1
|
Liu Z, Zhang X, Xu M, Hong JJ, Ciardiello A, Lei H, Shern JF, Thiele CJ. MYCN drives oncogenesis by cooperating with the histone methyltransferase G9a and the WDR5 adaptor to orchestrate global gene transcription. PLoS Biol 2024; 22:e3002240. [PMID: 38547242 PMCID: PMC11003700 DOI: 10.1371/journal.pbio.3002240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 04/09/2024] [Accepted: 02/28/2024] [Indexed: 04/11/2024] Open
Abstract
MYCN activates canonical MYC targets involved in ribosome biogenesis, protein synthesis, and represses neuronal differentiation genes to drive oncogenesis in neuroblastoma (NB). How MYCN orchestrates global gene expression remains incompletely understood. Our study finds that MYCN binds promoters to up-regulate canonical MYC targets but binds to both enhancers and promoters to repress differentiation genes. MYCN binding also increases H3K4me3 and H3K27ac on canonical MYC target promoters and decreases H3K27ac on neuronal differentiation gene enhancers and promoters. WDR5 facilitates MYCN promoter binding to activate canonical MYC target genes, whereas MYCN recruits G9a to enhancers to repress neuronal differentiation genes. Targeting both MYCN's active and repressive transcriptional activities using both WDR5 and G9a inhibitors synergistically suppresses NB growth. We demonstrate that MYCN cooperates with WDR5 and G9a to orchestrate global gene transcription. The targeting of both these cofactors is a novel therapeutic strategy to indirectly target the oncogenic activity of MYCN.
Collapse
Affiliation(s)
- Zhihui Liu
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Xiyuan Zhang
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Man Xu
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Jason J. Hong
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Amanda Ciardiello
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Haiyan Lei
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Jack F. Shern
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Carol J. Thiele
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, United States of America
| |
Collapse
|
2
|
Liu Z, Zhang X, Xu M, Hong JJ, Ciardiello A, Lei H, Shern JF, Thiele CJ. MYCN driven oncogenesis involves cooperation with WDR5 to activate canonical MYC targets and G9a to repress differentiation genes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.11.548643. [PMID: 37781575 PMCID: PMC10541123 DOI: 10.1101/2023.07.11.548643] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
MYCN activates canonical MYC targets involved in ribosome biogenesis, protein synthesis and represses neuronal differentiation genes to drive oncogenesis in neuroblastoma (NB). How MYCN orchestrates global gene expression remains incompletely understood. Our study finds that MYCN binds promoters to up-regulate canonical MYC targets but binds to both enhancers and promoters to repress differentiation genes. MYCN-binding also increases H3K4me3 and H3K27ac on canonical MYC target promoters and decreases H3K27ac on neuronal differentiation gene enhancers and promoters. WDR5 is needed to facilitate MYCN promoter binding to activate canonical MYC target genes, whereas MYCN recruits G9a to enhancers to repress neuronal differentiation genes. Targeting both MYCN's active and repressive transcriptional activities using both WDR5 and G9a inhibitors synergistically suppresses NB growth. We demonstrate that MYCN cooperates with WDR5 and G9a to orchestrate global gene transcription. The targeting of both these cofactors is a novel therapeutic strategy to indirectly target the oncogenic activity of MYCN.
Collapse
Affiliation(s)
- Zhihui Liu
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Xiyuan Zhang
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Man Xu
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Jason J. Hong
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Amanda Ciardiello
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Haiyan Lei
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Jack F. Shern
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Carol J. Thiele
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
3
|
Bhardwaj N, Das G, Srinivasan R. Neuroblastoma-derived v-myc avian myelocytomatosis viral related oncogene or MYCN gene. J Clin Pathol 2023:jcp-2022-208476. [PMID: 37221048 DOI: 10.1136/jcp-2022-208476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 04/13/2023] [Indexed: 05/25/2023]
Abstract
The MYCN gene belongs to the MYC family of transcription factors. Amplification of MYCN, first discovered in neuroblastoma cells, ushered in the era of cancer genomics. The MYCN gene and MYCN protein are extensively studied in the context of neuroblastoma. As demonstrated in transgenic mouse models, MYCN gene shows a restricted spatiotemporal expression predominantly in the neural crest cells which explains the associated neoplasms including neuroblastoma and central nervous system tumours. In neuroblastoma, MYCN amplification is a marker of aggressive tumours with poor prognosis and survival and forms the basis of risk stratification classifications.MYCN dysregulated expression occurs by several mechanisms at the transcriptional, translational and post-translational levels. These include massive gene amplification which occurs in an extrachromosomal location, upregulated transcription and stabilisation of the protein increasing its half-life. MYCN protein, a basic loop-helix-loop leucine zipper transcription factor, has many regions which bind to several proteins foremost of which is MAX forming the MYC:MAX heterodimer. Overall, MYCN controls multiple aspects of cell fate, foremost of which is cellular proliferation besides cell differentiation, apoptosis and cellular metabolism, all of which are the focus of this brief review. In addition to amplification, other mechanisms of MYCN overexpression include activating missense mutations as reported in basal cell carcinoma and Wilms tumour. A better understanding of this molecule will help in the discovery of novel strategies for its indirect targeting to improve the outcomes of patients with neuroblastoma and other MYCN-associated neoplasms.
Collapse
Affiliation(s)
- Neha Bhardwaj
- Department of Pathology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Gargi Das
- Medical Oncology (Pediatric Oncology), Cancer Institute-WIA, Chennai, Tamil Nadu, India
| | - Radhika Srinivasan
- Pathology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
4
|
Nataf S, Guillen M, Pays L. Irrespective of Plaque Activity, Multiple Sclerosis Brain Periplaques Exhibit Alterations of Myelin Genes and a TGF-Beta Signature. Int J Mol Sci 2022; 23:ijms232314993. [PMID: 36499320 PMCID: PMC9738407 DOI: 10.3390/ijms232314993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 11/23/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022] Open
Abstract
In a substantial share of patients suffering from multiple sclerosis (MS), neurological functions slowly deteriorate despite a lack of radiological activity. Such a silent progression, observed in either relapsing-remitting or progressive forms of MS, is driven by mechanisms that appear to be independent from plaque activity. In this context, we previously reported that, in the spinal cord of MS patients, periplaques cover large surfaces of partial demyelination characterized notably by a transforming growth factor beta (TGF-beta) molecular signature and a decreased expression of the oligodendrocyte gene NDRG1 (N-Myc downstream regulated 1). In the present work, we re-assessed a previously published RNA expression dataset in which brain periplaques were originally used as internal controls. When comparing the mRNA profiles obtained from brain periplaques with those derived from control normal white matter samples, we found that, irrespective of plaque activity, brain periplaques exhibited a TGF-beta molecular signature, an increased expression of TGFB2 (transforming growth factor beta 2) and a decreased expression of the oligodendrocyte genes NDRG1 (N-Myc downstream regulated 1) and MAG (myelin-associated glycoprotein). From these data obtained at the mRNA level, a survey of the human proteome allowed predicting a protein-protein interaction network linking TGFB2 to the down-regulation of both NDRG1 and MAG in brain periplaques. To further elucidate the role of NDRG1 in periplaque-associated partial demyelination, we then extracted the interaction network linking NDRG1 to proteins detected in human central myelin sheaths. We observed that such a network was highly significantly enriched in RNA-binding proteins that notably included several HNRNPs (heterogeneous nuclear ribonucleoproteins) involved in the post-transcriptional regulation of MAG. We conclude that both brain and spinal cord periplaques host a chronic process of tissue remodeling, during which oligodendrocyte myelinating functions are altered. Our findings further suggest that TGFB2 may fuel such a process. Overall, the present work provides additional evidence that periplaque-associated partial demyelination may drive the silent progression observed in a subset of MS patients.
Collapse
Affiliation(s)
- Serge Nataf
- Bank of Tissues and Cells, Hospices Civils de Lyon, Hôpital Edouard Herriot, Place d’Arsonval, F-69003 Lyon, France
- Stem-Cell and Brain Research Institute, 18 Avenue de Doyen Lépine, F-69500 Bron, France
- Lyon-Est School of Medicine, University Claude Bernard Lyon 1, 43 Bd du 11 Novembre 1918, F-69100 Villeurbanne, France
- Correspondence:
| | - Marine Guillen
- Bank of Tissues and Cells, Hospices Civils de Lyon, Hôpital Edouard Herriot, Place d’Arsonval, F-69003 Lyon, France
- Stem-Cell and Brain Research Institute, 18 Avenue de Doyen Lépine, F-69500 Bron, France
| | - Laurent Pays
- Bank of Tissues and Cells, Hospices Civils de Lyon, Hôpital Edouard Herriot, Place d’Arsonval, F-69003 Lyon, France
- Stem-Cell and Brain Research Institute, 18 Avenue de Doyen Lépine, F-69500 Bron, France
- Lyon-Est School of Medicine, University Claude Bernard Lyon 1, 43 Bd du 11 Novembre 1918, F-69100 Villeurbanne, France
| |
Collapse
|
5
|
Bartolucci D, Montemurro L, Raieli S, Lampis S, Pession A, Hrelia P, Tonelli R. MYCN Impact on High-Risk Neuroblastoma: From Diagnosis and Prognosis to Targeted Treatment. Cancers (Basel) 2022; 14:4421. [PMID: 36139583 PMCID: PMC9496712 DOI: 10.3390/cancers14184421] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/05/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022] Open
Abstract
Among childhood cancers, neuroblastoma is the most diffuse solid tumor and the deadliest in children. While to date, the pathology has become progressively manageable with a significant increase in 5-year survival for its less aggressive form, high-risk neuroblastoma (HR-NB) remains a major issue with poor outcome and little survivability of patients. The staging system has also been improved to better fit patient needs and to administer therapies in a more focused manner in consideration of pathology features. New and improved therapies have been developed; nevertheless, low efficacy and high toxicity remain a staple feature of current high-risk neuroblastoma treatment. For this reason, more specific procedures are required, and new therapeutic targets are also needed for a precise medicine approach. In this scenario, MYCN is certainly one of the most interesting targets. Indeed, MYCN is one of the most relevant hallmarks of HR-NB, and many studies has been carried out in recent years to discover potent and specific inhibitors to block its activities and any related oncogenic function. N-Myc protein has been considered an undruggable target for a long time. Thus, many new indirect and direct approaches have been discovered and preclinically evaluated for the interaction with MYCN and its pathways; a few of the most promising approaches are nearing clinical application for the investigation in HR-NB.
Collapse
Affiliation(s)
| | - Luca Montemurro
- Pediatric Oncology and Hematology Unit, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | | | | | - Andrea Pession
- Pediatric Unit, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Patrizia Hrelia
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Roberto Tonelli
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| |
Collapse
|
6
|
Perri P, Ponzoni M, Corrias MV, Ceccherini I, Candiani S, Bachetti T. A Focus on Regulatory Networks Linking MicroRNAs, Transcription Factors and Target Genes in Neuroblastoma. Cancers (Basel) 2021; 13:5528. [PMID: 34771690 PMCID: PMC8582685 DOI: 10.3390/cancers13215528] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/26/2021] [Accepted: 10/28/2021] [Indexed: 12/17/2022] Open
Abstract
Neuroblastoma (NB) is a tumor of the peripheral sympathetic nervous system that substantially contributes to childhood cancer mortality. NB originates from neural crest cells (NCCs) undergoing a defective sympathetic neuronal differentiation and although the starting events leading to the development of NB remain to be fully elucidated, the master role of genetic alterations in key oncogenes has been ascertained: (1) amplification and/or over-expression of MYCN, which is strongly associated with tumor progression and invasion; (2) activating mutations, amplification and/or over-expression of ALK, which is involved in tumor initiation, angiogenesis and invasion; (3) amplification and/or over-expression of LIN28B, promoting proliferation and suppression of neuroblast differentiation; (4) mutations and/or over-expression of PHOX2B, which is involved in the regulation of NB differentiation, stemness maintenance, migration and metastasis. Moreover, altered microRNA (miRNA) expression takes part in generating pathogenetic networks, in which the regulatory loops among transcription factors, miRNAs and target genes lead to complex and aberrant oncogene expression that underlies the development of a tumor. In this review, we have focused on the circuitry linking the oncogenic transcription factors MYCN and PHOX2B with their transcriptional targets ALK and LIN28B and the tumor suppressor microRNAs let-7, miR-34 and miR-204, which should act as down-regulators of their expression. We have also looked at the physiologic role of these genetic and epigenetic determinants in NC development, as well as in terminal differentiation, with their pathogenic dysregulation leading to NB oncogenesis.
Collapse
Affiliation(s)
- Patrizia Perri
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (M.P.); (M.V.C.)
| | - Mirco Ponzoni
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (M.P.); (M.V.C.)
| | - Maria Valeria Corrias
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (M.P.); (M.V.C.)
| | - Isabella Ceccherini
- Laboratory of Genetics and Genomics of Rare Diseases, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy;
| | - Simona Candiani
- Department of Earth, Environment and Life Sciences, University of Genoa, 16132 Genoa, Italy;
| | - Tiziana Bachetti
- Laboratory of Genetics and Genomics of Rare Diseases, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy;
- Department of Earth, Environment and Life Sciences, University of Genoa, 16132 Genoa, Italy;
| |
Collapse
|
7
|
Lourenco C, Resetca D, Redel C, Lin P, MacDonald AS, Ciaccio R, Kenney TMG, Wei Y, Andrews DW, Sunnerhagen M, Arrowsmith CH, Raught B, Penn LZ. MYC protein interactors in gene transcription and cancer. Nat Rev Cancer 2021; 21:579-591. [PMID: 34188192 DOI: 10.1038/s41568-021-00367-9] [Citation(s) in RCA: 145] [Impact Index Per Article: 48.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/04/2021] [Indexed: 02/07/2023]
Abstract
The transcription factor and oncoprotein MYC is a potent driver of many human cancers and can regulate numerous biological activities that contribute to tumorigenesis. How a single transcription factor can regulate such a diverse set of biological programmes is central to the understanding of MYC function in cancer. In this Perspective, we highlight how multiple proteins that interact with MYC enable MYC to regulate several central control points of gene transcription. These include promoter binding, epigenetic modifications, initiation, elongation and post-transcriptional processes. Evidence shows that a combination of multiple protein interactions enables MYC to function as a potent oncoprotein, working together in a 'coalition model', as presented here. Moreover, as MYC depends on its protein interactome for function, we discuss recent research that emphasizes an unprecedented opportunity to target protein interactors to directly impede MYC oncogenesis.
Collapse
Affiliation(s)
| | - Diana Resetca
- Princess Margaret Cancer Centre, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Cornelia Redel
- Princess Margaret Cancer Centre, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Peter Lin
- Princess Margaret Cancer Centre, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Alannah S MacDonald
- Princess Margaret Cancer Centre, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Roberto Ciaccio
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Tristan M G Kenney
- Princess Margaret Cancer Centre, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Yong Wei
- Princess Margaret Cancer Centre, Toronto, ON, Canada
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - David W Andrews
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Maria Sunnerhagen
- Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Cheryl H Arrowsmith
- Princess Margaret Cancer Centre, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Structural Genomics Consortium, Toronto, ON, Canada
| | - Brian Raught
- Princess Margaret Cancer Centre, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Linda Z Penn
- Princess Margaret Cancer Centre, Toronto, ON, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
8
|
A flexible microfluidic system for single-cell transcriptome profiling elucidates phased transcriptional regulators of cell cycle. Sci Rep 2021; 11:7918. [PMID: 33846365 PMCID: PMC8041752 DOI: 10.1038/s41598-021-86070-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 03/07/2021] [Indexed: 02/06/2023] Open
Abstract
Single cell transcriptome profiling has emerged as a breakthrough technology for the high-resolution understanding of complex cellular systems. Here we report a flexible, cost-effective and user-friendly droplet-based microfluidics system, called the Nadia Instrument, that can allow 3' mRNA capture of ~ 50,000 single cells or individual nuclei in a single run. The precise pressure-based system demonstrates highly reproducible droplet size, low doublet rates and high mRNA capture efficiencies that compare favorably in the field. Moreover, when combined with the Nadia Innovate, the system can be transformed into an adaptable setup that enables use of different buffers and barcoded bead configurations to facilitate diverse applications. Finally, by 3' mRNA profiling asynchronous human and mouse cells at different phases of the cell cycle, we demonstrate the system's ability to readily distinguish distinct cell populations and infer underlying transcriptional regulatory networks. Notably this provided supportive evidence for multiple transcription factors that had little or no known link to the cell cycle (e.g. DRAP1, ZKSCAN1 and CEBPZ). In summary, the Nadia platform represents a promising and flexible technology for future transcriptomic studies, and other related applications, at cell resolution.
Collapse
|
9
|
Catastrophic ATP loss underlies a metabolic combination therapy tailored for MYCN-amplified neuroblastoma. Proc Natl Acad Sci U S A 2021; 118:2009620118. [PMID: 33762304 DOI: 10.1073/pnas.2009620118] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
MYCN-amplified neuroblastoma is a lethal subset of pediatric cancer. MYCN drives numerous effects in the cell, including metabolic changes that are critical for oncogenesis. The understanding that both compensatory pathways and intrinsic redundancy in cell systems exists implies that the use of combination therapies for effective and durable responses is necessary. Additionally, the most effective targeted therapies exploit an "Achilles' heel" and are tailored to the genetics of the cancer under study. We performed an unbiased screen on select metabolic targeted therapy combinations and correlated sensitivity with over 20 subsets of cancer. We found that MYCN-amplified neuroblastoma is hypersensitive to the combination of an inhibitor of the lactate transporter MCT1, AZD3965, and complex I of the mitochondrion, phenformin. Our data demonstrate that MCT4 is highly correlated with resistance to the combination in the screen and lowly expressed in MYCN-amplified neuroblastoma. Low MCT4 combines with high expression of the MCT2 and MCT1 chaperone CD147 in MYCN-amplified neuroblastoma, altogether conferring sensitivity to the AZD3965 and phenformin combination. The result is simultaneous disruption of glycolysis and oxidative phosphorylation, resulting in dramatic disruption of adenosine triphosphate (ATP) production, endoplasmic reticulum stress, and cell death. In mouse models of MYCN-amplified neuroblastoma, the combination was tolerable at concentrations where it shrank tumors and did not increase white-blood-cell toxicity compared to single drugs. Therefore, we demonstrate that a metabolic combination screen can identify vulnerabilities in subsets of cancer and put forth a metabolic combination therapy tailored for MYCN-amplified neuroblastoma that demonstrates efficacy and tolerability in vivo.
Collapse
|
10
|
Nandan D, Rath CT, Reiner NE. Leishmania regulates host macrophage miRNAs expression by engaging transcription factor c-Myc. J Leukoc Biol 2020; 109:999-1007. [PMID: 33211335 DOI: 10.1002/jlb.4ru0920-614r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/26/2020] [Accepted: 10/28/2020] [Indexed: 12/19/2022] Open
Abstract
Parasites of Leishmania genus have developed sophisticated strategies allowing them to deactivate their host macrophage to promote their survival. It has become clear that miRNAs play important roles in shaping innate and adaptive immune responses toward pathogens. It is not surprising that several pathogens including Leishmania have evolved the ability to regulate host macrophage miRNA expression in order to manipulate host cell phenotypes to their advantage. However, very little is known about the mechanisms used by intracellular pathogens to drive changes in host cell miRNA abundance. In this review, Leishmania exploitation of macrophage transcription factor c-Myc as a critical proxy virulence factor to regulate abundance of macrophage miRNAs influencing macrophage physiology to promote its survival will be discussed.
Collapse
Affiliation(s)
- Devki Nandan
- Department of Medicine, Division of Infectious Diseases, University of British Columbia, Vancouver, British Columbia, Canada
| | - Carolina Torturella Rath
- Department of Medicine, Division of Infectious Diseases, University of British Columbia, Vancouver, British Columbia, Canada
| | - Neil E Reiner
- Department of Medicine, Division of Infectious Diseases, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
11
|
Bellamy J, Szemes M, Melegh Z, Dallosso A, Kollareddy M, Catchpoole D, Malik K. Increased Efficacy of Histone Methyltransferase G9a Inhibitors Against MYCN-Amplified Neuroblastoma. Front Oncol 2020; 10:818. [PMID: 32537432 PMCID: PMC7269128 DOI: 10.3389/fonc.2020.00818] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 04/27/2020] [Indexed: 01/09/2023] Open
Abstract
Targeted inhibition of proteins modulating epigenetic changes is an increasingly important priority in cancer therapeutics, and many small molecule inhibitors are currently being developed. In the case of neuroblastoma (NB), a pediatric solid tumor with a paucity of intragenic mutations, epigenetic deregulation may be especially important. In this study we validate the histone methyltransferase G9a/EHMT2 as being associated with indicators of poor prognosis in NB. Immunological analysis of G9a protein shows it to be more highly expressed in NB cell-lines with MYCN amplification, which is a primary determinant of dismal outcome in NB patients. Furthermore, G9a protein in primary tumors is expressed at higher levels in poorly differentiated/undifferentiated NB, and correlates with high EZH2 expression, a known co-operative oncoprotein in NB. Our functional analyses demonstrate that siRNA-mediated G9a depletion inhibits cell growth in all NB cell lines, but, strikingly, only triggers apoptosis in NB cells with MYCN amplification, suggesting a synthetic lethal relationship between G9a and MYCN. This pattern of sensitivity is also evident when using small molecule inhibitors of G9a, UNC0638, and UNC0642. The increased efficacy of G9a inhibition in the presence of MYCN-overexpression is also demonstrated in the SHEP-21N isogenic model with tet-regulatable MYCN. Finally, using RNA sequencing, we identify several potential tumor suppressor genes that are reactivated by G9a inhibition in NB, including the CLU, FLCN, AMHR2, and AKR1C1-3. Together, our study underlines the under-appreciated role of G9a in NB, especially in MYCN-amplified tumors.
Collapse
Affiliation(s)
- Jacob Bellamy
- Cancer Epigenetics Laboratory, School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Marianna Szemes
- Cancer Epigenetics Laboratory, School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Zsombor Melegh
- Cancer Epigenetics Laboratory, School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
- Department of Cellular Pathology, Southmead Hospital, Bristol, United Kingdom
| | - Anthony Dallosso
- Department of Cellular Pathology, Southmead Hospital, Bristol, United Kingdom
| | - Madhu Kollareddy
- Cancer Epigenetics Laboratory, School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Daniel Catchpoole
- The Kids Research Institute, The Children's Hospital at Westmead, Westmead, NSW, Australia
| | - Karim Malik
- Cancer Epigenetics Laboratory, School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
12
|
Upton K, Modi A, Patel K, Kendsersky NM, Conkrite KL, Sussman RT, Way GP, Adams RN, Sacks GI, Fortina P, Diskin SJ, Maris JM, Rokita JL. Epigenomic profiling of neuroblastoma cell lines. Sci Data 2020; 7:116. [PMID: 32286315 PMCID: PMC7156688 DOI: 10.1038/s41597-020-0458-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 03/12/2020] [Indexed: 12/19/2022] Open
Abstract
Understanding the aberrant transcriptional landscape of neuroblastoma is necessary to provide insight to the underlying influences of the initiation, progression and persistence of this developmental cancer. Here, we present chromatin immunoprecipitation sequencing (ChIP-Seq) data for the oncogenic transcription factors, MYCN and MYC, as well as regulatory histone marks H3K4me1, H3K4me3, H3K27Ac, and H3K27me3 in ten commonly used human neuroblastoma-derived cell line models. In addition, for all of the profiled cell lines we provide ATAC-Seq as a measure of open chromatin. We validate specificity of global MYCN occupancy in MYCN amplified cell lines and functional redundancy of MYC occupancy in MYCN non-amplified cell lines. Finally, we show with H3K27Ac ChIP-Seq that these cell lines retain expression of key neuroblastoma super-enhancers (SE). We anticipate this dataset, coupled with available transcriptomic profiling on the same cell lines, will enable the discovery of novel gene regulatory mechanisms in neuroblastoma.
Collapse
Affiliation(s)
- Kristen Upton
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA
| | - Apexa Modi
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA
- Genomics and Computational Biology Graduate Group, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Khushbu Patel
- Department of Bioinformatics and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA
| | - Nathan M Kendsersky
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA
- Pharmacology Graduate Group, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Karina L Conkrite
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA
| | - Robyn T Sussman
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA
| | - Gregory P Way
- Genomics and Computational Biology Graduate Group, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Rebecca N Adams
- Cancer Genomics and Bioinformatics Laboratory, Sidney Kimmel Cancer Center, Philadelphia, Pennsylvania, 19107, USA
| | - Gregory I Sacks
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA
| | - Paolo Fortina
- Cancer Genomics and Bioinformatics Laboratory, Sidney Kimmel Cancer Center, Philadelphia, Pennsylvania, 19107, USA
| | - Sharon J Diskin
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
| | - John M Maris
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA.
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA.
| | - Jo Lynne Rokita
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA.
- Department of Bioinformatics and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA.
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA.
| |
Collapse
|
13
|
A Wnt-BMP4 Signaling Axis Induces MSX and NOTCH Proteins and Promotes Growth Suppression and Differentiation in Neuroblastoma. Cells 2020; 9:cells9030783. [PMID: 32210188 PMCID: PMC7140810 DOI: 10.3390/cells9030783] [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: 02/10/2020] [Revised: 03/18/2020] [Accepted: 03/19/2020] [Indexed: 01/09/2023] Open
Abstract
The Wnt and bone morphogenetic protein (BMP) signaling pathways are known to be crucial in the development of neural crest lineages, including the sympathetic nervous system. Surprisingly, their role in paediatric neuroblastoma, the prototypic tumor arising from this lineage, remains relatively uncharacterised. We previously demonstrated that Wnt/b-catenin signaling can have cell-type-specific effects on neuroblastoma phenotypes, including growth inhibition and differentiation, and that BMP4 mRNA and protein were induced by Wnt3a/Rspo2. In this study, we characterised the phenotypic effects of BMP4 on neuroblastoma cells, demonstrating convergent induction of MSX homeobox transcription factors by Wnt and BMP4 signaling and BMP4-induced growth suppression and differentiation. An immunohistochemical analysis of BMP4 expression in primary neuroblastomas confirms a striking absence of BMP4 in poorly differentiated tumors, in contrast to a high expression in ganglion cells. These results are consistent with a tumor suppressive role for BMP4 in neuroblastoma. RNA sequencing following BMP4 treatment revealed induction of Notch signaling, verified by increases of Notch3 and Hes1 proteins. Together, our data demonstrate, for the first time, Wnt-BMP-Notch signaling crosstalk associated with growth suppression of neuroblastoma.
Collapse
|
14
|
Targeting metabolic activity in high-risk neuroblastoma through Monocarboxylate Transporter 1 (MCT1) inhibition. Oncogene 2020; 39:3555-3570. [PMID: 32123312 DOI: 10.1038/s41388-020-1235-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 02/13/2020] [Accepted: 02/19/2020] [Indexed: 12/13/2022]
Abstract
Amplification of the MYCN oncogene occurs in ~25% of primary neuroblastomas and is the single most powerful biological marker of poor prognosis in this disease. MYCN transcriptionally regulates a range of biological processes important for cancer, including cell metabolism. The MYCN-regulated metabolic gene SLC16A1, encoding the lactate transporter monocarboxylate transporter 1 (MCT1), is a potential therapeutic target. Treatment of neuroblastoma cells with the MCT1 inhibitor SR13800 increased intracellular lactate levels, disrupted the nicotinamide adenine dinucleotide (NADH/NAD+) ratio, and decreased intracellular glutathione levels. Metabolite tracing with 13C-glucose and 13C-glutamine following MCT1 inhibitor treatment revealed increased quantities of tricarboxylic acid (TCA) cycle intermediates and increased oxygen consumption rate. MCT1 inhibition was highly synergistic with vincristine and LDHA inhibition under cell culture conditions, but this combination was ineffective against neuroblastoma xenografts. Posttreatment xenograft tumors had increased synthesis of the MCT1 homolog MCT4/SLC16A, a known resistance factor to MCT1 inhibition. We found that MCT4 was negatively regulated by MYCN in luciferase reporter assays and its synthesis in neuroblastoma cells was increased under hypoxic conditions and following hypoxia-inducible factor (HIF1) induction, suggesting that MCT4 may contribute to resistance to MCT1 inhibitor treatment in hypoxic neuroblastoma tumors. Co-treatment of neuroblastoma cells with inhibitors of MCT1 and LDHA, the enzyme responsible for lactate production, resulted in a large increase in intracellular pyruvate and was highly synergistic in decreasing neuroblastoma cell viability. These results highlight the potential of targeting MCT1 in neuroblastoma in conjunction with strategies that involve disruption of pyruvate homeostasis and indicate possible resistance mechanisms.
Collapse
|
15
|
Hu X, Zheng W, Zhu Q, Gu L, Du Y, Han Z, Zhang X, Carter DR, Cheung BB, Qiu A, Jiang C. Increase in DNA Damage by MYCN Knockdown Through Regulating Nucleosome Organization and Chromatin State in Neuroblastoma. Front Genet 2019; 10:684. [PMID: 31396265 PMCID: PMC6667652 DOI: 10.3389/fgene.2019.00684] [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: 04/30/2019] [Accepted: 07/01/2019] [Indexed: 11/25/2022] Open
Abstract
As a transcription factor, MYCN regulates myriad target genes including the histone chaperone FACT. Moreover, FACT and MYCN expression form a forward feedback loop in neuroblastoma. It is unclear whether MYCN is involved in chromatin remodeling in neuroblastoma through regulation of its target genes. We showed here that MYCN knockdown resulted in loss of the nucleosome-free regions through nucleosome assembly in the promoters of genes functionally enriched for DNA repair. The active mark H3K9ac was removed or replaced by the repressive mark H3K27me3 in the promoters of double-strand break repair-related genes upon MYCN knockdown. Such chromatin state alterations occurred only in MYCN-bound promoters. Consistently, MYCN knockdown resulted in a marked increase in DNA damage in the treatment with hydroxyurea. In contrast, nucleosome reorganization and histone modification changes in the enhancers largely included target genes with tumorigenesis-related functions such as cell proliferation, cell migration, and cell–cell adhesion. The chromatin state significantly changed in both MYCN-bound and MYCN-unbound enhancers upon MYCN knockdown. Furthermore, MYCN knockdown independently regulated chromatin remodeling in the promoters and the enhancers. These findings reveal the novel epigenetic regulatory role of MYCN in chromatin remodeling and provide an alternative potential epigenetic strategy for MYCN-driven neuroblastoma treatment.
Collapse
Affiliation(s)
- Xinjie Hu
- Institute of Translational Research, Tongji Hospital, the School of Life Sciences and Technology, Shanghai Key Laboratory of Signaling and Disease Research, Tongji University, Shanghai, China
| | - Weisheng Zheng
- Institute of Translational Research, Tongji Hospital, the School of Life Sciences and Technology, Shanghai Key Laboratory of Signaling and Disease Research, Tongji University, Shanghai, China
| | - Qianshu Zhu
- Institute of Translational Research, Tongji Hospital, the School of Life Sciences and Technology, Shanghai Key Laboratory of Signaling and Disease Research, Tongji University, Shanghai, China
| | - Liang Gu
- Institute of Translational Research, Tongji Hospital, the School of Life Sciences and Technology, Shanghai Key Laboratory of Signaling and Disease Research, Tongji University, Shanghai, China
| | - Yanhua Du
- Institute of Translational Research, Tongji Hospital, the School of Life Sciences and Technology, Shanghai Key Laboratory of Signaling and Disease Research, Tongji University, Shanghai, China
| | - Zhe Han
- Institute of Translational Research, Tongji Hospital, the School of Life Sciences and Technology, Shanghai Key Laboratory of Signaling and Disease Research, Tongji University, Shanghai, China
| | - Xiaobai Zhang
- Institute of Translational Research, Tongji Hospital, the School of Life Sciences and Technology, Shanghai Key Laboratory of Signaling and Disease Research, Tongji University, Shanghai, China
| | - Daniel R Carter
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW, Australia.,School of Women's and Children's Health, UNSW Sydney, Randwick, NSW, Australia.,School of Biomedical Engineering, University of Technology, Sydney, NSW, Australia
| | - Belamy B Cheung
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW, Australia.,School of Women's and Children's Health, UNSW Sydney, Randwick, NSW, Australia
| | - Andong Qiu
- Institute of Translational Research, Tongji Hospital, the School of Life Sciences and Technology, Shanghai Key Laboratory of Signaling and Disease Research, Tongji University, Shanghai, China.,The Research Center of Stem Cells and Ageing, Tsingtao Advanced Research Institute, Tongji University, Tsingdao, China
| | - Cizhong Jiang
- Institute of Translational Research, Tongji Hospital, the School of Life Sciences and Technology, Shanghai Key Laboratory of Signaling and Disease Research, Tongji University, Shanghai, China.,The Research Center of Stem Cells and Ageing, Tsingtao Advanced Research Institute, Tongji University, Tsingdao, China
| |
Collapse
|
16
|
Szemes M, Greenhough A, Malik K. Wnt Signaling Is a Major Determinant of Neuroblastoma Cell Lineages. Front Mol Neurosci 2019; 12:90. [PMID: 31040767 PMCID: PMC6476918 DOI: 10.3389/fnmol.2019.00090] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 03/21/2019] [Indexed: 01/09/2023] Open
Abstract
The neural crest (NC), which has been referred to as the fourth germ layer, comprises a multipotent cell population which will specify diverse cells and tissues, including craniofacial cartilage and bones, melanocytes, the adrenal medulla and the peripheral nervous system. These cell fates are known to be determined by gene regulatory networks (GRNs) acting at various stages of NC development, such as induction, specification, and migration. Although transcription factor hierarchies and some of their interplay with morphogenetic signaling pathways have been characterized, the full complexity of activities required for regulated development remains uncharted. Deregulation of these pathways may contribute to tumorigenesis, as in the case of neuroblastoma, a frequently lethal embryonic cancer thought to arise from the sympathoadrenal lineage of the NC. In this “Hypothesis and Theory” article, we utilize the next generation sequencing data from neuroblastoma cells and tumors to evaluate the possible influences of Wnt signaling on NC GRNs and on neuroblastoma cell lineages. We propose that Wnt signaling is a major determinant of regulatory networks that underlie mesenchymal/neural crest cell (NCC)-like cell identities through PRRX1 and YAP/TAZ transcription factors. Furthermore, Wnt may also co-operate with Hedgehog signaling in driving proneural differentiation programmes along the adrenergic (ADRN) lineage. Elucidation of Signaling Regulatory Networks can augment and complement GRNs in characterizing cell identities, which may in turn contribute to the design of improved therapeutics tailored to primary and relapsing neuroblastoma.
Collapse
Affiliation(s)
- Marianna Szemes
- Cancer Epigenetics Laboratory, School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Alexander Greenhough
- Cancer Epigenetics Laboratory, School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Karim Malik
- Cancer Epigenetics Laboratory, School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
17
|
Nunes-Xavier CE, Zaldumbide L, Aurtenetxe O, López-Almaraz R, López JI, Pulido R. Dual-Specificity Phosphatases in Neuroblastoma Cell Growth and Differentiation. Int J Mol Sci 2019; 20:ijms20051170. [PMID: 30866462 PMCID: PMC6429076 DOI: 10.3390/ijms20051170] [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: 02/13/2019] [Revised: 03/01/2019] [Accepted: 03/01/2019] [Indexed: 12/19/2022] Open
Abstract
Dual-specificity phosphatases (DUSPs) are important regulators of neuronal cell growth and differentiation by targeting proteins essential to neuronal survival in signaling pathways, among which the MAP kinases (MAPKs) stand out. DUSPs include the MAPK phosphatases (MKPs), a family of enzymes that directly dephosphorylate MAPKs, as well as the small-size atypical DUSPs, a group of low molecular-weight enzymes which display more heterogeneous substrate specificity. Neuroblastoma (NB) is a malignancy intimately associated with the course of neuronal and neuroendocrine cell differentiation, and constitutes the source of more common extracranial solid pediatric tumors. Here, we review the current knowledge on the involvement of MKPs and small-size atypical DUSPs in NB cell growth and differentiation, and discuss the potential of DUSPs as predictive biomarkers and therapeutic targets in human NB.
Collapse
Affiliation(s)
- Caroline E Nunes-Xavier
- Biomarkers in Cancer Unit, Biocruces-Bizkaia Health Research Institute, Barakaldo, Bizkaia 48903, Spain.
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital HF Radiumhospitalet, Oslo 0424, Norway.
| | - Laura Zaldumbide
- Department of Pathology, Cruces University Hospital, University of the Basque Country (UPV/EHU), Barakaldo, Bizkaia 48903, Spain.
| | - Olaia Aurtenetxe
- Biomarkers in Cancer Unit, Biocruces-Bizkaia Health Research Institute, Barakaldo, Bizkaia 48903, Spain.
| | - Ricardo López-Almaraz
- Pediatric Oncology and Hematology, Cruces University Hospital, Barakaldo, Bizkaia 48903, Spain.
| | - José I López
- Biomarkers in Cancer Unit, Biocruces-Bizkaia Health Research Institute, Barakaldo, Bizkaia 48903, Spain.
- Department of Pathology, Cruces University Hospital, University of the Basque Country (UPV/EHU), Barakaldo, Bizkaia 48903, Spain.
| | - Rafael Pulido
- Biomarkers in Cancer Unit, Biocruces-Bizkaia Health Research Institute, Barakaldo, Bizkaia 48903, Spain.
- IKERBASQUE, Basque Foundation for Science, Bilbao 48011, Spain.
| |
Collapse
|
18
|
Saeki N, Saito A, Sugaya Y, Amemiya M, Sasaki H. Indirect Down-regulation of Tumor-suppressive let-7 Family MicroRNAs by LMO1 in Neuroblastoma. Cancer Genomics Proteomics 2018; 15:413-420. [PMID: 30194082 DOI: 10.21873/cgp.20100] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 07/19/2018] [Accepted: 07/23/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND/AIM Overall survival for the high-risk group of neuroblastoma (NB) patients still remains at 40-50%, necessitating the establishment of a curable treatment. LIM domain only 1 (LMO1) gene encoding a transcriptional regulator is an NB-susceptibility gene with a tumor-promoting activity. Previously we conducted chromatin immunoprecipitation and DNA sequencing analyses on NB cell lines and identified 3 protein-coding genes regulated by LMO1. In this study, we extended our analyses to capture microRNA genes directly or indirectly regulated by LMO1. MATERIALS AND METHODS Using microarrays, we conducted a comparative gene expression analysis on an NB cell line SK-N-SH; between the cells with and without LMO1 suppression. RESULTS Overall, 18 microRNAs were identified to be indirectly down-regulated by LMO1 including 7 microRNAs of the let-7 family, whose cell proliferation inhibitory activity was observed. CONCLUSION Target genes of the LMO1-regulated microRNAs and their relevant pathways may be a potential therapeutic target.
Collapse
Affiliation(s)
- Norihisa Saeki
- Division of Anatomy and Physiology, Okinawa Prefectural College of Nursing, Okinawa, Japan
| | - Akira Saito
- Statistical Genetics Analysis Division, StaGen Co. Ltd., Tokyo, Japan
| | - Yuki Sugaya
- Statistical Genetics Analysis Division, StaGen Co. Ltd., Tokyo, Japan
| | - Mitsuhiro Amemiya
- Statistical Genetics Analysis Division, StaGen Co. Ltd., Tokyo, Japan
| | - Hiroki Sasaki
- Department of Translational Oncology, National Cancer Center Research Institute, Tokyo, Japan
| |
Collapse
|
19
|
Szemes M, Greenhough A, Melegh Z, Malik S, Yuksel A, Catchpoole D, Gallacher K, Kollareddy M, Park JH, Malik K. Wnt Signalling Drives Context-Dependent Differentiation or Proliferation in Neuroblastoma. Neoplasia 2018; 20:335-350. [PMID: 29505958 PMCID: PMC5909736 DOI: 10.1016/j.neo.2018.01.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 01/17/2018] [Accepted: 01/21/2018] [Indexed: 01/09/2023]
Abstract
Neuroblastoma is one of the commonest and deadliest solid tumours of childhood, and is thought to result from disrupted differentiation of the developing sympathoadrenergic lineage of the neural crest. Neuroblastoma exhibits intra- and intertumoural heterogeneity, with high risk tumours characterised by poor differentiation, which can be attributable to MYCN-mediated repression of genes involved in neuronal differentiation. MYCN is known to co-operate with oncogenic signalling pathways such as Alk, Akt and MEK/ERK signalling, and, together with c-MYC has been shown to be activated by Wnt signalling in various tissues. However, our previous work demonstrated that Wnt3a/Rspo2 treatment of some neuroblastoma cell lines can, paradoxically, decrease c-MYC and MYCN proteins. This prompted us to define the neuroblastoma-specific Wnt3a/Rspo2-driven transcriptome using RNA sequencing, and characterise the accompanying changes in cell biology. Here we report the identification of ninety Wnt target genes, and show that Wnt signalling is upstream of numerous transcription factors and signalling pathways in neuroblastoma. Using live-cell imaging, we show that Wnt signalling can drive differentiation of SK-N-BE(2)-C and SH-SY5Y cell-lines, but, conversely, proliferation of SK-N-AS cells. We show that cell-lines that differentiate show induction of pro-differentiation BMP4 and EPAS1 proteins, which is not apparent in the SK-N-AS cells. In contrast, SK-N-AS cells show increased CCND1, phosphorylated RB and E2F1 in response to Wnt3a/Rspo2, consistent with their proliferative response, and these proteins are not increased in differentiating lines. By meta-analysis of the expression of our 90 genes in primary tumour gene expression databases, we demonstrate discrete expression patterns of our Wnt genes in patient cohorts with different prognosis. Furthermore our analysis reveals interconnectivity within subsets of our Wnt genes, with one subset comprised of novel putative drivers of neuronal differentiation repressed by MYCN. Assessment of β-catenin immunohistochemistry shows high levels of β-catenin in tumours with better differentiation, further supporting a role for canonical Wnt signalling in neuroblastoma differentiation.
Collapse
Key Words
- alk, anaplastic lymphoma kinase
- atra, all-trans-retinoic acid
- bmp4, bone morphogenetic protein 4
- ccnd1, cyclin d1
- egf, epidermal growth factor
- epas1, endothelial pas domain protein 1
- erk, extracellular signal-regulated kinases
- emt, epithelial-mesenchymal transition
- kegg, kyoto encyclopedia of genes and genomes
- mapk, mitogen-activated protein kinase
- mek, mitogen-activated protein kinase kinase
- pbs, phosphate-buffered saline
- qrt-pcr, quantitative reverse-transcriptase polymerase chain reaction
- rb, retinoblastoma
- rnaseq, rna sequencing
- rspo2, r-spondin-2
- sds-page, sodium-dodecyl sulphate-polyacrylamide gel electrophoresis
- tcf/lef, t-cell factor/lymphoid enhancer binding factor
Collapse
Affiliation(s)
- Marianna Szemes
- Cancer Epigenetics Laboratory, School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Alexander Greenhough
- Cancer Epigenetics Laboratory, School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Zsombor Melegh
- Department of Cellular Pathology, Southmead Hospital, Bristol, UK
| | - Sally Malik
- Cancer Epigenetics Laboratory, School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Aysen Yuksel
- The Kids Research Institute, The Children's Hospital at Westmead, Westmead, Westmead NSW, 2145, Australia
| | - Daniel Catchpoole
- The Kids Research Institute, The Children's Hospital at Westmead, Westmead, Westmead NSW, 2145, Australia
| | - Kelli Gallacher
- Cancer Epigenetics Laboratory, School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Madhu Kollareddy
- Cancer Epigenetics Laboratory, School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Ji Hyun Park
- Cancer Epigenetics Laboratory, School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Karim Malik
- Cancer Epigenetics Laboratory, School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK.
| |
Collapse
|
20
|
Ferrucci F, Ciaccio R, Monticelli S, Pigini P, di Giacomo S, Purgato S, Erriquez D, Bernardoni R, Norris M, Haber M, Milazzo G, Perini G. MAX to MYCN intracellular ratio drives the aggressive phenotype and clinical outcome of high risk neuroblastoma. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2018; 1861:235-245. [DOI: 10.1016/j.bbagrm.2018.01.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 12/22/2017] [Accepted: 01/04/2018] [Indexed: 12/17/2022]
|
21
|
Durinck K, Speleman F. Epigenetic regulation of neuroblastoma development. Cell Tissue Res 2018; 372:309-324. [PMID: 29350283 DOI: 10.1007/s00441-017-2773-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 12/16/2017] [Indexed: 02/07/2023]
Abstract
In recent years, technological advances have enabled a detailed landscaping of the epigenome and the mechanisms of epigenetic regulation that drive normal cell function, development and cancer. Rather than merely a structural entity to support genome compaction, we now look at chromatin as a very dynamic and essential constellation that is actively participating in the tight orchestration of transcriptional regulation as well as DNA replication and repair. The unique feature of chromatin flexibility enabling fast switches towards more or less restricted epigenetic cellular states is, not surprisingly, intimately connected to cancer development and treatment resistance, and the central role of epigenetic alterations in cancer is illustrated by the finding that up to 50% of all mutations across cancer entities affect proteins controlling the chromatin status. We summarize recent insights into epigenetic rewiring underlying neuroblastoma (NB) tumor formation ranging from changes in DNA methylation patterns and mutations in epigenetic regulators to global effects on transcriptional regulatory circuits that involve key players in NB oncogenesis. Insights into the disruption of the homeostatic epigenetic balance contributing to developmental arrest of sympathetic progenitor cells and subsequent NB oncogenesis are rapidly growing and will be exploited towards the development of novel therapeutic strategies to increase current survival rates of patients with high-risk NB.
Collapse
Affiliation(s)
- Kaat Durinck
- Center for Medical Genetics, Ghent University, Ghent, Belgium.
| | - Frank Speleman
- Center for Medical Genetics, Ghent University, Ghent, Belgium
| |
Collapse
|
22
|
Xue C, Yu DMT, Gherardi S, Koach J, Milazzo G, Gamble L, Liu B, Valli E, Russell AJ, London WB, Liu T, Cheung BB, Marshall GM, Perini G, Haber M, Norris MD. MYCN promotes neuroblastoma malignancy by establishing a regulatory circuit with transcription factor AP4. Oncotarget 2018; 7:54937-54951. [PMID: 27448979 PMCID: PMC5342392 DOI: 10.18632/oncotarget.10709] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 06/16/2016] [Indexed: 12/11/2022] Open
Abstract
Amplification of the MYCN oncogene, a member of the MYC family of transcriptional regulators, is one of the most powerful prognostic markers identified for poor outcome in neuroblastoma, the most common extracranial solid cancer in childhood. While MYCN has been established as a key driver of malignancy in neuroblastoma, the underlying molecular mechanisms are poorly understood. Transcription factor activating enhancer binding protein-4 (TFAP4) has been reported to be a direct transcriptional target of MYC. We show for the first time that high expression of TFAP4 in primary neuroblastoma patients is associated with poor clinical outcome. siRNA-mediated suppression of TFAP4 in MYCN-expressing neuroblastoma cells led to inhibition of cell proliferation and migration. Chromatin immunoprecipitation assay demonstrated that TFAP4 expression is positively regulated by MYCN. Microarray analysis identified genes regulated by both MYCN and TFAP4 in neuroblastoma cells, including Phosphoribosyl-pyrophosphate synthetase-2 (PRPS2) and Syndecan-1 (SDC1), which are involved in cancer cell proliferation and metastasis. Overall this study suggests a regulatory circuit in which MYCN by elevating TFAP4 expression, cooperates with it to control a specific set of genes involved in tumor progression. These findings highlight the existence of a MYCN-TFAP4 axis in MYCN-driven neuroblastoma as well as identifying potential therapeutic targets for aggressive forms of this disease.
Collapse
Affiliation(s)
- Chengyuan Xue
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia
| | - Denise M T Yu
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia
| | - Samuele Gherardi
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Jessica Koach
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia
| | - Giorgio Milazzo
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Laura Gamble
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia
| | - Bing Liu
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia
| | - Emanuele Valli
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia
| | - Amanda J Russell
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia
| | - Wendy B London
- Boston Children's Hospital and Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Tao Liu
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia
| | - Belamy B Cheung
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia
| | - Glenn M Marshall
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia.,Kids Cancer Centre, Sydney Children's Hospital, Sydney, Australia
| | - Giovanni Perini
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy.,CIRI Health Sciences and Technologies University of Bologna, Bologna, Italy
| | - Michelle Haber
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia
| | - Murray D Norris
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia.,University of New South Wales Centre for Childhood Cancer Research, Sydney, Australia
| |
Collapse
|
23
|
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: 1.3] [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
|
24
|
Hsu CL, Chang HY, Chang JY, Hsu WM, Huang HC, Juan HF. Unveiling MYCN regulatory networks in neuroblastoma via integrative analysis of heterogeneous genomics data. Oncotarget 2017; 7:36293-36310. [PMID: 27167114 PMCID: PMC5095001 DOI: 10.18632/oncotarget.9202] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 04/19/2016] [Indexed: 12/31/2022] Open
Abstract
MYCN, an oncogenic transcription factor of the Myc family, is a major driver of neuroblastoma tumorigenesis. Due to the difficulty in drugging MYCN directly, revealing the molecules in MYCN regulatory networks will help to identify effective therapeutic targets for neuroblastoma therapy. Here we perform ChIP-sequencing and small RNA-sequencing of neuroblastoma cells to determine the MYCN-binding sites and MYCN-associated microRNAs, and integrate various types of genomic data to construct MYCN regulatory networks. The overall analysis indicated that MYCN-regulated genes were involved in a wide range of biological processes and could be used as signatures to identify poor-prognosis MYCN-non-amplified patients. Analysis of the MYCN binding sites showed that MYCN principally served as an activator. Using a computational approach, we identified 32 MYCN co-regulators, and some of these findings are supported by previous studies. Moreover, we investigated the interplay between MYCN transcriptional and microRNA post-transcriptional regulations and identified several microRNAs, such as miR-124-3p and miR-93-5p, which may significantly contribute to neuroblastoma pathogenesis. We also found MYCN and its regulated microRNAs acted together to repress the tumor suppressor genes. This work provides a comprehensive view of MYCN regulations for exploring therapeutic targets in neuroblastoma, as well as insights into the mechanism of neuroblastoma tumorigenesis.
Collapse
Affiliation(s)
- Chia-Lang Hsu
- Department of Life Science, Institute of Molecular and Cellular Biology, Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei 106, Taiwan
| | - Hsin-Yi Chang
- Department of Life Science, Institute of Molecular and Cellular Biology, Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei 106, Taiwan
| | - Jen-Yun Chang
- Department of Life Science, Institute of Molecular and Cellular Biology, Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei 106, Taiwan
| | - Wen-Ming Hsu
- Department of Surgery, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Hsuan-Cheng Huang
- Institute of Biomedical Informatics, Center for Systems and Synthetic Biology, National Yang-Ming University, Taipei 112, 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 106, Taiwan
| |
Collapse
|
25
|
Nataf S, Barritault M, Pays L. A Unique TGFB1-Driven Genomic Program Links Astrocytosis, Low-Grade Inflammation and Partial Demyelination in Spinal Cord Periplaques from Progressive Multiple Sclerosis Patients. Int J Mol Sci 2017; 18:ijms18102097. [PMID: 28981455 PMCID: PMC5666779 DOI: 10.3390/ijms18102097] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 09/25/2017] [Accepted: 09/29/2017] [Indexed: 02/08/2023] Open
Abstract
We previously reported that, in multiple sclerosis (MS) patients with a progressive form of the disease, spinal cord periplaques extend distance away from plaque borders and are characterized by the co-occurrence of partial demyelination, astrocytosis and low-grade inflammation. However, transcriptomic analyses did not allow providing a comprehensive view of molecular events in astrocytes vs. oligodendrocytes. Here, we re-assessed our transcriptomic data and performed co-expression analyses to characterize astrocyte vs. oligodendrocyte molecular signatures in periplaques. We identified an astrocytosis-related co-expression module whose central hub was the astrocyte gene Cx43/GJA1 (connexin-43, also named gap junction protein α-1). Such a module comprised GFAP (glial fibrillary acidic protein) and a unique set of transcripts forming a TGFB/SMAD1/SMAD2 (transforming growth factor β/SMAD family member 1/SMAD family member 2) genomic signature. Partial demyelination was characterized by a co-expression network whose central hub was the oligodendrocyte gene NDRG1 (N-myc downstream regulated 1), a gene previously shown to be specifically silenced in the normal-appearing white matter (NAWM) of MS patients. Surprisingly, besides myelin genes, the NDRG1 co-expression module comprised a highly significant number of translation/elongation-related genes. To identify a putative cause of NDRG1 downregulation in periplaques, we then sought to identify the cytokine/chemokine genes whose mRNA levels inversely correlated with those of NDRG1. Following this approach, we found five candidate immune-related genes whose upregulation associated with NDRG1 downregulation: TGFB1(transforming growth factor β 1), PDGFC (platelet derived growth factor C), IL17D (interleukin 17D), IL33 (interleukin 33), and IL12A (interleukin 12A). From these results, we propose that, in the spinal cord periplaques of progressive MS patients, TGFB1 may limit acute inflammation but concurrently induce astrocytosis and an alteration of the translation/elongation of myelin genes in oligodendrocytes.
Collapse
Affiliation(s)
- Serge Nataf
- Univ Lyon, CarMeN laboratory, Inserm U1060, INRA U1397, Université Claude Bernard Lyon 1, INSA Lyon, Charles Merieux Medical School, F-69600 Oullins, France.
- Banque de Tissus et de Cellules des Hospices Civils de Lyon, Hôpital Edouard Herriot, Place d'Arsonval, F-69003 Lyon, France.
| | - Marc Barritault
- Univ Lyon, Department of Cancer Cell Plasticity, Cancer Research Center of Lyon, INSERMU1052, CNRS UMR5286, University Claude Bernard Lyon 1, 151 Cours Albert Thomas, 69003 Lyon, France.
- Service d'Anatomie Pathologique, Hospices Civils de Lyon, Groupement Hospitalier Est, 59 boulevard Pinel, 69677 Bron, France.
| | - Laurent Pays
- Univ Lyon, CarMeN laboratory, Inserm U1060, INRA U1397, Université Claude Bernard Lyon 1, INSA Lyon, Charles Merieux Medical School, F-69600 Oullins, France.
- Banque de Tissus et de Cellules des Hospices Civils de Lyon, Hôpital Edouard Herriot, Place d'Arsonval, F-69003 Lyon, France.
| |
Collapse
|
26
|
The MYCN Protein in Health and Disease. Genes (Basel) 2017; 8:genes8040113. [PMID: 28358317 PMCID: PMC5406860 DOI: 10.3390/genes8040113] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 03/23/2017] [Accepted: 03/27/2017] [Indexed: 12/22/2022] Open
Abstract
MYCN is a member of the MYC family of proto-oncogenes. It encodes a transcription factor, MYCN, involved in the control of fundamental processes during embryonal development. The MYCN protein is situated downstream of several signaling pathways promoting cell growth, proliferation and metabolism of progenitor cells in different developing organs and tissues. Conversely, deregulated MYCN signaling supports the development of several different tumors, mainly with a childhood onset, including neuroblastoma, medulloblastoma, rhabdomyosarcoma and Wilms’ tumor, but it is also associated with some cancers occurring during adulthood such as prostate and lung cancer. In neuroblastoma, MYCN-amplification is the most consistent genetic aberration associated with poor prognosis and treatment failure. Targeting MYCN has been proposed as a therapeutic strategy for the treatment of these tumors and great efforts have allowed the development of direct and indirect MYCN inhibitors with potential clinical use.
Collapse
|
27
|
Duffy DJ, Krstic A, Halasz M, Schwarzl T, Fey D, Iljin K, Mehta JP, Killick K, Whilde J, Turriziani B, Haapa-Paananen S, Fey V, Fischer M, Westermann F, Henrich KO, Bannert S, Higgins DG, Kolch W. Integrative omics reveals MYCN as a global suppressor of cellular signalling and enables network-based therapeutic target discovery in neuroblastoma. Oncotarget 2016; 6:43182-201. [PMID: 26673823 PMCID: PMC4791225 DOI: 10.18632/oncotarget.6568] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 11/23/2015] [Indexed: 12/12/2022] Open
Abstract
Despite intensive study, many mysteries remain about the MYCN oncogene's functions. Here we focus on MYCN's role in neuroblastoma, the most common extracranial childhood cancer. MYCN gene amplification occurs in 20% of cases, but other recurrent somatic mutations are rare. This scarcity of tractable targets has hampered efforts to develop new therapeutic options. We employed a multi-level omics approach to examine MYCN functioning and identify novel therapeutic targets for this largely un-druggable oncogene. We used systems medicine based computational network reconstruction and analysis to integrate a range of omic techniques: sequencing-based transcriptomics, genome-wide chromatin immunoprecipitation, siRNA screening and interaction proteomics, revealing that MYCN controls highly connected networks, with MYCN primarily supressing the activity of network components. MYCN's oncogenic functions are likely independent of its classical heterodimerisation partner, MAX. In particular, MYCN controls its own protein interaction network by transcriptionally regulating its binding partners. Our network-based approach identified vulnerable therapeutically targetable nodes that function as critical regulators or effectors of MYCN in neuroblastoma. These were validated by siRNA knockdown screens, functional studies and patient data. We identified β-estradiol and MAPK/ERK as having functional cross-talk with MYCN and being novel targetable vulnerabilities of MYCN-amplified neuroblastoma. These results reveal surprising differences between the functioning of endogenous, overexpressed and amplified MYCN, and rationalise how different MYCN dosages can orchestrate cell fate decisions and cancerous outcomes. Importantly, this work describes a systems-level approach to systematically uncovering network based vulnerabilities and therapeutic targets for multifactorial diseases by integrating disparate omic data types.
Collapse
Affiliation(s)
- David J Duffy
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland.,The Whitney Laboratory for Marine Bioscience, University of Florida, St. Augustine, Florida, USA
| | - Aleksandar Krstic
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland
| | - Melinda Halasz
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland
| | - Thomas Schwarzl
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland.,European Molecular Biology Laboratory (EMBL), Meyerhofstraße, Heidelberg, Germany
| | - Dirk Fey
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland
| | | | - Jai Prakash Mehta
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland
| | - Kate Killick
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland
| | - Jenny Whilde
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland
| | | | | | - Vidal Fey
- VTT Technical Research Centre of Finland, Espoo, Finland
| | - Matthias Fischer
- Department of Paediatric Haematology and Oncology and Center for Molecular Medicine Cologne (CMMC), University Hospital Cologne, Cologne, Germany
| | - Frank Westermann
- Division of NB Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kai-Oliver Henrich
- Division of NB Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Steffen Bannert
- Division of NB Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Desmond G Higgins
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland.,Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin, Ireland.,School of Medicine and Medical Science, University College Dublin, Belfield, Dublin, Ireland
| | - Walter Kolch
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland.,Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin, Ireland.,School of Medicine and Medical Science, University College Dublin, Belfield, Dublin, Ireland
| |
Collapse
|
28
|
Vadie N, Saayman S, Lenox A, Ackley A, Clemson M, Burdach J, Hart J, Vogt PK, Morris KV. MYCNOS functions as an antisense RNA regulating MYCN. RNA Biol 2016; 12:893-9. [PMID: 26156430 DOI: 10.1080/15476286.2015.1063773] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Amplification or overexpression of neuronal MYC (MYCN) is associated with poor prognosis of human neuroblastoma. Three isoforms of the MYCN protein have been described as well as a protein encoded by an antisense transcript (MYCNOS) that originates from the opposite strand at the MYCN locus. Recent findings suggest that some antisense long non-coding RNAs (lncRNAs) can play a role in epigenetically regulating gene expression. Here we report that MYCNOS transcripts function as a modulator of the MYCN locus, affecting MYCN promoter usage and recruiting various proteins, including the Ras GTPase-activating protein-binding protein G3BP1, to the upstream MYCN promoter. Overexpression of MYCNOS results in a reduction of upstream MYCN promoter usage and increased MYCN expression, suggesting that the protein-coding MYCNOS also functions as a regulator of MYCN ultimately controlling MYCN transcriptional variants. The observations presented here demonstrate that protein-coding transcripts can regulate gene transcription and can tether regulatory proteins to target loci.
Collapse
Affiliation(s)
- Nadia Vadie
- a Molecular and Experimental Medicine ; The Scripps Research Institute ; La Jolla , CA USA
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Amente S, Milazzo G, Sorrentino MC, Ambrosio S, Di Palo G, Lania L, Perini G, Majello B. Lysine-specific demethylase (LSD1/KDM1A) and MYCN cooperatively repress tumor suppressor genes in neuroblastoma. Oncotarget 2016; 6:14572-83. [PMID: 26062444 PMCID: PMC4546488 DOI: 10.18632/oncotarget.3990] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 04/15/2015] [Indexed: 01/23/2023] Open
Abstract
The chromatin-modifying enzyme lysine-specific demethylase 1, KDM1A/LSD1 is involved in maintaining the undifferentiated, malignant phenotype of neuroblastoma cells and its overexpression correlated with aggressive disease, poor differentiation and infaust outcome. Here, we show that LSD1 physically binds MYCN both in vitro and in vivo and that such an interaction requires the MYCN BoxIII. We found that LSD1 co-localizes with MYCN on promoter regions of CDKN1A/p21 and Clusterin (CLU) suppressor genes and cooperates with MYCN to repress the expression of these genes. KDM1A needs to engage with MYCN in order to associate with the CDKN1A and CLU promoters. The expression of CLU and CDKN1A can be restored in MYCN-amplified cells by pharmacological inhibition of LSD1 activity or knockdown of its expression. Combined pharmacological inhibition of MYCN and LSD1 through the use of small molecule inhibitors synergistically reduces MYCN-amplified Neuroblastoma cell viability in vitro. These findings demonstrate that LSD1 is a critical co-factor of the MYCN repressive function, and suggest that combination of LSD1 and MYCN inhibitors may have strong therapeutic relevance to counteract MYCN-driven oncogenesis.
Collapse
Affiliation(s)
- Stefano Amente
- Department of Biology, University of Naples 'Federico II', Naples, Italy.,Department of Molecular Medicine and Medical Biotechnologies, University of Naples 'Federico II', Naples, Italy
| | - Giorgio Milazzo
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | | | - Susanna Ambrosio
- Department of Biology, University of Naples 'Federico II', Naples, Italy
| | - Giacomo Di Palo
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples 'Federico II', Naples, Italy
| | - Luigi Lania
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples 'Federico II', Naples, Italy
| | - Giovanni Perini
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy.,CIRI Health Sciences and Technologies (HST), Bologna, Italy
| | - Barbara Majello
- Department of Biology, University of Naples 'Federico II', Naples, Italy
| |
Collapse
|
30
|
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: 4.1] [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
|
31
|
Cortés C, Kozma SC, Tauler A, Ambrosio S. MYCN concurrence with SAHA-induced cell death in human neuroblastoma cells. Cell Oncol (Dordr) 2015; 38:341-52. [PMID: 26306783 DOI: 10.1007/s13402-015-0233-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/22/2015] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND In the past, the HDAC inhibitor suberoylanilide hydroxamic acid (SAHA) has been shown to induce apoptosis in several human tumor types, including neuroblastomas. Amplification and over-expression of the MYCN oncogene is a diagnostic hallmark and a poor prognostic indicator in high-risk neuroblastomas. Here, we studied the relationship between MYCN amplification and over-expression and the anti-tumor effect of SAHA to assess whether this drug may serve as a treatment option for high-risk neuroblastomas. METHODS Different human neuroblastoma cell lines, over-expressing or not over-expressing MYCN, were used in this study. Targeted knockdown and exogenous over-expression of MYCN were employed to examine correlations between MYCN expression levels and SAHA responses. After various time periods and concentration exposures to the drug, cell viability was measured by MTS assay, and variations in MYCN mRNA and protein levels were assessed by qPCR and Western blotting, respectively. RESULTS We found that SAHA decreased cell viability in all cell lines tested through apoptosis induction, and that SAHA had a stronger effect on cell lines carrying an amplified MYCN gene. A decrease in MYCN mRNA and protein levels was observed in the SAHA treated cell lines. Subsequent silencing and exogenous over-expression of MYCN changed the proliferation rate of the cells, but did not have any significant impact on the effect of SAHA on the viability of the cells. We also found that SAHA blocked the expression of MYCN and, by doing so, reduced the effects mediated by this protein. CONCLUSIONS Our results suggest that SAHA may be used as a single-drug treatment option for neuroblastomas with an amplified MYCN gene, and as an adjuvant treatment option for all neuroblastomas.
Collapse
Affiliation(s)
- Constanza Cortés
- Departament de Bioquímica i Biologia Molecular, Facultat de Farmàcia, Universitat de Barcelona, 08028, Barcelona, Catalunya, Spain.,Laboratory of Cancer Metabolism, IDIBELL, Hospital Duran i Reynals, L'Hospitalet de Llobregat, Catalunya, 08907, Spain
| | - Sara C Kozma
- Laboratory of Cancer Metabolism, IDIBELL, Hospital Duran i Reynals, L'Hospitalet de Llobregat, Catalunya, 08907, Spain
| | - Albert Tauler
- Departament de Bioquímica i Biologia Molecular, Facultat de Farmàcia, Universitat de Barcelona, 08028, Barcelona, Catalunya, Spain.,Laboratory of Cancer Metabolism, IDIBELL, Hospital Duran i Reynals, L'Hospitalet de Llobregat, Catalunya, 08907, Spain
| | - Santiago Ambrosio
- Unitat de Bioquímica, Dep. Ciències Fisiològiques II, Facultat de Medicina, Campus Universitari de Bellvitge - IDIBELL, Universitat de Barcelona, L'Hospitalet de Llobregat, Catalunya, 08907, Spain.
| |
Collapse
|
32
|
Chemoresistance, cancer stem cells, and miRNA influences: the case for neuroblastoma. Anal Cell Pathol (Amst) 2015; 2015:150634. [PMID: 26258008 PMCID: PMC4516851 DOI: 10.1155/2015/150634] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 06/26/2015] [Accepted: 07/01/2015] [Indexed: 12/12/2022] Open
Abstract
Neuroblastoma is a type of cancer that develops most often in infants and children under the age of five years. Neuroblastoma originates within the peripheral sympathetic ganglia, with 30% of the cases developing within the adrenal medulla, although it can also occur within other regions of the body such as nerve tissue in the spinal cord, neck, chest, abdomen, and pelvis. MicroRNAs (miRNAs) regulate cellular pathways, differentiation, apoptosis, and stem cell maintenance. Such miRNAs regulate genes involved in cellular processes. Consequently, they are implicated in the regulation of a spectrum of signaling pathways within the cell. In essence, the role of miRNAs in the development of cancer is of utmost importance for the understanding of dysfunctional cellular pathways that lead to the conversion of normal cells into cancer cells. This review focuses on highlighting the recent, important implications of miRNAs within the context of neuroblastoma basic research efforts, particularly concerning miRNA influences on cancer stem cell pathology and chemoresistance pathology for this condition, together with development of translational medicine approaches for novel diagnostic tools and therapies for this neuroblastoma.
Collapse
|
33
|
Planells-Ferrer L, Urresti J, Soriano A, Reix S, Murphy DM, Ferreres JC, Borràs F, Gallego S, Stallings RL, Moubarak RS, Segura MF, Comella JX. MYCN repression of Lifeguard/FAIM2 enhances neuroblastoma aggressiveness. Cell Death Dis 2014; 5:e1401. [PMID: 25188511 PMCID: PMC4540192 DOI: 10.1038/cddis.2014.356] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 07/15/2014] [Accepted: 07/22/2014] [Indexed: 01/20/2023]
Abstract
Neuroblastoma (NBL) is the most common solid tumor in infants and accounts for 15% of all pediatric cancer deaths. Several risk factors predict NBL outcome: age at the time of diagnosis, stage, chromosome alterations and MYCN (V-Myc Avian Myelocytomatosis Viral Oncogene Neuroblastoma-Derived Homolog) amplification, which characterizes the subset of the most aggressive NBLs with an overall survival below 30%. MYCN-amplified tumors develop exceptional chemoresistance and metastatic capacity. These properties have been linked to defects in the apoptotic machinery, either by silencing components of the extrinsic apoptotic pathway (e.g. caspase-8) or by overexpression of antiapoptotic regulators (e.g. Bcl-2, Mcl-1 or FLIP). Very little is known on the implication of death receptors and their antagonists in NBL. In this work, the expression levels of several death receptor antagonists were analyzed in multiple human NBL data sets. We report that Lifeguard (LFG/FAIM2 (Fas apoptosis inhibitory molecule 2)/NMP35) is downregulated in the most aggressive and undifferentiated tumors. Intringuingly, although LFG has been initially characterized as an antiapoptotic protein, we have found a new association with NBL differentiation. Moreover, LFG repression resulted in reduced cell adhesion, increased sphere growth and enhanced migration, thus conferring a higher metastatic capacity to NBL cells. Furthermore, LFG expression was found to be directly repressed by MYCN at the transcriptional level. Our data, which support a new functional role for a hitherto undiscovered MYCN target, provide a new link between MYCN overexpression and increased NBL metastatic properties.
Collapse
Affiliation(s)
- L Planells-Ferrer
- Cell Signaling and Apoptosis Group, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - J Urresti
- Cell Signaling and Apoptosis Group, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - A Soriano
- Laboratory of Translational Research in Pediatric Cancer, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - S Reix
- Cell Signaling and Apoptosis Group, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - D M Murphy
- Molecular and Cellular Therapeutics, Royal College of Surgeons and National Children's Research Centre Our Lady's Children's Hospital, Dublin, Ireland
| | - J C Ferreres
- Hospital Vall d'Hebron, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - F Borràs
- Hospital Vall d'Hebron, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - S Gallego
- 1] Laboratory of Translational Research in Pediatric Cancer, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain [2] Hospital Vall d'Hebron, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - R L Stallings
- Molecular and Cellular Therapeutics, Royal College of Surgeons and National Children's Research Centre Our Lady's Children's Hospital, Dublin, Ireland
| | - R S Moubarak
- Cell Signaling and Apoptosis Group, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - M F Segura
- Laboratory of Translational Research in Pediatric Cancer, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - J X Comella
- Cell Signaling and Apoptosis Group, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
34
|
Hann SR. MYC cofactors: molecular switches controlling diverse biological outcomes. Cold Spring Harb Perspect Med 2014; 4:a014399. [PMID: 24939054 DOI: 10.1101/cshperspect.a014399] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The transcription factor MYC has fundamental roles in proliferation, apoptosis, tumorigenesis, and stem cell pluripotency. Over the last 30 years extensive information has been gathered on the numerous cofactors that interact with MYC and the target genes that are regulated by MYC as a means of understanding the molecular mechanisms controlling its diverse roles. Despite significant advances and perhaps because the amount of information learned about MYC is overwhelming, there has been little consensus on the molecular functions of MYC that mediate its critical biological roles. In this perspective, the major MYC cofactors that regulate the various transcriptional activities of MYC, including canonical and noncanonical transactivation and transcriptional repression, will be reviewed and a model of how these transcriptional mechanisms control MYC-mediated proliferation, apoptosis, and tumorigenesis will be presented. The basis of the model is that a variety of cofactors form dynamic MYC transcriptional complexes that can switch the molecular and biological functions of MYC to yield a diverse range of outcomes in a cell-type- and context-dependent fashion.
Collapse
Affiliation(s)
- Stephen R Hann
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-2175
| |
Collapse
|
35
|
Sala A. Editorial: Targeting MYCN in Pediatric Cancers. Front Oncol 2014; 4:330. [PMID: 26029658 PMCID: PMC4429566 DOI: 10.3389/fonc.2014.00330] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 10/31/2014] [Indexed: 01/09/2023] Open
Affiliation(s)
- Arturo Sala
- Brunel Institute of Cancer Genetics and Pharmacogenomics, College of Health and Life Sciences, Brunel University London , London , UK
| |
Collapse
|