1
|
Qiao S, Jia Y, Xie L, Jing W, Xia Y, Song Y, Zhang J, Cao T, Song H, Meng L, Shi L, Zhang X. KLF7 promotes neuroblastoma differentiation through the GTPase signaling pathway by upregulating neuroblast differentiation-associated protein AHNAKs and glycerophosphodiesterase GDPD5. FEBS J 2024; 291:3870-3888. [PMID: 38924469 DOI: 10.1111/febs.17208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 03/10/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024]
Abstract
The arrest of neural crest-derived sympathoadrenal neuroblast differentiation contributes to neuroblastoma formation, and overriding this blocked differentiation is a clear strategy for treating high-risk neuroblastoma. A better understanding of neuroblast or neuroblastoma differentiation is essential for developing new therapeutic approaches. It has been proposed that Krueppel-like factor 7 (KLF7) is a neuroblastoma super-enhancer-associated transcription factor gene. Moreover, KLF7 was found to be intensely active in postmitotic neuroblasts of the developing nervous system during embryogenesis. However, the role of KLF7 in the differentiation of neuroblast or neuroblastoma is unknown. Here, we find a strong association between high KLF7 expression and favorable clinical outcomes in neuroblastoma. KLF7 induces differentiation of neuroblastoma cells independently of the retinoic acid (RA) pathway and acts cooperatively with RA to induce neuroblastoma differentiation. KLF7 alters the GTPase activity and multiple differentiation-related genes by binding directly to the promoters of neuroblast differentiation-associated protein (AHNAK and AHNAK2) and glycerophosphodiester phosphodiesterase domain-containing protein 5 (GDPD5) and regulating their expression. Furthermore, we also observe that silencing KLF7 in neuroblastoma cells promotes the adrenergic-to-mesenchymal transition accompanied by changes in enhancer-mediated gene expression. Our results reveal that KLF7 is an inducer of neuroblast or neuroblastoma differentiation with prognostic significance and potential therapeutic value.
Collapse
Affiliation(s)
- Shupei Qiao
- Heilongjiang Province Key Laboratory of Child Development and Genetic Research, Harbin Medical University, China
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, China
| | - Ying Jia
- Heilongjiang Province Key Laboratory of Child Development and Genetic Research, Harbin Medical University, China
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, China
| | - Li Xie
- Heilongjiang Province Key Laboratory of Child Development and Genetic Research, Harbin Medical University, China
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, China
| | - Wenwen Jing
- Heilongjiang Province Key Laboratory of Child Development and Genetic Research, Harbin Medical University, China
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, China
| | - Yang Xia
- Heilongjiang Province Key Laboratory of Child Development and Genetic Research, Harbin Medical University, China
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, China
| | - Yue Song
- Heilongjiang Province Key Laboratory of Child Development and Genetic Research, Harbin Medical University, China
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, China
| | - Jiahui Zhang
- Heilongjiang Province Key Laboratory of Child Development and Genetic Research, Harbin Medical University, China
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, China
| | - Tianhua Cao
- Heilongjiang Province Key Laboratory of Child Development and Genetic Research, Harbin Medical University, China
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, China
| | - Huilin Song
- Heilongjiang Province Key Laboratory of Child Development and Genetic Research, Harbin Medical University, China
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, China
| | - Lingdi Meng
- Heilongjiang Province Key Laboratory of Child Development and Genetic Research, Harbin Medical University, China
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, China
| | - Lei Shi
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, China
| | - Xue Zhang
- Heilongjiang Province Key Laboratory of Child Development and Genetic Research, Harbin Medical University, China
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, China
| |
Collapse
|
2
|
Qin X, Chen B. Comprehensive analysis and validation reveal potential MYCN regulatory biomarkers associated with neuroblastoma prognosis. J Biomol Struct Dyn 2023; 41:8902-8917. [PMID: 36300516 DOI: 10.1080/07391102.2022.2138977] [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: 05/30/2022] [Accepted: 10/16/2022] [Indexed: 10/31/2022]
Abstract
Neuroblastoma (NB) is an embryonic malignant tumor that occurs in the sympathetic nervous system. The treatment results of patients in the high-risk group are poor, and relapse and treatment failure can occur even with multiple combination treatments. The proto-oncogene MYCN is a BHLH Transcription Factor used as an independent prognostic factor for NB. The proportion of MYCN amplification in tumor tissues of high-risk patients reaches 40-50%. Hence, exploring new MYCN target genes is a meaningful approach in developing treatment for high-risk NB patients. The microarray datasets were obtained from Gene Expression Omnibus (GEO), and differentially expressed genes (DEGs) were identified. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and miRPathDB were used for enrichment analysis. STRING and Cytoscape were used to construct a protein-protein interaction (PPI) network and for modular analysis. The miRNet and NetworkAnalyst databases were used to predict and construct gene-miRNA and gene-TFs networks. The R2 database was used for expression, correlation, and prognostic analyses. The diagnostic value of the biomarkers was predicted by ROC analysis, and RT-qPCR was used to validate the identified hub genes. Finally, using specific MYCN siRNA and overexpressing plasmids, the correlation between the identified hub genes and MYCN was investigated. Our results showed that FBXO9, HECW2, MIB2, RNF19B, RNF213, TRIM36, and ZBTB16 are novel biomarkers that affect the prognosis of the NB patients. In addition, FBXO9, RNF19B, and TRIM36 were preliminarily confirmed as potential target genes of MYCN. Overall, FBXO9, HECW2, MIB2, RNF19B, RNF213, TRIM36, and ZBTB16 are expected to become novel biomarkers for the treatment of high-risk NB patients.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Xiuni Qin
- Guangzhou Concord Cancer Center, Guangzhou, China
| | - Bo Chen
- Medical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
3
|
Massudi H, Luo JS, Holien JK, Gadde S, Krishan S, Herath M, Koach J, Stevenson BW, Gorman MA, Venkat P, Mayoh C, Luo XQ, Parker MW, Cheung BB, Marshall GM. Inhibitors of the Oncogenic PA2G4-MYCN Protein-Protein Interface. Cancers (Basel) 2023; 15:cancers15061822. [PMID: 36980710 PMCID: PMC10046377 DOI: 10.3390/cancers15061822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/01/2023] [Accepted: 03/14/2023] [Indexed: 03/22/2023] Open
Abstract
MYCN is a major oncogenic driver for neuroblastoma tumorigenesis, yet there are no direct MYCN inhibitors. We have previously identified PA2G4 as a direct protein-binding partner of MYCN and drive neuroblastoma tumorigenesis. A small molecule known to bind PA2G4, WS6, significantly decreased tumorigenicity in TH-MYCN neuroblastoma mice, along with the inhibition of PA2G4 and MYCN interactions. Here, we identified a number of novel WS6 analogues, with 80% structural similarity, and used surface plasmon resonance assays to determine their binding affinity. Analogues #5333 and #5338 showed direct binding towards human recombinant PA2G4. Importantly, #5333 and #5338 demonstrated a 70-fold lower toxicity for normal human myofibroblasts compared to WS6. Structure-activity relationship analysis showed that a 2,3 dimethylphenol was the most suitable substituent at the R1 position. Replacing the trifluoromethyl group on the phenyl ring at the R2 position, with a bromine or hydrogen atom, increased the difference between efficacy against neuroblastoma cells and normal myofibroblast toxicity. The WS6 analogues inhibited neuroblastoma cell phenotype in vitro, in part through effects on apoptosis, while their anti-cancer effects required both PA2G4 and MYCN expression. Collectively, chemical inhibition of PA2G4-MYCN binding by WS6 analogues represents a first-in-class drug discovery which may have implications for other MYCN-driven cancers.
Collapse
Affiliation(s)
- Hassina Massudi
- Children’s Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2750, Australia
| | - Jie-Si Luo
- Children’s Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2750, Australia
- Department of Paediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510060, China
| | - Jessica K. Holien
- School of Science, STEM College, RMIT University, Melbourne, VIC 3000, Australia
- ACRF Rational Drug Discovery Centre, St. Vincent’s Institute of Medical Research, Fitzroy, VIC 3065, Australia
| | - Satyanarayana Gadde
- Children’s Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2750, Australia
| | - Sukriti Krishan
- Children’s Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2750, Australia
| | - Mika Herath
- Children’s Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2750, Australia
| | - Jessica Koach
- Children’s Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2750, Australia
| | - Brendan W. Stevenson
- ACRF Rational Drug Discovery Centre, St. Vincent’s Institute of Medical Research, Fitzroy, VIC 3065, Australia
| | - Michael A. Gorman
- ACRF Rational Drug Discovery Centre, St. Vincent’s Institute of Medical Research, Fitzroy, VIC 3065, Australia
- ACRF Facility for Innovative Cancer Drug Discovery, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Pooja Venkat
- Children’s Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2750, Australia
| | - Chelsea Mayoh
- Children’s Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2750, Australia
- School of Women’s and Children’s Health, UNSW Sydney, Sydney, NSW 2750, Australia
| | - Xue-Qun Luo
- Department of Paediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510060, China
| | - Michael W. Parker
- ACRF Rational Drug Discovery Centre, St. Vincent’s Institute of Medical Research, Fitzroy, VIC 3065, Australia
- ACRF Facility for Innovative Cancer Drug Discovery, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Belamy B. Cheung
- Children’s Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2750, Australia
- School of Women’s and Children’s Health, UNSW Sydney, Sydney, NSW 2750, Australia
- Correspondence: (B.B.C.); (G.M.M.); Tel.: +61-(02)-9385-2450 (B.B.C.); +61-(02)-9382-1721 (G.M.M.); Fax: +61-(02)-9662-6584 (B.B.C.); +61-(02)-9382-1789 (G.M.M.)
| | - Glenn M. Marshall
- Children’s Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2750, Australia
- Kids Cancer Centre, Sydney Children’s Hospital, Randwick, NSW 2031, Australia
- Correspondence: (B.B.C.); (G.M.M.); Tel.: +61-(02)-9385-2450 (B.B.C.); +61-(02)-9382-1721 (G.M.M.); Fax: +61-(02)-9662-6584 (B.B.C.); +61-(02)-9382-1789 (G.M.M.)
| |
Collapse
|
4
|
Hu X, Liu R, Hou J, Peng W, Wan S, Xu M, Li Y, Zhang G, Zhai X, Liang P, Cui H. SMARCE1 promotes neuroblastoma tumorigenesis through assisting MYCN-mediated transcriptional activation. Oncogene 2022; 41:4295-4306. [PMID: 35978151 DOI: 10.1038/s41388-022-02428-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 07/21/2022] [Accepted: 07/26/2022] [Indexed: 02/07/2023]
Abstract
SMARCE1 gene, encoding a core subunit of SWI/SNF chromatin remodeling complex, is situated on chromosome 17q21-ter region that is frequently gained in neuroblastoma. However, its role in the tumorigenesis remains unknown. Here, we showed that high expression of SMARCE1 was associated with poor prognosis of patients with neuroblastoma, especially those with MYCN amplification. Knockdown of SMARCE1 reduced proliferation, colony formation, and tumorigenicity of neuroblastoma cells. Mechanistically, SMARCE1 directly interacted with MYCN, which was necessary for MYCN-mediated transcriptional activation of downstream target genes including PLK1, ODC1, and E2F2. Overexpression of PLK1, ODC1 or E2F2 significantly reversed the inhibiting effect of SMARCE1 knockdown on the proliferation, colony formation, and tumorigenicity of MYCN-amplified neuroblastoma cells. Moreover, we revealed that MYCN directly regulated SMARCE1 transcription through binding to a non-canonical E-box of SMARCE1 promoter, thus enhancing SMARCE1-MYCN cooperativity. These findings establish SMARCE1 is a critical oncogenic factor in neuroblastoma and provide a new potential target for treatment of neuroblastoma with 17q21-ter gain and MYCN amplification.
Collapse
Affiliation(s)
- Xiaosong Hu
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400716, China.,Cancer Center, Medical Research Institute, Southwest University, Chongqing, 400716, China
| | - Ruochen Liu
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400716, China.,Cancer Center, Medical Research Institute, Southwest University, Chongqing, 400716, China
| | - Jianbing Hou
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400716, China.,Cancer Center, Medical Research Institute, Southwest University, Chongqing, 400716, China
| | - Wen Peng
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400716, China.,Cancer Center, Medical Research Institute, Southwest University, Chongqing, 400716, China
| | - Sicheng Wan
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400716, China.,Cancer Center, Medical Research Institute, Southwest University, Chongqing, 400716, China
| | - Minghao Xu
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400716, China.,Cancer Center, Medical Research Institute, Southwest University, Chongqing, 400716, China
| | - Yongsen Li
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400716, China.,Cancer Center, Medical Research Institute, Southwest University, Chongqing, 400716, China
| | - Guanghui Zhang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400716, China.,Cancer Center, Medical Research Institute, Southwest University, Chongqing, 400716, China
| | - Xuan Zhai
- Department of Neurosurgery, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.,Chongqing Key Laboratory of Pediatrics, Chongqing, 400010, China
| | - Ping Liang
- Department of Neurosurgery, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China. .,Chongqing Key Laboratory of Pediatrics, Chongqing, 400010, China.
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400716, China. .,Cancer Center, Medical Research Institute, Southwest University, Chongqing, 400716, China.
| |
Collapse
|
5
|
Catecholamine excretion profiles identify clinical subgroups of neuroblastoma patients. Eur J Cancer 2019; 111:21-29. [DOI: 10.1016/j.ejca.2019.01.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 01/09/2019] [Accepted: 01/15/2019] [Indexed: 01/10/2023]
|
6
|
Zhang Q, Zhang Q, Jiang X, Ye Y, Liao H, Zhu F, Yan J, Luo L, Tian L, Jiang C, Chen Y, Liang X, Sun Y. Collaborative ISL1/GATA3 interaction in controlling neuroblastoma oncogenic pathways overlapping with but distinct from MYCN. Theranostics 2019; 9:986-1000. [PMID: 30867811 PMCID: PMC6401405 DOI: 10.7150/thno.30199] [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: 09/25/2018] [Accepted: 01/14/2019] [Indexed: 12/12/2022] Open
Abstract
Background: Transcription factor ISL1 plays a critical role in sympathetic neurogenesis. Expression of ISL1 has been associated with neuroblastoma, a pediatric tumor derived from sympatho-adrenal progenitors, however the role of ISL1 in neuroblastoma remains unexplored. Method: Here, we knocked down ISL1 (KD) in SH-SY5Y neuroblastoma cells and performed RNA-seq and ISL1 ChIP-seq analyses. Results: Analyses of these data revealed that ISL1 acts upstream of multiple oncogenic genes and pathways essential for neuroblastoma proliferation and differentiation, including LMO1 and LIN28B. ISL1 promotes expression of a number of cell cycle associated genes, but represses differentiation associated genes including RA receptors and the downstream target genes EPAS1 and CDKN1A. Consequently, Knockdown of ISL1 inhibits neuroblastoma cell proliferation and migration in vitro and impedes tumor growth in vivo, and enhances neuronal differentiation by RA treatment. Furthermore, genome-wide mapping revealed a substantial co-occupancy of binding regions by ISL1 and GATA3, and ISL1 physically interacts with GATA3, and together they synergistically regulate the aforementioned oncogenic pathways. In addition, analyses of the roles of ISL1 and MYCN in MYCN-amplified and MYCN non-amplified neuroblastoma cells revealed an epistatic relationship between ISL1 and MYCN. ISL1 and MYCN function in parallel to regulate common yet distinct oncogenic pathways in neuroblastoma. Conclusion: Our study has demonstrated that ISL1 plays an essential role in neuroblastoma regulatory networks and may serve as a potential therapeutic target in neuroblastoma.
Collapse
|
7
|
TBX2 is a neuroblastoma core regulatory circuitry component enhancing MYCN/FOXM1 reactivation of DREAM targets. Nat Commun 2018; 9:4866. [PMID: 30451831 PMCID: PMC6242972 DOI: 10.1038/s41467-018-06699-9] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 09/13/2018] [Indexed: 12/13/2022] Open
Abstract
Chromosome 17q gains are almost invariably present in high-risk neuroblastoma cases. Here, we perform an integrative epigenomics search for dosage-sensitive transcription factors on 17q marked by H3K27ac defined super-enhancers and identify TBX2 as top candidate gene. We show that TBX2 is a constituent of the recently established core regulatory circuitry in neuroblastoma with features of a cell identity transcription factor, driving proliferation through activation of p21-DREAM repressed FOXM1 target genes. Combined MYCN/TBX2 knockdown enforces cell growth arrest suggesting that TBX2 enhances MYCN sustained activation of FOXM1 targets. Targeting transcriptional addiction by combined CDK7 and BET bromodomain inhibition shows synergistic effects on cell viability with strong repressive effects on CRC gene expression and p53 pathway response as well as several genes implicated in transcriptional regulation. In conclusion, we provide insight into the role of the TBX2 CRC gene in transcriptional dependency of neuroblastoma cells warranting clinical trials using BET and CDK7 inhibitors. In high-risk neuroblastoma cases, gains in chromosome 17q are common. Here, the authors investigate the epigenomics and transcriptomics of neuroblastoma, identifying TBX2 as a core regulatory circuitry component enhancing the reactivation of DREAM targets by MYCN/FOXM1.
Collapse
|
8
|
Zhang Q, Huang R, Ye Y, Guo X, Lu J, Zhu F, Gong X, Zhang Q, Yan J, Luo L, Zhuang S, Chen Y, Zhao X, Evans SM, Jiang C, Liang X, Sun Y. Temporal requirements for ISL1 in sympathetic neuron proliferation, differentiation, and diversification. Cell Death Dis 2018; 9:247. [PMID: 29445148 PMCID: PMC5833373 DOI: 10.1038/s41419-018-0283-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 12/14/2017] [Accepted: 12/22/2017] [Indexed: 12/22/2022]
Abstract
Malformations of the sympathetic nervous system have been associated with cardiovascular instability, gastrointestinal dysfunction, and neuroblastoma. A better understanding of the factors regulating sympathetic nervous system development is critical to the development of potential therapies. Here, we have uncovered a temporal requirement for the LIM homeodomain transcription factor ISL1 during sympathetic nervous system development by the analysis of two mutant mouse lines: an Isl1 hypomorphic line and mice with Isl1 ablated in neural crest lineages. During early development, ISL1 is required for sympathetic neuronal fate determination, differentiation, and repression of glial differentiation, although it is dispensable for initial noradrenergic differentiation. ISL1 also plays an essential role in sympathetic neuron proliferation by controlling cell cycle gene expression. During later development, ISL1 is required for axon growth and sympathetic neuron diversification by maintaining noradrenergic differentiation, but repressing cholinergic differentiation. RNA-seq analyses of sympathetic ganglia from Isl1 mutant and control embryos, together with ISL1 ChIP-seq analysis on sympathetic ganglia, demonstrated that ISL1 regulates directly or indirectly several distinct signaling pathways that orchestrate sympathetic neurogenesis. A number of genes implicated in neuroblastoma pathogenesis are direct downstream targets of ISL1. Our study revealed a temporal requirement for ISL1 in multiple aspects of sympathetic neuron development, and suggested Isl1 as a candidate gene for neuroblastoma.
Collapse
Affiliation(s)
- Qingquan Zhang
- Key Laboratory of Arrhythmia, Ministry of Education, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ru Huang
- Key Laboratory of Arrhythmia, Ministry of Education, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Youqiong Ye
- School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xiaoxia Guo
- Key Laboratory of Arrhythmia, Ministry of Education, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jun Lu
- Key Laboratory of Systems Biomedicine, Ministry of Education, Shanghai Centre for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fugui Zhu
- School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xiaohui Gong
- Key Laboratory of Arrhythmia, Ministry of Education, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qitong Zhang
- Key Laboratory of Arrhythmia, Ministry of Education, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jie Yan
- Key Laboratory of Arrhythmia, Ministry of Education, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lina Luo
- Key Laboratory of Arrhythmia, Ministry of Education, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shaowei Zhuang
- Seventh People's Hospital of Shanghai University of TCM, Shanghai, China
| | - Yihan Chen
- Key Laboratory of Arrhythmia, Ministry of Education, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaodong Zhao
- Key Laboratory of Systems Biomedicine, Ministry of Education, Shanghai Centre for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Sylvia M Evans
- Department of Medicine, Department of Pharmacology, Skaggs School of Pharmacy, University of California San Diego, California, USA
| | - Cizhong Jiang
- School of Life Sciences and Technology, Tongji University, Shanghai, China.
| | - Xingqun Liang
- Key Laboratory of Arrhythmia, Ministry of Education, East Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Yunfu Sun
- Key Laboratory of Arrhythmia, Ministry of Education, East Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
9
|
Groß A, Schulz C, Kolb J, Koster J, Wehner S, Czaplinski S, Khilan A, Rohrer H, Harter PN, Klingebiel T, Langer JD, Geerts D, Schulte D. Tumorigenic and Antiproliferative Properties of the TALE-Transcription Factors MEIS2D and MEIS2A in Neuroblastoma. Cancer Res 2018; 78:1935-1947. [DOI: 10.1158/0008-5472.can-17-1860] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 11/15/2017] [Accepted: 01/24/2018] [Indexed: 11/16/2022]
|
10
|
Warren M, Matsuno R, Tran H, Shimada H. Utility of Phox2b immunohistochemical stain in neural crest tumours and non-neural crest tumours in paediatric patients. Histopathology 2017; 72:685-696. [DOI: 10.1111/his.13412] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 10/04/2017] [Indexed: 11/30/2022]
Affiliation(s)
- Mikako Warren
- Department of Pathology and Laboratory Medicine; Children's Hospital Los Angeles; Keck School of Medicine; University of Southern California; Los Angeles CA USA
| | - Ryosuke Matsuno
- Department of Pathology and Laboratory Medicine; Children's Hospital Los Angeles; Keck School of Medicine; University of Southern California; Los Angeles CA USA
| | - Henry Tran
- Department of Pathology and Laboratory Medicine; Children's Hospital Los Angeles; Keck School of Medicine; University of Southern California; Los Angeles CA USA
| | - Hiroyuki Shimada
- Department of Pathology and Laboratory Medicine; Children's Hospital Los Angeles; Keck School of Medicine; University of Southern California; Los Angeles CA USA
| |
Collapse
|
11
|
Risk Stratification of Pediatric Patients With Neuroblastoma Using Volumetric Parameters of 18F-FDG and 18F-DOPA PET/CT. Clin Nucl Med 2017; 42:e142-e148. [PMID: 28072621 DOI: 10.1097/rlu.0000000000001529] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
PURPOSE This study determined the prognostic value of volumetric parameters derived from pretreatment F-FDG and F-DOPA PET/CT of neuroblastoma and their correlation with clinical and histopathologic features. PATIENTS AND METHODS A total of 25 children with neuroblastoma underwent pretreatment F-FDG and F-DOPA PET/CT within 4 weeks. The SUVmax of primary tumors on F-FDG and F-DOPA PET were recorded as SUVFDG and SUVDOPA, respectively. For volumetric parameters of primary tumors, 40% of SUVmax was used to generate volume of interest. If the 40% of SUVmax was below 2.5, an SUV threshold of 2.5 was used instead. Metabolic tumor volume (MTV), total lesion glycolysis (TLG), dopaminergic tumor volume (DTV), and total lesion F-DOPA activity (TLDA) were recorded as F-FDG and F-DOPA volumetric parameters. All indices were compared between groups distinguished by survival status and clinical features, including bone marrow involvement, lymph node metastasis, amplification of the MYCN oncogene, invasive features on anatomic images, and risk categories. The Kaplan-Meier method and log-rank test were used to compare the survival curves between groups. RESULTS The median follow-up period was 28.2 months. Nonsurvivors (20%) tended to have lower SUVDOPA, DTV, and TLDA (P ≤ 0.05), and higher SUVFDG, MTV, and TLG (all P < 0.05). Lower F-DOPA uptake is associated with bone marrow and lymph node metastases (all P < 0.05). Higher F-FDG uptake is associated with MYCN amplification (all P < 0.05) and anatomic invasive features of tumors such as vascular encasement or adjacent organ invasion (TLG, P = 0.05). Only volumetric indices (DTV, TLDA, MTV, and TLG) significantly differed among risk groups (all P < 0.05). CONCLUSIONS Pretherapeutic F-DOPA and F-FDG PET provided complementary information, and both can be served for risk stratification. Volumetric indices of F-DOPA and F-FDG PET correlate more highly with risk grouping.
Collapse
|
12
|
Hernandez-Martinez JM, Forrest CM, Darlington LG, Smith RA, Stone TW. Quinolinic acid induces neuritogenesis in SH-SY5Y neuroblastoma cells independently of NMDA receptor activation. Eur J Neurosci 2017; 45:700-711. [PMID: 27973747 DOI: 10.1111/ejn.13499] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 11/29/2016] [Accepted: 11/29/2016] [Indexed: 12/25/2022]
Abstract
Glutamate and nicotinamide adenine dinucleotide (NAD+ ) have been implicated in neuronal development and several types of cancer. The kynurenine pathway of tryptophan metabolism includes quinolinic acid (QA) which is both a selective agonist at N-methyl-D-aspartate (NMDA) receptors and also a precursor for the formation of NAD+ . The effect of QA on cell survival and differentiation has therefore been examined on SH-SY5Y human neuroblastoma cells. Retinoic acid (RA, 10 μm) induced differentiation of SH-SY5Y cells into a neuronal phenotype showing neurite growth. QA (50-150 nm) also caused a concentration-dependent increase in the neurite/soma ratio, indicating differentiation. Both RA and QA increased expression of the neuronal marker β3-tubulin in whole-cell homogenates and in the neuritic fraction assessed using a neurite outgrowth assay. Expression of the neuronal proliferation marker doublecortin revealed that, unlike RA, QA did not decrease the number of mitotic cells. QA-induced neuritogenesis coincided with an increase in the generation of reactive oxygen species. Neuritogenesis was prevented by diphenylene-iodonium (an inhibitor of NADPH oxidase) and superoxide dismutase, supporting the involvement of reactive oxygen species. NMDA itself did not promote neuritogenesis and the NMDA antagonist dizocilpine (MK-801) did not prevent quinolinate-induced neuritogenesis, indicating that the effects of QA were independent of NMDA receptors. Nicotinamide caused a significant increase in the neurite/soma ratio and the expression of β3-tubulin in the neuritic fraction. Taken together, these results suggest that QA induces neuritogenesis by promoting oxidizing conditions and affecting the availability of NAD+ , independently of NMDA receptors.
Collapse
Affiliation(s)
- Juan-Manuel Hernandez-Martinez
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, West Medical Building, Glasgow, G12 8QQ, UK
| | - Caroline M Forrest
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, West Medical Building, Glasgow, G12 8QQ, UK
| | | | - Robert A Smith
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, West Medical Building, Glasgow, G12 8QQ, UK
| | - Trevor W Stone
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, West Medical Building, Glasgow, G12 8QQ, UK
| |
Collapse
|
13
|
Galli S, Naranjo A, Van Ryn C, Tilan JU, Trinh E, Yang C, Tsuei J, Hong SH, Wang H, Izycka-Swieszewska E, Lee YC, Rodriguez OC, Albanese C, Kitlinska J. Neuropeptide Y as a Biomarker and Therapeutic Target for Neuroblastoma. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:3040-3053. [PMID: 27743558 DOI: 10.1016/j.ajpath.2016.07.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 07/06/2016] [Accepted: 07/11/2016] [Indexed: 12/17/2022]
Abstract
Neuroblastoma (NB) is a pediatric malignant neoplasm of sympathoadrenal origin. Challenges in its management include stratification of this heterogeneous disease and a lack of both adequate treatments for high-risk patients and noninvasive biomarkers of disease progression. Our previous studies have identified neuropeptide Y (NPY), a sympathetic neurotransmitter expressed in NB, as a potential therapeutic target for these tumors by virtue of its Y5 receptor (Y5R)-mediated chemoresistance and Y2 receptor (Y2R)-mediated proliferative and angiogenic activities. The goal of this study was to determine the clinical relevance and utility of these findings. Expression of NPY and its receptors was evaluated in corresponding samples of tumor RNA, tissues, and sera from 87 patients with neuroblastic tumors and in tumor tissues from the TH-MYCN NB mouse model. Elevated serum NPY levels correlated with an adverse clinical presentation, poor survival, metastasis, and relapse, whereas strong Y5R immunoreactivity was a marker of angioinvasive tumor cells. In NB tissues from TH-MYCN mice, high immunoreactivity of both NPY and Y5R marked angioinvasive NB cells. Y2R was uniformly expressed in undifferentiated tumor cells, which supports its previously reported role in NB cell proliferation. Our findings validate NPY as a therapeutic target for advanced NB and implicate the NPY/Y5R axis in disease dissemination. The correlation between elevated systemic NPY and NB progression identifies serum NPY as a novel NB biomarker.
Collapse
Affiliation(s)
- Susana Galli
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, District of Columbia
| | - Arlene Naranjo
- Department of Biostatistics, Children's Oncology Group Statistics & Data Center, University of Florida, Gainesville, Florida
| | - Collin Van Ryn
- Department of Biostatistics, Children's Oncology Group Statistics & Data Center, University of Florida, Gainesville, Florida
| | - Jason U Tilan
- Department of Nursing, School of Nursing and Health Studies, Georgetown University, Washington, District of Columbia; Department of Human Science, School of Nursing and Health Studies, Georgetown University, Washington, District of Columbia
| | - Emily Trinh
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, District of Columbia
| | - Chao Yang
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, District of Columbia
| | - Jessica Tsuei
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, District of Columbia
| | - Sung-Hyeok Hong
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia
| | - Hongkun Wang
- Department of Biostatistics and Bioinformatics, Georgetown University Medical Center, Washington, District of Columbia
| | - Ewa Izycka-Swieszewska
- Department of Pathology and Neuropathology, Medical University of Gdańsk, Gdańsk, Poland
| | - Yi-Chien Lee
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia
| | - Olga C Rodriguez
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia
| | - Chris Albanese
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia; Department of Pathology, Georgetown University Medical Center, Washington, District of Columbia
| | - Joanna Kitlinska
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, District of Columbia.
| |
Collapse
|
14
|
Ikram F, Ackermann S, Kahlert Y, Volland R, Roels F, Engesser A, Hertwig F, Kocak H, Hero B, Dreidax D, Henrich KO, Berthold F, Nürnberg P, Westermann F, Fischer M. Transcription factor activating protein 2 beta (TFAP2B) mediates noradrenergic neuronal differentiation in neuroblastoma. Mol Oncol 2015; 10:344-59. [PMID: 26598443 DOI: 10.1016/j.molonc.2015.10.020] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 10/05/2015] [Accepted: 10/23/2015] [Indexed: 10/22/2022] Open
Abstract
Neuroblastoma is an embryonal pediatric tumor that originates from the developing sympathetic nervous system and shows a broad range of clinical behavior, ranging from fatal progression to differentiation into benign ganglioneuroma. In experimental neuroblastoma systems, retinoic acid (RA) effectively induces neuronal differentiation, and RA treatment has been therefore integrated in current therapies. However, the molecular mechanisms underlying differentiation are still poorly understood. We here investigated the role of transcription factor activating protein 2 beta (TFAP2B), a key factor in sympathetic nervous system development, in neuroblastoma pathogenesis and differentiation. Microarray analyses of primary neuroblastomas (n = 649) demonstrated that low TFAP2B expression was significantly associated with unfavorable prognostic markers as well as adverse patient outcome. We also found that low TFAP2B expression was strongly associated with CpG methylation of the TFAP2B locus in primary neuroblastomas (n = 105) and demethylation with 5-aza-2'-deoxycytidine resulted in induction of TFAP2B expression in vitro, suggesting that TFAP2B is silenced by genomic methylation. Tetracycline inducible re-expression of TFAP2B in IMR-32 and SH-EP neuroblastoma cells significantly impaired proliferation and cell cycle progression. In IMR-32 cells, TFAP2B induced neuronal differentiation, which was accompanied by up-regulation of the catecholamine biosynthesizing enzyme genes DBH and TH, and down-regulation of MYCN and REST, a master repressor of neuronal genes. By contrast, knockdown of TFAP2B by lentiviral transduction of shRNAs abrogated RA-induced neuronal differentiation of SH-SY5Y and SK-N-BE(2)c neuroblastoma cells almost completely. Taken together, our results suggest that TFAP2B is playing a vital role in retaining RA responsiveness and mediating noradrenergic neuronal differentiation in neuroblastoma.
Collapse
Affiliation(s)
- Fakhera Ikram
- Department of Pediatric Oncology and Hematology, University Children's Hospital of Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Germany; Cologne Center for Genomics (CCG), University of Cologne, Germany
| | - Sandra Ackermann
- Department of Pediatric Oncology and Hematology, University Children's Hospital of Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Germany
| | - Yvonne Kahlert
- Department of Pediatric Oncology and Hematology, University Children's Hospital of Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Germany
| | - Ruth Volland
- Department of Pediatric Oncology and Hematology, University Children's Hospital of Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Germany
| | - Frederik Roels
- Department of Pediatric Oncology and Hematology, University Children's Hospital of Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Germany
| | - Anne Engesser
- Department of Pediatric Oncology and Hematology, University Children's Hospital of Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Germany
| | - Falk Hertwig
- Department of Pediatric Oncology and Hematology, University Children's Hospital of Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Germany
| | - Hayriye Kocak
- Department of Pediatric Oncology and Hematology, University Children's Hospital of Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Germany
| | - Barbara Hero
- Department of Pediatric Oncology and Hematology, University Children's Hospital of Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Germany
| | - Daniel Dreidax
- Division Neuroblastoma Genomics (B087), German Cancer Research Center, Heidelberg, Germany
| | - Kai-Oliver Henrich
- Division Neuroblastoma Genomics (B087), German Cancer Research Center, Heidelberg, Germany
| | - Frank Berthold
- Department of Pediatric Oncology and Hematology, University Children's Hospital of Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Germany
| | - Peter Nürnberg
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Germany; Cologne Center for Genomics (CCG), University of Cologne, Germany
| | - Frank Westermann
- Division Neuroblastoma Genomics (B087), German Cancer Research Center, Heidelberg, Germany
| | - Matthias Fischer
- Department of Pediatric Oncology and Hematology, University Children's Hospital of Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Germany; Max Planck Institute for Metabolism Research, Cologne, Germany.
| |
Collapse
|
15
|
Lambertz I, Kumps C, Claeys S, Lindner S, Beckers A, Janssens E, Carter DR, Cazes A, Cheung BB, De Mariano M, De Bondt A, De Brouwer S, Delattre O, Gibbons J, Janoueix-Lerosey I, Laureys G, Liang C, Marchall GM, Porcu M, Takita J, Trujillo DC, Van Den Wyngaert I, Van Roy N, Van Goethem A, Van Maerken T, Zabrocki P, Cools J, Schulte JH, Vialard J, Speleman F, De Preter K. Upregulation of MAPK Negative Feedback Regulators and RET in Mutant ALK Neuroblastoma: Implications for Targeted Treatment. Clin Cancer Res 2015; 21:3327-39. [PMID: 25805801 DOI: 10.1158/1078-0432.ccr-14-2024] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 03/13/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE Activating ALK mutations are present in almost 10% of primary neuroblastomas and mark patients for treatment with small-molecule ALK inhibitors in clinical trials. However, recent studies have shown that multiple mechanisms drive resistance to these molecular therapies. We anticipated that detailed mapping of the oncogenic ALK-driven signaling in neuroblastoma can aid to identify potential fragile nodes as additional targets for combination therapies. EXPERIMENTAL DESIGN To achieve this goal, transcriptome profiling was performed in neuroblastoma cell lines with the ALK(F1174L) or ALK(R1275Q) hotspot mutations, ALK amplification, or wild-type ALK following pharmacologic inhibition of ALK using four different compounds. Next, we performed cross-species genomic analyses to identify commonly transcriptionally perturbed genes in MYCN/ALK(F1174L) double transgenic versus MYCN transgenic mouse tumors as compared with the mutant ALK-driven transcriptome in human neuroblastomas. RESULTS A 77-gene ALK signature was established and successfully validated in primary neuroblastoma samples, in a neuroblastoma cell line with ALK(F1174L) and ALK(R1275Q) regulable overexpression constructs and in other ALKomas. In addition to the previously established PI3K/AKT/mTOR, MAPK/ERK, and MYC/MYCN signaling branches, we identified that mutant ALK drives a strong upregulation of MAPK negative feedback regulators and upregulates RET and RET-driven sympathetic neuronal markers of the cholinergic lineage. CONCLUSIONS We provide important novel insights into the transcriptional consequences and the complexity of mutant ALK signaling in this aggressive pediatric tumor. The negative feedback loop of MAPK pathway inhibitors may affect novel ALK inhibition therapies, whereas mutant ALK induced RET signaling can offer novel opportunities for testing ALK-RET oriented molecular combination therapies.
Collapse
Affiliation(s)
- Irina Lambertz
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Candy Kumps
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Shana Claeys
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Sven Lindner
- Department of Pediatric Oncology and Haematology, University Children's Hospital Essen, Germany
| | - Anneleen Beckers
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Els Janssens
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Daniel R Carter
- Kids Cancer Center, Sydneys Children's Hospital, Children's Cancer Institute, Lowy Cancer Centre, University of New South Wales, Sydney, Australia
| | - Alex Cazes
- Unité Inserm U830, Centre de Recherche, Institut Curie, Paris, France
| | - Belamy B Cheung
- Kids Cancer Center, Sydneys Children's Hospital, Children's Cancer Institute, Lowy Cancer Centre, University of New South Wales, Sydney, Australia
| | - Marilena De Mariano
- Biotherapy Unit, IRCCS AOU San Martino-IST, Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - An De Bondt
- Oncology Discovery Research and Early Development, Johnson & Johnson, Beerse, Belgium
| | - Sara De Brouwer
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Olivier Delattre
- Unité Inserm U830, Centre de Recherche, Institut Curie, Paris, France
| | - Jay Gibbons
- VP Oncology, Xcovery LLC, West Palm Beach, Florida
| | | | - Geneviève Laureys
- Department of Pediatric Oncology and Haematology, Ghent University Hospital, Ghent, Belgium
| | - Chris Liang
- VP Oncology, Xcovery LLC, West Palm Beach, Florida
| | - Glenn M Marchall
- Kids Cancer Center, Sydneys Children's Hospital, Children's Cancer Institute, Lowy Cancer Centre, University of New South Wales, Sydney, Australia
| | - Michael Porcu
- Center for Human Genetics, K.U. Leuven-VIB, Leuven, Belgium
| | - Junko Takita
- Department of Pediatrics, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | | | - Ilse Van Den Wyngaert
- Oncology Discovery Research and Early Development, Johnson & Johnson, Beerse, Belgium
| | - Nadine Van Roy
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Alan Van Goethem
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Tom Van Maerken
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Piotr Zabrocki
- Center for Human Genetics, K.U. Leuven-VIB, Leuven, Belgium
| | - Jan Cools
- Center for Human Genetics, K.U. Leuven-VIB, Leuven, Belgium
| | - Johannes H Schulte
- Department of Pediatric Oncology and Haematology, University Children's Hospital Essen, Germany. German Cancer Consortium (DKTK), Germany. Translational Neuro-Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany. German Cancer Research Center (DKFZ), Heidelberg, Germany. Centre for Medical Biotechnology, University Duisburg-Essen, Essen, Germany
| | - Jorge Vialard
- Oncology Discovery Biology, Janssen Research & Development, a division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Frank Speleman
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium.
| | - Katleen De Preter
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
16
|
Yarmishyn AA, Batagov AO, Tan JZ, Sundaram GM, Sampath P, Kuznetsov VA, Kurochkin IV. HOXD-AS1 is a novel lncRNA encoded in HOXD cluster and a marker of neuroblastoma progression revealed via integrative analysis of noncoding transcriptome. BMC Genomics 2014; 15 Suppl 9:S7. [PMID: 25522241 PMCID: PMC4290621 DOI: 10.1186/1471-2164-15-s9-s7] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background Long noncoding RNAs (lncRNAs) constitute a major, but poorly characterized part of human transcriptome. Recent evidence indicates that many lncRNAs are involved in cancer and can be used as predictive and prognostic biomarkers. Significant fraction of lncRNAs is represented on widely used microarray platforms, however they have usually been ignored in cancer studies. Results We developed a computational pipeline to annotate lncRNAs on popular Affymetrix U133 microarrays, creating a resource allowing measurement of expression of 1581 lncRNAs. This resource can be utilized to interrogate existing microarray datasets for various lncRNA studies. We found that these lncRNAs fall into three distinct classes according to their statistical distribution by length. Remarkably, these three classes of lncRNAs were co-localized with protein coding genes exhibiting distinct gene ontology groups. This annotation was applied to microarray analysis which identified a 159 lncRNA signature that discriminates between localized and metastatic stages of neuroblastoma. Analysis of an independent patient cohort revealed that this signature differentiates also relapsing from non-relapsing primary tumors. This is the first example of the signature developed via the analysis of expression of lncRNAs solely. One of these lncRNAs, termed HOXD-AS1, is encoded in HOXD cluster. HOXD-AS1 is evolutionary conserved among hominids and has all bona fide features of a gene. Studying retinoid acid (RA) response of SH-SY5Y cell line, a model of human metastatic neuroblastoma, we found that HOXD-AS1 is a subject to morphogenic regulation, is activated by PI3K/Akt pathway and itself is involved in control of RA-induced cell differentiation. Knock-down experiments revealed that HOXD-AS1 controls expression levels of clinically significant protein-coding genes involved in angiogenesis and inflammation, the hallmarks of metastatic cancer. Conclusions Our findings greatly extend the number of noncoding RNAs functionally implicated in tumor development and patient treatment and highlight their role as potential prognostic biomarkers of neuroblastomas.
Collapse
|
17
|
Hu K, Zhao F, Wang Q. Mechanical characterization of living and dead undifferentiated human adipose-derived stem cells by using atomic force microscopy. Proc Inst Mech Eng H 2013; 227:1319-23. [DOI: 10.1177/0954411913503064] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
In this article, to map the mechanical properties of undifferentiated human adipose-derived stem cells, local mechanical characterization is carried out on the adipose-derived stem cells. In addition, to distinguish the living and dead human adipose-derived stem cells, mechanical characterization is also implemented on both living and dead adipose-derived stem cells. In this study, Young’s modulus of the cell membrane is used for representing the mechanical properties of cells. To obtain Young’s modulus of cell membrane, the force-spectroscopy mode of atomic force microscopy is employed to measure the atomic force microscopy tip indentation depth and force on the cells. Then, Young’s modulus is obtained through fitting these experimental data to the Hertzian contact mechanics model. The global Young’s moduli of living and dead undifferentiated adipose-derived stem cells are about 1.27 and 18.61 kPa, respectively. This displays obvious gap of Young’s modulus between the living and dead undifferentiated adipose-derived stem cells. Finally, comparison of the local Young’s modulus shows deviation of the local Young’s modulus for either living or dead undifferentiated adipose-derived stem cells, and the root-mean-square errors of the global Young’s modulus of living and dead undifferentiated adipose-derived stem cells are about 0.48 and 5.05 kPa, respectively.
Collapse
Affiliation(s)
- Kexiang Hu
- School of Electronics and Information, Hangzhou Dianzi University, Hangzhou, P.R. China
- National Key Laboratory of Micro/Nano Fabrication Technology, Key Laboratory for Thin Film and Microfabrication Technology of Ministry of Education, Research Institute of Micro/Nano Science and Technology, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Feihu Zhao
- National Centre for Biomedical Engineering and Sciences (NCBES) and Department of Mechanical and Biomedical Engineering, National University of Ireland, Galway, Ireland
| | - Qingkang Wang
- National Key Laboratory of Micro/Nano Fabrication Technology, Key Laboratory for Thin Film and Microfabrication Technology of Ministry of Education, Research Institute of Micro/Nano Science and Technology, Shanghai Jiao Tong University, Shanghai, P.R. China
| |
Collapse
|
18
|
Lu MY, Liu YL, Chang HH, Jou ST, Yang YL, Lin KH, Lin DT, Lee YL, Lee H, Wu PY, Luo TY, Shen LH, Huang SF, Liao YF, Hsu WM, Tzen KY. Characterization of Neuroblastic Tumors Using 18F-FDOPA PET. J Nucl Med 2012; 54:42-9. [DOI: 10.2967/jnumed.112.102772] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
19
|
Decock A, Ongenaert M, Hoebeeck J, De Preter K, Van Peer G, Van Criekinge W, Ladenstein R, Schulte JH, Noguera R, Stallings RL, Van Damme A, Laureys G, Vermeulen J, Van Maerken T, Speleman F, Vandesompele J. Genome-wide promoter methylation analysis in neuroblastoma identifies prognostic methylation biomarkers. Genome Biol 2012; 13:R95. [PMID: 23034519 PMCID: PMC3491423 DOI: 10.1186/gb-2012-13-10-r95] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 10/03/2012] [Indexed: 01/06/2023] Open
Abstract
Background Accurate outcome prediction in neuroblastoma, which is necessary to enable the optimal choice of risk-related therapy, remains a challenge. To improve neuroblastoma patient stratification, this study aimed to identify prognostic tumor DNA methylation biomarkers. Results To identify genes silenced by promoter methylation, we first applied two independent genome-wide methylation screening methodologies to eight neuroblastoma cell lines. Specifically, we used re-expression profiling upon 5-aza-2'-deoxycytidine (DAC) treatment and massively parallel sequencing after capturing with a methyl-CpG-binding domain (MBD-seq). Putative methylation markers were selected from DAC-upregulated genes through a literature search and an upfront methylation-specific PCR on 20 primary neuroblastoma tumors, as well as through MBD- seq in combination with publicly available neuroblastoma tumor gene expression data. This yielded 43 candidate biomarkers that were subsequently tested by high-throughput methylation-specific PCR on an independent cohort of 89 primary neuroblastoma tumors that had been selected for risk classification and survival. Based on this analysis, methylation of KRT19, FAS, PRPH, CNR1, QPCT, HIST1H3C, ACSS3 and GRB10 was found to be associated with at least one of the classical risk factors, namely age, stage or MYCN status. Importantly, HIST1H3C and GNAS methylation was associated with overall and/or event-free survival. Conclusions This study combines two genome-wide methylation discovery methodologies and is the most extensive validation study in neuroblastoma performed thus far. We identified several novel prognostic DNA methylation markers and provide a basis for the development of a DNA methylation-based prognostic classifier in neuroblastoma.
Collapse
|
20
|
PHOX2B immunolabeling: a novel tool for the diagnosis of undifferentiated neuroblastomas among childhood small round blue-cell tumors. Am J Surg Pathol 2012; 36:1141-9. [PMID: 22790854 DOI: 10.1097/pas.0b013e31825a6895] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Peripheral neuroblastic tumors are the most commonly occurring extracranial tumors in children. Although a reliable diagnosis is achievable in the majority of cases, diagnosis of a minority of peripheral neuroblastic tumor cases (especially undifferentiated neuroblastoma) poses a challenge compared with that of other pediatric small round blue-cell tumors. A panel of immunohistochemical markers and fusion transcripts is available for the diagnosis of such tumors, but the markers for neuroblastoma lack specificity and sensitivity. As the transcription factor PHOX2B is highly specific for the peripheral autonomic nervous system from which peripheral neuroblastic tumors are derived, we have assessed PHOX2B immunolabeling as a diagnostic tool in pediatric small round blue-cell tumors. We observed PHOX2B expression in all peripheral neuroblastic tumors, paragangliomas, and pheochromocytomas tested but in no other pediatric tumors among the 388 cases studied by expression microarray and the 109 cases studied by immunohistochemical analysis. We then assessed the results of PHOX2B immunohistochemistry in 12 cases of undifferentiated pediatric neoplasms: PHOX2B was expressed in 6/6 undifferentiated neuroblastomas and in no other small round blue-cell tumors. Finally, we showed that PHOX2B immunohistochemical analysis improves the diagnosis of undifferentiated neuroblastoma with high specificity and sensitivity.
Collapse
|
21
|
Vamvaca K, Lansbury PT, Stefanis L. N-terminal deletion does not affect α-synuclein membrane binding, self-association and toxicity in human neuroblastoma cells, unlike yeast. J Neurochem 2011; 119:389-97. [PMID: 21848810 DOI: 10.1111/j.1471-4159.2011.07431.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
α-Synuclein causes Parkinson's disease if mutated or aberrantly produced in neurons. α-Synuclein-lipid interactions are important for the normal function of the protein, but can also contribute to pathogenesis. We previously reported that deletion of the first 10 N-terminal amino acids dramatically reduced lipid binding in vitro, as well as membrane binding and toxicity in yeast. Here we extend this study to human neuroblastoma SHSY-5Y cells, and find that in these cells the first 10 N-terminal residues do not affect α-synuclein membrane binding, self-association and cell viability, contrary to yeast. Differences in lipid composition, membrane fluidity and cytosolic factors between yeast and neuronal cells may account for the distinct binding behavior of the truncated variant in these two systems. Retinoic acid promotes differentiation and α-synuclein oligomer formation in neuroblastoma cells, while addition of a proteasomal inhibitor induces neurite outgrowth and toxicity to certain wild-type and truncated α-synuclein clones. Yeast recapitulate several features of α-synuclein (patho)biology, but its simplicity sets limitations; verification of yeast results in more relevant model systems is, therefore, essential.
Collapse
Affiliation(s)
- Katherina Vamvaca
- Division of Basic Neurosciences, Biomedical Research Foundation of the Academy of Athens, Soranou Efesiou 4, Athens, Greece.
| | | | | |
Collapse
|
22
|
Benko E, Winkelmann A, Meier JC, Persson PB, Scholz H, Fähling M. Phorbol-Ester Mediated Suppression of hASH1 Synthesis: Multiple Ways to Keep the Level Down. Front Mol Neurosci 2011; 4:1. [PMID: 21441980 PMCID: PMC3057490 DOI: 10.3389/fnmol.2011.00001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Accepted: 01/20/2011] [Indexed: 11/13/2022] Open
Abstract
Human achaete-scute homolog-1 (hASH1), encoded by the human ASCL1 gene, belongs to the family of basic helix-loop-helix transcription factors. hASH1 and its mammalian homolog Mash1 are expressed in the central and peripheral nervous system during development, and promote early neuronal differentiation. Furthermore, hASH1 is involved in the specification of neuronal subtype identities. Misexpression of the transcription factor is correlated with a variety of tumors, including lung cancer and neuroendocrine tumors. To gain insights into the molecular mechanisms of hASH1 regulation, we screened for conditions causing changes in hASH1 gene expression rate. We found that treatment of human neuroblastoma-derived Kelly cells with phorbol 12-myristate 13-acetate (PMA) resulted in a fast, strong and long-lasting suppression of hASH1 synthesis. Reporter gene assays with constructs, in which the luciferase activity was controlled either by the ASCL1 promoter or by the hASH1 mRNA untranslated regions (UTRs), revealed a mainly UTR-dependent mechanism. The hASH1 promoter activity was decreased only after 48 h of PMA administration. Our data indicate that different mechanisms acting consecutively at the transcriptional and post-transcriptional level are responsible for hASH1 suppression after PMA treatment. We provide evidence that short term inhibition of hASH1 synthesis is attributed to hASH1 mRNA destabilization, which seems to depend mainly on protein kinase C activity. Under prolonged conditions (48 h), hASH1 suppression is mediated by decreased promoter activity and inhibition of mRNA translation.
Collapse
Affiliation(s)
- Edgar Benko
- Institut für Vegetative Physiologie, Charité - Universitätsmedizin Berlin Berlin, Germany
| | | | | | | | | | | |
Collapse
|
23
|
Henriksen JR, Haug BH, Buechner J, Tømte E, Løkke C, Flaegstad T, Einvik C. Conditional expression of retrovirally delivered anti-MYCN shRNA as an in vitro model system to study neuronal differentiation in MYCN-amplified neuroblastoma. BMC DEVELOPMENTAL BIOLOGY 2011; 11:1. [PMID: 21194500 PMCID: PMC3022612 DOI: 10.1186/1471-213x-11-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2010] [Accepted: 01/03/2011] [Indexed: 11/18/2022]
Abstract
Background Neuroblastoma is a childhood cancer derived from immature cells of the sympathetic nervous system. The disease is clinically heterogeneous, ranging from neuronal differentiated benign ganglioneuromas to aggressive metastatic tumours with poor prognosis. Amplification of the MYCN oncogene is a well established poor prognostic factor found in up to 40% of high risk neuroblastomas. Using neuroblastoma cell lines to study neuronal differentiation in vitro is now well established. Several protocols, including exposure to various agents and growth factors, will differentiate neuroblastoma cell lines into neuron-like cells. These cells are characterized by a neuronal morphology with long extensively branched neurites and expression of several neurospecific markers. Results In this study we use retrovirally delivered inducible short-hairpin RNA (shRNA) modules to knock down MYCN expression in MYCN-amplified (MNA) neuroblastoma cell lines. By addition of the inducer doxycycline, we show that the Kelly and SK-N-BE(2) neuroblastoma cell lines efficiently differentiate into neuron-like cells with an extensive network of neurites. These cells are further characterized by increased expression of the neuronal differentiation markers NFL and GAP43. In addition, we show that induced expression of retrovirally delivered anti-MYCN shRNA inhibits cell proliferation by increasing the fraction of MNA neuroblastoma cells in the G1 phase of the cell cycle and that the clonogenic growth potential of these cells was also dramatically reduced. Conclusion We have developed an efficient MYCN-knockdown in vitro model system to study neuronal differentiation in MNA neuroblastomas.
Collapse
Affiliation(s)
- Jørn R Henriksen
- Department of Pediatrics, University Hospital of North-Norway, 9038 Tromsø, Norway
| | | | | | | | | | | | | |
Collapse
|
24
|
Janoueix-Lerosey I, Schleiermacher G, Delattre O. Molecular pathogenesis of peripheral neuroblastic tumors. Oncogene 2010; 29:1566-79. [PMID: 20101209 DOI: 10.1038/onc.2009.518] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Neuroblastoma (NB) is an embryonal cancer of the sympathetic nervous system observed in early childhood, characterized by a broad spectrum of clinical behaviors, ranging from spontaneous regression to fatal outcome despite aggressive therapies. NB accounts for 8-10% of pediatric cancers and 15% of the deaths attributable to malignant conditions in children. Interestingly, NB may occur in various contexts, being mostly sporadic but also familial or syndromic. This review focuses on recent advances in the identification of the genes and mechanisms implicated in NB pathogenesis. Although the extensive characterization of the genomic aberrations recurrently observed in sporadic NBs provides important insights into the understanding of the clinical heterogeneity of this neoplasm, analysis of familial and syndromic cases also unravels essential clues on the genetic bases of NB. Recently, the ALK gene emerged as an important NB gene, being implicated both in sporadic and familial cases. The identification of gene expression signatures associated with patient's outcome points out the potential of using gene expression profiling to improve clinical management of patients suffering from NB. Finally, based on recent observations integrating genomic analyses, biological data and clinical information, we discuss possible evolution/progression schemes in NB.
Collapse
Affiliation(s)
- I Janoueix-Lerosey
- INSERM U830, Laboratoire de Génétique et Biologie des Cancers, Institut Curie, Paris Cedex 05, France.
| | | | | |
Collapse
|