1
|
Yoo D, Wu S, Choi S, Huh SO, Sadra A. STK33 as the functional substrate of miR-454-3p for suppression and apoptosis in neuroblastoma. Mol Cells 2024; 47:100145. [PMID: 39515612 DOI: 10.1016/j.mocell.2024.100145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 10/31/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
miR-454-3p has been reported to be a tumor-suppressive microRNA (miRNA) in multiple cancer types. We identified the kinase STK33 mRNA, which is a high-risk factor for survival in neuroblastoma (NB) patients, as being a substrate of miR-454-3p in NB. Even though STK33 is an attractive target for several cancers, the development of inhibitors of STK33 has been challenging. For the various cell lines tested, we demonstrated reduced growth and viability with the miR-454-3p mimic. From among the candidate NB-associated miRNAs, miR-454-3p mimic and its antagonist had the most profound effect on STK33 mRNA and protein-level changes. Under various conditions of growth and external stress for the cells, the RNA levels for miR-454-3p and STK33 also negatively correlated. Luciferase reporter assays demonstrated STK33 as a substrate for miR-454-3p, and recombinant versions of STK33 resistant to miR-454-3p significantly blunted the suppressive effect of the miR-454-3p and established STK33 as the major functional substrate of miR-454-3p. Overexpression of miR-454-3p or knockdown of STK33 mRNA promoted autophagy and at the same time, increased the apoptotic markers in the tested NB cells, indicating a mechanism for the suppressive effect of the agents. Given the difficult-to-drug targets such as STK33 and the recent successes in RNA delivery methods for cancer treatment, it is thought that targeting cancer cells with a suppressive miRNA such as miR-454-3p for STK33-dependent cancer types may be an alternative means of NB therapy.
Collapse
Affiliation(s)
- Dongkwan Yoo
- Department of Pharmacology, College of Medicine, Institute of Natural Medicine, Hallym University, Chuncheon, Gangwon State, Republic of Korea
| | - Sichen Wu
- Department of Pharmacology, College of Medicine, Institute of Natural Medicine, Hallym University, Chuncheon, Gangwon State, Republic of Korea
| | - Seunghyuk Choi
- Department of Pharmacology, College of Medicine, Institute of Natural Medicine, Hallym University, Chuncheon, Gangwon State, Republic of Korea
| | - Sung-Oh Huh
- Department of Pharmacology, College of Medicine, Institute of Natural Medicine, Hallym University, Chuncheon, Gangwon State, Republic of Korea.
| | - Ali Sadra
- Department of Pharmacology, College of Medicine, Institute of Natural Medicine, Hallym University, Chuncheon, Gangwon State, Republic of Korea.
| |
Collapse
|
2
|
Ataei A, Tahsili M, Hayadokht G, Daneshvar M, Mohammadi Nour S, Soofi A, Masoudi A, Kabiri M, Natami M. Targeting long noncoding RNAs in neuroblastoma: Progress and prospects. Chem Biol Drug Des 2023; 102:640-652. [PMID: 37291742 DOI: 10.1111/cbdd.14263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 04/10/2023] [Accepted: 04/18/2023] [Indexed: 06/10/2023]
Abstract
Neuroblastoma (NB) is the third most prevalent tumor that mostly influences infants and young children. Although different treatments have been developed for the treatment of NB, high-risk patients have been reported to have low survival rates. Currently, long noncoding RNAs (lncRNAs) have shown an attractive potential in cancer research and a party of investigations have been performed to understand mechanisms underlying tumor development through lncRNA dysregulation. Researchers have just newly initiated to exhibit the involvement of lncRNAs in NB pathogenesis. In this review article, we tried to clarify the point we stand with respect to the involvement of lncRNAs in NB. Moreover, implications for the pathologic roles of lncRNAs in the development of NB have been discussed. It seems that some of these lncRNAs have promising potential to be applied as biomarkers for NB prognosis and treatment.
Collapse
Affiliation(s)
- Ali Ataei
- School of Medicine, Bam University of Medical Sciences, Bam, Iran
| | | | - Golsa Hayadokht
- School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | | | | | - Asma Soofi
- Department of Physical Chemistry, School of Chemistry, College of Sciences, University of Tehran, Tehran, Iran
| | - Alireza Masoudi
- Department of Laboratory Sciences, Faculty of Alied Medical Sciences, Qom University of Medical Sciences, Qom, Iran
| | - Maryam Kabiri
- Faculty of Medicine, Islamic Azad University of Medical Sciences, Tehran, Iran
| | - Mohammad Natami
- Department of Urology, Faculty of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
3
|
Anoushirvani AA, Jafarian Yazdi A, Amirabadi S, Asouri SA, Shafabakhsh R, Sheida A, Hosseini Khabr MS, Jafari A, Tamehri Zadeh SS, Hamblin MR, Kalantari L, Talaei Zavareh SA, Mirzaei H. Role of non-coding RNAs in neuroblastoma. Cancer Gene Ther 2023; 30:1190-1208. [PMID: 37217790 DOI: 10.1038/s41417-023-00623-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 03/25/2023] [Accepted: 05/04/2023] [Indexed: 05/24/2023]
Abstract
Neuroblastoma is known as the most prevalent extracranial malignancy in childhood with a neural crest origin. It has been widely accepted that non-coding RNAs (ncRNAs) play important roles in many types of cancer, including glioma and gastrointestinal cancers. They may regulate the cancer gene network. According to recent sequencing and profiling studies, ncRNAs genes are deregulated in human cancers via deletion, amplification, abnormal epigenetic, or transcriptional regulation. Disturbances in the expression of ncRNAs may act either as oncogenes or as anti-tumor suppressor genes, and can lead to the induction of cancer hallmarks. ncRNAs can be secreted from tumor cells inside exosomes, where they can be transferred to other cells to affect their function. However, these topics still need more study to clarify their exact roles, so the present review addresses different roles and functions of ncRNAs in neuroblastoma.
Collapse
Affiliation(s)
- Ali Arash Anoushirvani
- Department of Internal Medicine, Firoozgar Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Sanaz Amirabadi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Sahar Ahmadi Asouri
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University, Kashan, Iran
| | - Rana Shafabakhsh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University, Kashan, Iran
| | - Amirhossein Sheida
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Maryam Sadat Hosseini Khabr
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Ameneh Jafari
- ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, P.O. BOX: 15179/64311, Tehran, Iran
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, South Africa
| | - Leila Kalantari
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran.
| | | | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University, Kashan, Iran.
| |
Collapse
|
4
|
Kciuk M, Yahya EB, Mohamed MMI, Abdulsamad MA, Allaq AA, Gielecińska A, Kontek R. Insights into the Role of LncRNAs and miRNAs in Glioma Progression and Their Potential as Novel Therapeutic Targets. Cancers (Basel) 2023; 15:3298. [PMID: 37444408 DOI: 10.3390/cancers15133298] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Accumulating evidence supports that both long non-coding and micro RNAs (lncRNAs and miRNAs) are implicated in glioma tumorigenesis and progression. Poor outcome of gliomas has been linked to late-stage diagnosis and mostly ineffectiveness of conventional treatment due to low knowledge about the early stage of gliomas, which are not possible to observe with conventional diagnostic approaches. The past few years witnessed a revolutionary advance in biotechnology and neuroscience with the understanding of tumor-related molecules, including non-coding RNAs that are involved in the angiogenesis and progression of glioma cells and thus are used as prognostic biomarkers as well as novel therapeutic targets. The emerging research on lncRNAs and miRNAs highlights their crucial role in glioma progression, offering new insights into the disease. These non-coding RNAs hold significant potential as novel therapeutic targets, paving the way for innovative treatment approaches against glioma. This review encompasses a comprehensive discussion about the role of lncRNAs and miRNAs in gene regulation that is responsible for the promotion or the inhibition of glioma progression and collects the existing links between these key cancer-related molecules.
Collapse
Affiliation(s)
- Mateusz Kciuk
- Department of Molecular Biotechnology and Genetics, University of Lodz, 90-237 Lodz, Poland
- Doctoral School of Exact and Natural Sciences, University of Lodz, 90-237 Lodz, Poland
| | - Esam Bashir Yahya
- Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, Penang 11800, Malaysia
| | | | - Muhanad A Abdulsamad
- Department of Molecular Biology, Faculty of Science, Sabratha University, Sabratha 00218, Libya
| | - Abdulmutalib A Allaq
- Faculty of Applied Science, Universiti Teknologi MARA, Shah Alam 40450, Malaysia
| | - Adrianna Gielecińska
- Department of Molecular Biotechnology and Genetics, University of Lodz, 90-237 Lodz, Poland
- Doctoral School of Exact and Natural Sciences, University of Lodz, 90-237 Lodz, Poland
| | - Renata Kontek
- Department of Molecular Biotechnology and Genetics, University of Lodz, 90-237 Lodz, Poland
| |
Collapse
|
5
|
Chadda KR, Blakey EE, Coleman N, Murray MJ. The clinical utility of dysregulated microRNA expression in paediatric solid tumours. Eur J Cancer 2022; 176:133-154. [PMID: 36215946 DOI: 10.1016/j.ejca.2022.09.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 09/10/2022] [Indexed: 12/15/2022]
Abstract
MicroRNAs (miRNAs) are short, non-protein-coding genes that regulate the expression of numerous protein-coding genes. Their expression is dysregulated in cancer, where they may function as oncogenes or tumour suppressor genes. As miRNAs are highly resistant to degradation, they are ideal biomarker candidates to improve the diagnosis and clinical management of cancer, including prognostication. Furthermore, miRNAs dysregulated in malignancy represent potential therapeutic targets. The use of miRNAs for these purposes is a particularly attractive option to explore for paediatric malignancies, where the mutational burden is typically low, in contrast to cancers affecting adult patients. As childhood cancers are rare, it has taken time to accumulate the necessary body of evidence showing the potential for miRNAs to improve clinical management across this group of tumours. Here, we review the current literature regarding the potential clinical utility of miRNAs in paediatric solid tumours, which is now both timely and justified. Exploring such avenues is warranted to improve the management and outcomes of children affected by cancer.
Collapse
Affiliation(s)
- Karan R Chadda
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Ellen E Blakey
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Nicholas Coleman
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK; Department of Paediatric Histopathology, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge, CB2 0QQ, UK.
| | - Matthew J Murray
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK; Department of Paediatric Haematology and Oncology, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
6
|
Segura MF, Soriano A, Roma J, Piskareva O, Jiménez C, Boloix A, Fletcher JI, Haber M, Gray JC, Cerdá-Alberich L, Martínez de Las Heras B, Cañete A, Gallego S, Moreno L. Methodological advances in the discovery of novel neuroblastoma therapeutics. Expert Opin Drug Discov 2021; 17:167-179. [PMID: 34807782 DOI: 10.1080/17460441.2022.2002297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Neuroblastoma is a cancer of the sympathetic nervous system that causes up to 15% of cancer-related deaths among children. Among the ~1,000 newly diagnosed cases per year in Europe, more than half are classified as high-risk, with a 5-year survival rate <50%. Current multimodal treatments have improved survival among these patients, but relapsed and refractory tumors remain a major therapeutic challenge. A number of new methodologies are paving the way for the development of more effective and safer therapies to ultimately improve outcomes for high-risk patients. AREAS COVERED The authors provide a critical review on methodological advances aimed at providing new therapeutic opportunities for neuroblastoma patients, including preclinical models of human disease, generation of omics data to discover new therapeutic targets, and artificial intelligence-based technologies to implement personalized treatments. EXPERT OPINION While survival of childhood cancer has improved over the past decades, progress has been uneven. Still, survival is dismal for some cancers, including high-risk neuroblastoma. Embracing new technologies (e.g. molecular profiling of tumors, 3D in vitro models, etc.), international collaborative efforts and the incorporation of new therapies (e.g. RNA-based therapies, epigenetic therapies, immunotherapy) will ultimately lead to more effective and safer therapies for these subgroups of neuroblastoma patients.
Collapse
Affiliation(s)
- Miguel F Segura
- Pediatric Oncology and Hematology Department, Translational Research in Child and Adolescent Cancer, Vall d'Hebron Institut de Recerca (VHIR), Barcelona. Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Aroa Soriano
- Pediatric Oncology and Hematology Department, Translational Research in Child and Adolescent Cancer, Vall d'Hebron Institut de Recerca (VHIR), Barcelona. Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Josep Roma
- Pediatric Oncology and Hematology Department, Translational Research in Child and Adolescent Cancer, Vall d'Hebron Institut de Recerca (VHIR), Barcelona. Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Olga Piskareva
- Cancer Bioengineering Group, Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland.,Department of Anatomy and Regenerative Medicine, Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland.,National Children's Research Centre, OLCHC, Dublin, Ireland School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Carlos Jiménez
- Pediatric Oncology and Hematology Department, Translational Research in Child and Adolescent Cancer, Vall d'Hebron Institut de Recerca (VHIR), Barcelona. Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Ariadna Boloix
- Pediatric Oncology and Hematology Department, Translational Research in Child and Adolescent Cancer, Vall d'Hebron Institut de Recerca (VHIR), Barcelona. Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Jamie I Fletcher
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Kensington, Australia
| | - Michelle Haber
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Kensington, Australia
| | - Juliet C Gray
- Antibody and Vaccine Group, Centre for Cancer Immunology, University of Southampton Faculty of Medicine, Southampton, UK
| | - Leonor Cerdá-Alberich
- Grupo de Investigación Biomédica En Imagen, Instituto de Investigación Sanitaria La Fe, Spain
| | | | - Adela Cañete
- Unidad de Oncohematología Pediátrica, Hospital Universitario y Politécnico La Fe, Spain
| | - Soledad Gallego
- Pediatric Oncology and Hematology Department, Translational Research in Child and Adolescent Cancer, Vall d'Hebron Institut de Recerca (VHIR), Barcelona. Universitat Autònoma de Barcelona, Bellaterra, Spain.,Pediatric Oncology and Hematology Department, Vall d'Hebron University Hospital-UAB, Barcelona, Spain
| | - Lucas Moreno
- Pediatric Oncology and Hematology Department, Translational Research in Child and Adolescent Cancer, Vall d'Hebron Institut de Recerca (VHIR), Barcelona. Universitat Autònoma de Barcelona, Bellaterra, Spain.,Pediatric Oncology and Hematology Department, Vall d'Hebron University Hospital-UAB, Barcelona, Spain
| |
Collapse
|
7
|
Misiak D, Hagemann S, Bell JL, Busch B, Lederer M, Bley N, Schulte JH, Hüttelmaier S. The MicroRNA Landscape of MYCN-Amplified Neuroblastoma. Front Oncol 2021; 11:647737. [PMID: 34026620 PMCID: PMC8138323 DOI: 10.3389/fonc.2021.647737] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 04/15/2021] [Indexed: 02/01/2023] Open
Abstract
MYCN gene amplification and upregulated expression are major hallmarks in the progression of high-risk neuroblastoma. MYCN expression and function in modulating gene synthesis in neuroblastoma is controlled at virtually every level, including poorly understood regulation at the post-transcriptional level. MYCN modulates the expression of various microRNAs including the miR-17-92 cluster. MYCN mRNA expression itself is subjected to the control by miRNAs, most prominently the miR-17-92 cluster that balances MYCN expression by feed-back regulation. This homeostasis seems disturbed in neuroblastoma where MYCN upregulation coincides with severely increased expression of the miR-17-92 cluster. In the presented study, we applied high-throughput next generation sequencing to unravel the miRNome in a cohort of 97 neuroblastomas, representing all clinical stages. Aiming to reveal the MYCN-dependent miRNome, we evaluate miRNA expression in MYCN-amplified as well as none amplified tumor samples. In correlation with survival data analysis of differentially expressed miRNAs, we present various putative oncogenic as well as tumor suppressive miRNAs in neuroblastoma. Using microRNA trapping by RNA affinity purification, we provide a comprehensive view of MYCN-regulatory miRNAs in neuroblastoma-derived cells, confirming a pivotal role of the miR-17-92 cluster and moderate association by the let-7 miRNA family. Attempting to decipher how MYCN expression escapes elevated expression of inhibitory miRNAs, we present evidence that RNA-binding proteins like the IGF2 mRNA binding protein 1 reduce miRNA-directed downregulation of MYCN in neuroblastoma. Our findings emphasize the potency of post-transcriptional regulation of MYCN in neuroblastoma and unravel new avenues to pursue inhibition of this potent oncogene.
Collapse
Affiliation(s)
- Danny Misiak
- Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Sven Hagemann
- Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Jessica L. Bell
- Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Bianca Busch
- Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Marcell Lederer
- Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Nadine Bley
- Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Johannes H. Schulte
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- German Consortium for Translational Cancer Research (DKTK), Partner Site Charité Berlin, Berlin, Germany
| | - Stefan Hüttelmaier
- Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| |
Collapse
|
8
|
Abstract
Neuroblastoma (NB) is a pediatric cancer of the sympathetic nervous system and one of the most common solid tumors in infancy. Amplification of MYCN, copy number alterations, numerical and segmental chromosomal aberrations, mutations, and rearrangements on a handful of genes, such as ALK, ATRX, TP53, RAS/MAPK pathway genes, and TERT, are attributed as underlying causes that give rise to NB. However, the heterogeneous nature of the disease-along with the relative paucity of recurrent somatic mutations-reinforces the need to understand the interplay of genetic factors and epigenetic alterations in the context of NB. Epigenetic mechanisms tightly control gene expression, embryogenesis, imprinting, chromosomal stability, and tumorigenesis, thereby playing a pivotal role in physio- and pathological settings. The main epigenetic alterations include aberrant DNA methylation, disrupted patterns of posttranslational histone modifications, alterations in chromatin composition and/or architecture, and aberrant expression of non-coding RNAs. DNA methylation and demethylation are mediated by DNA methyltransferases (DNMTs) and ten-eleven translocation (TET) proteins, respectively, while histone modifications are coordinated by histone acetyltransferases and deacetylases (HATs, HDACs), and histone methyltransferases and demethylases (HMTs, HDMs). This article focuses predominately on the crosstalk between the epigenome and NB, and the implications it has on disease diagnosis and treatment.
Collapse
Affiliation(s)
- Irfete S Fetahu
- St. Anna Children's Cancer Research Institute, Zimmermannplatz 10, 1090, Vienna, Austria.
| | - Sabine Taschner-Mandl
- St. Anna Children's Cancer Research Institute, Zimmermannplatz 10, 1090, Vienna, Austria.
| |
Collapse
|
9
|
Marengo B, Pulliero A, Corrias MV, Leardi R, Farinini E, Fronza G, Menichini P, Monti P, Monteleone L, Valenti GE, Speciale A, Perri P, Madia F, Izzotti A, Domenicotti C. Potential Role of miRNAs in the Acquisition of Chemoresistance in Neuroblastoma. J Pers Med 2021; 11:jpm11020107. [PMID: 33562297 PMCID: PMC7916079 DOI: 10.3390/jpm11020107] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/20/2021] [Accepted: 02/04/2021] [Indexed: 02/06/2023] Open
Abstract
Neuroblastoma (NB) accounts for about 8–10% of pediatric cancers, and the main causes of death are the presence of metastases and the acquisition of chemoresistance. Metastatic NB is characterized by MYCN amplification that correlates with changes in the expression of miRNAs, which are small non-coding RNA sequences, playing a crucial role in NB development and chemoresistance. In the present study, miRNA expression was analyzed in two human MYCN-amplified NB cell lines, one sensitive (HTLA-230) and one resistant to Etoposide (ER-HTLA), by microarray and RT-qPCR techniques. These analyses showed that miRNA-15a, -16-1, -19b, -218, and -338 were down-regulated in ER-HTLA cells. In order to validate the presence of this down-regulation in vivo, the expression of these miRNAs was analyzed in primary tumors, metastases, and bone marrow of therapy responder and non-responder pediatric patients. Principal component analysis data showed that the expression of miRNA-19b, -218, and -338 influenced metastases, and that the expression levels of all miRNAs analyzed were higher in therapy responders in respect to non-responders. Collectively, these findings suggest that these miRNAs might be involved in the regulation of the drug response, and could be employed for therapeutic purposes.
Collapse
Affiliation(s)
- Barbara Marengo
- Department of Experimental Medicine, University of Genova, 16100 Genova, Italy; (L.M.); (G.E.V.); (A.I.); (C.D.)
- Correspondence: ; Tel.: +39-010-3538831
| | | | - Maria Valeria Corrias
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, 16100 Genova, Italy; (M.V.C.); (P.P.)
| | - Riccardo Leardi
- Department of Pharmacy, University of Genova, 16100 Genova, Italy; (R.L.); (E.F.)
| | - Emanuele Farinini
- Department of Pharmacy, University of Genova, 16100 Genova, Italy; (R.L.); (E.F.)
| | - Gilberto Fronza
- UOC Mutagenesis and Cancer Prevention, IRCCS Ospedale Policlinico San Martino, 16100 Genova, Italy; (G.F.); (P.M.); (P.M.); (A.S.)
| | - Paola Menichini
- UOC Mutagenesis and Cancer Prevention, IRCCS Ospedale Policlinico San Martino, 16100 Genova, Italy; (G.F.); (P.M.); (P.M.); (A.S.)
| | - Paola Monti
- UOC Mutagenesis and Cancer Prevention, IRCCS Ospedale Policlinico San Martino, 16100 Genova, Italy; (G.F.); (P.M.); (P.M.); (A.S.)
| | - Lorenzo Monteleone
- Department of Experimental Medicine, University of Genova, 16100 Genova, Italy; (L.M.); (G.E.V.); (A.I.); (C.D.)
| | - Giulia Elda Valenti
- Department of Experimental Medicine, University of Genova, 16100 Genova, Italy; (L.M.); (G.E.V.); (A.I.); (C.D.)
| | - Andrea Speciale
- UOC Mutagenesis and Cancer Prevention, IRCCS Ospedale Policlinico San Martino, 16100 Genova, Italy; (G.F.); (P.M.); (P.M.); (A.S.)
| | - Patrizia Perri
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, 16100 Genova, Italy; (M.V.C.); (P.P.)
| | - Francesca Madia
- Medical Genetics Unit, IRCCS Giannina Gaslini Institute, 16100 Genova, Italy;
| | - Alberto Izzotti
- Department of Experimental Medicine, University of Genova, 16100 Genova, Italy; (L.M.); (G.E.V.); (A.I.); (C.D.)
- UOC Mutagenesis and Cancer Prevention, IRCCS Ospedale Policlinico San Martino, 16100 Genova, Italy; (G.F.); (P.M.); (P.M.); (A.S.)
| | - Cinzia Domenicotti
- Department of Experimental Medicine, University of Genova, 16100 Genova, Italy; (L.M.); (G.E.V.); (A.I.); (C.D.)
| |
Collapse
|
10
|
Synergism of Proneurogenic miRNAs Provides a More Effective Strategy to Target Glioma Stem Cells. Cancers (Basel) 2021; 13:cancers13020289. [PMID: 33466745 PMCID: PMC7831004 DOI: 10.3390/cancers13020289] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/08/2021] [Accepted: 01/11/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary miRNAs function as critical regulators of gene expression and have been defined as contributors of cancer phenotypes by acting as oncogenes or tumor suppressors. Based on these findings, miRNA-based therapies have been explored in the treatment of many different malignancies. The use of single miRNAs has faced some challenges and showed limited success. miRNAs cooperate to regulate distinct biological processes and pathways and, therefore, combination of related miRNAs could amplify the repression of oncogenic factors and the effect on cancer relevant pathways. We established that the combination of tumor suppressor miRNAs miR-124, miR-128, and miR-137 is much more effective than single miRNAs in disrupting proliferation and survival of glioma stem cells and neuroblastoma lines and promoting differentiation and response to radiation. Subsequent genomic analyses showed that other combinations of tumor suppressor miRNAs could be equally effective, and its use could provide new routes to target in special cancer-initiating cell populations. Abstract Tumor suppressor microRNAs (miRNAs) have been explored as agents to target cancer stem cells. Most strategies use a single miRNA mimic and present many disadvantages, such as the amount of reagent required and the diluted effect on target genes. miRNAs work in a cooperative fashion to regulate distinct biological processes and pathways. Therefore, we propose that miRNA combinations could provide more efficient ways to target cancer stem cells. We have previously shown that miR-124, miR-128, and miR-137 function synergistically to regulate neurogenesis. We used a combination of these three miRNAs to treat glioma stem cells and showed that this treatment was much more effective than single miRNAs in disrupting cell proliferation and survival and promoting differentiation and response to radiation. Transcriptomic analyses indicated that transcription regulation, angiogenesis, metabolism, and neuronal differentiation are among the main biological processes affected by transfection of this miRNA combination. In conclusion, we demonstrated the value of using combinations of neurogenic miRNAs to disrupt cancer phenotypes and glioma stem cell growth. The synergistic effect of these three miRNA amplified the repression of oncogenic factors and the effect on cancer relevant pathways. Future therapeutic approaches would benefit from utilizing miRNA combinations, especially when targeting cancer-initiating cell populations.
Collapse
|
11
|
Deng D, Yang S, Wang X. Long non-coding RNA SNHG16 regulates cell behaviors through miR-542-3p/HNF4α axis via RAS/RAF/MEK/ERK signaling pathway in pediatric neuroblastoma cells. Biosci Rep 2020; 40:BSR20200723. [PMID: 32412051 PMCID: PMC7251324 DOI: 10.1042/bsr20200723] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/14/2020] [Accepted: 05/14/2020] [Indexed: 01/27/2023] Open
Abstract
Neuroblastoma (NB) is an extracranial solid tumor in children with complex mechanism. Increasing reports indicated that long non-coding RNA (lncRNA) small nucleolar RNA host gene 16 (SNHG16) account for the pathogenesis of NB. Nevertheless, the precise functions of SNHG16 needed to be further exposed in NB progression. Our data revealed that SNHG16 and hepatocyte nuclear factor 4 α (HNF4α) were up-regulated, but miR-542-3p was down-regulated in NB. Knockdown of SNHG16 or HNF4α could impede cell proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT) in vitro. Interestingly, the role of SNHG16 detetion in cell behaviors was rescued by HNF4α overexpression in NB cells. Mechanically, SNHG16 modulated the progression of tumor growth via miR-542-3p/HNF4α axis in NB. Also, SNHG16 knockdown inactivated rat sarcoma/effector of RAS/mitogen-activated extracellular signal-regulated kinase/extracellular regulated protein kinases (RAS/RAF/MEK/ERK) signaling pathway through HNF4α. Therefore, SNHG16/miR-542-3p/HNF4α axis modified NB progression via RAS/RAF/MEK/ERK signaling pathway, might highlight a novel therapeutic approach for NB.
Collapse
Affiliation(s)
- Defeng Deng
- Department of Hematology, Kaifeng Children’s Hospital, Kaifeng, Henan, China
| | - Shuangjie Yang
- Department of Hematology, Kaifeng Children’s Hospital, Kaifeng, Henan, China
| | - Xiang Wang
- Department of Hematology, Kaifeng Children’s Hospital, Kaifeng, Henan, China
| |
Collapse
|
12
|
Wei Q, Guo Z, Chen D, Jia X. MiR-542-3p Suppresses Neuroblastoma Cell Proliferation and Invasion by Downregulation of KDM1A and ZNF346. Open Life Sci 2020; 15:173-184. [PMID: 33987474 PMCID: PMC8114778 DOI: 10.1515/biol-2020-0018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 12/02/2019] [Indexed: 12/20/2022] Open
Abstract
Neuroblastoma is one of the most common malignancies in infants and children. MicroRNAs (miRNAs) have been reported as significant regulators that play important roles in neuroblastoma development. This research aimed to analyze the functional mechanism of miR-542-3p in neuroblastoma. Here, we found that miR-542-3p was downregulated and KDM1A as well as ZNF346 were upregulated in neuroblastoma tissues and cells. Both overexpression of miR-542-3p and the knockdown of KDM1A suppressed cell proliferation and invasion in neuroblastomas. Moreover, miR-542-3p reduced the levels of KDM1A and ZNF346 through interaction. Both KDM1A overexpression and ZNF346 upregulation weakened the effect of miR-542-3p on neuroblastoma cells. Besides, miR-542-3p negatively regulated tumor growth in vivo. Our results suggested that miR-542-3p suppressed cell proliferation and invasion by targeting KDM1A and ZNF346 in neuroblastomas, providing a theoretical basis for the treatment of neuroblastoma.
Collapse
Affiliation(s)
- Qiang Wei
- Department II of General Surgery, Xi'an Children's Hospital, Xi'an, Shaanxi, China
| | - Zhao Guo
- Department II of General Surgery, Xi'an Children's Hospital, Xi'an, Shaanxi, China
| | - Dong Chen
- Department II of General Surgery, Xi'an Children's Hospital, Xi'an, Shaanxi, China
| | - Xinjian Jia
- Department II of General Surgery, Xi'an Children's Hospital, Xi'an, Shaanxi, China
| |
Collapse
|
13
|
Abstract
Abdominal tumors (AT) in children account for approximately 17% of all pediatric solid tumor cases, and frequently exhibit embryonal histological features that differentiate them from adult cancers. Current molecular approaches have greatly improved the understanding of the distinctive pathology of each tumor type and enabled the characterization of novel tumor biomarkers. As seen in abdominal adult tumors, microRNAs (miRNAs) have been increasingly implicated in either the initiation or progression of childhood cancer. Moreover, besides predicting patient prognosis, they represent valuable diagnostic tools that may also assist the surveillance of tumor behavior and treatment response, as well as the identification of the primary metastatic sites. Thus, the present study was undertaken to compile up-to-date information regarding the role of dysregulated miRNAs in the most common histological variants of AT, including neuroblastoma, nephroblastoma, hepatoblastoma, hepatocarcinoma, and adrenal tumors. Additionally, the clinical implications of dysregulated miRNAs as potential diagnostic tools or indicators of prognosis were evaluated.
Collapse
|
14
|
Nanotechnological based miRNA intervention in the therapeutic management of neuroblastoma. Semin Cancer Biol 2019; 69:100-108. [PMID: 31562954 DOI: 10.1016/j.semcancer.2019.09.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 08/29/2019] [Accepted: 09/24/2019] [Indexed: 01/07/2023]
Abstract
Neuroblastoma (NB) is a widely diagnosed cancer in children, characterized by amplification of the gene encoding the MYCN transcription factor, which is highly predictive of poor clinical outcome and metastatic disease. microRNAs (a class of small non-coding RNAs) are regulated by MYCN transcription factor in neuroblastoma cells. The current research is focussed on identifying differential role of miRNAs and their interactions with signalling proteins, which are intricately linked with cellular processes like apoptosis, proliferation or metastasis. However, the therapeutic success of miRNAs is limited by pharmaco-technical issues which are well counteracted by nanotechnological advancements. The nanoformulated miRNAs unload anti-cancer drugs in a controlled and prespecified manner at target sites, to influence the activity of target protein in amelioration of NB. Recent advances and developments in the field of miRNAs-based systems for clinical management of NBs and the role of nanotechnology to overcome challenges with drug delivery of miRNAs have been reviewed in this paper.
Collapse
|
15
|
Yerukala Sathipati S, Sahu D, Huang HC, Lin Y, Ho SY. Identification and characterization of the lncRNA signature associated with overall survival in patients with neuroblastoma. Sci Rep 2019; 9:5125. [PMID: 30914706 PMCID: PMC6435792 DOI: 10.1038/s41598-019-41553-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 03/05/2019] [Indexed: 01/16/2023] Open
Abstract
Neuroblastoma (NB) is a commonly occurring cancer among infants and young children. Recently, long non-coding RNAs (lncRNAs) have been using as prognostic biomarkers for therapeutics and interventions in various cancers. Considering the poor survival of NB, the lncRNA-based therapeutic strategies must be improved. This work proposes an overall survival time estimator called SVR-NB to identify the lncRNA signature that is associated with the overall survival of patients with NB. SVR-NB is an optimized support vector regression (SVR)-based method that uses an inheritable bi-objective combinatorial genetic algorithm for feature selection. The dataset of 231 NB patients that contains overall survival information and expression profiles of 783 lncRNAs was used to design and evaluate SVR-NB from the database of gene expression omnibus accession GSE62564. SVR-NB identified a signature of 35 lncRNAs and achieved a mean squared correlation coefficient of 0.85 and a mean absolute error of 0.56 year between the actual and estimated overall survival time using 10-fold cross-validation. Further, we ranked and characterized the 35 lncRNAs according to their contribution towards the estimation accuracy. Functional annotations and co-expression gene analysis of LOC440896, LINC00632, and IGF2-AS revealed the association of co-expressed genes in Kyoto Encyclopedia of Genes and Genomes pathways.
Collapse
Grants
- This work was funded by Ministry of Science and Technology ROC under the contract numbers MOST 106-2634-F-075-001-, 106-2218-E-009-031-, 107-2221-E-009-154-, 107-2218-E-029-001-, and 107-2314-B-039-025-. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
- This work was funded by Ministry of Science and Technology ROC under the contract numbers MOST 107-2221-E-009 -154 &#x2013;, 107-2634-F-075 -001 &#x2013;, 107-2218-E-009 -005 &#x2013;, 107-2218-E-029 -001 &#x2013;, and 107-2319-B-400 -001 &#x2013;, and was financially supported by the &#x201C;Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B)&#x201D; from The Featured Areas Research Center Program within the framework of the Higher Education Sprout Project by the Ministry of Education (MOE) in Taiwan. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
Collapse
Affiliation(s)
| | - Divya Sahu
- Institute of Biomedical Informatics, Center for Systems and Synthetic Biology, National Yang-Ming University, Taipei, Taiwan
| | - Hsuan-Cheng Huang
- Institute of Biomedical Informatics, Center for Systems and Synthetic Biology, National Yang-Ming University, Taipei, Taiwan
- Bioinformatics Program, Taiwan International Graduate Program, Institute of Information Science, Academia Sinica, Taipei, Taiwan
| | - Yenching Lin
- Interdisciplinary Neuroscience Ph.D. Program, National Chiao Tung University, Hsinchu, Taiwan
| | - Shinn-Ying Ho
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, Taiwan.
- Bioinformatics Program, Taiwan International Graduate Program, Institute of Information Science, Academia Sinica, Taipei, Taiwan.
- Interdisciplinary Neuroscience Ph.D. Program, National Chiao Tung University, Hsinchu, Taiwan.
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan.
- Center For Intelligent Drug Systems and Smart Bio-devices (IDSB), National Chiao Tung University, Hsinchu, Taiwan.
| |
Collapse
|
16
|
Zhao Z, Partridge V, Sousares M, Shelton SD, Holland CL, Pertsemlidis A, Du L. microRNA-2110 functions as an onco-suppressor in neuroblastoma by directly targeting Tsukushi. PLoS One 2018; 13:e0208777. [PMID: 30550571 PMCID: PMC6294380 DOI: 10.1371/journal.pone.0208777] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 11/21/2018] [Indexed: 11/18/2022] Open
Abstract
microRNA-2110 (miR-2110) was previously identified as inducing neurite outgrowth in a neuroblastoma cell lines BE(2)-C, suggesting its differentiation-inducing and oncosuppressive function in neuroblastoma. In this study, we demonstrated that synthetic miR-2110 mimic had a generic effect on reducing cell survival in neuroblastoma cell lines with distinct genetic backgrounds, although the induction of cell differentiation traits varied between cell lines. In investigating the mechanisms underlying such functions of miR-2110, we identified that among its predicted target genes down-regulated by miR-2110, knockdown of Tsukushi (TSKU) expression showed the most potent effect in inducing cell differentiation and reducing cell survival, suggesting that TSKU protein plays a key role in mediating the functions of miR-2110. In investigating the clinical relevance of miR-2110 and TSKU expression in neuroblastoma patients, we found that low tumor miR-2110 levels were significantly correlated with high tumor TSKU mRNA levels, and that both low miR-2110 and high TSKU mRNA levels were significantly correlated with poor patient survival. These findings altogether support the oncosuppressive function of miR-2110 and suggest an important role for miR-2110 and its target TSKU in neuroblastoma tumorigenesis and in determining patient prognosis.
Collapse
Affiliation(s)
- Zhenze Zhao
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, Texas, United States of America
| | - Veronica Partridge
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, Texas, United States of America
| | - Michaela Sousares
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, Texas, United States of America
| | - Spencer D. Shelton
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, Texas, United States of America
| | - Cory L. Holland
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, Texas, United States of America
| | - Alexander Pertsemlidis
- Greehey Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, Texas, United States of America
- Department of Cell Systems and Anatomy, The University of Texas Health, San Antonio, Texas, United States of America
- Department of Pediatrics, The University of Texas Health, San Antonio, Texas, United States of America
| | - Liqin Du
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, Texas, United States of America
- * E-mail:
| |
Collapse
|
17
|
miR-542-5p Attenuates Fibroblast Activation by Targeting Integrin α6 in Silica-Induced Pulmonary Fibrosis. Int J Mol Sci 2018; 19:ijms19123717. [PMID: 30467286 PMCID: PMC6320929 DOI: 10.3390/ijms19123717] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 11/12/2018] [Accepted: 11/17/2018] [Indexed: 11/28/2022] Open
Abstract
Silicosis is a very serious occupational disease and it features pathological manifestations of inflammatory infiltration, excessive proliferation of fibroblasts and massive depositions of the extracellular matrix in the lungs. Recent studies described the roles of a variety of microRNAs (miRNAs) in fibrotic diseases. Here, we aimed to explore the potential mechanism of miR-542-5p in the activation of lung fibroblasts. To induce a pulmonary fibrosis mouse model, silica suspension and the miR-542-5p agomir were administered to mice by intratracheal instillation and tail vein injection. We found that miR-542-5p was significantly decreased in mouse fibrotic lung tissues and up-regulation of miR-542-5p visually attenuated a series of fibrotic lesions, including alveolar structural damage, alveolar interstitial thickening and silica-induced nodule formation. The down-regulation of miR-542-5p was also observed in mouse fibroblast (NIH-3T3) treated with transforming growth factor β1 (TGF-β1). The proliferation and migration ability of NIH-3T3 cells were also inhibited by the transfection of miR-542-5p mimic. Integrin α6 (Itga6), reported as a cell surface protein associated with fibroblast proliferation, was confirmed to be a direct target of miR-542-5p. The knockdown of Itga6 significantly inhibited the phosphorylation of FAK/PI3K/AKT. In conclusion, miR-542-5p has a potential function for reducing the proliferation of fibroblasts and inhibiting silica-induced pulmonary fibrosis, which might be partially realized by directly binding to Itga6. Our data suggested that miR-542-5p might be a new therapeutic target for silicosis or other pulmonary fibrosis.
Collapse
|
18
|
Li Q, Song S, Ni G, Li Y, Wang X. Serum miR-542-3p as a prognostic biomarker in osteosarcoma. Cancer Biomark 2018; 21:521-526. [PMID: 29103020 DOI: 10.3233/cbm-170255] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Emerging evidence has suggested that circulating microRNAs (miRNAs) in body fluids have novel diagnostic and prognostic significance for patients with malignant diseases. The lack of useful biomarkers is a crucial problem of osteosarcoma (OS); Previous study has reported that miR-542-3p was significantly upregulated in osteosarcoma tissues and miR-542-3p may be as an oncogene in osteosarcoma pathogenesis. In our study, we investigated the circulating miR-542-3p and its clinical relevance in osteosarcoma. METHODS Serum MiR-542-3p levels were determined by quantitative real-time PCR assays (qRT-PCR) in 76 patients with OS and 76 healthy volunteers. Patient survival analyses were performed by Kaplan-Meier analyses and Cox regression models. All statistical tests were two-sided. RESULTS It was observed that the serum levels of miR-542-3p was significantly higher in patients with OS compared with the control groups (P< 0.01). High serum of miR-542-3p was significantly associated with advanced tumor stage and shorter survival (P< 0.01). ROC curve analysis calculated the ideal miR-542-3p cut-off value of 0.84 in prediction of OS, with a sensitivity of 53.8%, specificity of 93.6%, positive predictive value of 87.3% and negative predictive value of 63.7%. CONCLUSIONS The results showed that serum miR-542-3p levels could serve as a non-invasive blood biomarker for tumor monitoring and prognostic prediction in osteosarcoma patients.
Collapse
Affiliation(s)
- Qicai Li
- Department of Traumatology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Shirong Song
- Department of Traumatology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.,Department of Clinical Laboratory, People's Hospital of Weifang, Weifang, Shandong, China
| | - Guangzhen Ni
- Department of Clinical Laboratory, People's Hospital of Weifang, Weifang, Shandong, China
| | - Yu Li
- Department of Traumatology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xiaohui Wang
- Department of Traumatology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.,Department of Traumatology, The Central Hospital of Linyi, Linyi, Yishui, Shandong, China
| |
Collapse
|
19
|
Sahu D, Ho SY, Juan HF, Huang HC. High-risk, Expression-Based Prognostic Long Noncoding RNA Signature in Neuroblastoma. JNCI Cancer Spectr 2018; 2:pky015. [PMID: 31360848 PMCID: PMC6649748 DOI: 10.1093/jncics/pky015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 03/13/2018] [Accepted: 03/29/2018] [Indexed: 12/19/2022] Open
Abstract
Background Current clinical risk factors stratify patients with neuroblastoma (NB) for appropriate treatments, yet patients with similar clinical behaviors evoke variable responses. MYCN amplification is one of the established drivers of NB and, when combined with high-risk displays, worsens outcomes. Growing high-throughput transcriptomics studies suggest long noncoding RNA (lncRNA) dysregulation in cancers, including NB. However, expression-based lncRNA signatures are altered by MYCN amplification, which is associated with high-risk, and patient prognosis remains limited. Methods We investigated RNA-seq-based expression profiles of lncRNAs in MYCN status and risk status in a discovery cohort (n = 493) and validated them in three independent cohorts. In the discovery cohort, a prognostic association of lncRNAs was determined by univariate Cox regression and integrated into a signature using the risk score method. A novel risk score threshold selection criterion was developed to stratify patients into risk groups. Outcomes by risk group and clinical subgroup were assessed using Kaplan-Meier survival curves and multivariable Cox regression. The performance of lncRNA signatures was evaluated by receiver operating characteristic curve. All statistical tests were two-sided. Results In the discovery cohort, 16 lncRNAs that were differentially expressed (fold change ≥ 2 and adjusted P ≤ 0.01) integrated into a prognostic signature. A high risk score group of lncRNA signature had poor event-free survival (EFS; P < 1E-16). Notably, lncRNA signature was independent of other clinical risk factors when predicting EFS (hazard ratio = 3.21, P = 5.95E-07). The findings were confirmed in independent cohorts (P = 2.86E-02, P = 6.18E-03, P = 9.39E-03, respectively). Finally, the lncRNA signature had higher accuracy for EFS prediction (area under the curve = 0.788, 95% confidence interval = 0.746 to 0.831). Conclusions Here, we report the first (to our knowledge) RNA-seq 16-lncRNA prognostic signature for NB that may contribute to precise clinical stratification and EFS prediction.
Collapse
Affiliation(s)
- Divya Sahu
- Institute of Bioinformatics and Systems Biology.,Bioinformatics Program, Taiwan International Graduate Program, Institute of Information Science, Academia Sinica, Taipei, Taiwan.,Institute of Biomedical Informatics, Center for Systems and Synthetic Biology, National Yang-Ming University, Taipei, Taiwan
| | - Shinn-Ying Ho
- Institute of Bioinformatics and Systems Biology.,Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan.,Bioinformatics Program, Taiwan International Graduate Program, Institute of Information Science, Academia Sinica, Taipei, Taiwan
| | - Hsueh-Fen Juan
- Department of Life Science, Institute of Molecular and Cellular Biology, Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | - Hsuan-Cheng Huang
- Bioinformatics Program, Taiwan International Graduate Program, Institute of Information Science, Academia Sinica, Taipei, Taiwan.,Institute of Biomedical Informatics, Center for Systems and Synthetic Biology, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
20
|
Li SH, Li JP, Chen L, Liu JL. miR-146a induces apoptosis in neuroblastoma cells by targeting BCL11A. Med Hypotheses 2018; 117:21-27. [PMID: 30077189 DOI: 10.1016/j.mehy.2018.05.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/13/2018] [Accepted: 05/25/2018] [Indexed: 12/27/2022]
Abstract
Aberrant expression of miR-146a has been reported to be involved in the progression and metastasis of various types of human cancers; however, its potential role in human neuroblastoma is still poorly understood. The purpose of our study was to investigate the molecular mechanism and possible role of miR-146a in human neuroblastoma. In this study, targeted genes were predicted by bioinformatic analysis and confirmed by dual-Luciferase reporter assay. The expression level of miR-146a in the human neuroblastoma SK-N-SH cell line was detected by quantitative RT-PCR. We used flow cytometric analysis to determine apoptosis and necrosis of SK-N-SH cells after transfection with miR-146a inhibitor, miR-146a mimic, and negative controls. The expression level of target genes was detected by RT-PCR and Western blotting. We identified BCL11A as a target of miR-146a. Thus, miR-146a targets the 3'UTR of BCL11A and inhibits its mRNA and protein expression. Overexpression of miR-146a can inhibit the growth and promote the apoptosis of human neuroblastoma SK-N-SH cells through inhibiting the expression of BCL11A. Furthermore, we found that upregulation of BCL11A by miR-146a inhibitor can promote SK-N-SH cells growth and protect SK-N-SH cells against apoptosis. Our results showed that miR-146a is a potential tumor suppressor gene in human neuroblastoma via directly targeting BCL11A. These findings suggest that miR-146a might be a new candidate target for treatment of human neuroblastoma.
Collapse
Affiliation(s)
- Sheng-Hua Li
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University in Nanning, China
| | - Jin-Pin Li
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University in Nanning, China
| | - Lan Chen
- Department of Internal Medicine, The Second Affiliated Hospital of Guangxi Medical University in Nanning, China
| | - Jing-Li Liu
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University in Nanning, China.
| |
Collapse
|
21
|
Stigliani S, Morandi F, Persico L, Lagazio C, Erminio G, Scaruffi P, Corrias MV. miRNA expression profile of bone marrow resident cells from children with neuroblastoma is not significantly different from that of healthy children. Oncotarget 2018; 9:19014-19025. [PMID: 29721180 PMCID: PMC5922374 DOI: 10.18632/oncotarget.24874] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 03/02/2018] [Indexed: 12/29/2022] Open
Abstract
The miRNA expression profiles of bone marrow resident cells from children with neuroblastoma were compared to that of healthy children. No significant difference was found between localized and metastatic neuroblastoma, or between children with neuroblastoma and healthy children. By considering the fold change we identified six miRNAs over-expressed by more than 150 fold in neuroblastoma. Validation confirmed miR-221 over-expression in BM resident cells from children with neuroblastoma, regardless of localized or metastatic disease. MiR-221 over-expression was unlikely derived from neuroblastoma primary tumors or from bone marrow-infiltrating metastatic cells, since neuroblastoma cells expressed lower or similar amount of miR-221 than BM cells, respectively. To get insight on the genes potentially regulated by miR-221 we merged the list of miR-221 potential targets with the genes under-expressed by BM resident cells from children with neuroblastoma, as compared with healthy children. In silico analysis demonstrated that none of the miR-221 target genes belonged to heme biosynthetic processes found altered in children with neuroblastoma, whereas two genes associated with mitochondria. However, the encoded proteins were not under-expressed in children with neuroblastoma, making unlikely that altered erythrocyte maturation in children with neuroblastoma was mediated by miR-221. In conclusion, miRNA expression profiles of BM resident cells from children with localized and metastatic neuroblastoma were similar to that of BM resident cells from healthy children. Moreover, miRNAs expressed by neuroblastoma primary tumors or by BM-infiltrating NB cells do not appear to be involved in mediating the functional defect of erythrocyte maturation recently observed in children with neuroblastoma.
Collapse
Affiliation(s)
- Sara Stigliani
- Physiopathology of Human Reproduction, Ospedale Policlinico San Martino, Genoa, Italy
| | - Fabio Morandi
- Experimental Therapy in Oncology, IRCCS Istituto Giannina Gaslini, Genoa, Italy.,Present address: Stem Cell Laboratory and Cell Therapy Center, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Luca Persico
- Department of Economy, University of Genoa, Genoa, Italy
| | | | - Giovanni Erminio
- Epidemiology, Biostatistics and Committees, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Paola Scaruffi
- Physiopathology of Human Reproduction, Ospedale Policlinico San Martino, Genoa, Italy
| | | |
Collapse
|
22
|
MicroRNA-193b-3p represses neuroblastoma cell growth via downregulation of Cyclin D1, MCL-1 and MYCN. Oncotarget 2018; 9:18160-18179. [PMID: 29719597 PMCID: PMC5915064 DOI: 10.18632/oncotarget.24793] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 02/28/2018] [Indexed: 12/29/2022] Open
Abstract
Neuroblastoma is the most common diagnosed tumor in infants and the second most common extracranial tumor of childhood. The survival rate of patients with high-risk neuroblastoma is still very low despite intensive multimodal treatments. Therefore, new treatment strategies are needed. In recent years, miRNA-based anticancer therapy has received growing attention. Advances in this novel treatment strategy strongly depends on the identification of candidate miRNAs with broad-spectrum antitumor activity. Here, we identify miR-193b as a miRNA with tumor suppressive properties. We show that miR-193b is expressed at low levels in neuroblastoma cell lines and primary tumor samples. Introduction of miR-193b mimics into nine neuroblastoma cell lines with distinct genetic characteristics significantly reduces cell growth in vitro independent of risk factors such as p53 functionality or MYCN amplification. Functionally, miR-193b induces a G1 cell cycle arrest and cell death in neuroblastoma cell lines by reducing the expression of MYCN, Cyclin D1 and MCL-1, three important oncogenes in neuroblastoma of which inhibition has shown promising results in preclinical testing. Therefore, we suggest that miR-193b may represent a new candidate for miRNA-based anticancer therapy in neuroblastoma.
Collapse
|
23
|
Which statistical significance test best detects oncomiRNAs in cancer tissues? An exploratory analysis. Oncotarget 2018; 7:85613-85623. [PMID: 27784000 PMCID: PMC5356763 DOI: 10.18632/oncotarget.12828] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 10/14/2016] [Indexed: 01/09/2023] Open
Abstract
MicroRNAs(miRNAs) often exert their oncogenic and tumor suppressor functions by suppressing protein-coding genes expressions in cancers and thus have a strong association with cancers' generation, development and metastasis. Through comprehensively understanding differentially expressed miRNAs (oncomiRNA) in tumor tissues, we can elucidate the underlying molecular mechanisms in tumorigenesis and develop novel strategies for cancer diagnosis and treatment. The differential expression of miRNAs can now be analyzed through numerous statistical significance tests based on different principles, which are also available in various R packages. However, the results can be notably different. In this study, we compared miRNAs obtained from 6 common significance tests/R packages (t-test, Limma, DESeq, edgeR, LRT and MARS) with the miRNAs archived in two databases; HMDD 2.0 database, which collects experimentally validated differentially expressed miRNAs, and Infer microRNA-disease association database, which contains the potential disease-associated miRNAs by network forecasting. Finally, we sought the MARS method in DEGseq package more effectively searched out differentially expressed miRNAs than other common methods.
Collapse
|
24
|
Zhu QN, Renaud H, Guo Y. Bioinformatics-based identification of miR-542-5p as a predictive biomarker in breast cancer therapy. Hereditas 2018; 155:17. [PMID: 29371858 PMCID: PMC5769523 DOI: 10.1186/s41065-018-0055-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 01/04/2018] [Indexed: 01/04/2023] Open
Abstract
Background Tamoxifen is the first-line hormone therapy for estrogen receptor alpha positive (ERα+) breast cancer. However, about 40% of patients with ERα + breast cancer who receive tamoxifen therapy eventually develop resistance resulting in a poor prognosis. The aim of this study was to mine available data sets in the Gene Expression Omnibus (GEO) database, including in vitro (cell lines) and in vivo (tissue samples), and to identify all miRNAs associated with tamoxifen resistance (TamR) in breast cancer. Secondly, this study aimed to predict the key gene regulatory networks of newly found TamR-related miRNAs and evaluate the potential role of the miRNAs and targets as potential prognosis biomarkers for breast cancer patients. Result Microarray data sets from two different studies were used from the GEO database: 1. GSE66607: miRNA of MCF-7 TamR cells; 2. GSE37405: TamR tissues. Differentially expressed microRNAs (miRNAs) were identified in both data sets and 5 differentially expressed miRNAs were found to overlap between the two data sets. Profiles of GSE37405 and data from the Kaplan-Meier Plotter Database (KMPD) along with Gene Expression Profiling Interactive Analysis (GEPIA) were used to reveal the relationship between these 5 miRNAs and overall survival. The results showed that has-miR-542-5p was the only miRNA associated with overall survival of ERα + breast cancer patients who received adjuvant tamoxifen. Targets of has-miR-542-5p were predicted by miRanda and TargetScan, and the mRNA expression of the three 3 target gene, Tyrosine 3-Monooxygenase/Tryptophan 5-Monooxygenase Activation Protein Beta (YWHAB), Lymphocyte Antigen 9 (LY9), and Secreted Frizzled Related Protein 1 (SFRP1) were associated with overall survival in 2 different databases. Copy-number alterations (CNAs) of SFRP1 confer survival disadvantage to breast cancer patients and alter the mRNA expression of SFRP1 in cBioPortal database. Conclusion This study indicates that miRNA has-miR-542-5p is associated with TamR and can predict prognosis of breast cancer patients. Furthermore, has-miR-542-5p may be acting through a mechanism involving the target genes YWHAB, LY9, and SFRP1. Overall, has-miR-542-5p is a predictive biomarker and potential target for therapy of breast cancer patients. Electronic supplementary material The online version of this article (10.1186/s41065-018-0055-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Qiong-Ni Zhu
- 1Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008 People's Republic of China.,2Institute of Clinical Pharmacology, Central South University, Changsha, 410078 People's Republic of China.,3Hunan Key Laboratory of Pharmacogenetics, Changsha, 410078 People's Republic of China
| | - Helen Renaud
- 4University of Kansas Medical Center, Kansas City, KS 66160 USA
| | - Ying Guo
- 1Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008 People's Republic of China.,2Institute of Clinical Pharmacology, Central South University, Changsha, 410078 People's Republic of China.,3Hunan Key Laboratory of Pharmacogenetics, Changsha, 410078 People's Republic of China
| |
Collapse
|
25
|
Zammit V, Baron B, Ayers D. MiRNA Influences in Neuroblast Modulation: An Introspective Analysis. Genes (Basel) 2018; 9:genes9010026. [PMID: 29315268 PMCID: PMC5793179 DOI: 10.3390/genes9010026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 12/22/2017] [Accepted: 12/29/2017] [Indexed: 02/07/2023] Open
Abstract
Neuroblastoma (NB) is the most common occurring solid paediatric cancer in children under the age of five years. Whether of familial or sporadic origin, chromosome abnormalities contribute to the development of NB and cause dysregulation of microRNAs (miRNAs). MiRNAs are small non-coding, single stranded RNAs that target messenger RNAs at the post-transcriptional levels by repressing translation within all facets of human physiology. Such gene 'silencing' activities by miRNAs allows the development of regulatory feedback loops affecting multiple functions within the cell, including the possible differentiation of neural stem cell (NSC) lineage selection. Neurogenesis includes stages of self-renewal and fate specification of NSCs, migration and maturation of young neurones, and functional integration of new neurones into the neural circuitry, all of which are regulated by miRNAs. The role of miRNAs and their interaction in cellular processes are recognised aspects of cancer genetics, and miRNAs are currently employed as biomarkers for prognosis and tumour characterisation in multiple cancer models. Consequently, thorough understanding of the mechanisms of how these miRNAs interplay at the transcriptomic level will definitely lead to the development of novel, bespoke and efficient therapeutic measures, with this review focusing on the influences of miRNAs on neuroblast modulations leading to neuroblastoma.
Collapse
Affiliation(s)
- Vanessa Zammit
- National Blood Transfusion Service, St. Luke's Hospital, PTA1010 G'Mangia, Malta.
- School of Biomedical Science and Physiology, University of Wolverhampton, Wolverhampton WV1 1LY, UK.
| | - Byron Baron
- Centre for Molecular Medicine and Biobanking, Faculty of Medicine and Surgery, University of Malta, MSD2080 Msida, Malta.
| | - Duncan Ayers
- Centre for Molecular Medicine and Biobanking, Faculty of Medicine and Surgery, University of Malta, MSD2080 Msida, Malta.
- School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK.
| |
Collapse
|
26
|
Naraparaju K, Kolla V, Zhuang T, Higashi M, Iyer R, Kolla S, Okawa ER, Blobel GA, Brodeur GM. Role of microRNAs in epigenetic silencing of the CHD5 tumor suppressor gene in neuroblastomas. Oncotarget 2017; 7:15977-85. [PMID: 26895110 PMCID: PMC4941291 DOI: 10.18632/oncotarget.7434] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 02/05/2016] [Indexed: 02/07/2023] Open
Abstract
Neuroblastoma (NB), a tumor of the sympathetic nervous system, is the most common extracranial solid tumor of childhood. We and others have identified distinct patterns of genomic change that underlie diverse clinical behaviors, from spontaneous regression to relentless progression. We first identified CHD5 as a tumor suppressor gene that is frequently deleted in NBs. Mutation of the remaining CHD5 allele is rare in these tumors, yet expression is very low or absent, so expression is likely regulated by epigenetic mechanisms. In order to understand the potential role of miRNA regulation of CHD5 protein expression in NBs, we examined all miRNAs that are predicted to target the 3′-UTR using miRanda, TargetScan and other algorithms. We identified 18 miRNAs that were predicted by 2 or more programs: miR-204, -211, -216b, -17, -19ab, -20ab, -93, -106ab, -130ab, -301ab, -454, -519d, -3666. We then performed transient transfections in two NB cell lines, NLF (MYCN amplified) and SY5Y (MYCN non-amplified), with the reporter plasmid and miRNA mimic, as well as appropriate controls. We found seven miRNAs that significantly downregulated CHD5 expression in NB: miR-211, 17, -93, -20b, -106b, -204, and -3666. Interestingly, MYCN upregulates several of the candidates we identified: miR-17, -93, -106b & -20b. This suggests that miRNAs driven by MYCN and other genes represent a potential epigenetic mechanism to regulate CHD5 expression.
Collapse
Affiliation(s)
- Koumudi Naraparaju
- Division of Oncology and Hematology, The Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA
| | - Venkatadri Kolla
- Division of Oncology and Hematology, The Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA
| | - Tiangang Zhuang
- Division of Oncology and Hematology, The Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA
| | - Mayumi Higashi
- Division of Oncology and Hematology, The Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA
| | - Radhika Iyer
- Division of Oncology and Hematology, The Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA
| | - Sriharsha Kolla
- Division of Oncology and Hematology, The Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA
| | - Erin R Okawa
- Division of Oncology and Hematology, The Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA
| | - Gerd A Blobel
- Department of Pediatrics, The Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA
| | - Garrett M Brodeur
- Division of Oncology and Hematology, The Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
27
|
Moreno-Smith M, Lakoma A, Chen Z, Tao L, Scorsone KA, Schild L, Aviles-Padilla K, Nikzad R, Zhang Y, Chakraborty R, Molenaar JJ, Vasudevan SA, Sheehan V, Kim ES, Paust S, Shohet JM, Barbieri E. p53 Nongenotoxic Activation and mTORC1 Inhibition Lead to Effective Combination for Neuroblastoma Therapy. Clin Cancer Res 2017; 23:6629-6639. [PMID: 28821555 DOI: 10.1158/1078-0432.ccr-17-0668] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 06/26/2017] [Accepted: 08/11/2017] [Indexed: 12/14/2022]
Abstract
Purpose: mTORC1 inhibitors are promising agents for neuroblastoma therapy; however, they have shown limited clinical activity as monotherapy, thus rational drug combinations need to be explored to improve efficacy. Importantly, neuroblastoma maintains both an active p53 and an aberrant mTOR signaling.Experimental Design: Using an orthotopic xenograft model and modulating p53 levels, we investigated the antitumor effects of the mTORC1 inhibitor temsirolimus in neuroblastoma expressing normal, decreased, or mutant p53, both as single agent and in combination with first- and second-generation MDM2 inhibitors to reactivate p53.Results: Nongenotoxic p53 activation suppresses mTOR activity. Moreover, p53 reactivation via RG7388, a second-generation MDM2 inhibitor, strongly enhances the in vivo antitumor activity of temsirolimus. Single-agent temsirolimus does not elicit apoptosis, and tumors rapidly regrow after treatment suspension. In contrast, our combination therapy triggers a potent apoptotic response in wild-type p53 xenografts and efficiently blocks tumor regrowth after treatment completion. We also found that this combination uniquely led to p53-dependent suppression of survivin whose ectopic expression is sufficient to rescue the apoptosis induced by our combination.Conclusions: Our study supports a novel highly effective strategy that combines RG7388 and temsirolimus in wild-type p53 neuroblastoma, which warrants testing in early-phase clinical trials. Clin Cancer Res; 23(21); 6629-39. ©2017 AACR.
Collapse
Affiliation(s)
- Myrthala Moreno-Smith
- Department of Pediatrics, Section of Hematology-Oncology, Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas
| | - Anna Lakoma
- Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
| | - Zaowen Chen
- Department of Pediatrics, Section of Hematology-Oncology, Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas
| | - Ling Tao
- Department of Pediatrics, Section of Hematology-Oncology, Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas
| | - Kathleen A Scorsone
- Department of Pediatrics, Section of Hematology-Oncology, Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas
| | - Linda Schild
- Department of Oncogenomics, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Kevin Aviles-Padilla
- Department of Pediatrics, Center for Human Immunobiology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - Rana Nikzad
- Department of Pediatrics, Center for Human Immunobiology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - Yankai Zhang
- Department of Pediatrics, Section of Hematology-Oncology, Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas
| | - Rikhia Chakraborty
- Department of Pediatrics, Section of Hematology-Oncology, Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas
| | - Jan J Molenaar
- Department of Oncogenomics, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Sanjeev A Vasudevan
- Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
| | - Vivien Sheehan
- Department of Pediatrics, Section of Hematology-Oncology, Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas
| | - Eugene S Kim
- Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
| | - Silke Paust
- Department of Pediatrics, Center for Human Immunobiology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - Jason M Shohet
- Department of Pediatrics, Section of Hematology-Oncology, Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas
| | - Eveline Barbieri
- Department of Pediatrics, Section of Hematology-Oncology, Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
28
|
Samaraweera L, Spengler BA, Ross RA. Reciprocal antagonistic regulation of N-myc mRNA by miR‑17 and the neuronal-specific RNA-binding protein HuD. Oncol Rep 2017; 38:545-550. [PMID: 28560387 PMCID: PMC5492688 DOI: 10.3892/or.2017.5664] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 11/15/2016] [Indexed: 12/12/2022] Open
Abstract
Neuroblastoma is a childhood cancer originating from embryonic neural crest cells. Amplification of the proto-oncogene N-myc, seen in ~30% of neuroblastoma tumors, is a marker for poor prognosis. Recently discovered small regulatory RNAs, microRNAs (miRNAs), are implicated in cancers, including neuroblastoma. miRNAs downregulate the expression of genes by binding to the 3-untranslated regions (3′-UTRs), thereby inhibiting translation or inducing degradation of cognate mRNAs. Our study sought to identify miRNAs that regulate N-myc expression and thereby malignancy in neuroblastoma. miRNAs whose expression negatively correlates with N-myc expression were identified from a miRNA microarray of 4 N-myc-amplified neuroblastoma cell lines. Three of these miRNAs (miR-17, miR-20a and miR-18a) belong to the miR-17-92 cluster, previously shown to be upregulated by N-myc. qPCR validation of these miRNAs in a larger panel of cell lines revealed that levels of miR-17 were inversely proportional to N-myc mRNA amounts in the N-myc-amplified cell lines. Notably, miR-17 also downregulated N-myc protein synthesis in the N-myc-amplified cells, thereby generating a negative feedback regulatory loop between the proto-oncogene and this miRNA. Moreover, the neuronal-specific RNA-binding protein HuD (ELAVL4), which regulates the processing/stability of N-myc mRNA, competes with miR-17 for a binding site in the 3′-UTR of N-myc. Thus, N-myc levels appear to be modulated by the antagonistic interactions of both miR-17, as a negative regulator, and HuD, as a positive regulator, providing further evidence of the complex cellular control mechanisms of this oncogene in N-myc-amplified neuroblastoma cells.
Collapse
Affiliation(s)
- Leleesha Samaraweera
- Department of Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Barbara A Spengler
- Department of Biological Sciences, Fordham University, Bronx, NY 10458, USA
| | - Robert A Ross
- Department of Biological Sciences, Fordham University, Bronx, NY 10458, USA
| |
Collapse
|
29
|
Althoff K, Schulte JH, Schramm A. Towards diagnostic application of non-coding RNAs in neuroblastoma. Expert Rev Mol Diagn 2016; 16:1307-1313. [PMID: 27813435 DOI: 10.1080/14737159.2016.1256207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Neuroblastoma is a solid cancer of childhood, which is devastating upon recurrence. Markers for minimal residual disease and early detection of relapse are eagerly awaited to improve the outcome of affected patients. Several miRNAs have been identified as key regulators of neuroblastoma pathogenesis. Areas covered: Here, we focus on miRNAs that have been linked to MYCN, a prominent oncogenic driver, and we review the hitherto known interactions between miRNAs and other important players in neuroblastoma. Expert commentary: Existing diagnostic miRNA signatures remain to be established in clinical settings. Moreover, inhibition of individual oncogenic miRNAs or enhancement of tumor suppressive miRNA function could represent a new therapeutic approach in cancer treatment, including NB.
Collapse
Affiliation(s)
- Kristina Althoff
- a Department of Pediatric Oncology and Hematology , University Children's Hospital Essen , Essen , Germany
| | - Johannes H Schulte
- b Department of Pediatric Oncology and Hematology , Charité University Medicine , Berlin , Germany.,c Berlin Institute of Health (BIH) , Germany.,d German Cancer Consortium (DKTK Berlin) , Germany
| | - Alexander Schramm
- a Department of Pediatric Oncology and Hematology , University Children's Hospital Essen , Essen , Germany
| |
Collapse
|
30
|
Stigliani S, Scaruffi P, Lagazio C, Persico L, Carlini B, Varesio L, Morandi F, Morini M, Gigliotti AR, Esposito MR, Viscardi E, Cecinati V, Conte M, Corrias MV. Deregulation of focal adhesion pathway mediated by miR-659-3p is implicated in bone marrow infiltration of stage M neuroblastoma patients. Oncotarget 2016; 6:13295-308. [PMID: 25980492 PMCID: PMC4537015 DOI: 10.18632/oncotarget.3745] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 04/08/2015] [Indexed: 11/25/2022] Open
Abstract
To get insights on the metastatic process of human neuroblastoma (NB), the miRNA expression profile of bone marrow (BM)-infiltrating cells has been determined and compared to that of primary tumors.Twenty-two BM-infiltrating cells, 22 primary tumors, and 4 paired samples from patients with metastatic NB aged > 12 months were analyzed for the expression of 670 miRNAs by stem-loop RT-qPCR. The miRNAs whose expression was significantly different were subjected to selection criteria, and 20 selected miRNAs were tested in 10 additional BM-infiltrating cells and primary tumors. Among the miRNAs confirmed to be differentially expressed, miR-659-3p was further analyzed. Transfection of miR-659-3p mimic and inhibitor demonstrated the specific suppression and over-expression, respectively, of the miR-659-3p target gene CNOT1, a regulator of transcription of genes containing AU-rich element (ARE) sequence. Among the ARE-containing genes, miR-659-3p mimic and inhibitor specifically modified the expression of AKT3, BCL2, CYR61 and THSB2, belonging to the focal adhesion pathway. Most importantly, in BM-infiltrating cells CNOT1 expression was significantly higher, and that of AKT3, BCL2, THSB2 and CYR61 was significantly lower than in primary tumors. Thus, our study suggests a role of the focal adhesion pathway, regulated by miR-659-3p through CNOT1, in the human NB metastatic process.
Collapse
Affiliation(s)
- Sara Stigliani
- U.O.S. Physiopathology of Human Reproduction, IRCCS A.O.U. San Martino-IST, Genova, Italy
| | - Paola Scaruffi
- U.O.S. Physiopathology of Human Reproduction, IRCCS A.O.U. San Martino-IST, Genova, Italy
| | | | - Luca Persico
- Department of Economy, University of Genoa, Genova, Italy
| | - Barbara Carlini
- Laboratory of Oncology, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Luigi Varesio
- Laboratory of Molecular Biology, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Fabio Morandi
- Laboratory of Oncology, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Martina Morini
- Laboratory of Molecular Biology, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Anna Rita Gigliotti
- Epidemiology, Biostatistics and Committees Unit, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Maria Rosaria Esposito
- Neuroblastoma Laboratory, Pediatric Research Institute, Fondazione Città della Speranza, Padova, Italy
| | - Elisabetta Viscardi
- Pediatric Hematology and Oncology Division, Padova University Hospital, Padova, Italy
| | - Valerio Cecinati
- UOS Divisione Oncoematologia Pediatrica, Ospedale Civile di Pescara, Pescara, Italy
| | - Massimo Conte
- Oncology Unit, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | | |
Collapse
|
31
|
Beckers A, Van Peer G, Carter DR, Mets E, Althoff K, Cheung BB, Schulte JH, Mestdagh P, Vandesompele J, Marshall GM, De Preter K, Speleman F. MYCN-targeting miRNAs are predominantly downregulated during MYCN‑driven neuroblastoma tumor formation. Oncotarget 2016; 6:5204-16. [PMID: 25294817 PMCID: PMC4467143 DOI: 10.18632/oncotarget.2477] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 09/15/2014] [Indexed: 12/25/2022] Open
Abstract
MYCN is a transcription factor that plays key roles in both normal development and cancer. In neuroblastoma, MYCN acts as a major oncogenic driver through pleiotropic effects regulated by multiple protein encoding genes as well as microRNAs (miRNAs). MYCN activity is tightly controlled at the level of transcription and protein stability through various mechanisms. Like most genes, MYCN is further controlled by miRNAs, but the full complement of all miRNAs implicated in this process has not been determined through an unbiased approach. To elucidate the role of miRNAs in regulation of MYCN, we thus explored the MYCN-miRNA interactome to establish miRNAs controlling MYCN expression levels. We combined results from an unbiased and genome-wide high-throughput miRNA target reporter screen with miRNA and mRNA expression data from patients and a murine neuroblastoma progression model. We identified 29 miRNAs targeting MYCN, of which 12 miRNAs are inversely correlated with MYCN expression or activity in neuroblastoma tumor tissue. The majority of MYCN-targeting miRNAs in neuroblastoma showed a decrease in expression during murine MYCN-driven neuroblastoma tumor development. Therefore, we provide evidence that MYCN-targeting miRNAs are preferentially downregulated in MYCN-driven neuroblastoma, suggesting that MYCN negatively controls the expression of these miRNAs, to safeguard its expression.
Collapse
Affiliation(s)
- Anneleen Beckers
- Center for Medical Genetics (CMGG), Ghent University, Ghent, Belgium
| | - Gert Van Peer
- Center for Medical Genetics (CMGG), Ghent University, Ghent, Belgium
| | - Daniel R Carter
- Children's Cancer Institute, University of New South Wales, Sydney, Australia
| | - Evelien Mets
- Center for Medical Genetics (CMGG), Ghent University, Ghent, Belgium
| | - Kristina Althoff
- Department of Pediatric Oncology and Hematology, University Children's Hospital Essen, Essen, Germany.,German Cancer Consortium (DKTK), Germany
| | - Belamy B Cheung
- Children's Cancer Institute, University of New South Wales, Sydney, Australia
| | - Johannes H Schulte
- Department of Pediatric Oncology and Hematology, University Children's Hospital Essen, Essen, Germany.,German Cancer Consortium (DKTK), Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Translational Neuro-Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Pieter Mestdagh
- Center for Medical Genetics (CMGG), Ghent University, Ghent, Belgium
| | - Jo Vandesompele
- Center for Medical Genetics (CMGG), Ghent University, Ghent, Belgium
| | - Glenn M Marshall
- Children's Cancer Institute, University of New South Wales, Sydney, Australia.,Kids Cancer Centre, Sydney Children's Hospital, Sydney, Australia
| | - Katleen De Preter
- Center for Medical Genetics (CMGG), Ghent University, Ghent, Belgium
| | - Frank Speleman
- Center for Medical Genetics (CMGG), Ghent University, Ghent, Belgium
| |
Collapse
|
32
|
Jones TA, Jeyapalan JN, Forshew T, Tatevossian RG, Lawson ARJ, Patel SN, Doctor GT, Mumin MA, Picker SR, Phipps KP, Michalski A, Jacques TS, Sheer D. Molecular analysis of pediatric brain tumors identifies microRNAs in pilocytic astrocytomas that target the MAPK and NF-κB pathways. Acta Neuropathol Commun 2015; 3:86. [PMID: 26682910 PMCID: PMC4683939 DOI: 10.1186/s40478-015-0266-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Accepted: 12/05/2015] [Indexed: 12/17/2022] Open
Abstract
Introduction Pilocytic astrocytomas are slow-growing tumors that usually occur in the cerebellum or in the midline along the hypothalamic/optic pathways. The most common genetic alterations in pilocytic astrocytomas activate the ERK/MAPK signal transduction pathway, which is a major driver of proliferation but is also believed to induce senescence in these tumors. Here, we have conducted a detailed investigation of microRNA and gene expression, together with pathway analysis, to improve our understanding of the regulatory mechanisms in pilocytic astrocytomas. Results Pilocytic astrocytomas were found to have distinctive microRNA and gene expression profiles compared to normal brain tissue and a selection of other pediatric brain tumors. Several microRNAs found to be up-regulated in pilocytic astrocytomas are predicted to target the ERK/MAPK and NF-κB signaling pathways as well as genes involved in senescence-associated inflammation and cell cycle control. Furthermore, IGFBP7 and CEBPB, which are transcriptional inducers of the senescence-associated secretory phenotype (SASP), were also up-regulated together with the markers of senescence and inflammation, CDKN1A (p21), CDKN2A (p16) and IL1B. Conclusion These findings provide further evidence of a senescent phenotype in pilocytic astrocytomas. In addition, they suggest that the ERK/MAPK pathway, which is considered the major driver of these tumors, is regulated not only by genetic aberrations but also by microRNAs. Electronic supplementary material The online version of this article (doi:10.1186/s40478-015-0266-3) contains supplementary material, which is available to authorized users.
Collapse
|
33
|
De Falco G, Ambrosio MR, Fuligni F, Onnis A, Bellan C, Rocca BJ, Navari M, Etebari M, Mundo L, Gazaneo S, Facchetti F, Pileri SA, Leoncini L, Piccaluga PP. Burkitt lymphoma beyond MYC translocation: N-MYC and DNA methyltransferases dysregulation. BMC Cancer 2015; 15:668. [PMID: 26453442 PMCID: PMC4600215 DOI: 10.1186/s12885-015-1661-7] [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: 01/26/2015] [Accepted: 09/28/2015] [Indexed: 12/04/2022] Open
Abstract
Background The oncogenic transcription factor MYC is pathologically activated in many human malignancies. A paradigm for MYC dysregulation is offered by Burkitt lymphoma, where chromosomal translocations leading to Immunoglobulin gene-MYC fusion are the crucial initiating oncogenic events. However, Burkitt lymphoma cases with no detectable MYC rearrangement but maintaining MYC expression have been identified and alternative mechanisms can be involved in MYC dysregulation in these cases. Methods We studied the microRNA profile of MYC translocation-positive and MYC translocation-negative Burkitt lymphoma cases in order to uncover possible differences at the molecular level. Data was validated at the mRNA and protein level by quantitative Real-Time polymerase chain reaction and immunohistochemistry, respectively. Results We identified four microRNAs differentially expressed between the two groups. The impact of these microRNAs on the expression of selected genes was then investigated. Interestingly, in MYC translocation-negative cases we found over-expression of DNA-methyl transferase family members, consistent to hypo-expression of the hsa-miR-29 family. This finding suggests an alternative way for the activation of lymphomagenesis in these cases, based on global changes in methylation landscape, aberrant DNA hypermethylation, lack of epigenetic control on transcription of targeted genes, and increase of genomic instability. In addition, we observed an over-expression of another MYC family gene member, MYCN that may therefore represent a cooperating mechanism of MYC in driving the malignant transformation in those cases lacking an identifiable MYC translocation but expressing the gene at the mRNA and protein levels. Conclusions Collectively, our results showed that MYC translocation-positive and MYC translocation-negative Burkitt lymphoma cases are slightly different in terms of microRNA and gene expression. MYC translocation-negative Burkitt lymphoma, similarly to other aggressive B-cell non Hodgkin’s lymphomas, may represent a model to understand the intricate molecular pathway responsible for MYC dysregulation in cancer. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1661-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Giulia De Falco
- Department of Medical Biotechnologies, University of Siena, Italy - Via delle Scotte, 6 - 53100, Siena, Italy. .,School of Biological and Chemical Sciences, Queen Mary University of London, London, UK.
| | - Maria Raffaella Ambrosio
- Department of Medical Biotechnologies, University of Siena, Italy - Via delle Scotte, 6 - 53100, Siena, Italy.
| | - Fabio Fuligni
- Department of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, Via Zamboni, 33, 40126, Bologna, Italy.
| | - Anna Onnis
- Department of Medical Biotechnologies, University of Siena, Italy - Via delle Scotte, 6 - 53100, Siena, Italy.
| | - Cristiana Bellan
- Department of Medical Biotechnologies, University of Siena, Italy - Via delle Scotte, 6 - 53100, Siena, Italy.
| | - Bruno Jim Rocca
- Department of Medical Biotechnologies, University of Siena, Italy - Via delle Scotte, 6 - 53100, Siena, Italy.
| | - Mohsen Navari
- Department of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, Via Zamboni, 33, 40126, Bologna, Italy.
| | - Maryam Etebari
- Department of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, Via Zamboni, 33, 40126, Bologna, Italy.
| | - Lucia Mundo
- Department of Medical Biotechnologies, University of Siena, Italy - Via delle Scotte, 6 - 53100, Siena, Italy.
| | - Sara Gazaneo
- Department of Medical Biotechnologies, University of Siena, Italy - Via delle Scotte, 6 - 53100, Siena, Italy.
| | - Fabio Facchetti
- Unit of Pathology, Brescia University, Piazza del Mercato, 15, Brescia, Italy.
| | - Stefano A Pileri
- Department of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, Via Zamboni, 33, 40126, Bologna, Italy.
| | - Lorenzo Leoncini
- Department of Medical Biotechnologies, University of Siena, Italy - Via delle Scotte, 6 - 53100, Siena, Italy.
| | - Pier Paolo Piccaluga
- Department of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, Via Zamboni, 33, 40126, Bologna, Italy.
| |
Collapse
|
34
|
Liu J, Li Y. Trichostatin A and Tamoxifen inhibit breast cancer cell growth by miR-204 and ERα reducing AKT/mTOR pathway. Biochem Biophys Res Commun 2015; 467:242-7. [PMID: 26436206 DOI: 10.1016/j.bbrc.2015.09.182] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 09/29/2015] [Indexed: 02/07/2023]
Abstract
MicroRNAs (miRs) are small non-coding RNAs aberrantly expressed in human tumors. Increasing evidence suggests that miRNAs are functionally important in cancers. We demonstrated miR-204 exerts its function by targeting gene involved in tumor growth and chemotherapy drugs reactivity. Here, we show that Trichostatin A (TSA) could increase ERα expression in MCF-7 and MDA-MB-231 cells by reducing miR204. Analysis of tumors growth inhibition shows that TSA promotes ERα expression, which could be reversed by miR-204 mimic transfection. When miR-204 is down regulated, the inhibition of TAM on breast cancer cells is enhanced. Caspase 3 activity is also increased. TSA and TAM combination inhibits Mcl-1 expression by decreasing phosphorylation of AKT induced by ERα increase in vivo and in vitro.
Collapse
Affiliation(s)
- Junbiao Liu
- Department of Oncology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| | - Yan Li
- Department of Oncology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| |
Collapse
|
35
|
Schwarzl T, Higgins DG, Kolch W, Duffy DJ. Measuring Transcription Rate Changes via Time-Course 4-Thiouridine Pulse-Labelling Improves Transcriptional Target Identification. J Mol Biol 2015; 427:3368-74. [PMID: 26362006 DOI: 10.1016/j.jmb.2015.09.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 09/01/2015] [Accepted: 09/02/2015] [Indexed: 10/23/2022]
Abstract
Identifying changes in the transcriptional regulation of target genes from high-throughput studies is important for unravelling molecular mechanisms controlled by a given perturbation. When measuring global transcript levels only, the effect of the perturbation [e.g., transcription factor (TF) overexpression or drug treatment] on its target genes is often obscured by delayed feedback and secondary effects until the changes are fully propagated. As a proof of principle, we show that selective measuring of transcripts that are only synthesised after a perturbation [4-thiouridine (4sU) sequencing (4sU-seq)] is a more sensitive method to identify targets and time-dependent transcriptional responses than global transcript profiling. By metabolically labelling RNA in a time-course setup, we could vastly increase the sensitivity of MYCN target gene detection compared to traditional RNA sequencing. The validity of targets identified by 4sU-seq was demonstrated using chromatin immunoprecipitation sequencing and neuroblastoma microarray tumour data. Here, we describe the methodology, both molecular biology and computational aspects, required to successfully apply this 4sU-seq approach.
Collapse
Affiliation(s)
- Thomas Schwarzl
- Systems Biology Ireland, Conway Institute of Biomolecular and Biomedical Research and School of Medicine and Medical Science, University College Dublin Conway Institute, Belfield, Dublin 4, Ireland.
| | - Desmond G Higgins
- Systems Biology Ireland, Conway Institute of Biomolecular and Biomedical Research and School of Medicine and Medical Science, University College Dublin Conway Institute, Belfield, Dublin 4, Ireland
| | - Walter Kolch
- Systems Biology Ireland, Conway Institute of Biomolecular and Biomedical Research and School of Medicine and Medical Science, University College Dublin Conway Institute, Belfield, Dublin 4, Ireland
| | - David J Duffy
- Systems Biology Ireland, Conway Institute of Biomolecular and Biomedical Research and School of Medicine and Medical Science, University College Dublin Conway Institute, Belfield, Dublin 4, Ireland
| |
Collapse
|
36
|
Pinto NR, Applebaum MA, Volchenboum SL, Matthay KK, London WB, Ambros PF, Nakagawara A, Berthold F, Schleiermacher G, Park JR, Valteau-Couanet D, Pearson ADJ, Cohn SL. Advances in Risk Classification and Treatment Strategies for Neuroblastoma. J Clin Oncol 2015; 33:3008-17. [PMID: 26304901 DOI: 10.1200/jco.2014.59.4648] [Citation(s) in RCA: 596] [Impact Index Per Article: 59.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Risk-based treatment approaches for neuroblastoma have been ongoing for decades. However, the criteria used to define risk in various institutional and cooperative groups were disparate, limiting the ability to compare clinical trial results. To mitigate this problem and enhance collaborative research, homogenous pretreatment patient cohorts have been defined by the International Neuroblastoma Risk Group classification system. During the past 30 years, increasingly intensive, multimodality approaches have been developed to treat patients who are classified as high risk, whereas patients with low- or intermediate-risk neuroblastoma have received reduced therapy. This treatment approach has resulted in improved outcome, although survival for high-risk patients remains poor, emphasizing the need for more effective treatments. Increased knowledge regarding the biology and genetic basis of neuroblastoma has led to the discovery of druggable targets and promising, new therapeutic approaches. Collaborative efforts of institutions and international cooperative groups have led to advances in our understanding of neuroblastoma biology, refinements in risk classification, and stratified treatment strategies, resulting in improved outcome. International collaboration will be even more critical when evaluating therapies designed to treat small cohorts of patients with rare actionable mutations.
Collapse
Affiliation(s)
- Navin R Pinto
- Navin R. Pinto, Mark A. Applebaum, Samuel L. Volchenboum, and Susan L. Cohn, Comer Children's Hospital, University of Chicago, Chicago, IL; Katherine K. Matthay, University of California San Francisco (UCSF) Benioff Children's Hospital, UCSF School of Medicine, San Francisco, CA; Wendy B. London, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA; Peter F. Ambros, Children's Cancer Research Institute, St Anna Kinderkrebsforschung, Vienna, Austria; Akira Nakagawara, Saga Medical Center Koseikan, Saga, Japan; Frank Berthold, Children's Hospital, University of Cologne, Koln, Germany; Gudrun Schleiermacher, Institut Curie, Paris; Dominique Valteau-Couanet, Gustave Roussy, Villejuif, France; Julie R. Park, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; and Andrew D.J. Pearson, Institute of Cancer Research and Royal Marsden Hospital, Surrey, United Kingdom
| | - Mark A Applebaum
- Navin R. Pinto, Mark A. Applebaum, Samuel L. Volchenboum, and Susan L. Cohn, Comer Children's Hospital, University of Chicago, Chicago, IL; Katherine K. Matthay, University of California San Francisco (UCSF) Benioff Children's Hospital, UCSF School of Medicine, San Francisco, CA; Wendy B. London, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA; Peter F. Ambros, Children's Cancer Research Institute, St Anna Kinderkrebsforschung, Vienna, Austria; Akira Nakagawara, Saga Medical Center Koseikan, Saga, Japan; Frank Berthold, Children's Hospital, University of Cologne, Koln, Germany; Gudrun Schleiermacher, Institut Curie, Paris; Dominique Valteau-Couanet, Gustave Roussy, Villejuif, France; Julie R. Park, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; and Andrew D.J. Pearson, Institute of Cancer Research and Royal Marsden Hospital, Surrey, United Kingdom
| | - Samuel L Volchenboum
- Navin R. Pinto, Mark A. Applebaum, Samuel L. Volchenboum, and Susan L. Cohn, Comer Children's Hospital, University of Chicago, Chicago, IL; Katherine K. Matthay, University of California San Francisco (UCSF) Benioff Children's Hospital, UCSF School of Medicine, San Francisco, CA; Wendy B. London, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA; Peter F. Ambros, Children's Cancer Research Institute, St Anna Kinderkrebsforschung, Vienna, Austria; Akira Nakagawara, Saga Medical Center Koseikan, Saga, Japan; Frank Berthold, Children's Hospital, University of Cologne, Koln, Germany; Gudrun Schleiermacher, Institut Curie, Paris; Dominique Valteau-Couanet, Gustave Roussy, Villejuif, France; Julie R. Park, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; and Andrew D.J. Pearson, Institute of Cancer Research and Royal Marsden Hospital, Surrey, United Kingdom
| | - Katherine K Matthay
- Navin R. Pinto, Mark A. Applebaum, Samuel L. Volchenboum, and Susan L. Cohn, Comer Children's Hospital, University of Chicago, Chicago, IL; Katherine K. Matthay, University of California San Francisco (UCSF) Benioff Children's Hospital, UCSF School of Medicine, San Francisco, CA; Wendy B. London, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA; Peter F. Ambros, Children's Cancer Research Institute, St Anna Kinderkrebsforschung, Vienna, Austria; Akira Nakagawara, Saga Medical Center Koseikan, Saga, Japan; Frank Berthold, Children's Hospital, University of Cologne, Koln, Germany; Gudrun Schleiermacher, Institut Curie, Paris; Dominique Valteau-Couanet, Gustave Roussy, Villejuif, France; Julie R. Park, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; and Andrew D.J. Pearson, Institute of Cancer Research and Royal Marsden Hospital, Surrey, United Kingdom
| | - Wendy B London
- Navin R. Pinto, Mark A. Applebaum, Samuel L. Volchenboum, and Susan L. Cohn, Comer Children's Hospital, University of Chicago, Chicago, IL; Katherine K. Matthay, University of California San Francisco (UCSF) Benioff Children's Hospital, UCSF School of Medicine, San Francisco, CA; Wendy B. London, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA; Peter F. Ambros, Children's Cancer Research Institute, St Anna Kinderkrebsforschung, Vienna, Austria; Akira Nakagawara, Saga Medical Center Koseikan, Saga, Japan; Frank Berthold, Children's Hospital, University of Cologne, Koln, Germany; Gudrun Schleiermacher, Institut Curie, Paris; Dominique Valteau-Couanet, Gustave Roussy, Villejuif, France; Julie R. Park, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; and Andrew D.J. Pearson, Institute of Cancer Research and Royal Marsden Hospital, Surrey, United Kingdom
| | - Peter F Ambros
- Navin R. Pinto, Mark A. Applebaum, Samuel L. Volchenboum, and Susan L. Cohn, Comer Children's Hospital, University of Chicago, Chicago, IL; Katherine K. Matthay, University of California San Francisco (UCSF) Benioff Children's Hospital, UCSF School of Medicine, San Francisco, CA; Wendy B. London, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA; Peter F. Ambros, Children's Cancer Research Institute, St Anna Kinderkrebsforschung, Vienna, Austria; Akira Nakagawara, Saga Medical Center Koseikan, Saga, Japan; Frank Berthold, Children's Hospital, University of Cologne, Koln, Germany; Gudrun Schleiermacher, Institut Curie, Paris; Dominique Valteau-Couanet, Gustave Roussy, Villejuif, France; Julie R. Park, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; and Andrew D.J. Pearson, Institute of Cancer Research and Royal Marsden Hospital, Surrey, United Kingdom
| | - Akira Nakagawara
- Navin R. Pinto, Mark A. Applebaum, Samuel L. Volchenboum, and Susan L. Cohn, Comer Children's Hospital, University of Chicago, Chicago, IL; Katherine K. Matthay, University of California San Francisco (UCSF) Benioff Children's Hospital, UCSF School of Medicine, San Francisco, CA; Wendy B. London, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA; Peter F. Ambros, Children's Cancer Research Institute, St Anna Kinderkrebsforschung, Vienna, Austria; Akira Nakagawara, Saga Medical Center Koseikan, Saga, Japan; Frank Berthold, Children's Hospital, University of Cologne, Koln, Germany; Gudrun Schleiermacher, Institut Curie, Paris; Dominique Valteau-Couanet, Gustave Roussy, Villejuif, France; Julie R. Park, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; and Andrew D.J. Pearson, Institute of Cancer Research and Royal Marsden Hospital, Surrey, United Kingdom
| | - Frank Berthold
- Navin R. Pinto, Mark A. Applebaum, Samuel L. Volchenboum, and Susan L. Cohn, Comer Children's Hospital, University of Chicago, Chicago, IL; Katherine K. Matthay, University of California San Francisco (UCSF) Benioff Children's Hospital, UCSF School of Medicine, San Francisco, CA; Wendy B. London, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA; Peter F. Ambros, Children's Cancer Research Institute, St Anna Kinderkrebsforschung, Vienna, Austria; Akira Nakagawara, Saga Medical Center Koseikan, Saga, Japan; Frank Berthold, Children's Hospital, University of Cologne, Koln, Germany; Gudrun Schleiermacher, Institut Curie, Paris; Dominique Valteau-Couanet, Gustave Roussy, Villejuif, France; Julie R. Park, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; and Andrew D.J. Pearson, Institute of Cancer Research and Royal Marsden Hospital, Surrey, United Kingdom
| | - Gudrun Schleiermacher
- Navin R. Pinto, Mark A. Applebaum, Samuel L. Volchenboum, and Susan L. Cohn, Comer Children's Hospital, University of Chicago, Chicago, IL; Katherine K. Matthay, University of California San Francisco (UCSF) Benioff Children's Hospital, UCSF School of Medicine, San Francisco, CA; Wendy B. London, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA; Peter F. Ambros, Children's Cancer Research Institute, St Anna Kinderkrebsforschung, Vienna, Austria; Akira Nakagawara, Saga Medical Center Koseikan, Saga, Japan; Frank Berthold, Children's Hospital, University of Cologne, Koln, Germany; Gudrun Schleiermacher, Institut Curie, Paris; Dominique Valteau-Couanet, Gustave Roussy, Villejuif, France; Julie R. Park, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; and Andrew D.J. Pearson, Institute of Cancer Research and Royal Marsden Hospital, Surrey, United Kingdom
| | - Julie R Park
- Navin R. Pinto, Mark A. Applebaum, Samuel L. Volchenboum, and Susan L. Cohn, Comer Children's Hospital, University of Chicago, Chicago, IL; Katherine K. Matthay, University of California San Francisco (UCSF) Benioff Children's Hospital, UCSF School of Medicine, San Francisco, CA; Wendy B. London, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA; Peter F. Ambros, Children's Cancer Research Institute, St Anna Kinderkrebsforschung, Vienna, Austria; Akira Nakagawara, Saga Medical Center Koseikan, Saga, Japan; Frank Berthold, Children's Hospital, University of Cologne, Koln, Germany; Gudrun Schleiermacher, Institut Curie, Paris; Dominique Valteau-Couanet, Gustave Roussy, Villejuif, France; Julie R. Park, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; and Andrew D.J. Pearson, Institute of Cancer Research and Royal Marsden Hospital, Surrey, United Kingdom
| | - Dominique Valteau-Couanet
- Navin R. Pinto, Mark A. Applebaum, Samuel L. Volchenboum, and Susan L. Cohn, Comer Children's Hospital, University of Chicago, Chicago, IL; Katherine K. Matthay, University of California San Francisco (UCSF) Benioff Children's Hospital, UCSF School of Medicine, San Francisco, CA; Wendy B. London, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA; Peter F. Ambros, Children's Cancer Research Institute, St Anna Kinderkrebsforschung, Vienna, Austria; Akira Nakagawara, Saga Medical Center Koseikan, Saga, Japan; Frank Berthold, Children's Hospital, University of Cologne, Koln, Germany; Gudrun Schleiermacher, Institut Curie, Paris; Dominique Valteau-Couanet, Gustave Roussy, Villejuif, France; Julie R. Park, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; and Andrew D.J. Pearson, Institute of Cancer Research and Royal Marsden Hospital, Surrey, United Kingdom
| | - Andrew D J Pearson
- Navin R. Pinto, Mark A. Applebaum, Samuel L. Volchenboum, and Susan L. Cohn, Comer Children's Hospital, University of Chicago, Chicago, IL; Katherine K. Matthay, University of California San Francisco (UCSF) Benioff Children's Hospital, UCSF School of Medicine, San Francisco, CA; Wendy B. London, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA; Peter F. Ambros, Children's Cancer Research Institute, St Anna Kinderkrebsforschung, Vienna, Austria; Akira Nakagawara, Saga Medical Center Koseikan, Saga, Japan; Frank Berthold, Children's Hospital, University of Cologne, Koln, Germany; Gudrun Schleiermacher, Institut Curie, Paris; Dominique Valteau-Couanet, Gustave Roussy, Villejuif, France; Julie R. Park, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; and Andrew D.J. Pearson, Institute of Cancer Research and Royal Marsden Hospital, Surrey, United Kingdom
| | - Susan L Cohn
- Navin R. Pinto, Mark A. Applebaum, Samuel L. Volchenboum, and Susan L. Cohn, Comer Children's Hospital, University of Chicago, Chicago, IL; Katherine K. Matthay, University of California San Francisco (UCSF) Benioff Children's Hospital, UCSF School of Medicine, San Francisco, CA; Wendy B. London, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA; Peter F. Ambros, Children's Cancer Research Institute, St Anna Kinderkrebsforschung, Vienna, Austria; Akira Nakagawara, Saga Medical Center Koseikan, Saga, Japan; Frank Berthold, Children's Hospital, University of Cologne, Koln, Germany; Gudrun Schleiermacher, Institut Curie, Paris; Dominique Valteau-Couanet, Gustave Roussy, Villejuif, France; Julie R. Park, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; and Andrew D.J. Pearson, Institute of Cancer Research and Royal Marsden Hospital, Surrey, United Kingdom.
| |
Collapse
|
37
|
Wu K, Yang L, Chen J, Zhao H, Wang J, Xu S, Huang Z. miR-362-5p inhibits proliferation and migration of neuroblastoma cells by targeting phosphatidylinositol 3-kinase-C2β. FEBS Lett 2015; 589:1911-9. [PMID: 26073258 DOI: 10.1016/j.febslet.2015.05.056] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 05/24/2015] [Accepted: 05/26/2015] [Indexed: 02/03/2023]
Abstract
miR-362-5p is down-regulated in high-risk neuroblastoma and can function as a tumor suppressor. However, its role remains poorly understood. We show that miR-362-5p is down-regulated in metastatic neuroblastoma compared with primary neuroblastoma. Overexpression of miR-362-5p inhibits cell proliferation, migration and invasion of neuroblastoma cells in vitro and suppresses tumor growth of neuroblastoma in vivo. Phosphatidylinositol 3-kinase (PI3K)-C2β is a target of miR-362-5p. Knockdown of PI3K-C2β by siRNA had a similar effect to overexpression of miR-362-5p on SH-SY5Y cells. Overexpression of PI3K-C2β partially reversed tumor-suppressive effects of miR-362-5p. We suggest that miR-362-5p suppresses neuroblastoma cell growth and motility, partially by targeting PI3K-C2β.
Collapse
Affiliation(s)
- Kai Wu
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China.
| | - Liucheng Yang
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China.
| | - Jianfeng Chen
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Haijun Zhao
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China.
| | - Jianjun Wang
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Shuai Xu
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China.
| | - Zonghai Huang
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China.
| |
Collapse
|
38
|
Yáñez Y, Grau E, Rodríguez-Cortez VC, Hervás D, Vidal E, Noguera R, Hernández M, Segura V, Cañete A, Conesa A, Font de Mora J, Castel V. Two independent epigenetic biomarkers predict survival in neuroblastoma. Clin Epigenetics 2015; 7:16. [PMID: 25767620 PMCID: PMC4357365 DOI: 10.1186/s13148-015-0054-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 02/09/2015] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Neuroblastoma (NB) is the most common extracranial pediatric solid tumor with a highly variable clinical course, ranging from spontaneous regression to life-threatening disease. Survival rates for high-risk NB patients remain disappointingly low despite multimodal treatment. Thus, there is an urgent clinical need for additional biomarkers to improve risk stratification, treatment management, and survival rates in children with aggressive NB. RESULTS Using gene promoter methylation analysis in 48 neuroblastoma tumors with microarray technology, we found a strong association between survival and gene promoter hypermethylation (P = 0.036). Hypermethylation of 70 genes significantly differentiated high-risk survivor patients from those who died during follow-up time. Sixteen genes with relevant roles in cancer biology were further validated in an additional cohort of 83 neuroblastoma tumors by bisulfite pyrosequencing. High promoter methylation rates of these genes were found in patients with metastatic tumors (either stage metastatic (M) or metastatic special (MS)), 18 months or older at first diagnosis, MYCN amplification, relapsed, and dead. Notably, the degree of methylation of retinoblastoma 1 (RB1) and teratocarcinoma-derived growth factor 1 (TDGF1) predicts event-free and overall survival independently of the established risk factors. In addition, low RB1 mRNA expression levels associate with poor prognosis suggesting that promoter methylation could contribute to the transcriptional silencing of this gene in NB. CONCLUSIONS We found a new epigenetic signature predictive for NB patients' outcome: the methylation status of RB1 and TDGF1 associate with poorer survival. This information is useful to assess prognosis and improve treatment selection.
Collapse
Affiliation(s)
- Yania Yáñez
- Pediatric Oncology Unit, Hospital Universitari i Politècnic La Fe, Avda Fernando Abril Martorell, Valencia, 46026 Spain
| | - Elena Grau
- Pediatric Oncology Unit, Hospital Universitari i Politècnic La Fe, Avda Fernando Abril Martorell, Valencia, 46026 Spain
| | - Virginia C Rodríguez-Cortez
- Chromatin and Disease Group, Cancer Epigenetics and Biology Programme (PEBC) Bellvitge Biomedical Research Institute (IDIBELL), Gran Via de L'Hospitalet, Barcelona, 08908 Spain
| | - David Hervás
- Biostatistics Unit, Instituto de Investigación Sanitaria La Fe, Avda Fernando Abril Martorell, Valencia, 46026 Spain
| | - Enrique Vidal
- Genomics of Gene Expression Lab, Centro de Investigaciones Príncipe Felipe, Carrer d'Eduardo Primo Yúfera, Valencia, 46012 Spain
| | - Rosa Noguera
- Department of Pathology, Medical School, University of Valencia, Avda Blasco Ibáñez, Valencia, 46010 Spain
| | - Miguel Hernández
- Department of Pathology, Hospital Universitari i Politècnic La Fe, Avda Fernando Abril Martorell, Valencia, 46026 Spain
| | - Vanessa Segura
- Pediatric Oncology Unit, Hospital Universitari i Politècnic La Fe, Avda Fernando Abril Martorell, Valencia, 46026 Spain
| | - Adela Cañete
- Pediatric Oncology Unit, Hospital Universitari i Politècnic La Fe, Avda Fernando Abril Martorell, Valencia, 46026 Spain
| | - Ana Conesa
- Genomics of Gene Expression Lab, Centro de Investigaciones Príncipe Felipe, Carrer d'Eduardo Primo Yúfera, Valencia, 46012 Spain
| | - Jaime Font de Mora
- Laboratory of Cellular and Molecular Biology, Instituto de Investigación Sanitaria La Fe, Avda Fernando Abril Martorell, Valencia, 46026 Spain
| | - Victoria Castel
- Pediatric Oncology Unit, Hospital Universitari i Politècnic La Fe, Avda Fernando Abril Martorell, Valencia, 46026 Spain
| |
Collapse
|
39
|
Schleiermacher G, Janoueix-Lerosey I, Delattre O. Recent insights into the biology of neuroblastoma. Int J Cancer 2014; 135:2249-61. [PMID: 25124476 DOI: 10.1002/ijc.29077] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 05/08/2014] [Indexed: 01/24/2023]
Abstract
Neuroblastoma (NB) is an embryonal tumor of the sympathetic nervous system which accounts for 8-10% of pediatric cancers. It is characterized by a broad spectrum of clinical behaviors from spontaneous regression to fatal outcome despite aggressive therapies. Considerable progress has been made recently in the germline and somatic genetic characterization of patients and tumors. Indeed, predisposition genes that account for a significant proportion of familial and syndromic cases have been identified and genome-wide association studies have retrieved a number of susceptibility loci. In addition, genome-wide sequencing, copy-number and expression studies have been conducted on tumors and have detected important gene modifications, profiles and signatures that have strong implications for the therapeutic stratification of patients. The identification of major players in NB oncogenesis, including MYCN, ALK, PHOX2B and LIN28B, has enabled the development of new animal models. Our review focuses on these recent advances, on the insights they provide on the mechanisms involved in NB development and their applications for the clinical management of patients.
Collapse
Affiliation(s)
- Gudrun Schleiermacher
- Equipe SIRIC Recherche Translationnelle en Oncologie Pédiatrique, Département de Recherche Translationnelle et Inserm U830, Centre de Recherche, Paris Cedex, 05, France; Département de pédiatrie, Institut Curie, Paris Cedex, 05, France; Unité Génétique et Biologie des Cancers, Inserm U830, Centre de Recherche, Institut Curie, Paris Cedex, 05, France
| | | | | |
Collapse
|
40
|
Althoff K, Lindner S, Odersky A, Mestdagh P, Beckers A, Karczewski S, Molenaar JJ, Bohrer A, Knauer S, Speleman F, Epple M, Kozlova D, Yoon S, Baek K, Vandesompele J, Eggert A, Schramm A, Schulte JH. miR-542-3p exerts tumor suppressive functions in neuroblastoma by downregulating Survivin. Int J Cancer 2014; 136:1308-20. [DOI: 10.1002/ijc.29091] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 05/21/2014] [Accepted: 06/12/2014] [Indexed: 12/31/2022]
Affiliation(s)
- Kristina Althoff
- Department of Pediatric Oncology and Hematology; University Children's Hospital Essen; Essen Germany
- German Cancer Consortium (DKTK); Germany
| | - Sven Lindner
- Department of Pediatric Oncology and Hematology; University Children's Hospital Essen; Essen Germany
- German Cancer Consortium (DKTK); Germany
- German Cancer Research Center (DKFZ); Heidelberg Germany
| | - Andrea Odersky
- Department of Pediatric Oncology and Hematology; University Children's Hospital Essen; Essen Germany
- German Cancer Consortium (DKTK); Germany
| | - Pieter Mestdagh
- Center for Medical Genetics Ghent (CMGG); Ghent University Hospital; Ghent Belgium
| | - Anneleen Beckers
- Center for Medical Genetics Ghent (CMGG); Ghent University Hospital; Ghent Belgium
| | - Sarah Karczewski
- Institute for Molecular Biology II, Center for Medical Biotechnology (ZMB); University Duisburg-Essen; Essen Germany
| | - Jan J. Molenaar
- Department of Human Genetics, Academic Medical Centre; University of Amsterdam; Amsterdam The Netherlands
| | - Anna Bohrer
- Department of Neurosurgery; University Hospital Essen; Essen Germany
| | - Shirley Knauer
- Institute for Molecular Biology II, Center for Medical Biotechnology (ZMB); University Duisburg-Essen; Essen Germany
| | - Frank Speleman
- Center for Medical Genetics Ghent (CMGG); Ghent University Hospital; Ghent Belgium
| | - Matthias Epple
- Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE); University of Duisburg-Essen; Essen Germany
| | - Diana Kozlova
- Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE); University of Duisburg-Essen; Essen Germany
| | - Sena Yoon
- Graduate School of Biotechnology; Kyung Hee University; Yongin Republic of Korea
| | - Kwanghee Baek
- Graduate School of Biotechnology; Kyung Hee University; Yongin Republic of Korea
| | - Jo Vandesompele
- Center for Medical Genetics Ghent (CMGG); Ghent University Hospital; Ghent Belgium
| | - Angelika Eggert
- Department of Pediatric Oncology and Hematology; University Children's Hospital Essen; Essen Germany
| | - Alexander Schramm
- Department of Pediatric Oncology and Hematology; University Children's Hospital Essen; Essen Germany
| | - Johannes H. Schulte
- Department of Pediatric Oncology and Hematology; University Children's Hospital Essen; Essen Germany
- German Cancer Consortium (DKTK); Germany
- German Cancer Research Center (DKFZ); Heidelberg Germany
- Translational Neuro-Oncology, West German Cancer Center; University Hospital Essen, University Duisburg-Essen; Essen Germany
- Center for Medical Biotechnology; University Duisburg-Essen; Essen Germany
| |
Collapse
|
41
|
Significance and therapeutic value of miRNAs in embryonal neural tumors. Molecules 2014; 19:5821-62. [PMID: 24806581 PMCID: PMC6271640 DOI: 10.3390/molecules19055821] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 04/25/2014] [Accepted: 04/28/2014] [Indexed: 02/07/2023] Open
Abstract
Embryonal tumors of the nervous system are the leading cause of childhood cancer-related morbidity and mortality. Medulloblastoma, supratentorial primitive neuroectodermal tumors, atypical teratoid/rhabdoid tumor and neuroblastoma account for more than 20% of childhood malignancies and typify the current neural embryonal tumor model in pediatric oncology. Mechanisms driving the formation of these tumors point towards impaired differentiation of neuronal and neuron-associated cells during the development of the nervous system as an important factor. The importance of microRNAs (miRNAs) for proper embryonic cell function has been confirmed and their aberrant expressions have been linked to tumor development. The role of miRNAs in controlling essential regulators of key pathways implicated in tumor development makes their use in diagnostics a powerful tool to be used for early detection of cancer, risk assessment and prognosis, as well as for the design of innovative therapeutic strategies. In this review we focus on the significance of miRNAs involved in the biology of embryonal neural tumors, delineate their clinical significance and discuss their potential as a novel therapeutic target.
Collapse
|
42
|
Wang Y, Zhang Y, Huang J, Chen X, Gu X, Wang Y, Zeng L, Yang GY. Increase of circulating miR-223 and insulin-like growth factor-1 is associated with the pathogenesis of acute ischemic stroke in patients. BMC Neurol 2014; 14:77. [PMID: 24708646 PMCID: PMC4234389 DOI: 10.1186/1471-2377-14-77] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 04/04/2014] [Indexed: 12/13/2022] Open
Abstract
Background The relationship between circulating microRNA-223 and pathogenesis of acute ischemic stroke is unknown. Here we investigated the roles and possible targets of circulating microRNA-223 in human ischemic stroke within the first 72 hours. Methods Blood samples were collected from patients within 72 hours after cerebral ischemia (n = 79) and compared with healthy control samples (n = 75). The level of possible downstream factors of microRNA-223 including insulin-like growth factor-1, insulin-like growth factor-1 receptor and interleukin-6 was examined by ELISA assay. The relationship between the microRNA-223 level and NIHSS scores, TOAST subtypes, and infarct volume was analyzed respectively. In addition, twelve adult male CD-1 mice underwent middle cerebral artery occlusion using the suture technique. Circulating blood and brain tissue in the ischemic ipsilateral hemisphere were collected at 24 hours after middle cerebral artery occlusion. microRNA-223 was detected by real-time polymerase chain reactions. Results microRNA-223 levels in the circulating blood of acute ischemic stroke patients were greatly increased compared to the control (p < 0.05). microRNA-223, which were negatively correlated with NIHSS scores (r = −0.531, p < 0.01) and infarct volume (r = −0.265, p = 0.039), was significantly up-regulated in large artery and small artery strokes. The plasma level of insulin-like growth factor-1 was positively associated with that of microRNA-223 (r = 0.205, p = 0.022). Moreover, microRNA-223 in blood and brain were positively correlated (r = 0.834, p < 0.05), and they were up-regulated significantly in mice that underwent middle cerebral artery occlusion (p < 0.05). Conclusions Our results suggest that microRNA-223 is associated with acute ischemic stroke and possibly plays a role in stroke through up-regulating growth factor such as insulin-like growth factor-1 gene.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Lili Zeng
- Department of neurology, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, 197 Ruijin Er Road, 200025 Shanghai, China.
| | | |
Collapse
|
43
|
Stricker TP, Morales La Madrid A, Chlenski A, Guerrero L, Salwen HR, Gosiengfiao Y, Perlman EJ, Furman W, Bahrami A, Shohet JM, Zage PE, Hicks MJ, Shimada H, Suganuma R, Park JR, So S, London WB, Pytel P, Maclean KH, Cohn SL. Validation of a prognostic multi-gene signature in high-risk neuroblastoma using the high throughput digital NanoString nCounter™ system. Mol Oncol 2014; 8:669-78. [PMID: 24560446 DOI: 10.1016/j.molonc.2014.01.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 12/24/2013] [Accepted: 01/21/2014] [Indexed: 10/25/2022] Open
Abstract
Microarray-based molecular signatures have not been widely integrated into neuroblastoma diagnostic classification systems due to the complexities of the assay and requirement for high-quality RNA. New digital technologies that accurately quantify gene expression using RNA isolated from formalin-fixed paraffin embedded (FFPE) tissues are now available. In this study, we describe the first use of a high-throughput digital system to assay the expression of genes in an "ultra-high risk" microarray classifier in FFPE high-risk neuroblastoma tumors. Customized probes corresponding to the 42 genes in a published multi-gene neuroblastoma signature were hybridized to RNA isolated from 107 FFPE high-risk neuroblastoma samples using the NanoString nCounter™ Analysis System. For classification of each patient, the Pearson's correlation coefficient was calculated between the standardized nCounter™ data and the molecular signature from the microarray data. We demonstrate that the nCounter™ 42-gene panel sub-stratified the high-risk cohort into two subsets with statistically significantly different overall survival (p = 0.0027) and event-free survival (p = 0.028). In contrast, none of the established prognostic risk markers (age, stage, tumor histology, MYCN status, and ploidy) were significantly associated with survival. We conclude that the nCounter™ System can reproducibly quantify expression levels of signature genes in FFPE tumor samples. Validation of this microarray signature in our high-risk patient cohort using a completely different technology emphasizes the prognostic relevance of this classifier. Prospective studies testing the prognostic value of molecular signatures in high-risk neuroblastoma patients using FFPE tumor samples and the nCounter™ System are warranted.
Collapse
Affiliation(s)
- Thomas P Stricker
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN, USA
| | | | - Alexandre Chlenski
- Department of Pediatrics, Comer Children's Hospital, University of Chicago, Chicago, IL, USA
| | - Lisa Guerrero
- Department of Pediatrics, Comer Children's Hospital, University of Chicago, Chicago, IL, USA
| | - Helen R Salwen
- Department of Pediatrics, Comer Children's Hospital, University of Chicago, Chicago, IL, USA
| | - Yasmin Gosiengfiao
- Department of Pediatrics, Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Elizabeth J Perlman
- Department of Pathology, Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Wayne Furman
- Department of Hematology/Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Armita Bahrami
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jason M Shohet
- Department of Pediatrics, Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Peter E Zage
- Department of Pediatrics, Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - M John Hicks
- Department of Pathology, Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Hiroyuki Shimada
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Rie Suganuma
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Julie R Park
- Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA, USA; Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Sara So
- Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA, USA; Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Wendy B London
- Children's Oncology Group Statistics and Data Center, Boston, MA, USA; Boston Children's Hospital/Dana-Farber Cancer Institute, Boston, MA, USA
| | - Peter Pytel
- Department of Pathology, Comer Children's Hospital, University of Chicago, Chicago, IL, USA
| | | | - Susan L Cohn
- Department of Pediatrics, Comer Children's Hospital, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
44
|
Zhi F, Wang R, Wang Q, Xue L, Deng D, Wang S, Yang Y. MicroRNAs in neuroblastoma: small-sized players with a large impact. Neurochem Res 2014; 39:613-23. [PMID: 24477657 DOI: 10.1007/s11064-014-1247-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 12/22/2013] [Accepted: 01/21/2014] [Indexed: 12/23/2022]
Abstract
Neuroblastoma, a malignant embryonal tumor of the sympathetic nervous system, is the most common solid extracranial malignancy of childhood and accounts for 15 % of all childhood cancer deaths. The biological behavior of neuroblastoma is extensively heterogeneous, ranging from spontaneous regression to rapid progression despite multimodal aggressive therapy. Although the molecular basis of neuroblastoma has received considerable attention over the past decade, elucidating the mechanisms for the aggressive progression of neuroblastoma is needed for improving the efficacy of treatment. miRNAs (microRNAs) are small non-coding RNA molecules generally 19-22 nucleotides in length. miRNAs regulate 60 % of human gene expression at the post-transcriptional level by targeting regions of sequence complementarity on the 3'-untranslated regions (3'-UTRs) of specific mRNAs. miRNAs can either cause degradation of mRNAs or can inhibit their translation and therefore play major roles in normal growth and development. miRNA dysregulation has oncogenic or tumor-suppressive functions in virtually all forms of cancer, including neuroblastoma. The present review highlights the current insights on dysregulated miRNAs in neuroblastoma and on their roles in the diagnosis, prognosis, and treatment of this malignancy. As a rapidly evolving field of basic and biomedical sciences, miRNA research holds a great potential to impact on the management of neuroblastoma.
Collapse
Affiliation(s)
- Feng Zhi
- Modern Medical Research Center, Third Affiliated Hospital of Soochow University, 185#, Juqian Road, Changzhou, 213003, Jiangsu, China
| | | | | | | | | | | | | |
Collapse
|
45
|
Gustafson WC, Matthay KK. Progress towards personalized therapeutics: biologic- and risk-directed therapy for neuroblastoma. Expert Rev Neurother 2014; 11:1411-23. [DOI: 10.1586/ern.11.103] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
46
|
miR-329 suppresses the growth and motility of neuroblastoma by targeting KDM1A. FEBS Lett 2013; 588:192-7. [DOI: 10.1016/j.febslet.2013.11.036] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 11/23/2013] [Indexed: 11/23/2022]
|
47
|
Oberthuer A. Genomic markers for neuroblastoma risk estimation: superseding tumor stage, age and MYCN? Biomark Med 2013; 7:905-8. [PMID: 24266822 DOI: 10.2217/bmm.13.97] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Affiliation(s)
- André Oberthuer
- University of Cologne Children's Hospital, Department of Pediatric Oncology & Hematology, Germany, Kerpener Strasse 62, D-50931 Köln, Germany.
| |
Collapse
|
48
|
miR-137 regulates the constitutive androstane receptor and modulates doxorubicin sensitivity in parental and doxorubicin-resistant neuroblastoma cells. Oncogene 2013; 33:3717-29. [PMID: 23934188 DOI: 10.1038/onc.2013.330] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 06/14/2013] [Accepted: 06/28/2013] [Indexed: 12/31/2022]
Abstract
Chemotherapy is the most common treatment for cancer. However, multidrug resistance (MDR) remains a major obstacle to effective chemotherapy, limiting the efficacy of both conventional chemotherapeutic and novel biologic agents. The constitutive androstane receptor (CAR), a xenosensor, is a key regulator of MDR. It functions in xenobiotic detoxification by regulating the expression of phase I drug-metabolizing enzymes and ATP-binding cassette (ABC) transporters, whose overexpression in cancers and whose role in drug resistance make them potential therapeutic targets for reducing MDR. MicroRNAs (miRNAs) are endogenous negative regulators of gene expression and have been implicated in most cellular processes, including drug resistance. Here, we report the inversely related expression of miR-137 and CAR in parental and doxorubicin-resistant neuroblastoma cells, wherein miR-137 is downregulated in resistant cells. miR-137 overexpression resulted in downregulation of CAR protein and mRNA (via mRNA degradation); it sensitized doxorubicin-resistant cells to doxorubicin (as shown by reduced proliferation, increased apoptosis and increased G2-phase cell cycle arrest) and reduced the in vivo growth rate of neuroblastoma xenografts. We observed similar results in cellular models of hepatocellular and colon cancers, indicating that the doxorubicin-sensitizing effect of miR-137 is not tumor type-specific. Finally, we show for the first time a negative feedback loop whereby miR-137 downregulates CAR expression and CAR downregulates miR-137 expression. Hypermethylation of the miR-137 promoter and negative regulation of miR-137 by CAR contribute in part to reduced miR-137 expression and increased CAR and MDR1 expression in doxorubicin-resistant neuroblastoma cells. These findings demonstrate that miR-137 is a crucial regulator of cancer response to doxorubicin treatment, and they identify miR-137 as a highly promising target to reduce CAR-driven doxorubicin resistance.
Collapse
|
49
|
Wilzén A, Krona C, Sveinbjörnsson B, Kristiansson E, Dalevi D, Øra I, De Preter K, Stallings RL, Maris J, Versteeg R, Nilsson S, Kogner P, Abel F. ERBB3 is a marker of a ganglioneuroblastoma/ganglioneuroma-like expression profile in neuroblastic tumours. Mol Cancer 2013; 12:70. [PMID: 23835063 PMCID: PMC3766266 DOI: 10.1186/1476-4598-12-70] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 06/25/2013] [Indexed: 12/21/2022] Open
Abstract
Background Neuroblastoma (NB) tumours are commonly divided into three cytogenetic subgroups. However, by unsupervised principal components analysis of gene expression profiles we recently identified four distinct subgroups, r1-r4. In the current study we characterized these different subgroups in more detail, with a specific focus on the fourth divergent tumour subgroup (r4). Methods Expression microarray data from four international studies corresponding to 148 neuroblastic tumour cases were subject to division into four expression subgroups using a previously described 6-gene signature. Differentially expressed genes between groups were identified using Significance Analysis of Microarray (SAM). Next, gene expression network modelling was performed to map signalling pathways and cellular processes representing each subgroup. Findings were validated at the protein level by immunohistochemistry and immunoblot analyses. Results We identified several significantly up-regulated genes in the r4 subgroup of which the tyrosine kinase receptor ERBB3 was most prominent (fold change: 132–240). By gene set enrichment analysis (GSEA) the constructed gene network of ERBB3 (n = 38 network partners) was significantly enriched in the r4 subgroup in all four independent data sets. ERBB3 was also positively correlated to the ErbB family members EGFR and ERBB2 in all data sets, and a concurrent overexpression was seen in the r4 subgroup. Further studies of histopathology categories using a fifth data set of 110 neuroblastic tumours, showed a striking similarity between the expression profile of r4 to ganglioneuroblastoma (GNB) and ganglioneuroma (GN) tumours. In contrast, the NB histopathological subtype was dominated by mitotic regulating genes, characterizing unfavourable NB subgroups in particular. The high ErbB3 expression in GN tumour types was verified at the protein level, and showed mainly expression in the mature ganglion cells. Conclusions Conclusively, this study demonstrates the importance of performing unsupervised clustering and subtype discovery of data sets prior to analyses to avoid a mixture of tumour subtypes, which may otherwise give distorted results and lead to incorrect conclusions. The current study identifies ERBB3 as a clear-cut marker of a GNB/GN-like expression profile, and we suggest a 7-gene expression signature (including ERBB3) as a complement to histopathology analysis of neuroblastic tumours. Further studies of ErbB3 and other ErbB family members and their role in neuroblastic differentiation and pathogenesis are warranted.
Collapse
Affiliation(s)
- Annica Wilzén
- Department of Clinical Genetics, Institution of Biomedicine, Box 413, S- 405 30, Gothenburg University, Gothenburg, Sweden.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Extension of microRNA expression pattern associated with high-risk neuroblastoma. Tumour Biol 2013; 34:2315-9. [DOI: 10.1007/s13277-013-0777-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 03/26/2013] [Indexed: 01/21/2023] Open
|