díaz
- Grupo Medicina Molecular y de Translación - Facultad de Medicina, Universidad de Antioquia, Medellín, 050010, Colombia.
| | - Aimer Alonso Gutiérrez-Díaz
- RNómica Teórica y Computacional - Facultad de Ciencias, Universidad Nacional de Colombia, Bogotá, 111321, Colombia.
| | - Elizabeth Orozco-García
- Grupo Medicina Molecular y de Translación - Facultad de Medicina, Universidad de Antioquia, Medellín, 050010, Colombia.
| | - Andrés Puerta-González
- Grupo Medicina Molecular y de Translación - Facultad de Medicina, Universidad de Antioquia, Medellín, 050010, Colombia; RNómica Teórica y Computacional - Facultad de Ciencias, Universidad Nacional de Colombia, Bogotá, 111321, Colombia.
| | | | - Juan Carlos Gallego-Gómez
- Grupo Medicina Molecular y de Translación - Facultad de Medicina, Universidad de Antioquia, Medellín, 050010, Colombia.
| |
Collapse
|
14
|
Barros-Silva D, Costa-Pinheiro P, Duarte H, Sousa EJ, Evangelista AF, Graça I, Carneiro I, Martins AT, Oliveira J, Carvalho AL, Marques MM, Henrique R, Jerónimo C. MicroRNA-27a-5p regulation by promoter methylation and MYC signaling in prostate carcinogenesis. Cell Death Dis 2018; 9:167. [PMID: 29415999 PMCID: PMC5833437 DOI: 10.1038/s41419-017-0241-y] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 12/10/2017] [Accepted: 12/18/2017] [Indexed: 12/14/2022]
Abstract
Upregulation of MYC and miRNAs deregulation are common in prostate cancer (PCa). Overactive MYC may cause miRNAs’ expression deregulation through transcriptional and post-transcriptional mechanisms and epigenetic alterations are also involved in miRNAs dysregulation. Herein, we aimed to elucidate the role of regulatory network between MYC and miRNAs in prostate carcinogenesis. MYC expression was found upregulated in PCa cases and matched precursor lesions. MicroRNA’s microarray analysis of PCa samples with opposed MYC levels identified miRNAs significantly overexpressed in high-MYC PCa. However, validation of miR-27a-5p in primary prostate tissues disclosed downregulation in PCa, instead, correlating with aberrant promoter methylation. In a series of castration-resistant PCa (CRPC) cases, miR-27a-5p was upregulated, along with promoter hypomethylation. MYC and miR-27a-5p expression levels in LNCaP and PC3 cells mirrored those observed in hormone-naíve PCa and CRPC, respectively. ChIP analysis showed that miR-27a-5p expression is only regulated by c-Myc in the absence of aberrant promoter methylation. MiR-27a-5p knockdown in PC3 cells promoted cell growth, whereas miRNA forced expression in LNCaP and stable MYC-knockdown PC3 cells attenuated the malignant phenotype, suggesting a tumor suppressive role for miR-27a-5p. Furthermore, miR-27a-5p upregulation decreased EGFR/Akt1/mTOR signaling. We concluded that miR-27a-5p is positively regulated by MYC, and its silencing due to aberrant promoter methylation occurs early in prostate carcinogenesis, concomitantly with loss of MYC regulatory activity. Our results further suggest that along PCa progression, miR-27a-5p promoter becomes hypomethylated, allowing for MYC to resume its regulatory activity. However, the altered cellular context averts miR-27a-5p from successfully accomplishing its tumor suppressive function at this stage of disease.
Collapse
Affiliation(s)
- Daniela Barros-Silva
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal.,Master in Oncology, Institute of Biomedical Sciences Abel Salazar-University of Porto (ICBAS-UP), Porto, Portugal
| | - Pedro Costa-Pinheiro
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal
| | - Henrique Duarte
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal
| | - Elsa Joana Sousa
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal
| | | | - Inês Graça
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal
| | - Isa Carneiro
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal.,Master in Oncology, Institute of Biomedical Sciences Abel Salazar-University of Porto (ICBAS-UP), Porto, Portugal
| | - Ana Teresa Martins
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal.,Master in Oncology, Institute of Biomedical Sciences Abel Salazar-University of Porto (ICBAS-UP), Porto, Portugal
| | - Jorge Oliveira
- Department of Urology, Portuguese Oncology Institute of Porto (IPO Porto), Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal
| | - André L Carvalho
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil
| | - Márcia M Marques
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil.,Barretos School of Health Sciences, Barretos, São Paulo, Brazil
| | - Rui Henrique
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal.,Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal.,Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar (ICBAS)-University of Porto, Porto, Portugal
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal. .,Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar (ICBAS)-University of Porto, Porto, Portugal.
| |
Collapse
|
15
|
Masliah-Planchon J, Garinet S, Pasmant E. RAS-MAPK pathway epigenetic activation in cancer: miRNAs in action. Oncotarget 2018; 7:38892-38907. [PMID: 26646588 PMCID: PMC5122439 DOI: 10.18632/oncotarget.6476] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Accepted: 11/22/2015] [Indexed: 01/17/2023] Open
Abstract
The highly conserved RAS-mitogen activated protein kinase (MAPK) signaling pathway is involved in a wide range of cellular processes including differentiation, proliferation, and survival. Somatic mutations in genes encoding RAS-MAPK components frequently occur in many tumors, making the RAS-MAPK a critical pathway in human cancer. Since the pioneering study reporting that let-7 miRNA acted as tumor suppressor by repressing the RAS oncogene, growing evidence has suggested the importance of miRNAs targeting the RAS-MAPK in oncogenesis. MiRNAs alterations in human cancers may act as a rheostat of the oncogenic RAS signal that is often amplified as cancers progress. However, specific mechanisms leading to miRNAs deregulation and their functional consequences in cancer are far from being fully elucidated. In this review, we provide an experimental-validated map of RAS-MAPK oncomiRs and tumor suppressor miRNAs from transmembrane receptor to downstream ERK proteins. MiRNAs could be further considered as potential genetic biomarkers for diagnosis, prognosis, or therapeutic purpose.
Collapse
Affiliation(s)
- Julien Masliah-Planchon
- Unité de Génétique Somatique, Département de Génétique Oncologique, Institut Curie, Paris, France.,INSERM_U830, Institut Curie, Paris, France
| | - Simon Garinet
- Service de Biochimie et Génétique Moléculaire, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Eric Pasmant
- Service de Biochimie et Génétique Moléculaire, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris, France.,EA7331, Université Paris Descartes, Sorbonne Paris Cité, Faculté des Sciences Pharmaceutiques et Biologiques, Paris, France
| |
Collapse
|
16
|
Weak Regulation of Many Targets Is Cumulatively Powerful—An Evolutionary Perspective on microRNA Functionality. Mol Biol Evol 2017; 34:3041-3046. [DOI: 10.1093/molbev/msx260] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
17
|
Chang YJ, Tseng CY, Lin PY, Chuang YC, Chao MW. Acute exposure to DEHP metabolite, MEHP cause genotoxicity, mutagenesis and carcinogenicity in mammalian Chinese hamster ovary cells. Carcinogenesis 2017; 38:336-345. [PMID: 28426879 DOI: 10.1093/carcin/bgx009] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 01/15/2017] [Indexed: 12/20/2022] Open
Abstract
Di-(2-ethylhexyl) phthalate (DEHP), the common plasticizer used in the production of polyvinyl chloride, can be converted to the more potent metabolite mono-ethylhexyl phthalate (MEHP). Epidemiological studies have shown an association with elevated induction of rat hepatic cancer and reproductive toxicity in response to MEHP exposure. However, the mechanism of genotoxicity and carcinogenicity induced by MEHP treatment remains unclear. As a means to elucidate the mechanisms of action, lethality and mutagenicity in the adenine phosphoribosyltransferase (aprt+/-) gene induced in several CHO cell types by MEHP were assessed. Dose-response relationships were determined in the parental AA8 cell line, its nucleotide repair-deficient UV5 and base repair-deficient EM9 subclones, and also in AS52 cells harboring the bacterial guanine-hypoxanthine phosphoribosyltransferase (gpt) gene and its derived AS52-XPD-knockdown and AS52-PARP-1-knockdown cells. Treatment of AS52 with MEHP led to intracellular production of reactive oxygen species (ROS) and DNA strand breaks in a dose-dependent manner. Separately, mutations in the gpt gene of AS52 cells were characterized and found to be dominated by G:C to A:T and A:T to G:C transitions. Independent AS52-mutant cell (ASMC) clones were collected for the sequential in vivo xenograft tumorigenic studies, 4 of total 20 clones had aggressive tumor growth. Moreover, microarray analysis indicated miR-let-7a and miR-125b downregulated in ASMC, which might raise oncogenic MYC and RAS level and activate ErbB pathway. Comparative evaluation of the results indicates that the principal mechanism of this mutagenic action is probably to be through generation of ROS, causing base excision damage resulting in carcinogenicity.
Collapse
Affiliation(s)
- Yu-Jung Chang
- Department of Bioscience Technology, College of Science
| | - Chia-Yi Tseng
- Department of Biomedical Engineering, College of Engineering and.,Center of Nanotechnology, Chung Yuan Christian University, Taoyuan 32023, Taiwan
| | - Pei-Ying Lin
- Department of Bioscience Technology, College of Science
| | - Yu-Chen Chuang
- Department of Biomedical Engineering, College of Engineering and
| | - Ming-Wei Chao
- Department of Bioscience Technology, College of Science.,Center of Nanotechnology, Chung Yuan Christian University, Taoyuan 32023, Taiwan
| |
Collapse
|
18
|
Mizuno K, Mataki H, Arai T, Okato A, Kamikawaji K, Kumamoto T, Hiraki T, Hatanaka K, Inoue H, Seki N. The microRNA expression signature of small cell lung cancer: tumor suppressors of miR-27a-5p and miR-34b-3p and their targeted oncogenes. J Hum Genet 2017; 62:671-678. [DOI: 10.1038/jhg.2017.27] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 02/02/2017] [Accepted: 02/07/2017] [Indexed: 12/12/2022]
|
19
|
Li J, Zhao Y, Lu Y, Ritchie W, Grau G, Vadas MA, Gamble JR. The Poly-cistronic miR-23-27-24 Complexes Target Endothelial Cell Junctions: Differential Functional and Molecular Effects of miR-23a and miR-23b. MOLECULAR THERAPY. NUCLEIC ACIDS 2016; 5:e354. [PMID: 27741223 PMCID: PMC5023406 DOI: 10.1038/mtna.2016.62] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 06/20/2016] [Indexed: 01/09/2023]
Abstract
The regulation of function of endothelial cell-cell junctions is fundamental in sustaining vascular integrity. The polycistronic microRNA (miR) complexes containing miR-23a-27a-24-2, and 23b-27b-24-1 are predicted to target the majority of major endothelial junctional proteins. We focus on miR-23a and miR-23b, and investigate the functional effects of these miRs on junctions. While miR-23a and 23b only differ by 1 nucleotide (g19) outside the seed region and thus are predicted to have the same targets, they function differently with miR-23a inhibiting permeability and miR-23b inhibiting angiogenesis. Both miRs target the junctional attractive molecule (tight junction protein 2) ZO-2 and the repulsive molecule junctional adhesion molecule C (JAM-C), although the inhibition of JAM-C by miR-23a is more profound than by miR-23b. The difference in potency is attributable to differences at g19 since a mutation of the t17, the g19 binding site of miR-23b in the 3'UTR of JAM-C restores identity. We also show that the pattern of expression of miR-23a and miR-23b and their targets are different. Thus, the paralogues miR-23a and miR-23b can have profoundly different effects on endothelial cell function due at least partially to selective effects on target proteins and differences in expression patterns of the miRs. This work exposes a hitherto unappreciated complexity in therapeutically targeting miRs.
Collapse
Affiliation(s)
- Jia Li
- Centre for the Endothelium, Vascular Biology Program, Centenary Institute, University of Sydney, Sydney, Australia
| | - Yang Zhao
- Centre for the Endothelium, Vascular Biology Program, Centenary Institute, University of Sydney, Sydney, Australia
| | - Ying Lu
- Centre for the Endothelium, Vascular Biology Program, Centenary Institute, University of Sydney, Sydney, Australia
| | - William Ritchie
- Bioinformatics Laboratory, Centenary Institute, University of Sydney, Sydney, Australia
| | - Georges Grau
- Department of Pathology, Faculty of Medicine, School of Medical Sciences, University of Sydney, Sydney, Australia
| | - Mathew A Vadas
- Centre for the Endothelium, Vascular Biology Program, Centenary Institute, University of Sydney, Sydney, Australia
| | - Jennifer R Gamble
- Centre for the Endothelium, Vascular Biology Program, Centenary Institute, University of Sydney, Sydney, Australia
| |
Collapse
|
20
|
Tormo E, Pineda B, Serna E, Guijarro A, Ribas G, Fores J, Chirivella E, Climent J, Lluch A, Eroles P. MicroRNA Profile in Response to Doxorubicin Treatment in Breast Cancer. J Cell Biochem 2016; 116:2061-73. [PMID: 25802200 DOI: 10.1002/jcb.25162] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 03/10/2015] [Indexed: 12/16/2022]
Abstract
UNLABELLED Chemotherapy treatment is the standard in triple negative breast cancers, a cancer subgroup which lacks a specific target. The mechanisms leading to the response, as well as any markers that allow the differentiation between responder and non-responder groups prior to treatment are unknown. In parallel, miRNAs can act as oncogenes or tumor suppressors and there is evidence of their involvement in promoting resistance to anticancer drugs. Therefore we hypothesized that changes in miRNA expression after doxorubicin treatment may also be relevant in treatment response. OBJECTIVE To study miRNAs that are differentially expressed in response to doxorubicin treatment. METHODS One luminal-A and two triple negative, breast cancer cell lines were exposed to doxorubicin. Microarray analysis was performed to identify the common and differentially modified miRNAs. Genes and pathways that are theoretically regulated by these miRNAs were analyzed. RESULTS Thirteen miRNAs common to all three lines were modified, in addition to 25 that were specific to triple negative cell lines, and 69 that changed only in the luminal-A cell line. This altered expression pattern seemed to be more strongly related to the breast cancer subgroup than to the treatment. The analysis of target genes revealed that cancer related pathways were the most affected by these miRNAs, moreover many of them had been previously related to chemotherapy resistance; thus suggesting follow-up studies. Additionally, through functional assays, we showed that miR-548c-3p is implicated in doxorubicin-treated MCF-7 cell viability, suggesting a role for this miRNA in resistance.
Collapse
Affiliation(s)
- Eduardo Tormo
- INCLIVA Biomedical Research Institute, Valencia, Spain
| | - Begoña Pineda
- INCLIVA Biomedical Research Institute, Valencia, Spain
| | - Eva Serna
- Unidad Central de, Investigación en Medicina-INCLIVA, Universitat de Valencia, Valencia, Spain
| | - Alba Guijarro
- INCLIVA Biomedical Research Institute, Valencia, Spain
| | - Gloria Ribas
- INCLIVA Biomedical Research Institute, Valencia, Spain
| | - Jaume Fores
- INCLIVA Biomedical Research Institute, Valencia, Spain
| | | | - Joan Climent
- INCLIVA Biomedical Research Institute, Valencia, Spain
| | - Ana Lluch
- INCLIVA Biomedical Research Institute, Valencia, Spain.,Department of Hematology and Medical Oncology, Hospital Clínico Universitario de, Valencia, Spain
| | - Pilar Eroles
- INCLIVA Biomedical Research Institute, Valencia, Spain
| |
Collapse
|
21
|
Che H, Yan Y, Kang XH, Guo F, Yan ML, Liu HL, Hou X, Liu T, Zong DK, Sun LL, Bao YN, Sun LH, Yang BF, Ai J. MicroRNA-27a Promotes Inefficient Lysosomal Clearance in the Hippocampi of Rats Following Chronic Brain Hypoperfusion. Mol Neurobiol 2016; 54:2595-2610. [DOI: 10.1007/s12035-016-9856-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Accepted: 03/11/2016] [Indexed: 12/21/2022]
|
22
|
Yang S, Krug SM, Heitmann J, Hu L, Reinhold AK, Sauer S, Bosten J, Sommer C, Fromm M, Brack A, Rittner HL. Analgesic drug delivery via recombinant tissue plasminogen activator and microRNA-183-triggered opening of the blood-nerve barrier. Biomaterials 2016; 82:20-33. [DOI: 10.1016/j.biomaterials.2015.11.053] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 11/15/2015] [Accepted: 11/29/2015] [Indexed: 01/07/2023]
|
23
|
miR-27a and miR-27a* contribute to metastatic properties of osteosarcoma cells. Oncotarget 2016; 6:4920-35. [PMID: 25749032 PMCID: PMC4467124 DOI: 10.18632/oncotarget.3025] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 01/01/2015] [Indexed: 12/20/2022] Open
Abstract
Osteosarcoma (OS) is the most common primary malignant bone tumor in adolescents and young adults. The essential mechanisms underlying osteosarcomagenesis and progression continue to be obscure. MicroRNAs (miRNAs) have far-reaching effects on the cellular biology of development and cancer. We recently reported that unique miRNA signatures associate with the pathogenesis and progression of OS. Of particular interest, we found that higher expression of miR-27a is associated with clinical metastatic disease. We report here that overexpression of miR-27a/miR-27a*, a microRNA pair derived from a single precursor, promotes pulmonary OS metastases formation. By contrast, sequestering miR-27a/miR-27a* by sponge technology suppressed OS cells invasion and metastases formation. miR-27a/miR-27a* directly repressed CBFA2T3 expression among other target genes. We demonstrated that CBFA2T3 is downregulated in majority of OS samples and its over expression significantly attenuated OS metastatic process mediated by miR-27a/miR-27a* underscoring CBFA2T3 functions as a tumor suppressor in OS. These findings establish that miR-27a/miR-27a* pair plays a significant role in OS metastasis and proposes it as a potential diagnostic and therapeutic target in managing OS metastases.
Collapse
|
24
|
Yeh LY, Liu CJ, Wong YK, Chang C, Lin SC, Chang KW. miR-372 inhibits p62 in head and neck squamous cell carcinoma in vitro and in vivo. Oncotarget 2016; 6:6062-75. [PMID: 25714028 PMCID: PMC4467422 DOI: 10.18632/oncotarget.3340] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 01/05/2015] [Indexed: 01/18/2023] Open
Abstract
Here we showed that exogenous miR-372 expression and knockdown of p62 (sequestosome1 or SQSTM1), both increased migration of head and neck squamous cell carcinoma (HNSCC) cells. p62 induced phase II detoxification enzyme NADPH quinone oxidoreductase 1 (NQO1), which decreased ROS levels and cell migration. Also, miR-372 decreased p62 during hypoxia, thus increasing cell migration. Levels of miR-372 and p62 inversely correlated in human HNSCC tissues. Plasma levels of miR-372 was associated with advanced tumor stage and patient mortality. Both plasma and salivary miR-372 levels were decreased after tumor resection. We conclude that miR-372 decreases p62, thus increasing ROS and motility in HNSCC cells.
Collapse
Affiliation(s)
- Li-Yin Yeh
- Institute of Oral Biology, National Yang-Ming University, Taipei, Taiwan
| | - Chung-Ji Liu
- Department of Dentistry, National Yang-Ming University, Taipei, Taiwan.,Department of Dentistry, Taipei Mackay Memorial Hospital, Taipei, Taiwan
| | - Yong-Kie Wong
- Department of Dentistry, National Yang-Ming University, Taipei, Taiwan.,Department of Dentistry, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Christine Chang
- Institute of Oral Biology, National Yang-Ming University, Taipei, Taiwan
| | - Shu-Chun Lin
- Institute of Oral Biology, National Yang-Ming University, Taipei, Taiwan.,Department of Dentistry, National Yang-Ming University, Taipei, Taiwan.,Department of Stomatology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Kuo-Wei Chang
- Institute of Oral Biology, National Yang-Ming University, Taipei, Taiwan.,Department of Dentistry, National Yang-Ming University, Taipei, Taiwan.,Department of Stomatology, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
25
|
MicroRNA-873 mediates multidrug resistance in ovarian cancer cells by targeting ABCB1. Tumour Biol 2016; 37:10499-506. [DOI: 10.1007/s13277-016-4944-y] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 01/29/2016] [Indexed: 01/08/2023] Open
|
26
|
Janiszewska J, Szaumkessel M, Kostrzewska-Poczekaj M, Bednarek K, Paczkowska J, Jackowska J, Grenman R, Szyfter K, Wierzbicka M, Giefing M, Jarmuz-Szymczak M. Global miRNA Expression Profiling Identifies miR-1290 as Novel Potential oncomiR in Laryngeal Carcinoma. PLoS One 2015; 10:e0144924. [PMID: 26694163 PMCID: PMC4692263 DOI: 10.1371/journal.pone.0144924] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 10/21/2015] [Indexed: 12/30/2022] Open
Abstract
Background Laryngeal squamous cell carcinoma (LSCC) is the most common group among head and neck cancers. LSCC is characterized by a high incidence in Europe. With the aim of better understanding its genetic background we performed global miRNA expression profiling of LSCC cell lines and primary specimens. By this approach we identified a cohort of 33 upregulated and 9 downregulated miRNA genes in LSCC as compared to epithelial no tumor controls. Results Within this group we identified overexpression of the novel miR-1290 gene not reported in the context of LSCC before. Using a combined bioinformatical approach in connection with functional analysis we delineated two putative target genes of miR-1290 namely ITPR2 and MAF which are significantly downregulated in LSCC. They are interesting candidates for tumor suppressor genes as they are implicated in apoptosis and other processes deregulated in cancer. Conclusion Taken together, we propose miR-1290 as the new oncomiR involved in LSCC pathogenesis. Additionally, we suggest that the oncogenic potential of miR-1290 might be expressed by the involvement in downregulation of its target genes MAF and ITPR2.
Collapse
Affiliation(s)
- Joanna Janiszewska
- Institute of Human Genetics, Polish Academy of Sciences, Department of Cancer Genetics, Poznan, Poland
- * E-mail:
| | - Marcin Szaumkessel
- Institute of Human Genetics, Polish Academy of Sciences, Department of Cancer Genetics, Poznan, Poland
| | | | - Kinga Bednarek
- Institute of Human Genetics, Polish Academy of Sciences, Department of Cancer Genetics, Poznan, Poland
| | - Julia Paczkowska
- Institute of Human Genetics, Polish Academy of Sciences, Department of Cancer Genetics, Poznan, Poland
| | - Joanna Jackowska
- Department of Otolaryngology and Laryngological Oncology, University of Medical Sciences, Poznan, Poland
| | - Reidar Grenman
- Department of Otorhinolaryngology, Head and Neck Surgery and Department of Medical Biochemistry, Turku University Hospital and University of Turku, Turku, Finland
| | - Krzysztof Szyfter
- Institute of Human Genetics, Polish Academy of Sciences, Department of Cancer Genetics, Poznan, Poland
- Department of Audiology and Phoniatry, University of Medical Sciences, Poznan, Poland
| | - Malgorzata Wierzbicka
- Department of Otolaryngology and Laryngological Oncology, University of Medical Sciences, Poznan, Poland
| | - Maciej Giefing
- Institute of Human Genetics, Polish Academy of Sciences, Department of Cancer Genetics, Poznan, Poland
- Department of Otolaryngology and Laryngological Oncology, University of Medical Sciences, Poznan, Poland
| | - Malgorzata Jarmuz-Szymczak
- Institute of Human Genetics, Polish Academy of Sciences, Department of Cancer Genetics, Poznan, Poland
- Department of Hematology, K. Marcinkowski University of Medical Sciences, Poznan, Poland
| |
Collapse
|
27
|
MicroRNA-92b promotes tumor growth and activation of NF-κB signaling via regulation of NLK in oral squamous cell carcinoma. Oncol Rep 2015; 34:2961-8. [DOI: 10.3892/or.2015.4323] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 07/23/2015] [Indexed: 11/05/2022] Open
|
28
|
MicroRNAs and Growth Factors: An Alliance Propelling Tumor Progression. J Clin Med 2015; 4:1578-99. [PMID: 26287249 PMCID: PMC4555078 DOI: 10.3390/jcm4081578] [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: 07/02/2015] [Revised: 07/30/2015] [Accepted: 07/31/2015] [Indexed: 02/06/2023] Open
Abstract
Tumor progression requires cancer cell proliferation, migration, invasion, and attraction of blood and lymph vessels. These processes are tightly regulated by growth factors and their intracellular signaling pathways, which culminate in transcriptional programs. Hence, oncogenic mutations often capture growth factor signaling, and drugs able to intercept the underlying biochemical routes might retard cancer spread. Along with messenger RNAs, microRNAs play regulatory roles in growth factor signaling and in tumor progression. Because growth factors regulate abundance of certain microRNAs and the latter modulate the abundance of proteins necessary for growth factor signaling, the two classes of molecules form a dense web of interactions, which are dominated by a few recurring modules. We review specific examples of the alliance formed by growth factors and microRNAs and refer primarily to the epidermal growth factor (EGF) pathway. Clinical applications of the crosstalk between microRNAs and growth factors are described, including relevance to cancer therapy and to emergence of resistance to specific drugs.
Collapse
|
29
|
Quantitative Analysis of MicroRNAs in Vaccinia virus Infection Reveals Diversity in Their Susceptibility to Modification and Suppression. PLoS One 2015; 10:e0131787. [PMID: 26161560 PMCID: PMC4498801 DOI: 10.1371/journal.pone.0131787] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2015] [Accepted: 06/08/2015] [Indexed: 12/26/2022] Open
Abstract
Vaccinia virus (VACV) is a large cytoplasmic DNA virus that causes dramatic alterations to many cellular pathways including microRNA biogenesis. The virus encodes a poly(A) polymerase which was previously shown to add poly(A) tails to the 3’ end of cellular miRNAs, resulting in their degradation by 24 hours post infection (hpi). Here we used small RNA sequencing to quantify the impact of VACV infection on cellular miRNAs in human cells at both early (6 h) and late (24 h) times post infection. A detailed quantitative analysis of individual miRNAs revealed marked diversity in the extent of their modification and relative change in abundance during infection. Some miRNAs became highly modified (e.g. miR-29a-3p, miR-27b-3p) whereas others appeared resistant (e.g. miR-16-5p). Furthermore, miRNAs that were highly tailed at 6 hpi were not necessarily among the most reduced at 24 hpi. These results suggest that intrinsic features of human cellular miRNAs cause them to be differentially polyadenylated and altered in abundance during VACV infection. We also demonstrate that intermediate and late VACV gene expression are required for optimal repression of some miRNAs including miR-27-3p. Overall this work reveals complex and varied consequences of VACV infection on host miRNAs and identifies miRNAs which are largely resistant to VACV-induced polyadenylation and are therefore present at functional levels during the initial stages of infection and replication.
Collapse
|
30
|
Le X, Huang AT, Chen Y, Lai SY. Regulation of receptor tyrosine kinases by miRNA: overexpression of miRNA using lentiviral inducible expression vectors. Methods Mol Biol 2015; 1233:135-47. [PMID: 25319896 DOI: 10.1007/978-1-4939-1789-1_13] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
MicroRNAs have the ability to alter and regulate multiple genes, including RTK family members, making them an attractive approach for molecular therapeutic development. We use a pCDNA6.2-EmGFP-microRNA expression vector to overexpress individual mature microRNA and then transfer the expression cassette into a single, inducible lentiviral vector (pINDUCER20). We successfully use this system to create a pINDUCER-EmGFP-miRNA27a expression vector and generate a stable head and neck cancer cell line (UM-SCC-22A) that inducibly expresses miRNA-27a, resulting in targeted epidermal growth factor receptor down regulation. In this chapter, we describe the protocol for engineering the pINDUCER-EmGFP-microRNA expression vector, producing lentiviral particles for target cell infection, and evaluating downregulation of gene expression.
Collapse
Affiliation(s)
- XiangDong Le
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | | | | | | |
Collapse
|
31
|
Role of MicroRNAs in Renin-Angiotensin-Aldosterone System-Mediated Cardiovascular Inflammation and Remodeling. Int J Inflam 2015; 2015:101527. [PMID: 26064773 PMCID: PMC4438140 DOI: 10.1155/2015/101527] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 04/18/2015] [Indexed: 12/27/2022] Open
Abstract
MicroRNAs are endogenous regulators of gene expression either by inhibiting translation or protein degradation. Recent studies indicate that microRNAs play a role in cardiovascular disease and renin-angiotensin-aldosterone system- (RAAS-) mediated cardiovascular inflammation, either as mediators or being targeted by RAAS pharmacological inhibitors. The exact role(s) of microRNAs in RAAS-mediated cardiovascular inflammation and remodeling is/are still in early stage of investigation. However, few microRNAs have been shown to play a role in RAAS signaling, particularly miR-155, miR-146a/b, miR-132/122, and miR-483-3p. Identification of specific microRNAs and their targets and elucidating microRNA-regulated mechanisms associated RAS-mediated cardiovascular inflammation and remodeling might lead to the development of novel pharmacological strategies to target RAAS-mediated vascular pathologies. This paper reviews microRNAs role in inflammatory factors mediating cardiovascular inflammation and RAAS genes and the effect of RAAS pharmacological inhibition on microRNAs and the resolution of RAAS-mediated cardiovascular inflammation and remodeling. Also, this paper discusses the advances on microRNAs-based therapeutic approaches that may be important in targeting RAAS signaling.
Collapse
|
32
|
Xie F, Zhang B. microRNA evolution and expression analysis in polyploidized cotton genome. PLANT BIOTECHNOLOGY JOURNAL 2015; 13:421-34. [PMID: 25561162 DOI: 10.1111/pbi.12295] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 10/16/2014] [Accepted: 10/20/2014] [Indexed: 05/28/2023]
Abstract
Cotton (Gossypium hirsutum L.), the most important fibre plant in the world, is a tetraploid species, originating from the reunion of two ancestral cotton species ~1-2 million years ago. It has been reported that a great number of genes were quickly erased or preferentially remained after whole-genome duplication, ultimately leading to morphogenesis evolution. However, microRNAs (miRNAs), a new class of gene regulators, have not been well studied in polyploidization. Here, we systematically investigated miRNA evolution amongst cultivated upland cotton G. hirsutum (AADD) and its two ancestors, G. arboreum (AA) and G. raimondii (DD). Our results show that certain highly conserved miRNAs were likely to be lost, whereas certain were remained after genome polyploidization. Cotton-specific miRNAs might undergo remarkably expansion, resulting in overall miRNA increase in upland cotton. Based on the sequenced genomes of G. arboreum and G. raimondii, we are capable for the first time to categorize the origin of miRNAs and coding genes in upland cotton. Different genome-derived miRNAs and miRNA*s displayed asymmetric expression pattern, implicating their diverse functions in upland cotton. No miRNA targeting preference was observed between different genome-derived miRNAs. The origin of miRNAs and coding genes has no impact on becoming miRNAs and their targets, despite some miRNAs and their targets are extremely conserved in the three cotton species. GO- and KEGG-based analysis of conserved miRNAs show that conserved miRNAs and their targets participate in a series of important biological processes and metabolism pathways. Additionally, A-derived miRNAs might be more responsible for ovule and fibre development.
Collapse
Affiliation(s)
- Fuliang Xie
- Department of Biology, East Carolina University, Greenville, NC, USA
| | | |
Collapse
|
33
|
Wang WM, Zhao ZL, Ma SR, Yu GT, Liu B, Zhang L, Zhang WF, Kulkarni AB, Sun ZJ, Zhao YF. Epidermal growth factor receptor inhibition reduces angiogenesis via hypoxia-inducible factor-1α and Notch1 in head neck squamous cell carcinoma. PLoS One 2015; 10:e0119723. [PMID: 25723392 PMCID: PMC4344331 DOI: 10.1371/journal.pone.0119723] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 01/27/2015] [Indexed: 11/18/2022] Open
Abstract
Angiogenesis, a marker of cancer development, affects response to radiotherapy sensibility. This preclinical study aims to understand the receptor tyrosine kinase-mediated angiogenesis in head neck squamous cell carcinoma (HNSCC). The receptor tyrosine kinase activity in a transgenic mouse model of HNSCC was assessed. The anti-tumorigenetic and anti-angiogenetic effects of cetuximab-induced epidermal growth factor receptor (EGFR) inhibition were investigated in xenograft and transgenic mouse models of HNSCC. The signaling transduction of Notch1 and hypoxia-inducible factor-1α (HIF-1α) was also analyzed. EGFR was overexpressed and activated in the Tgfbr1/Pten deletion (2cKO) mouse model of HNSCC. Cetuximab significantly delayed tumor onset by reducing tumor angiogenesis. This drug exerted similar effects on heterotopic xenograft tumors. In the human HNSCC tissue array, increased EGFR expression correlated with increased HIF-1α and micro vessel density. Cetuximab inhibited tumor-induced angiogenesis in vitro and in vivo by significantly downregulating HIF-1α and Notch1. EGFR is involved in the tumor angiogenesis of HNSCC via the HIF-1α and Notch1 pathways. Therefore, targeting EGFR by suppressing hypoxia- and Notch-induced angiogenesis may benefit HNSCC therapy.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/metabolism
- Cell Line, Tumor
- Cetuximab/pharmacology
- Cetuximab/therapeutic use
- ErbB Receptors/antagonists & inhibitors
- ErbB Receptors/genetics
- ErbB Receptors/metabolism
- Head and Neck Neoplasms/drug therapy
- Head and Neck Neoplasms/metabolism
- Human Umbilical Vein Endothelial Cells/drug effects
- Human Umbilical Vein Endothelial Cells/metabolism
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Mice
- Mice, Nude
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/metabolism
- Receptor, Notch1/genetics
- Receptor, Notch1/metabolism
Collapse
Affiliation(s)
- Wei-Ming Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial-Head and Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhi-Li Zhao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial-Head and Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Si-Rui Ma
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial-Head and Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Guang-Tao Yu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China
| | - Bing Liu
- Department of Oral and Maxillofacial-Head and Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Lu Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China
| | - Wen-Feng Zhang
- Department of Oral and Maxillofacial-Head and Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Ashok B. Kulkarni
- Functional Genomics Section, Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Zhi-Jun Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial-Head and Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- * E-mail: (ZJS); (YFZ)
| | - Yi-Fang Zhao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial-Head and Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- * E-mail: (ZJS); (YFZ)
| |
Collapse
|
34
|
Sampson VB, Yoo S, Kumar A, Vetter NS, Kolb EA. MicroRNAs and Potential Targets in Osteosarcoma: Review. Front Pediatr 2015; 3:69. [PMID: 26380245 PMCID: PMC4547013 DOI: 10.3389/fped.2015.00069] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 07/20/2015] [Indexed: 12/13/2022] Open
Abstract
Osteosarcoma is the most common bone cancer in children and young adults. Surgery and multi-agent chemotherapy are the standard treatment regimens for this disease. New therapies are being investigated to improve overall survival in patients. Molecular targets that actively modulate cell processes, such as cell-cycle control, cell proliferation, metabolism, and apoptosis, have been studied, but it remains a challenge to develop novel, effective-targeted therapies to treat this heterogeneous and complex disease. MicroRNAs (miRNAs) are small non-coding RNAs that play critical roles in regulating cell processes including growth, development, and disease. miRNAs function as oncogenes or tumor suppressors to regulate gene and protein expression. Several studies have demonstrated the involvement of miRNAs in the pathogenesis of osteosarcoma with the potential for development in disease diagnostics and therapeutics. In this review, we discuss the current knowledge on the role of miRNAs and their target genes and evaluate their potential use as therapeutic agents in osteosarcoma. We also summarize the efficacy of inhibition of oncogenic miRNAs or expression of tumor suppressor miRNAs in preclinical models of osteosarcoma. Recent progress on systemic delivery as well as current applications for miRNAs as therapeutic agents has seen the advancement of miR-34a in clinical trials for adult patients with non-resectable primary liver cancer or metastatic cancer with liver involvement. We suggest a global approach to the understanding of the pathogenesis of osteosarcoma may identify candidate miRNAs as promising biomarkers for this rare disease.
Collapse
Affiliation(s)
- Valerie B Sampson
- Nemours Center for Cancer and Blood Disorders, Alfred I. duPont Hospital for Children , Wilmington, DE , USA
| | - Soonmoon Yoo
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children , Wilmington, DE , USA
| | - Asmita Kumar
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children , Wilmington, DE , USA
| | - Nancy S Vetter
- Nemours Center for Cancer and Blood Disorders, Alfred I. duPont Hospital for Children , Wilmington, DE , USA
| | - E Anders Kolb
- Nemours Center for Cancer and Blood Disorders, Alfred I. duPont Hospital for Children , Wilmington, DE , USA
| |
Collapse
|
35
|
Romay MC, Che N, Becker SN, Pouldar D, Hagopian R, Xiao X, Lusis AJ, Berliner JA, Civelek M. Regulation of NF-κB signaling by oxidized glycerophospholipid and IL-1β induced miRs-21-3p and -27a-5p in human aortic endothelial cells. J Lipid Res 2014; 56:38-50. [PMID: 25327529 DOI: 10.1194/jlr.m052670] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Exposure of endothelial cells (ECs) to agents such as oxidized glycerophospholipids (oxGPs) and cytokines, known to accumulate in atherosclerotic lesions, perturbs the expression of hundreds of genes in ECs involved in inflammatory and other biological processes. We hypothesized that microRNAs (miRNAs) are involved in regulating the inflammatory response in human aortic endothelial cells (HAECs) in response to oxGPs and interleukin 1β (IL-1β). Using next-generation sequencing and RT-quantitative PCR, we characterized the profile of expressed miRNAs in HAECs pre- and postexposure to oxGPs. Using this data, we identified miR-21-3p and miR-27a-5p to be induced 3- to 4-fold in response to oxGP and IL-1β treatment compared with control treatment. Transient overexpression of miR-21-3p and miR-27a-5p resulted in the downregulation of 1,253 genes with 922 genes overlapping between the two miRNAs. Gene Ontology functional enrichment analysis predicted that the two miRNAs were involved in the regulation of nuclear factor κB (NF-κB) signaling. Overexpression of these two miRNAs leads to changes in p65 nuclear translocation. Using 3' untranslated region luciferase assay, we identified 20 genes within the NF-κB signaling cascade as putative targets of miRs-21-3p and -27a-5p, implicating these two miRNAs as modulators of NF-κB signaling in ECs.
Collapse
Affiliation(s)
- Milagros C Romay
- Departments of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095
| | - Nam Che
- Medicine, University of California, Los Angeles, Los Angeles, CA 90095
| | - Scott N Becker
- Departments of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095
| | - Delila Pouldar
- Medicine, University of California, Los Angeles, Los Angeles, CA 90095
| | - Raffi Hagopian
- Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095
| | - Xinshu Xiao
- Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095
| | - Aldons J Lusis
- Departments of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095 Medicine, University of California, Los Angeles, Los Angeles, CA 90095 Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095
| | - Judith A Berliner
- Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA 90095
| | - Mete Civelek
- Medicine, University of California, Los Angeles, Los Angeles, CA 90095
| |
Collapse
|
36
|
Masterson L, Moualed D, Liu ZW, Howard JEF, Dwivedi RC, Tysome JR, Benson R, Sterling JC, Sudhoff H, Jani P, Goon PKC. De-escalation treatment protocols for human papillomavirus-associated oropharyngeal squamous cell carcinoma: a systematic review and meta-analysis of current clinical trials. Eur J Cancer 2014; 50:2636-48. [PMID: 25091798 DOI: 10.1016/j.ejca.2014.07.001] [Citation(s) in RCA: 221] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 06/27/2014] [Accepted: 07/01/2014] [Indexed: 11/26/2022]
Abstract
Iatrogenic complications associated with current treatment protocols for oropharyngeal squamous cell carcinoma are noted to cause high rates of acute and chronic morbidity. The aims of this study are to provide an overview of the current de-escalation trials for human papillomavirus positive (HPV+) oropharyngeal carcinoma and to evaluate the evidence supporting improved response to treatment of patients within this viral cohort. This study reviewed all completed or in progress randomised controlled trials (RCTs) assessing clinical interventions for human papillomavirus-associated locally advanced oropharyngeal squamous cell carcinoma. We utilised a validated 'risk of bias' tool to assess study quality. We identified nine RCTs that met the full inclusion criteria for this review (all of which are currently on-going and will report from 2015 onwards). Five RCTs performed a post hoc analysis by HPV status, which allowed meta-analysis of 1130 patients. The data reveal a significant difference in overall survival (hazard ratio (HR) 0.49 [95% confidence interval (CI) 0.35-0.69]), loco-regional failure (HR 0.43 [95% CI 0.17-1.11]) and disease specific survival (0.41 [95% 0.3-0.56]) in favour of the HPV+ category. In considering de-escalation treatment protocols, nine studies are currently ongoing. Our meta-analysis provides strong evidence for an improved prognosis in the viral associated cohort when treated by platinum based chemotherapy in combination with radiotherapy or primary radiotherapy. So far, one trial (with moderate to high risk of bias) suggests a reduced survival outcome for the HPV+ population when using the epidermal growth factor receptor (EGFR) inhibitor cetuximab.
Collapse
Affiliation(s)
- Liam Masterson
- ENT Department, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK; Department of Pathology, University of Cambridge, UK.
| | - Daniel Moualed
- ENT Department, Milton Keynes Hospital NHS Foundation Trust, Milton Keynes, UK
| | - Zi Wei Liu
- ENT Department, Barts Health NHS Trust, London, UK
| | - James E F Howard
- Oncology Centre, University College Hospital NHS Trust, London, UK
| | - Raghav C Dwivedi
- ENT Department, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - James R Tysome
- ENT Department, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | | | | | - Holger Sudhoff
- Department of Otolaryngology, Head and Neck Surgery, Bielefeld Academic Teaching Hospital, Bielefeld, Germany
| | - Piyush Jani
- ENT Department, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | | |
Collapse
|
37
|
Bao Y, Chen Z, Guo Y, Feng Y, Li Z, Han W, Wang J, Zhao W, Jiao Y, Li K, Wang Q, Wang J, Zhang H, Wang L, Yang W. Tumor suppressor microRNA-27a in colorectal carcinogenesis and progression by targeting SGPP1 and Smad2. PLoS One 2014; 9:e105991. [PMID: 25166914 PMCID: PMC4148394 DOI: 10.1371/journal.pone.0105991] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 07/26/2014] [Indexed: 02/07/2023] Open
Abstract
The aberrant expression of microRNAs (miRNAs) is associated with colorectal carcinogenesis, but the underlying mechanisms are not clear. This study showed that the miRNA-27a (miR-27a) was significantly reduced in colorectal cancer tissues and colorectal cancer cell lines, and that the reduced miR-27a was associated with distant metastasis and colorectal cancer clinical pathological stages-miR-27a was lower at stages III/IV than that at stage II. Bioinformatic and systemic biological analysis predicted several targets of miR-27a, among them SGPP1 and Smad2 were significantly affected. SGPP1 and Smad2 at mRNA and protein levels were negatively correlated with miR-27a in human colorectal cancer tissues and cancer cell lines. Increased miR-27a significantly repressed SGPP1 and Smad2 at transcriptional and translational levels. Functional studies showed that increasing miR-27a inhibited colon cancer cell proliferation, promoted apoptosis and attenuated cell migration, which were also linked to downregulation of p-STAT3 and upregulation of cleaved caspase 3. In vivo, miR-27a inhibited colon cancer cell growth in tumor-bearing mice. Taken together, this study has revealed miR-27a as a tumor suppressor and has identified SGPP1 and Smad2 as novel targets of miR-27a, linking to STAT3 for regulating cancer cell proliferation, apoptosis and migration in colorectal cancer. Therefore, miR-27a could be a useful biomarker for monitoring colorectal cancer development and progression, and also could have a therapeutic potential by targeting SGPP1, Smad2 and STAT3 for colorectal cancer therapy.
Collapse
Affiliation(s)
- Yonghua Bao
- Department of Immunology, Xinxiang Medical University, Xinxiang, China
| | - Zhiguo Chen
- Department of Pathology, Xinxiang Medical University, Xinxiang, China
| | - Yongchen Guo
- Department of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Yansheng Feng
- Department of Pathophysiology, Xinxiang Medical University, Xinxiang, China
| | - Zexin Li
- Department of Surgery, the First Affiliated Hospital, Xinxiang Medical University, Weihui, China
| | - Wenliang Han
- Department of Gastroenterology, Xinxiang Central Hospital, Xinxiang Medical University, Xinxiang, China
| | - Jianguo Wang
- Department of Surgery, the First Affiliated Hospital, Xinxiang Medical University, Weihui, China
| | - Weixing Zhao
- Department of Pathology, Xinxiang Medical University, Xinxiang, China
| | - Yunjuan Jiao
- Department of Pathology, Xinxiang Medical University, Xinxiang, China
| | - Kai Li
- Department of Pathology, Xinxiang Medical University, Xinxiang, China
| | - Qian Wang
- Department of Immunology, Xinxiang Medical University, Xinxiang, China
| | - Jiaqi Wang
- Department of Pathology, Xinxiang Medical University, Xinxiang, China
| | - Huijuan Zhang
- Department of Pathology, Xinxiang Medical University, Xinxiang, China
| | - Liang Wang
- Department of Pathology, Xinxiang Medical University, Xinxiang, China,Department of Pathology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Wancai Yang
- Department of Pathology, Xinxiang Medical University, Xinxiang, China,Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, United States of America,* E-mail:
| |
Collapse
|
38
|
Xiong XD, Luo XP, Cheng J, Liu X, Li EM, Zeng LQ. A genetic variant in pre-miR-27a is associated with a reduced cervical cancer risk in southern Chinese women. Gynecol Oncol 2014; 132:450-4. [DOI: 10.1016/j.ygyno.2013.12.030] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 12/14/2013] [Accepted: 12/19/2013] [Indexed: 12/19/2022]
|