1
|
Sundaramoorthy S, Colombo DF, Sanalkumar R, Broye L, Balmas Bourloud K, Boulay G, Cironi L, Stamenkovic I, Renella R, Kuttler F, Turcatti G, Rivera MN, Mühlethaler-Mottet A, Bardet AF, Riggi N. Preclinical spheroid models identify BMX as a therapeutic target for metastatic MYCN nonamplified neuroblastoma. JCI Insight 2024; 9:e169647. [PMID: 39133652 PMCID: PMC11383371 DOI: 10.1172/jci.insight.169647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 06/10/2024] [Indexed: 09/11/2024] Open
Abstract
The development of targeted therapies offers new hope for patients affected by incurable cancer. However, multiple challenges persist, notably in controlling tumor cell plasticity in patients with refractory and metastatic illness. Neuroblastoma (NB) is an aggressive pediatric malignancy originating from defective differentiation of neural crest-derived progenitors with oncogenic activity due to genetic and epigenetic alterations and remains a clinical challenge for high-risk patients. To identify critical genes driving NB aggressiveness, we performed combined chromatin and transcriptome analyses on matched patient-derived xenografts (PDXs), spheroids, and differentiated adherent cultures derived from metastatic MYCN nonamplified tumors. Bone marrow kinase on chromosome X (BMX) was identified among the most differentially regulated genes in PDXs and spheroids versus adherent models. BMX expression correlated with high tumor stage and poor patient survival and was crucial to the maintenance of the self-renewal and tumorigenic potential of NB spheroids. Moreover, BMX expression positively correlated with the mesenchymal NB cell phenotype, previously associated with increased chemoresistance. Finally, BMX inhibitors readily reversed this cellular state, increased the sensitivity of NB spheroids toward chemotherapy, and partially reduced tumor growth in a preclinical NB model. Altogether, our study identifies BMX as a promising innovative therapeutic target for patients with high-risk MYCN nonamplified NB.
Collapse
Affiliation(s)
| | | | - Rajendran Sanalkumar
- Experimental Pathology Service, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Liliane Broye
- Experimental Pathology Service, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Katia Balmas Bourloud
- Department Woman-Mother-Child, Division of Pediatrics, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Gaylor Boulay
- Department of Pathology and Cancer Center, Massachusetts General Hospital and Harvard Medical School
| | - Luisa Cironi
- Experimental Pathology Service, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Ivan Stamenkovic
- Experimental Pathology Service, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Raffaele Renella
- Department Woman-Mother-Child, Division of Pediatrics, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Fabien Kuttler
- Biomolecular Screening Facility, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Gerardo Turcatti
- Biomolecular Screening Facility, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Miguel N Rivera
- Department of Pathology and Cancer Center, Massachusetts General Hospital and Harvard Medical School
| | - Annick Mühlethaler-Mottet
- Department Woman-Mother-Child, Division of Pediatrics, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Anaïs Flore Bardet
- Biotechnology and Cell Signaling (BSC), CNRS UMR7242, University of Strasbourg, Illkirch, France
- Institute of Genetics and Molecular and Cellular Biology (IGBMC), CNRS UMR7104, University of Strasbourg, INSERM U1258, Illkirch, France
| | - Nicolò Riggi
- Experimental Pathology Service, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
2
|
Pomaville M, Chennakesavalu M, Wang P, Jiang Z, Sun HL, Ren P, Borchert R, Gupta V, Ye C, Ge R, Zhu Z, Brodnik M, Zhong Y, Moore K, Salwen H, George RE, Krajewska M, Chlenski A, Applebaum MA, He C, Cohn SL. Small-molecule inhibition of the METTL3/METTL14 complex suppresses neuroblastoma tumor growth and promotes differentiation. Cell Rep 2024; 43:114165. [PMID: 38691450 PMCID: PMC11181463 DOI: 10.1016/j.celrep.2024.114165] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 03/10/2024] [Accepted: 04/12/2024] [Indexed: 05/03/2024] Open
Abstract
The N6-methyladenosine (m6A) RNA modification is an important regulator of gene expression. m6A is deposited by a methyltransferase complex that includes methyltransferase-like 3 (METTL3) and methyltransferase-like 14 (METTL14). High levels of METTL3/METTL14 drive the growth of many types of adult cancer, and METTL3/METTL14 inhibitors are emerging as new anticancer agents. However, little is known about the m6A epitranscriptome or the role of the METTL3/METTL14 complex in neuroblastoma, a common pediatric cancer. Here, we show that METTL3 knockdown or pharmacologic inhibition with the small molecule STM2457 leads to reduced neuroblastoma cell proliferation and increased differentiation. These changes in neuroblastoma phenotype are associated with decreased m6A deposition on transcripts involved in nervous system development and neuronal differentiation, with increased stability of target mRNAs. In preclinical studies, STM2457 treatment suppresses the growth of neuroblastoma tumors in vivo. Together, these results support the potential of METTL3/METTL14 complex inhibition as a therapeutic strategy against neuroblastoma.
Collapse
Affiliation(s)
- Monica Pomaville
- Department of Pediatrics, University of Chicago Comer Children's Hospital, Chicago, IL 60637, USA
| | | | - Pingluan Wang
- Department of Chemistry, University of Chicago, Chicago, IL 60637, USA
| | - Zhiwei Jiang
- Department of Chemistry, University of Chicago, Chicago, IL 60637, USA
| | - Hui-Lung Sun
- Department of Chemistry, University of Chicago, Chicago, IL 60637, USA
| | - Peizhe Ren
- Department of Chemistry, University of Chicago, Chicago, IL 60637, USA
| | - Ryan Borchert
- Department of Pediatrics, University of Chicago Comer Children's Hospital, Chicago, IL 60637, USA
| | - Varsha Gupta
- Department of Pediatrics, University of Chicago Comer Children's Hospital, Chicago, IL 60637, USA
| | - Chang Ye
- Department of Chemistry, University of Chicago, Chicago, IL 60637, USA
| | - Ruiqi Ge
- Department of Chemistry, University of Chicago, Chicago, IL 60637, USA
| | - Zhongyu Zhu
- Department of Chemistry, University of Chicago, Chicago, IL 60637, USA
| | - Mallory Brodnik
- Department of Pediatrics, University of Chicago Comer Children's Hospital, Chicago, IL 60637, USA
| | - Yuhao Zhong
- Department of Chemistry, University of Chicago, Chicago, IL 60637, USA
| | - Kelley Moore
- Department of Pediatrics, University of Chicago Comer Children's Hospital, Chicago, IL 60637, USA
| | - Helen Salwen
- Department of Pediatrics, University of Chicago Comer Children's Hospital, Chicago, IL 60637, USA
| | - Rani E George
- Department of Pediatric Hematology/Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Malgorzata Krajewska
- School of Biochemistry and Cell Biology, Biosciences Institute, University College Cork, Cork, Ireland
| | - Alexandre Chlenski
- Department of Pediatrics, University of Chicago Comer Children's Hospital, Chicago, IL 60637, USA
| | - Mark A Applebaum
- Department of Pediatrics, University of Chicago Comer Children's Hospital, Chicago, IL 60637, USA
| | - Chuan He
- Department of Chemistry, University of Chicago, Chicago, IL 60637, USA; Howard Hughes Medical Institute, University of Chicago, Chicago, Il 60637 USA
| | - Susan L Cohn
- Department of Pediatrics, University of Chicago Comer Children's Hospital, Chicago, IL 60637, USA.
| |
Collapse
|
3
|
Zhang X, Wang B, Lin L, Zhou C, Zhu J, Wu H, He J. TET3 gene rs828867 G>A polymorphism reduces neuroblastoma risk in Chinese children. Heliyon 2024; 10:e27988. [PMID: 38509981 PMCID: PMC10951652 DOI: 10.1016/j.heliyon.2024.e27988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 02/25/2024] [Accepted: 03/09/2024] [Indexed: 03/22/2024] Open
Abstract
Objective Neuroblastoma (NB) is a prevalent pediatric tumor originating from primordial neural crest cells. As one of the latest epigenetics investigations focuses, RNA 5-methylcytosine (m5C) is closely related to cancer risk. TET methylcytosine dioxygenase 3 (TET3) is a demethylase for m5C modification. Whether there is an association between TET3 gene polymorphisms and neuroblastoma risk remains unclear. Methods We conducted an epidemiological study in 402 patients and 473 controls to evaluate the relationship between TET3 gene SNPs (rs7560668 T > C, rs828867 G > A, and rs6546891 A > G) and NB susceptibility. Results Our results showed that rs828867 G > A significantly reduced NB risk in Chinese children [GA vs. GG, adjusted odds ratio (OR) = 0.72, 95% confidence interval (CI) = 0.52-0.98, P=0.040; GA/AA vs. GG, adjusted OR = 0.74, 95% CI = 0.55-0.998, P=0.048]. Individuals with 2-3 risk genotypes had a significantly higher NB risk than those with 0-1 risk genotypes (adjusted OR = 1.40, 95% CI = 1.04-1.88, P=0.027). The stratified analysis showed that the rs828867 G > A associated with decreased NB risk is remarkable among children aged >18 months (adjusted OR = 0.67, 95% CI = 0.46-0.96, P=0.029) and patients at clinical III + IV stages (adjusted OR = 0.67, 95% CI = 0.45-0.98, P=0.040). Compared with the 0-1 risk genotype, the concurrence of 2-3 risk genotypes significantly increased NB risk in the following subgroups: children aged >18 months and patients at clinical III + IV stages. GTEx analysis suggested that rs828867 G > A was significantly associated with RP11-287D1.4 and POLE4 mRNA expression. Conclusions Overall, our results revealed that rs828867 G > A in the TET3 gene is significantly associated with predisposition to NB.
Collapse
Affiliation(s)
- Xinxin Zhang
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Bo Wang
- Department of Clinical Laboratory, Qingdao Eighth People's Hospital, Qingdao 266100, Shandong, China
| | - Lei Lin
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Chunlei Zhou
- Department of Pathology, Children's Hospital of Nanjing Medical University, Nanjing 210008, Jiangsu, China
| | - Jinhong Zhu
- Department of Clinical Laboratory, Biobank, Harbin Medical University Cancer Hospital, Harbin 150040, Heilongjiang, China
| | - Haiyan Wu
- Department of Pathology, Children's Hospital of Nanjing Medical University, Nanjing 210008, Jiangsu, China
| | - Jing He
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| |
Collapse
|
4
|
Zheng M, Kumar A, Sharma V, Behl T, Sehgal A, Wal P, Shinde NV, Kawaduji BS, Kapoor A, Anwer MK, Gulati M, Shen B, Singla RK, Bungau SG. Revolutionizing pediatric neuroblastoma treatment: unraveling new molecular targets for precision interventions. Front Cell Dev Biol 2024; 12:1353860. [PMID: 38601081 PMCID: PMC11004261 DOI: 10.3389/fcell.2024.1353860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 03/13/2024] [Indexed: 04/12/2024] Open
Abstract
Neuroblastoma (NB) is the most frequent solid tumor in pediatric cases, contributing to around 15% of childhood cancer-related deaths. The wide-ranging genetic, morphological, and clinical diversity within NB complicates the success of current treatment methods. Acquiring an in-depth understanding of genetic alterations implicated in the development of NB is essential for creating safer and more efficient therapies for this severe condition. Several molecular signatures are being studied as potential targets for developing new treatments for NB patients. In this article, we have examined the molecular factors and genetic irregularities, including those within insulin gene enhancer binding protein 1 (ISL1), dihydropyrimidinase-like 3 (DPYSL3), receptor tyrosine kinase-like orphan receptor 1 (ROR1) and murine double minute 2-tumor protein 53 (MDM2-P53) that play an essential role in the development of NB. A thorough summary of the molecular targeted treatments currently being studied in pre-clinical and clinical trials has been described. Recent studies of immunotherapeutic agents used in NB are also studied in this article. Moreover, we explore potential future directions to discover new targets and treatments to enhance existing therapies and ultimately improve treatment outcomes and survival rates for NB patients.
Collapse
Affiliation(s)
- Min Zheng
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Ankush Kumar
- Amity School of Pharmaceutical Sciences, Amity University, Mohali, Punjab, India
| | - Vishakha Sharma
- Amity School of Pharmaceutical Sciences, Amity University, Mohali, Punjab, India
| | - Tapan Behl
- Amity School of Pharmaceutical Sciences, Amity University, Mohali, Punjab, India
| | - Aayush Sehgal
- GHG Khalsa College of Pharmacy, Ludhiana, Punjab, India
| | - Pranay Wal
- Pranveer Singh Institute of Technology, Pharmacy, Kanpur, Uttar Pradesh, India
| | | | | | - Anupriya Kapoor
- School of Pharmaceutical Sciences, Chhatrapati Shahu Ji Maharaj University, Kanpur, Uttar Pradesh, India
| | - Md. Khalid Anwer
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
- Australian Research Consortium in Complementary and Integrative Medicine, Faculty of Health, University of Technology Sydney, Ultimo, NSW, Australia
| | - Bairong Shen
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Rajeev K. Singla
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Simona Gabriela Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
- Doctoral School of Biomedical Sciences, University of Oradea, Oradea, Romania
| |
Collapse
|
5
|
Jiang H, Tiche SJ, He CJ, Jedoui M, Forgo B, Zhao M, He B, Li Y, Li AM, Truong AT, Ho J, Simmermaker C, Yang Y, Zhou MN, Hu Z, Cuthbertson DJ, Svensson KJ, Hazard FK, Shimada H, Chiu B, Ye J. Mitochondrial uncoupler and retinoic acid synergistically induce differentiation and inhibit proliferation in neuroblastoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.22.576741. [PMID: 38328117 PMCID: PMC10849550 DOI: 10.1101/2024.01.22.576741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Neuroblastoma is a leading cause of death in childhood cancer cases. Unlike adult malignancies, which typically develop from aged cells through accumulated damage and mutagenesis, neuroblastoma originates from neural crest cells with disrupted differentiation. This distinct feature provides novel therapeutic opportunities beyond conventional cytotoxic methods. Previously, we reported that the mitochondrial uncoupler NEN (niclosamide ethanolamine) activated mitochondria respiration to reprogram the epigenome, promoting neuronal differentiation. In the current study, we further combine NEN with retinoic acid (RA) to promote neural differentiation both in vitro and in vivo. The treatment increased the expression of RA signaling and neuron differentiation-related genes, resulting in a global shift in the transcriptome towards a more favorable prognosis. Overall, these results suggest that the combination of a mitochondrial uncoupler and the differentiation agent RA is a promising therapeutic strategy for neuroblastoma.
Collapse
Affiliation(s)
- Haowen Jiang
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | | | - Clifford JiaJun He
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Mohamed Jedoui
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Balint Forgo
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Meng Zhao
- Department of Pathology, Stanford University, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Bo He
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Yang Li
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Albert M. Li
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | | | - Jestine Ho
- Agilent Technologies, Inc., Santa Clara, CA, USA
| | | | - Yanan Yang
- Agilent Technologies, Inc., Santa Clara, CA, USA
| | - Meng-Ning Zhou
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Zhen Hu
- Olivia Consulting Service, Redwood City, CA, USA
| | | | - Katrin J. Svensson
- Department of Pathology, Stanford University, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | | | | | - Bill Chiu
- Department of Surgery, Stanford University, Stanford, CA, USA
| | - Jiangbin Ye
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| |
Collapse
|
6
|
Jahangiri L. Predicting Neuroblastoma Patient Risk Groups, Outcomes, and Treatment Response Using Machine Learning Methods: A Review. Med Sci (Basel) 2024; 12:5. [PMID: 38249081 PMCID: PMC10801560 DOI: 10.3390/medsci12010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 12/28/2023] [Accepted: 01/03/2024] [Indexed: 01/23/2024] Open
Abstract
Neuroblastoma, a paediatric malignancy with high rates of cancer-related morbidity and mortality, is of significant interest to the field of paediatric cancers. High-risk NB tumours are usually metastatic and result in survival rates of less than 50%. Machine learning approaches have been applied to various neuroblastoma patient data to retrieve relevant clinical and biological information and develop predictive models. Given this background, this study will catalogue and summarise the literature that has used machine learning and statistical methods to analyse data such as multi-omics, histological sections, and medical images to make clinical predictions. Furthermore, the question will be turned on its head, and the use of machine learning to accurately stratify NB patients by risk groups and to predict outcomes, including survival and treatment response, will be summarised. Overall, this study aims to catalogue and summarise the important work conducted to date on the subject of expression-based predictor models and machine learning in neuroblastoma for risk stratification and patient outcomes including survival, and treatment response which may assist and direct future diagnostic and therapeutic efforts.
Collapse
Affiliation(s)
- Leila Jahangiri
- School of Science and Technology, Nottingham Trent University, Clifton Site, Nottingham NG11 8NS, UK;
- Division of Cellular and Molecular Pathology, Addenbrookes Hospital, University of Cambridge, Cambridge CB2 0QQ, UK
| |
Collapse
|
7
|
Chennakesavalu M, Moore K, Chaves G, Veeravalli S, TerHaar R, Wu T, Lyu R, Chlenski A, He C, Piunti A, Applebaum MA. 5-Hydroxymethylcytosine Profiling of Cell-Free DNA Identifies Bivalent Genes That Are Prognostic of Survival in High-Risk Neuroblastoma. JCO Precis Oncol 2024; 8:e2300297. [PMID: 38295320 PMCID: PMC10843272 DOI: 10.1200/po.23.00297] [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: 06/11/2023] [Revised: 11/01/2023] [Accepted: 11/27/2023] [Indexed: 02/02/2024] Open
Abstract
PURPOSE Neuroblastoma is the most common extracranial solid tumor in childhood. We previously showed that circulating cell-free DNA (cfDNA) and tumor biopsy derived 5-hydroxymethylcytosime (5-hmC) profiles identified patients with neuroblastoma who experienced subsequent relapse. Here, we hypothesized that 5-hmC modifications selectively enriched in cfDNA compared with tumor biopsy samples would identify epigenetic changes associated with aggressive tumor behavior and identify novel biomarkers of outcome in patients with high-risk neuroblastoma. METHODS 5-hmC profiles from cfDNA (n = 64) and tumor biopsies (n = 48) were compared. Two neuroblastoma cell lines underwent chromatin immunoprecipitation followed by sequencing (ChIP-Seq) for H3K27me3, H3K4me3, and H3K27ac; kethoxal-associated single-stranded DNA sequencing; hmC-Seal for 5-hmC; and RNA-sequencing (RNA-Seq). Genes enriched for both H3K27me3 and H3K4me3 in the included cell lines were defined as bivalent. Using bivalent genes defined in vitro, a bivalent signature was established in three publicly available cohorts of patients with neuroblastoma through gene set variation analysis. Differences between tumors with high or low bivalent signatures were assessed by the Kaplan-Meier method and Cox proportional hazards models. RESULTS In cfDNA compared with tumor biopsy derived 5-hmC profiles, we found increased 5-hmC deposition on Polycomb Repressive Complex 2 target genes, a finding previously described in the context of bivalent genes. We identified 313 genes that bore bivalent chromatin marks, were enriched for mediators of neuronal differentiation, and were transcriptionally repressed across a panel of heterogeneous neuroblastoma cell lines. In three distinct clinical cohorts, low bivalent signature was significantly and independently associated with worse clinical outcome in patients with high-risk neuroblastoma. CONCLUSION Low expression of bivalent genes is a biomarker of worse outcome in patients with high-risk neuroblastoma.
Collapse
Affiliation(s)
| | - Kelley Moore
- Department of Pediatrics, University of Chicago, Chicago, IL
| | | | | | - Rachel TerHaar
- Department of Pediatrics, University of Chicago, Chicago, IL
| | - Tong Wu
- Department of Chemistry, University of Chicago, Chicago, IL
| | - Ruitu Lyu
- Department of Chemistry, University of Chicago, Chicago, IL
| | | | - Chuan He
- Department of Chemistry, University of Chicago, Chicago, IL
- Howard Hughes Medical Institute, University of Chicago, Chicago, IL
| | - Andrea Piunti
- Department of Pediatrics, University of Chicago, Chicago, IL
| | | |
Collapse
|
8
|
Chennakesavalu M, Moore K, Chaves G, Veeravalli S, TerHaar R, Wu T, Lyu R, Chlenski A, He C, Piunti A, Applebaum MA. 5-hydroxymethylcytosine profiling of cell-free DNA identifies bivalent genes that are prognostic of survival in high-risk neuroblastoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.27.538309. [PMID: 37163024 PMCID: PMC10168384 DOI: 10.1101/2023.04.27.538309] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Neuroblastoma is the most common extra-cranial solid tumor in childhood and epigenetic dysregulation is a key driver of this embryonal disease. In cell-free DNA from neuroblastoma patients with high-risk disease, we found increased 5-hydroxymethylcytosine (5-hmC) deposition on Polycomb Repressive Complex 2 (PRC2) target genes, a finding previously described in the context of bivalent genes. As bivalent genes, defined as genes bearing both activating (H3K4me3) and repressive (H3K27me3) chromatin modifications, have been shown to play an important role in development and cancer, we investigated the potential role of bivalent genes in maintaining a de-differentiated state in neuroblastoma and their potential use as a biomarker. We identified 313 genes that bore bivalent chromatin marks, were enriched for mediators of neuronal differentiation, and were transcriptionally repressed across a panel of heterogenous neuroblastoma cell lines. Through gene set variance analysis, we developed a clinically implementable bivalent signature. In three distinct clinical cohorts, low bivalent signature was significantly and independently associated with worse clinical outcome in high-risk neuroblastoma patients. Thus, low expression of bivalent genes is a biomarker of ultra-high-risk disease and may represent a therapeutic opportunity in neuroblastoma.
Collapse
|
9
|
Ahmad MH, Ghosh B, Rizvi MA, Ali M, Kaur L, Mondal AC. Neural crest cells development and neuroblastoma progression: Role of Wnt signaling. J Cell Physiol 2023; 238:306-328. [PMID: 36502519 DOI: 10.1002/jcp.30931] [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: 05/12/2022] [Revised: 09/19/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022]
Abstract
Neuroblastoma (NB) is one of the most common heterogeneous extracranial cancers in infancy that arises from neural crest (NC) cells of the sympathetic nervous system. The Wnt signaling pathway, both canonical and noncanonical pathway, is a highly conserved signaling pathway that regulates the development and differentiation of the NC cells during embryogenesis. Reports suggest that aberrant activation of Wnt ligands/receptors in Wnt signaling pathways promote progression and relapse of NB. Wnt signaling pathways regulate NC induction and migration in a similar manner; it regulates proliferation and metastasis of NB. Inhibiting the Wnt signaling pathway or its ligands/receptors induces apoptosis and abrogates proliferation and tumorigenicity in all major types of NB cells. Here, we comprehensively discuss the Wnt signaling pathway and its mechanisms in regulating the development of NC and NB pathogenesis. This review highlights the implications of aberrant Wnt signaling in the context of etiology, progression, and relapse of NB. We have also described emerging strategies for Wnt-based therapies against the progression of NB that will provide new insights into the development of Wnt-based therapeutic strategies for NB.
Collapse
Affiliation(s)
- Mir Hilal Ahmad
- School of Life Sciences, Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India.,Genome Biology Lab, Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Balaram Ghosh
- Department of Clinical Pharmacology, Midnapore Medical College & Hospital, West Bengal, Medinipur, India
| | - Moshahid Alam Rizvi
- Genome Biology Lab, Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Mansoor Ali
- School of Life Sciences, Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Loveleena Kaur
- Division of Cancer Pharmacology, Indian Institute of Integrative Medicine (IIIM), Srinagar, India
| | - Amal Chandra Mondal
- School of Life Sciences, Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
10
|
Zhuo Z, Lin L, Miao L, Li M, He J. Advances in liquid biopsy in neuroblastoma. FUNDAMENTAL RESEARCH 2022; 2:903-917. [PMID: 38933377 PMCID: PMC11197818 DOI: 10.1016/j.fmre.2022.08.005] [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: 04/27/2022] [Revised: 07/18/2022] [Accepted: 08/09/2022] [Indexed: 10/15/2022] Open
Abstract
Even with intensive treatment of high-risk neuroblastoma (NB) patients, half of high-risk NB patients still relapse. New therapies targeting the biological characteristics of NB have important clinical value for the personalized treatment of NB. However, the current biological markers for NB are mainly analyzed by tissue biopsy. In recent years, circulating biomarkers of NB based on liquid biopsy have attracted more and more attention. This review summarizes the analytes and methods for liquid biopsy of NB. We focus on the application of liquid biopsy in the diagnosis, prognosis assessment, and monitoring of NB. Finally, we discuss the prospects and challenges of liquid biopsy in NB.
Collapse
Affiliation(s)
- Zhenjian Zhuo
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
- Laboratory Animal Center, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Lei Lin
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Lei Miao
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Meng Li
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Jing He
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| |
Collapse
|
11
|
PRMT5 activates AKT via methylation to promote tumor metastasis. Nat Commun 2022; 13:3955. [PMID: 35803962 PMCID: PMC9270419 DOI: 10.1038/s41467-022-31645-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 06/24/2022] [Indexed: 11/08/2022] Open
Abstract
Protein arginine methyltransferase 5 (PRMT5) is the primary methyltransferase generating symmetric-dimethyl-arginine marks on histone and non-histone proteins. PRMT5 dysregulation is implicated in multiple oncogenic processes. Here, we report that PRMT5-mediated methylation of protein kinase B (AKT) is required for its subsequent phosphorylation at Thr308 and Ser473. Moreover, pharmacologic or genetic inhibition of PRMT5 abolishes AKT1 arginine 15 methylation, thereby preventing AKT1 translocation to the plasma membrane and subsequent recruitment of its upstream activating kinases PDK1 and mTOR2. We show that PRMT5/AKT signaling controls the expression of the epithelial-mesenchymal-transition transcription factors ZEB1, SNAIL, and TWIST1. PRMT5 inhibition significantly attenuates primary tumor growth and broadly blocks metastasis in multiple organs in xenograft tumor models of high-risk neuroblastoma. Collectively, our results suggest that PRMT5 inhibition augments anti-AKT or other downstream targeted therapeutics in high-risk metastatic cancers.
Collapse
|
12
|
Holliday H, Yang J, Dodson E, Nikolic I, Kamili A, Wheatley M, Deng N, Alexandrou S, Davis TP, Kavallaris M, Caldon CE, McCarroll J, De Preter K, Mestdagh P, Marshall GM, Simpson KJ, Fletcher J, Swarbrick A. miR-99b-5p, miR-380-3p, and miR-485-3p are novel chemosensitizing miRNAs in high-risk neuroblastoma. Mol Ther 2022; 30:1119-1134. [PMID: 34998954 PMCID: PMC8899605 DOI: 10.1016/j.ymthe.2022.01.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 11/17/2021] [Accepted: 01/03/2022] [Indexed: 12/12/2022] Open
Abstract
Neuroblastoma is a deadly childhood cancer arising in the developing sympathetic nervous system. High-risk patients are currently treated with intensive chemotherapy, which is curative in only 50% of children and leaves some surviving patients with life-long side effects. microRNAs (miRNAs) are critical regulators of neural crest development and are deregulated during neuroblastoma tumorigenesis, making miRNA-based drugs an attractive therapeutic avenue. A functional screen of >1,200 miRNA mimics was conducted in neuroblastoma cell lines to discover miRNAs that sensitized cells to low doses (30% inhibitory concentration [IC30]) of doxorubicin and vincristine chemotherapy used in the treatment of the disease. Three miRNAs, miR-99b-5p, miR-380-3p, and miR-485-3p, had potent chemosensitizing activity with doxorubicin in multiple models of high-risk neuroblastoma. These miRNAs underwent genomic loss in a subset of neuroblastoma patients, and low expression predicted poor survival outcome. In vitro functional assays revealed each of these miRNAs enhanced the anti-proliferative and pro-apoptotic effects of doxorubicin. We used RNA sequencing (RNA-seq) to show that miR-99b-5p represses neuroblastoma dependency genes LIN28B and PHOX2B both in vitro and in patient-derived xenograft (PDX) tumors. Luciferase reporter assays demonstrate that PHOX2B is a direct target of miR-99b-5p. We anticipate that restoring the function of the tumor-suppressive miRNAs discovered here may be a valuable therapeutic strategy for the treatment of neuroblastoma patients.
Collapse
Affiliation(s)
- Holly Holliday
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia; Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2031, Australia; School of Women's and Children's Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Jessica Yang
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Eoin Dodson
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Iva Nikolic
- Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC 3002, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC 3002, Australia
| | - Alvin Kamili
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2031, Australia; School of Women's and Children's Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Madeleine Wheatley
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2031, Australia
| | - Niantao Deng
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia
| | - Sarah Alexandrou
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia
| | - Thomas P Davis
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Australian Institute for Bioengineering, The University of Queensland, Brisbane, QLD 2072, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3052, Australia
| | - Maria Kavallaris
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2031, Australia; School of Women's and Children's Health, UNSW Sydney, Sydney, NSW 2052, Australia; Australian Centre for Nanomedicine, UNSW Sydney, Sydney, NSW 2052, Australia
| | - C Elizabeth Caldon
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia
| | - Joshua McCarroll
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2031, Australia; School of Women's and Children's Health, UNSW Sydney, Sydney, NSW 2052, Australia; Australian Centre for Nanomedicine, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Katleen De Preter
- Cancer Research Institute Ghent, Ghent University, Ghent B-9000, Belgium
| | - Pieter Mestdagh
- Cancer Research Institute Ghent, Ghent University, Ghent B-9000, Belgium
| | - Glenn M Marshall
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2031, Australia; Kids Cancer Centre, Sydney Children's Hospital, Sydney, NSW 2031, Australia
| | - Kaylene J Simpson
- Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC 3002, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC 3002, Australia
| | - Jamie Fletcher
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2031, Australia; School of Women's and Children's Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Alexander Swarbrick
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia.
| |
Collapse
|
13
|
Li L, Chen S, Li J, Rong G, Yang J, Li Y. Characterization of m6A-related lncRNA signature in neuroblastoma. Front Pediatr 2022; 10:927885. [PMID: 36324814 PMCID: PMC9618704 DOI: 10.3389/fped.2022.927885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 09/20/2022] [Indexed: 11/28/2022] Open
Abstract
N6-methyladenosine (m6A) constitutes one of the most common modifications in mRNA, rRNA, tRNA, microRNA, and long-chain noncoding RNA. The influence of modifications of m6A on the stability of RNA depends upon the expression of methyltransferase ("writer") and demethylase ("eraser") and m6A binding protein ("reader"). In this study, we identified a set of m6A-related lncRNA expression profiles in neuroblastoma (NBL) based on the Therapeutically Applicable Research to Generate Effective Treatments (TARGET) program. Thereupon, we identified two subgroups of neuroblastoma (high-risk group and low-risk group) by applying consensus clustering to m6A RNA methylation regulators ("Readers,", "Writer," and "Erase"). Relative to the low-risk group, the high-risk group correlates with a poorer prognosis. Moreover, the present study also revealed that the high-risk group proves to be significantly positively enriched in the tumor-related signaling pathways, including the P53 signaling pathway, cell cycle, and DNA repair. This finding indicates that these molecular prognostic markers may also be potentially valuable in early diagnosis, which provides a new research direction for the study of molecular mechanisms underlying the development of NBL. In conclusion, this study constructed a new model of NBL prognosis based on m6a-associated lncRNAs. Ultimately, this model is helpful for stratification of prognosis and development of treatment strategies.
Collapse
Affiliation(s)
- Liming Li
- Department of Pediatric Surgery, GuiPing People's Hospital, Guangxi, China
| | - Sisi Chen
- Department of Pediatric Surgery, GuiPing People's Hospital, Guangxi, China
| | - Jianhong Li
- Department of Pediatric Surgery, GuiPing People's Hospital, Guangxi, China
| | - Guochou Rong
- Department of Pediatric Surgery, GuiPing People's Hospital, Guangxi, China
| | - Juchao Yang
- Department of Pediatric Surgery, GuiPing People's Hospital, Guangxi, China
| | - Yunquan Li
- Department of Pediatric Surgery, GuiPing People's Hospital, Guangxi, China
| |
Collapse
|
14
|
Pan C, Liang L, Wang Z, Zhang B, Li Q, Tian Y, Yu Y, Chen Z, Wang X, Liu H. Expression and significance of SOX B1 genes in glioblastoma multiforme patients. J Cell Mol Med 2021; 26:789-799. [PMID: 34953010 PMCID: PMC8817144 DOI: 10.1111/jcmm.17120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/18/2021] [Accepted: 12/01/2021] [Indexed: 12/21/2022] Open
Abstract
The overall survival of glioblastoma multiforme (GBM) patients remains poor. To improve patient outcomes, effective diagnostic and prognostic biomarkers for GBM are needed. In this study, we first applied bioinformatic analyses to identify biomarkers for GBM, focusing on SOX (sex‐determining region on the Y chromosome (SRY)‐related high mobility group (HMG) box) B1 family members. The ONCOMINE, GEPIA, LinkedOmics and CCLE databases were used to assess mRNA expression levels of the SOX B1 family members in different cancers and normal tissue. Further bioinformatic analysis was performed using the ONCOMINE database in combination with the LinkedOmics data set to identify the prognostic value of SOX B1 family members for GBM. We found mRNA expression levels of all tested SOX B1 genes were significantly increased in GBM. In the LinkedOmics database, increased expression of SOX3 indicated a better overall survival. In GEPIA databases, increased expression of all SOX B1 family members suggested an improved overall survival, but none of them were statistically different. Then, Transwell assays and wound healing were employed to evaluate the motility and invasive captivity of U251 cells when silencing SOX2 and SOX3. We found exogenous inhibition of SOX2 appeared to reduce the migration and invasion of U251 cells in vitro. Collectively, our research suggested that SOX2 might serve as a cancer‐promoting gene to identify high‐risk GBM patients, and SOX3 had the potential to be a prognostic biomarker for GBM patients.
Collapse
Affiliation(s)
- Cunyao Pan
- School of Public Health Lanzhou University, Lanzhou, China
| | - Lanlan Liang
- School of Public Health Lanzhou University, Lanzhou, China
| | - Zirou Wang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Baoyi Zhang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Qionglin Li
- School of Public Health Lanzhou University, Lanzhou, China
| | - Yingrui Tian
- School of Public Health Lanzhou University, Lanzhou, China
| | - Yijing Yu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Zhaoli Chen
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Xinxing Wang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Hui Liu
- School of Public Health Lanzhou University, Lanzhou, China
| |
Collapse
|
15
|
Jahangiri L, Pucci P, Ishola T, Pereira J, Cavanagh ML, Turner SD. Deep analysis of neuroblastoma core regulatory circuitries using online databases and integrated bioinformatics shows their pan-cancer roles as prognostic predictors. Discov Oncol 2021; 12:56. [PMID: 35201514 PMCID: PMC8777518 DOI: 10.1007/s12672-021-00452-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 11/16/2021] [Indexed: 12/29/2022] Open
Abstract
AIM Neuroblastoma is a heterogeneous childhood cancer derived from the neural crest. The dual cell identities of neuroblastoma include Mesenchymal (MES) and Adrenergic (ADRN). These identities are conferred by a small set of tightly-regulated transcription factors (TFs) binding super enhancers, collectively forming core regulatory circuitries (CRCs). The purpose of this study was to gain a deep understanding of the role of MES and ADRN TFs in neuroblastoma and other cancers as potential indicators of disease prognosis, progression, and relapse. METHODS To that end, we first investigated the expression and mutational profile of MES and ADRN TFs in neuroblastoma. Moreover, we established their correlation with neuroblastoma risk groups and overall survival while establishing their extended networks with long non-coding RNAs (lncRNAs). Furthermore, we analysed the pan-cancer expression and mutational profile of these TFs and their correlation with patient survival and finally their network connectivity, using a panel of bioinformatic tools including GEPIA2, human pathology atlas, TIMER2, Omicsnet, and Cytoscape. RESULTS We show the association of multiple MES and ADRN TFs with neuroblastoma risk groups and overall survival and find significantly higher expression of various MES and ADRN TFs compared to normal tissues and their association with overall survival and disease-free survival in multiple cancers. Moreover, we report the strong correlation of the expression of these TFs with the infiltration of stromal and immune cells in the tumour microenvironment and with stemness and metastasis-related genes. Furthermore, we reveal extended pan-cancer networks comprising these TFs that influence the tumour microenvironment and metastasis and may be useful indicators of cancer prognosis and patient survival. CONCLUSION Our meta-analysis shows the significance of MES and ADRN TFs as indicators of patient prognosis and the putative utility of these TFs as potential novel biomarkers.
Collapse
Affiliation(s)
- Leila Jahangiri
- Department of Life Sciences, Birmingham City University, Birmingham, UK
- School of Science & Technology, Nottingham Trent University, Clifton Lane, Nottingham, NG11 8NS UK
- Division of Cellular and Molecular Pathology, Addenbrooke’s Hospital, University of Cambridge, Cambridge, UK
| | - Perla Pucci
- Division of Cellular and Molecular Pathology, Addenbrooke’s Hospital, University of Cambridge, Cambridge, UK
| | - Tala Ishola
- Department of Life Sciences, Birmingham City University, Birmingham, UK
| | - Joao Pereira
- Department of Neurology, Massachusetts General Hospital, Boston, MA USA
| | - Megan L. Cavanagh
- Department of Life Sciences, Birmingham City University, Birmingham, UK
| | - Suzanne D. Turner
- Division of Cellular and Molecular Pathology, Addenbrooke’s Hospital, University of Cambridge, Cambridge, UK
- CEITEC, Masaryk University, Brno, Czech Republic
| |
Collapse
|
16
|
Ernsberger U, Deller T, Rohrer H. The sympathies of the body: functional organization and neuronal differentiation in the peripheral sympathetic nervous system. Cell Tissue Res 2021; 386:455-475. [PMID: 34757495 PMCID: PMC8595186 DOI: 10.1007/s00441-021-03548-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/20/2021] [Indexed: 02/06/2023]
Abstract
During the last 30 years, our understanding of the development and diversification of postganglionic sympathetic neurons has dramatically increased. In parallel, the list of target structures has been critically extended from the cardiovascular system and selected glandular structures to metabolically relevant tissues such as white and brown adipose tissue, lymphoid tissues, bone, and bone marrow. A critical question now emerges for the integration of the diverse sympathetic neuron classes into neural circuits specific for these different target tissues to achieve the homeostatic regulation of the physiological ends affected.
Collapse
Affiliation(s)
- Uwe Ernsberger
- Institute for Clinical Neuroanatomy, Goethe University, Frankfurt/Main, Germany.
| | - Thomas Deller
- Institute for Clinical Neuroanatomy, Goethe University, Frankfurt/Main, Germany
| | - Hermann Rohrer
- Institute for Clinical Neuroanatomy, Goethe University, Frankfurt/Main, Germany.
| |
Collapse
|
17
|
Jiménez C, Antonelli R, Masanas M, Soriano A, Devis-Jauregui L, Camacho J, Magdaleno A, Guillén G, Hladun R, Jubierre L, Roma J, Llobet-Navas D, Sánchez de Toledo J, Moreno L, Gallego S, Segura MF. Neuronal Differentiation-Related Epigenetic Regulator ZRF1 Has Independent Prognostic Value in Neuroblastoma but Is Functionally Dispensable In Vitro. Cancers (Basel) 2021; 13:cancers13194845. [PMID: 34638328 PMCID: PMC8508520 DOI: 10.3390/cancers13194845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/17/2021] [Accepted: 09/25/2021] [Indexed: 11/27/2022] Open
Abstract
Simple Summary Neuroblastoma is the most common pediatric solid tumor occurring outside the brain, and it is thought to arise from cells that acquire errors during the normal process of embryonal development. Today, we know that embryonal development is regulated by epigenetics, a mechanism that determines which genes need to be expressed in each cell type and developmental step. Epigenetic errors, therefore, are considered contributory to the appearance and progression of tumors such as neuroblastoma. Here, we aimed at finding whether ZRF1, a known epigenetic regulator, could play a significant role in the aggressiveness of neuroblastoma. Our results suggest that ZRF1 does not seem to have any relevant function in neuroblastoma cells; however, the levels of this epigenetic regulator are related to the prognostic of neuroblastoma patients and could be used to predict their progression and improve the diagnosis. Abstract Neuroblastoma is a pediatric tumor of the peripheral nervous system that accounts for up to ~15% of all cancer-related deaths in children. Recently, it has become evident that epigenetic deregulation is a relevant event in pediatric tumors such as high-risk neuroblastomas, and a determinant for processes, such as cell differentiation blockade and sustained proliferation, which promote tumor progression and resistance to current therapies. Thus, a better understanding of epigenetic factors implicated in the aggressive behavior of neuroblastoma cells is crucial for the development of better treatments. In this study, we characterized the role of ZRF1, an epigenetic activator recruited to genes involved in the maintenance of the identity of neural progenitors. We combined analysis of patient sample expression datasets with loss- and gain-of-function studies on neuroblastoma cell lines. Functional analyses revealed that ZRF1 is functionally dispensable for those cellular functions related to cell differentiation, proliferation, migration, and invasion, and does not affect the cellular response to chemotherapeutic agents. However, we found that high levels of ZRF1 mRNA expression are associated to shorter overall survival of neuroblastoma patients, even when those patients with the most common molecular alterations used as prognostic factors are removed from the analyses, thereby suggesting that ZRF1 expression could be used as an independent prognostic factor in neuroblastoma.
Collapse
Affiliation(s)
- Carlos Jiménez
- Group of Translational Research in Child and Adolescent Cancer, Vall d’Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; (C.J.); (R.A.); (M.M.); (A.S.); (A.M.); (G.G.); (R.H.); (L.J.); (J.R.); (J.S.d.T.); (L.M.); (S.G.)
| | - Roberta Antonelli
- Group of Translational Research in Child and Adolescent Cancer, Vall d’Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; (C.J.); (R.A.); (M.M.); (A.S.); (A.M.); (G.G.); (R.H.); (L.J.); (J.R.); (J.S.d.T.); (L.M.); (S.G.)
| | - Marc Masanas
- Group of Translational Research in Child and Adolescent Cancer, Vall d’Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; (C.J.); (R.A.); (M.M.); (A.S.); (A.M.); (G.G.); (R.H.); (L.J.); (J.R.); (J.S.d.T.); (L.M.); (S.G.)
| | - Aroa Soriano
- Group of Translational Research in Child and Adolescent Cancer, Vall d’Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; (C.J.); (R.A.); (M.M.); (A.S.); (A.M.); (G.G.); (R.H.); (L.J.); (J.R.); (J.S.d.T.); (L.M.); (S.G.)
| | - Laura Devis-Jauregui
- Molecular Mechanisms and Experimental Therapy in Oncology-Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), 08908 L’Hospitalet de Llobregat, Spain; (L.D.-J.); (D.L.-N.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Jessica Camacho
- Pathology Department, Vall d’Hebron University Hospital—UAB, 08035 Barcelona, Spain;
| | - Ainara Magdaleno
- Group of Translational Research in Child and Adolescent Cancer, Vall d’Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; (C.J.); (R.A.); (M.M.); (A.S.); (A.M.); (G.G.); (R.H.); (L.J.); (J.R.); (J.S.d.T.); (L.M.); (S.G.)
| | - Gabriela Guillén
- Group of Translational Research in Child and Adolescent Cancer, Vall d’Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; (C.J.); (R.A.); (M.M.); (A.S.); (A.M.); (G.G.); (R.H.); (L.J.); (J.R.); (J.S.d.T.); (L.M.); (S.G.)
- Surgery Department, Vall d’Hebron University Hospital—UAB, 08035 Barcelona, Spain
| | - Raquel Hladun
- Group of Translational Research in Child and Adolescent Cancer, Vall d’Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; (C.J.); (R.A.); (M.M.); (A.S.); (A.M.); (G.G.); (R.H.); (L.J.); (J.R.); (J.S.d.T.); (L.M.); (S.G.)
- Pediatric Oncology and Hematology Department, Vall d’Hebron University Hospital—UAB, 08035 Barcelona, Spain
| | - Luz Jubierre
- Group of Translational Research in Child and Adolescent Cancer, Vall d’Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; (C.J.); (R.A.); (M.M.); (A.S.); (A.M.); (G.G.); (R.H.); (L.J.); (J.R.); (J.S.d.T.); (L.M.); (S.G.)
| | - Josep Roma
- Group of Translational Research in Child and Adolescent Cancer, Vall d’Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; (C.J.); (R.A.); (M.M.); (A.S.); (A.M.); (G.G.); (R.H.); (L.J.); (J.R.); (J.S.d.T.); (L.M.); (S.G.)
| | - David Llobet-Navas
- Molecular Mechanisms and Experimental Therapy in Oncology-Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), 08908 L’Hospitalet de Llobregat, Spain; (L.D.-J.); (D.L.-N.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Josep Sánchez de Toledo
- Group of Translational Research in Child and Adolescent Cancer, Vall d’Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; (C.J.); (R.A.); (M.M.); (A.S.); (A.M.); (G.G.); (R.H.); (L.J.); (J.R.); (J.S.d.T.); (L.M.); (S.G.)
- Catalan Institute of Oncology (ICO), 08908 L’Hospitalet de Llobregat, Spain
| | - Lucas Moreno
- Group of Translational Research in Child and Adolescent Cancer, Vall d’Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; (C.J.); (R.A.); (M.M.); (A.S.); (A.M.); (G.G.); (R.H.); (L.J.); (J.R.); (J.S.d.T.); (L.M.); (S.G.)
- Pediatric Oncology and Hematology Department, Vall d’Hebron University Hospital—UAB, 08035 Barcelona, Spain
| | - Soledad Gallego
- Group of Translational Research in Child and Adolescent Cancer, Vall d’Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; (C.J.); (R.A.); (M.M.); (A.S.); (A.M.); (G.G.); (R.H.); (L.J.); (J.R.); (J.S.d.T.); (L.M.); (S.G.)
- Pediatric Oncology and Hematology Department, Vall d’Hebron University Hospital—UAB, 08035 Barcelona, Spain
| | - Miguel F. Segura
- Group of Translational Research in Child and Adolescent Cancer, Vall d’Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; (C.J.); (R.A.); (M.M.); (A.S.); (A.M.); (G.G.); (R.H.); (L.J.); (J.R.); (J.S.d.T.); (L.M.); (S.G.)
- Correspondence:
| |
Collapse
|
18
|
Clark RA, Newton M, Qiao J, Lee S, Chung DH. Reactivation of silenced α-N-catenin induces retinoic acid sensitivity in neuroblastoma cells. Surgery 2021; 170:1546-1553. [PMID: 34092372 DOI: 10.1016/j.surg.2021.04.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 04/08/2021] [Accepted: 04/20/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND High-risk neuroblastoma remains the most difficult pediatric solid tumor to treat and is associated with chemotherapy and radiation resistance that may be secondary to epigenetic modifications. We have previously found that α-N-catenin, a cell-adhesion protein encoded by the gene CTNNA2, plays a tumor suppressor role in neuroblastoma by inhibiting the NF-κB signaling pathway. A subset of neuroblastoma tumors that lack α-N-catenin are resistant to all-trans retinoic acid. However, the mechanism of CTNNA2 silencing in neuroblastoma remains unknown. Herein, we sought to determine the mechanism of α-N-catenin silencing in neuroblastoma. METHODS Two human neuroblastoma cell lines, SK-N-AS and BE(2)-C, were stably transfected with a plasmid expressing CTNNA2. Both cell lines were treated with the histone deacetylase inhibitor Trichostatin A alone and in combination with retinoic acid. Cell survival and colony formation were measured. Cellular differentiation and expression of cell survival signaling pathways were analyzed. Immunoblotting and reverse transcription quantitative polymerase chain reaction were used to examine protein and messenger RNA expression. RESULTS Retinoic acid treatment induced cellular differentiation and inhibited cellular proliferation in BE(2)-C cells but did not induce differentiation in SK-N-AS cells. Re-expression of α-N-catenin enhanced the sensitivity to retinoic acid-induced cell growth arrest and downregulated key cell survival pathways in both cell lines. Trichostatin A treatment induced CTNNA2 expression in SK-N-AS cells, and combination treatment with Trichostatin A induced retinoic acid sensitivity in retinoic acid-resistant cells. CONCLUSION Re-expression of α-N-catenin in retinoic acid-resistant cells induced sensitivity to retinoic acid treatment and is controlled epigenetically via histone deacetylase. α-N-catenin is a potential biomarker for retinoic acid sensitivity and combination treatment with Trichostatin A and retinoic acid may improve survival among children with high-risk, retinoic acid-resistant neuroblastoma.
Collapse
Affiliation(s)
- Rachael A Clark
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX
| | - Micah Newton
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX
| | - Jingbo Qiao
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX
| | - Sora Lee
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX
| | - Dai H Chung
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX.
| |
Collapse
|
19
|
Krstic A, Konietzny A, Halasz M, Cain P, Oppermann U, Kolch W, Duffy DJ. A Chemo-Genomic Approach Identifies Diverse Epigenetic Therapeutic Vulnerabilities in MYCN-Amplified Neuroblastoma. Front Cell Dev Biol 2021; 9:612518. [PMID: 33968920 PMCID: PMC8097097 DOI: 10.3389/fcell.2021.612518] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 03/25/2021] [Indexed: 11/13/2022] Open
Abstract
Although a rare disease, neuroblastoma accounts for the highest proportion of childhood cancer deaths. There is a lack of recurrent somatic mutations in neuroblastoma embryonal tumours, suggesting a possible role for epigenetic alterations in driving this cancer. While an increasing number of reports suggest an association of MYCN with epigenetic machinery, the mechanisms of these interactions are poorly understood in the neuroblastoma setting. Utilising chemo-genomic approaches we revealed global MYCN-epigenetic interactions and identified numerous epigenetic proteins as MYCN targets. The epigenetic regulators HDAC2, CBX8 and CBP (CREBBP) were all MYCN target genes and also putative MYCN interactors. MYCN-related epigenetic genes included SMARCs, HDACs, SMYDs, BRDs and CREBBP. Expression levels of the majority of MYCN-related epigenetic genes showed predictive ability for neuroblastoma patient outcome. Furthermore, a compound library screen targeting epigenetic proteins revealed broad susceptibility of neuroblastoma cells to all classes of epigenetic regulators, belonging to families of bromodomains, HDACs, HATs, histone methyltransferases, DNA methyltransferases and lysin demethylases. Ninety-six percent of the compounds reduced MYCN-amplified neuroblastoma cell viability. We show that the C646 (CBP-bromodomain targeting compound) exhibits switch-like temporal and dose response behaviour and is effective at reducing neuroblastoma viability. Responsiveness correlates with MYCN expression, with MYCN-amplified cells being more susceptible to C646 treatment. Thus, exploiting the broad vulnerability of neuroblastoma cells to epigenetic targeting compounds represents an exciting strategy in neuroblastoma treatment, particularly for high-risk MYCN-amplified tumours.
Collapse
Affiliation(s)
- Aleksandar Krstic
- Systems Biology Ireland and Precision Oncology Ireland, School of Medicine, University College Dublin, Dublin, Ireland
| | - Anja Konietzny
- Systems Biology Ireland and Precision Oncology Ireland, School of Medicine, University College Dublin, Dublin, Ireland.,Centre for Molecular Neurobiology Hamburg (ZMNH), Emmy-Noether Group "Neuronal Protein Transport", University Medical Centre Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Melinda Halasz
- Systems Biology Ireland and Precision Oncology Ireland, School of Medicine, University College Dublin, Dublin, Ireland
| | - Peter Cain
- Botnar Research Centre, NIHR Oxford Biomedical Research Unit, Institute of Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom.,Centre for Medicines Discovery, University of Oxford, Oxford, United Kingdom
| | - Udo Oppermann
- Botnar Research Centre, NIHR Oxford Biomedical Research Unit, Institute of Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom.,Centre for Medicines Discovery, University of Oxford, Oxford, United Kingdom
| | - Walter Kolch
- Systems Biology Ireland and Precision Oncology Ireland, School of Medicine, University College Dublin, Dublin, Ireland.,Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland
| | - David J Duffy
- Systems Biology Ireland and Precision Oncology Ireland, School of Medicine, University College Dublin, Dublin, Ireland.,The Whitney Laboratory for Marine Bioscience and Sea Turtle Hospital, University of Florida, St. Augustine, FL, United States.,Department of Biology, University of Florida, Gainesville, FL, United States
| |
Collapse
|
20
|
Erfani S, Hua H, Pan Y, Zhou BP, Yang XH. The Context-Dependent Impact of Integrin-Associated CD151 and Other Tetraspanins on Cancer Development and Progression: A Class of Versatile Mediators of Cellular Function and Signaling, Tumorigenesis and Metastasis. Cancers (Basel) 2021; 13:cancers13092005. [PMID: 33919420 PMCID: PMC8122392 DOI: 10.3390/cancers13092005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 03/18/2021] [Accepted: 04/01/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Tetraspanins are a family of molecules abundantly expressed on the surface of normal or tumor cells. They have been implicated in recruiting or sequestering key molecular regulators of malignancy of a variety of human cancers, including breast and lung cancers, glioblastoma and leukemia. Yet, how their actions take place remains mysterious due to a lack of traditional platform for molecular interactions. The current review digs into this mystery by examining findings from recent studies of multiple tetraspanins, particularly CD151. The molecular basis for differential impact of tetraspanins on tumor development, progression, and spreading to secondary sites is highlighted, and the complexity and plasticity of their control over tumor cell activities and interaction with their surroundings is discussed. Finally, an outlook is provided regarding tetraspanins as candidate biomarkers and targets for the diagnosis and treatment of human cancer. Abstract As a family of integral membrane proteins, tetraspanins have been functionally linked to a wide spectrum of human cancers, ranging from breast, colon, lung, ovarian, prostate, and skin carcinomas to glioblastoma. CD151 is one such prominent member of the tetraspanin family recently suggested to mediate tumor development, growth, and progression in oncogenic context- and cell lineage-dependent manners. In the current review, we summarize recent advances in mechanistic understanding of the function and signaling of integrin-associated CD151 and other tetraspanins in multiple cancer types. We also highlight emerging genetic and epigenetic evidence on the intrinsic links between tetraspanins, the epithelial-mesenchymal transition (EMT), cancer stem cells (CSCs), and the Wnt/β-catenin pathway, as well as the dynamics of exosome and cellular metabolism. Finally, we discuss the implications of the highly plastic nature and epigenetic susceptibility of CD151 expression, function, and signaling for clinical diagnosis and therapeutic intervention for human cancer.
Collapse
Affiliation(s)
- Sonia Erfani
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40536, USA;
- Markey Cancer Center, University of Kentucky Medical Center, Lexington, KY 40536, USA
- Pharmacy Department, St. Elizabeth Healthcare, Edgewood, KY 41017, USA
| | - Hui Hua
- The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui 230001, China; (H.H.); (Y.P.)
- Provincial Hospital, Hefei, Anhui 230001, China
| | - Yueyin Pan
- The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui 230001, China; (H.H.); (Y.P.)
- Provincial Hospital, Hefei, Anhui 230001, China
| | - Binhua P. Zhou
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, KY 40536, USA;
| | - Xiuwei H. Yang
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40536, USA;
- Markey Cancer Center, University of Kentucky Medical Center, Lexington, KY 40536, USA
- Correspondence: ; Tel.: +1-859-323-1996
| |
Collapse
|
21
|
Consales C, Butera A, Merla C, Pasquali E, Lopresto V, Pinto R, Pierdomenico M, Mancuso M, Marino C, Benassi B. Exposure of the SH-SY5Y Human Neuroblastoma Cells to 50-Hz Magnetic Field: Comparison Between Two-Dimensional (2D) and Three-Dimensional (3D) In Vitro Cultures. Mol Neurobiol 2020; 58:1634-1649. [PMID: 33230715 PMCID: PMC7932966 DOI: 10.1007/s12035-020-02192-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 10/30/2020] [Indexed: 12/13/2022]
Abstract
We here characterize the response to the extremely low-frequency (ELF) magnetic field (MF, 50 Hz, 1 mT) of SH-SY5Y human neuroblastoma cells, cultured in a three-dimensional (3D) Alvetex® scaffold compared to conventional two-dimensional (2D) monolayers. We proved that the growing phenotype of proliferating SH-SY5Y cells is not affected by the culturing conditions, as morphology, cell cycle distribution, proliferation/differentiation gene expression of 3D-cultures overlap what reported in 2D plates. In response to 72-h exposure to 50-Hz MF, we demonstrated that no proliferation change and apoptosis activation occur in both 2D and 3D cultures. Consistently, no modulation of Ki67, MYCN, CCDN1, and Nestin, of invasiveness and neo-angiogenesis-controlling genes (HIF-1α, VEGF, and PDGF) and of microRNA epigenetic signature (miR-21-5p, miR-222-3p and miR-133b) is driven by ELF exposure. Conversely, intracellular glutathione content and SOD1 expression are exclusively impaired in 3D-culture cells in response to the MF, whereas no change of such redox modulators is observed in SH-SY5Y cells if grown on 2D monolayers. Moreover, ELF-MF synergizes with the differentiating agents to stimulate neuroblastoma differentiation into a dopaminergic (DA) phenotype in the 3D-scaffold culture only, as growth arrest and induction of p21, TH, DAT, and GAP43 are reported in ELF-exposed SH-SY5Y cells exclusively if grown on 3D scaffolds. As overall, our findings prove that 3D culture is a more reliable experimental model for studying SH-SY5Y response to ELF-MF if compared to 2D conventional monolayer, and put the bases for promoting 3D systems in future studies addressing the interaction between electromagnetic fields and biological systems.
Collapse
Affiliation(s)
- Claudia Consales
- Division of Health Protection Technologies, ENEA-Casaccia Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Via Anguillarese 301, 00123, Rome, Italy
| | - Alessio Butera
- Experimental Medicine and Surgery, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Caterina Merla
- Division of Health Protection Technologies, ENEA-Casaccia Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Via Anguillarese 301, 00123, Rome, Italy
| | - Emanuela Pasquali
- Division of Health Protection Technologies, ENEA-Casaccia Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Via Anguillarese 301, 00123, Rome, Italy
| | - Vanni Lopresto
- Division of Health Protection Technologies, ENEA-Casaccia Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Via Anguillarese 301, 00123, Rome, Italy
| | - Rosanna Pinto
- Division of Health Protection Technologies, ENEA-Casaccia Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Via Anguillarese 301, 00123, Rome, Italy
| | - Maria Pierdomenico
- Division of Health Protection Technologies, ENEA-Casaccia Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Via Anguillarese 301, 00123, Rome, Italy
| | - Mariateresa Mancuso
- Division of Health Protection Technologies, ENEA-Casaccia Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Via Anguillarese 301, 00123, Rome, Italy
| | - Carmela Marino
- Division of Health Protection Technologies, ENEA-Casaccia Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Via Anguillarese 301, 00123, Rome, Italy
| | - Barbara Benassi
- Division of Health Protection Technologies, ENEA-Casaccia Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Via Anguillarese 301, 00123, Rome, Italy.
| |
Collapse
|
22
|
NBPF1 independently determine the risk stratification and prognosis of patients with neuroblastoma. Genomics 2020; 112:3951-3957. [DOI: 10.1016/j.ygeno.2020.06.038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 06/22/2020] [Indexed: 02/07/2023]
|
23
|
Weiser DA, West-Szymanski DC, Fraint E, Weiner S, Rivas MA, Zhao CWT, He C, Applebaum MA. Progress toward liquid biopsies in pediatric solid tumors. Cancer Metastasis Rev 2020; 38:553-571. [PMID: 31836951 DOI: 10.1007/s10555-019-09825-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Pediatric solid tumors have long been known to shed tumor cells, DNA, RNA, and proteins into the blood. Recent technological advances have allowed for improved capture and analysis of these typically scant circulating materials. Efforts are ongoing to develop "liquid biopsy" assays as minimally invasive tools to address diagnostic, prognostic, and disease monitoring needs in childhood cancer care. Applying these highly sensitive technologies to serial liquid biopsies is expected to advance understanding of tumor biology, heterogeneity, and evolution over the course of therapy, thus opening new avenues for personalized therapy. In this review, we outline the latest technologies available for liquid biopsies and describe the methods, pitfalls, and benefits of the assays that are being developed for children with extracranial solid tumors. We discuss what has been learned in several of the most common pediatric solid tumors including neuroblastoma, sarcoma, Wilms tumor, and hepatoblastoma and highlight promising future directions for the field.
Collapse
Affiliation(s)
- Daniel A Weiser
- Department of Pediatrics, Albert Einstein College of Medicine and Children's Hospital at Montefiore, Bronx, NY, USA
| | | | - Ellen Fraint
- Department of Pediatrics, Albert Einstein College of Medicine and Children's Hospital at Montefiore, Bronx, NY, USA
| | - Shoshana Weiner
- Department of Pediatrics, Weill Cornell Medical Center, New York, NY, USA
| | - Marco A Rivas
- Department of Chemistry, Department of Biochemistry and Molecular Biology, and Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, USA
| | - Carolyn W T Zhao
- Department of Chemistry, Department of Biochemistry and Molecular Biology, and Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, USA
| | - Chuan He
- Department of Chemistry, Department of Biochemistry and Molecular Biology, and Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, USA.,Howard Hughes Medical Institute, The University of Chicago, Chicago, IL, USA
| | - Mark A Applebaum
- Department of Pediatrics, The University of Chicago, 900 E. 57th St., KCBD 5116, Chicago, IL, 60637, USA.
| |
Collapse
|
24
|
Li S, Zhuo Z, Chang X, Ma Y, Zhou H, Zhang J, Cheng J, He J, Li Y. NRAS rs2273267 A>T polymorphism reduces neuroblastoma risk in Chinese children. Gene 2020; 727:144262. [PMID: 31759987 DOI: 10.1016/j.gene.2019.144262] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 10/21/2019] [Accepted: 11/07/2019] [Indexed: 02/07/2023]
Abstract
Neuroblastoma is an extracranial solid tumor that mainly occurs in childhood. Mutations of NRAS gene have been described in several cancers. However, whether NRAS gene polymorphisms can predict the risk of neuroblastoma have not been investigated. We hypothesized that variations of NRAS gene contribute to neuroblastoma predisposition. Therefore, we conducted a multi-center case-control study using 263 cases and 715 controls to examine the association of NRAS gene rs2273267 A>T polymorphism and neuroblastoma risk. We calculated odds ratios (ORs) and corresponding 95% confidence intervals (CIs) to assess the strength of the associations. Relative to those with AA genotype, subjects with AT/TT genotype had reduced neuroblastoma risk (adjusted OR = 0.72, 95% CI = 0.54-0.96, P = 0.024). Stratified analysis revealed that rs2273267 AT/TT carriers were less likely to develop neuroblastoma for patients with tumor originating from the adrenal gland (adjusted OR = 0.67, 95% CI = 0.45-0.99, P = 0.047) and clinical stages III + IV (adjusted OR = 0.57, 95% CI = 0.36-0.90, P = 0.015). Our findings underline the likely importance of NRAS gene rs2273267 A>T in the risk of neuroblastoma. Further independent case-control studies with functional analysis are needed to verify the role of NRAS gene rs2273267 A>T polymorphism in the risk of neuroblastoma.
Collapse
Affiliation(s)
- Suhong Li
- Department of Pathology, Children Hospital and Women Health Center of Shanxi, Taiyuan, 030013 Shannxi, China.
| | - Zhenjian Zhuo
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong 510623, Guangzhou, China
| | - Xinghong Chang
- Department of Clinical Laboratory, Children Hospital and Women Health Center of Shanxi, Taiyuan 030013, Shannxi, China
| | - Yan Ma
- Department of Pathology, Children Hospital and Women Health Center of Shanxi, Taiyuan, 030013 Shannxi, China
| | - Haixia Zhou
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
| | - Jiao Zhang
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Jiwen Cheng
- Department of Pediatric Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi, China
| | - Jing He
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong 510623, Guangzhou, China.
| | - Yangyang Li
- Department of Pathology, Children Hospital and Women Health Center of Shanxi, Taiyuan, 030013 Shannxi, China
| |
Collapse
|
25
|
Li Y, Gruber JJ, Litzenburger UM, Zhou Y, Miao YR, LaGory EL, Li AM, Hu Z, Yip M, Hart LS, Maris JM, Chang HY, Giaccia AJ, Ye J. Acetate supplementation restores chromatin accessibility and promotes tumor cell differentiation under hypoxia. Cell Death Dis 2020; 11:102. [PMID: 32029721 PMCID: PMC7005271 DOI: 10.1038/s41419-020-2303-9] [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: 09/23/2019] [Revised: 01/23/2020] [Accepted: 01/23/2020] [Indexed: 12/11/2022]
Abstract
Despite the fact that Otto H. Warburg discovered the Warburg effect almost one hundred years ago, why cancer cells waste most of the glucose carbon as lactate remains an enigma. Warburg proposed a connection between the Warburg effect and cell dedifferentiation. Hypoxia is a common tumor microenvironmental stress that induces the Warburg effect and blocks tumor cell differentiation. The underlying mechanism by which this occurs is poorly understood, and no effective therapeutic strategy has been developed to overcome this resistance to differentiation. Using a neuroblastoma differentiation model, we discovered that hypoxia repressed cell differentiation through reducing cellular acetyl-CoA levels, leading to reduction of global histone acetylation and chromatin accessibility. The metabolic switch triggering this global histone hypoacetylation was the induction of pyruvate dehydrogenase kinases (PDK1 and PDK3). Inhibition of PDKs using dichloroacetate (DCA) restored acetyl-CoA generation and histone acetylation under hypoxia. Knocking down PDK1 induced neuroblastoma cell differentiation, highlighting the critical role of PDK1 in cell fate control. Importantly, acetate or glycerol triacetate (GTA) supplementation restored differentiation markers expression and neuron differentiation under hypoxia. Moreover, ATAC-Seq analysis demonstrated that hypoxia treatment significantly reduced chromatin accessibility at RAR/RXR binding sites, which can be restored by acetate supplementation. In addition, hypoxia-induced histone hypermethylation by increasing 2-hydroxyglutarate (2HG) and reducing α-ketoglutarate (αKG). αKG supplementation reduced histone hypermethylation upon hypoxia, but did not restore histone acetylation or differentiation markers expression. Together, these findings suggest that diverting pyruvate flux away from acetyl-CoA generation to lactate production is the key mechanism that Warburg effect drives dedifferentiation and tumorigenesis. We propose that combining differentiation therapy with acetate/GTA supplementation might represent an effective therapy against neuroblastoma.
Collapse
Affiliation(s)
- Yang Li
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Joshua J Gruber
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Ulrike M Litzenburger
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Yiren Zhou
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Yu Rebecca Miao
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Edward L LaGory
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Albert M Li
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Zhen Hu
- Olivia Consulting Service, Redwood City, CA, 94063, USA
| | - Michaela Yip
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Lori S Hart
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia and the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - John M Maris
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia and the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, 94305, USA
| | - Amato J Giaccia
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Jiangbin Ye
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
26
|
Wang Z, Cheng H, Xu H, Yu X, Sui D. A five-gene signature derived from m6A regulators to improve prognosis prediction of neuroblastoma. Cancer Biomark 2020; 28:275-284. [PMID: 32176634 DOI: 10.3233/cbm-191196] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND OBJECTIVE N6-methyladenosine (m6a) is the most abundant form of methylated modification in eukaryotic mRNA. However, the role of m6A-related genes in neuroblastoma (NB), one of the most common paediatric malignant tumours, is not well known. This study aimed to determine the prognostic role of m6A-related genes in neuroblastoma. METHODS We analysed the expression of 20 published m6A methylation regulators in 498 patients with NB from the Gene Expression Omnibus database. To determine the independent prognostic factors, we used univariate Cox analysis, the least absolute shrinkage and selection operator (LASSO) regression. The multivariate Cox analysis was used to construct a prognostic risk prediction model. 120 NB tissues from "Therapeutically Applicable Research To Generate Effective Treatments" (TARGET ) database was used to test the prognostic value. Gene set enrichment analysis was performed to discover the potential biological function of the m6A signature. RESULTS The risk prediction model consisted of five genes (METT14, WTAP, HNRNPC, YTHDF1 and IGF2BP2). The receiving operating characteristic curve showed the high exactitude of the risk model. Cox regression analysis revealed that the risk model was an independent prognostic factor of overall survival. These results were reproduced using another published independent dataset. Further functional enrichment analysis suggested the involvement of the 5-gene signature in several malignancies. CONCLUSION The five m6A regulatory genes identified in this study enable clinical prognosis of NB and may serve as novel therapeutic targets for NB.
Collapse
Affiliation(s)
- Zhichao Wang
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
- Department of Pediatric Surgery, First Hospital of Jilin University, Changchun, Jilin, China
| | - Huiyan Cheng
- Department of Gynecology and Obstetrics, First Hospital of Jilin University, Changchun, Jilin, China
| | - Huali Xu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| | - Xiaofeng Yu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| | - Dayun Sui
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| |
Collapse
|
27
|
Prajapati B, Fatma M, Fatima M, Khan MT, Sinha S, Seth PK. Identification of lncRNAs Associated With Neuroblastoma in Cross-Sectional Databases: Potential Biomarkers. Front Mol Neurosci 2019; 12:293. [PMID: 31920530 PMCID: PMC6920248 DOI: 10.3389/fnmol.2019.00293] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 11/14/2019] [Indexed: 12/31/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) have emerged as an important regulatory control in biological systems. Though the field of lncRNA has been progressing rapidly, a complete understanding of the role of lncRNAs in neuroblastoma pathogenesis is still lacking. To identify the abrogated lncRNAs in primary neuroblastoma and in the metastasized as well as the relapsed form of neuroblastoma, we analyzed an RNA-seq dataset on neuroblastoma that is available online to identify the lncRNAs that could potentially be contributing to the biology of neuroblastoma. The identified lncRNAs were further scrutinized using a publicly available epigenetic dataset of neuroblastoma and a cancer database. After this cross-sectional study, we were able to identify three significant lncRNAs, CASC15, PPP1R26-AS1, and USP3-AS1, which could serve as potential biomarkers in clinical studies of neuroblastoma pathogenesis.
Collapse
Affiliation(s)
| | - Mena Fatma
- National Brain Research Centre, Gurgaon, India
| | | | | | - Subrata Sinha
- National Brain Research Centre, Gurgaon, India.,Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | | |
Collapse
|
28
|
Epigenetic deregulation of GATA3 in neuroblastoma is associated with increased GATA3 protein expression and with poor outcomes. Sci Rep 2019; 9:18934. [PMID: 31831790 PMCID: PMC6908619 DOI: 10.1038/s41598-019-55382-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 11/19/2019] [Indexed: 01/04/2023] Open
Abstract
To discover epigenetic changes that may underly neuroblastoma pathogenesis, we identified differentially methylated genes in neuroblastoma cells compared to neural crest cells, the presumptive precursors cells for neuroblastoma, by using genome-wide DNA methylation analysis. We previously described genes that were hypermethylated in neuroblastoma; in this paper we report on 67 hypomethylated genes, which were filtered to select genes that showed transcriptional over-expression and an association with poor prognosis in neuroblastoma, highlighting GATA3 for detailed studies. Specific methylation assays confirmed the hypomethylation of GATA3 in neuroblastoma, which correlated with high expression at both the RNA and protein level. Demethylation with azacytidine in cultured sympathetic ganglia cells led to increased GATA3 expression, suggesting a mechanistic link between GATA3 expression and DNA methylation. Neuroblastomas that had completely absent GATA3 methylation and/or very high levels of protein expression, were associated with poor prognosis. Knock-down of GATA3 in neuroblastoma cells lines inhibited cell proliferation and increased apoptosis but had no effect on cellular differentiation. These results identify GATA3 as an epigenetically regulated component of the neuroblastoma transcriptional control network, that is essential for neuroblastoma proliferation. This suggests that the GATA3 transcriptional network is a promising target for novel neuroblastoma therapies.
Collapse
|
29
|
Ruan Y, Shi P, Lei Y, Weng S, Li S, Huang L, Lin X, Yao H. Polyvinyl butyral/graphene oxide nanocomposite modified electrode for the integrate determination of terminal metabolites of catecholamines in human urine. J Electroanal Chem (Lausanne) 2019. [DOI: 10.1016/j.jelechem.2019.113267] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
30
|
Qiao J, Rellinger EJ, Kim KW, Powers CM, Lee S, Correa H, Chung DH. Identification of α-N-catenin as a novel tumor suppressor in neuroblastoma. Oncotarget 2019; 10:5028-5040. [PMID: 31489113 PMCID: PMC6707940 DOI: 10.18632/oncotarget.27096] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 06/14/2019] [Indexed: 12/14/2022] Open
Abstract
The lost expression of α-catenin has been found in cancers, and reinstalling α-catenin inhibits tumor growth. Here we hypothesized that the α-N-catenin, a homologous member of α-catenin and neural-specific expressed, functions as a novel tumor suppressor in neural crest-derived tumor, neuroblastoma. We correlated CTNNA2 (encodes α-N-catenin) expression to neuroblastoma disease relapse-free survival probability using publicly accessible human neuroblastoma datasets in R2 platform. The result showed that it negatively correlated to relapse-free survival probability significantly in patients with neuroblastoma with non-MYCN amplified tumor. Conversely, overexpressing CTNNA2 suppressed the neuroblastoma cell proliferation as measuring by the clonogenesis, inhibited anchorage-independent growth with soft agar colony formation assay. Forced expression of CTNNA2 decreased cell migration and invasion. Further, overexpression of CTNNA2 reduced the secretion of angiogenic factor IL-8 and HUVEC tubule formation. Our results show, for the first time, that α-N-catenin is a tumor suppressor in neuroblastoma cells. These findings were further corroborated with in vivo tumor xenograft study, in which α-N-catenin inhibited tumor growth and reduced tumor blood vessel formation. Interestingly, this is only observed in SK-N-AS xenografts lacking MYCN expression, and not in BE(2)-C xenografts with MYCN amplification. Mechanistically, α-N-catenin attenuated NF-κB responsive genes by inhibiting NF-κB transcriptional activity. In conclusion, these data demonstrate that α-N-catenin is a tumor suppressor in non-MYCN-amplified neuroblastomas and it inhibits NF-κB signaling pathway to suppress tumor growth in human neuroblastomas. Therefore, restoring the expression of α-N-catenin can be a novel therapeutic approach for neuroblastoma patients who have the deletion of CTNNA2 and lack of MYCN amplification.
Collapse
Affiliation(s)
- Jingbo Qiao
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Eric J. Rellinger
- Department of Pediatric Surgery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Kwang Woon Kim
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Camille M. Powers
- Department of Pediatric Surgery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Sora Lee
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Hernan Correa
- Department of Pathology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Dai H. Chung
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
31
|
Applebaum MA, Barr EK, Karpus J, Nie J, Zhang Z, Armstrong AE, Uppal S, Sukhanova M, Zhang W, Chlenski A, Salwen HR, Wilkinson E, Dobratic M, Grossman R, Godley LA, Stranger BE, He C, Cohn SL. 5-Hydroxymethylcytosine Profiles Are Prognostic of Outcome in Neuroblastoma and Reveal Transcriptional Networks That Correlate With Tumor Phenotype. JCO Precis Oncol 2019; 3. [PMID: 31179414 PMCID: PMC6553657 DOI: 10.1200/po.18.00402] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
PURPOSE Whole-genome profiles of the epigenetic modification 5-hydroxymethylcytosine (5-hmC) are robust diagnostic biomarkers in adult patients with cancer. We investigated if 5-hmC profiles would serve as novel prognostic markers in neuroblastoma, a clinically heterogeneous pediatric cancer. Because this DNA modification facilitates active gene expression, we hypothesized that 5-hmC profiles would identify transcriptomic networks driving the clinical behavior of neuroblastoma. PATIENTS AND METHODS Nano-hmC-Seal sequencing was performed on DNA from Discovery (n = 51), Validation (n = 38), and Children’s Oncology Group (n = 20) cohorts of neuroblastoma tumors. RNA was isolated from 48 tumors for RNA sequencing. Genes with differential 5-hmC or expression between clusters were identified using DESeq2. A 5-hmC model predicting outcome in high-risk patients was established using linear discriminant analysis. RESULTS Comparison of low- versus high-risk tumors in the Discovery cohort revealed 577 genes with differential 5-hmC. Hierarchical clustering of tumors from the Discovery and Validation cohorts using these genes identified two main clusters highly associated with established prognostic markers, clinical risk group, and outcome. Genes with increased 5-hmC and expression in the favorable cluster were enriched for pathways of neuronal differentiation and KRAS activation, whereas genes involved in inflammation and the PRC2 complex were identified in the unfavorable cluster. The linear discriminant analysis model trained on high-risk Discovery cohort tumors was prognostic of outcome when applied to high-risk tumors from the Validation and Children’s Oncology Group cohorts (hazard ratio, 3.8). CONCLUSION 5-hmC profiles may be optimal DNA-based biomarkers in neuroblastoma. Analysis of transcriptional networks regulated by these epigenomic modifications may lead to a deeper understanding of drivers of neuroblastoma phenotype.
Collapse
Affiliation(s)
| | - Erin K Barr
- Texas Tech University Health Sciences, Lubbock, TX
| | | | - Ji Nie
- University of Chicago, Chicago, IL
| | | | | | | | | | - Wei Zhang
- Northwestern University, Chicago, IL
| | | | | | | | | | | | | | | | - Chuan He
- University of Chicago, Chicago, IL.,Howard Hughes Medical Institute, Chevy Chase, MD
| | | |
Collapse
|
32
|
5-Methylcytosine and 5-Hydroxymethylcytosine Signatures Underlying Pediatric Cancers. EPIGENOMES 2019; 3:epigenomes3020009. [PMID: 34968232 PMCID: PMC8594703 DOI: 10.3390/epigenomes3020009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 05/06/2019] [Indexed: 12/12/2022] Open
Abstract
In addition to the genetic variations, recent evidence has shown that DNA methylation of both 5-methylcytosine (5mC) and 5-hydroxymethylcytosine (5hmC) underlies the pathogenesis of pediatric cancer. Given the high mortality rate, there is an urgent need to study the mechanisms contributing to the pathogenicity of pediatric cancer. Over the past decades, next-generation sequencing (NGS) has enabled us to perform genome-wide screening to study the complex regulatory mechanisms of 5mC and 5hmC underlying pediatric tumorigenesis. To shed light on recent developments on pediatric cancer predisposition and tumor progression, here we discuss the role of both genome-wide and locus-specific dysregulation of 5mC and 5hmC in hematopoiesis malignancy and neuroblastoma, the most common types of pediatric cancer, together with their therapeutic potential.
Collapse
|
33
|
Li W, Cao J, Liu J, Chu W, Zhang C, Chen S, Kang Z. Downregulation of CDKL1 suppresses neuroblastoma cell proliferation, migration and invasion. Cell Mol Biol Lett 2019; 24:19. [PMID: 30891073 PMCID: PMC6407201 DOI: 10.1186/s11658-019-0139-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Accepted: 02/11/2019] [Indexed: 01/28/2023] Open
Abstract
Background Cyclin-dependent kinase-like 1 (CDKL1) is a member of the cell division control protein 2-related serine–threonine protein kinase family. It is known to occur in various malignant tumors, but its role in neuroblastoma (NB) remains unclear. Methods We constructed a CDKL1-silenced NB cell strain (SH-SY5Y) and used real-time PCR and western blotting to confirm the silencing. Functional analyses were performed using the MTT, colony-formation, FACS, wound-healing and transwell invasion assays. Results The expression of CDKL1 was significantly upregulated in NB tissue as compared to the adjacent normal tissue. CDKL1 knockdown significantly suppressed cell viability and colony formation ability. It also induced cell cycle G0/G1 phase arrest and apoptosis, and suppressed the migration and invasion ability of SH-SY5Y cells. CDKL1 knockdown decreased the CDK4, cyclin D1 and vimentin expression levels, and increased the caspase-3, PARP and E-cadherin expression levels in SH-SY5Y cells. Conclusions Our findings suggest that CDKL1 plays an important role in NB cell proliferation, migration and invasion. It might serve as a potential target for NB therapy.
Collapse
Affiliation(s)
- Weiyi Li
- 1Eye Hospital, China Academy of Chinese Medical Sciences, No 33 Lugu Road, Shijingshan district, Beijing, 100040 China
| | - Jing Cao
- Yinan Branch of Qilu Hospital of Shandong University, Linyi, Shandong China
| | - Jian Liu
- 1Eye Hospital, China Academy of Chinese Medical Sciences, No 33 Lugu Road, Shijingshan district, Beijing, 100040 China
| | - Wenli Chu
- 1Eye Hospital, China Academy of Chinese Medical Sciences, No 33 Lugu Road, Shijingshan district, Beijing, 100040 China
| | - Congqing Zhang
- 1Eye Hospital, China Academy of Chinese Medical Sciences, No 33 Lugu Road, Shijingshan district, Beijing, 100040 China
| | - Shuiling Chen
- 1Eye Hospital, China Academy of Chinese Medical Sciences, No 33 Lugu Road, Shijingshan district, Beijing, 100040 China
| | - Zefeng Kang
- 1Eye Hospital, China Academy of Chinese Medical Sciences, No 33 Lugu Road, Shijingshan district, Beijing, 100040 China
| |
Collapse
|
34
|
Huber K, Janoueix-Lerosey I, Kummer W, Rohrer H, Tischler AS. The sympathetic nervous system: malignancy, disease, and novel functions. Cell Tissue Res 2019; 372:163-170. [PMID: 29623426 DOI: 10.1007/s00441-018-2831-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Katrin Huber
- Department of Medicine, University of Fribourg, Route-Albert-Gockel 1, 1700, Fribourg, Switzerland.
| | - Isabelle Janoueix-Lerosey
- SIREDO Oncology Center (Care, Innovation and research for children and AYA with cancer), Inserm U830, PSL Research University, Equipe labellisée Ligue Nationale contre le cancer, Institut Curie, 26 rue d'Ulm, 75005, Paris, France
| | - Wolfgang Kummer
- Institute for Anatomy and Cell Biology, Justus Liebig University Giessen, Aulweg 123, 35385, Giessen, Germany
| | - Hermann Rohrer
- Institute for Clinical Neuroanatomy, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt/M, Germany
| | - Arthur S Tischler
- Department of Pathology and Laboratory Medicine, Tufts Medical Center and Tufts University School of Medicine, Boston, MA, 02111, USA
| |
Collapse
|
35
|
Li Y, Zhuo ZJ, Zhou H, Liu J, Xiao Z, Xiao Y, He J, Liu Z. miR-34b/c rs4938723 T>C Decreases Neuroblastoma Risk: A Replication Study in the Hunan Children. DISEASE MARKERS 2019; 2019:6514608. [PMID: 31583029 PMCID: PMC6754906 DOI: 10.1155/2019/6514608] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 07/30/2019] [Accepted: 08/13/2019] [Indexed: 02/07/2023]
Abstract
Neuroblastoma is the most common seen solid neural tumor in children less than age one. As mutation in the miR-34b/c gene is observed in several types of human malignancies, there likely to be similar events that contribute to the pathogenesis of neuroblastoma. We hypothesize that polymorphism in the miR-34b/c gene might predispose to neuroblastoma. Here, we conducted this replication study by genotyping rs4938723 T>C from miR-34b/c in Hunan children (162 subjects with neuroblastoma and 270 control subjects) and examined its effect on the risk of neuroblastoma. We determined such association using logistic regression, adjusted for age and gender. Relative to those with TT genotype, subjects with C allele had reduced neuroblastoma risk (TC vs. TT: adjusted OR = 0.46, 95%CI = 0.30-0.71; additive model: adjusted OR = 0.64, 95%CI = 0.47-0.88; TC/CC vs. TT: adjusted OR = 0.49, 95%CI = 0.33-0.73). Stratified analysis revealed that rs4938723 TC/CC carriers were less likely to develop neuroblastoma for patients in the subgroups of age ≤ 18 months, age > 18 months, females, males, tumors in retroperitoneal, tumors in other sites, and clinical stages II, III, IV, and III+IV. Our findings verified miR-34b/c rs4938723 C variant allele as a protective factor for the risk of neuroblastoma. Further investigation of how miR-34b/c rs4938723 T>C might modify neuroblastoma risk is warranted.
Collapse
Affiliation(s)
- Yong Li
- 1Department of Pediatric Surgery, Hunan Children's Hospital, Changsha, 410004 Hunan, China
| | - Zhen-Jian Zhuo
- 2Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623 Guangdong, China
| | - Haiyan Zhou
- 3Department of Pathology, Xiang-ya School of Medicine, Central South University, Changsha, 410013 Hunan, China
| | - Jiabin Liu
- 2Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623 Guangdong, China
| | - Zhenghui Xiao
- 4Emergency Center of Hunan Children's Hospital, Changsha, 410004 Hunan, China
| | - Yaling Xiao
- 1Department of Pediatric Surgery, Hunan Children's Hospital, Changsha, 410004 Hunan, China
| | - Jing He
- 2Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623 Guangdong, China
| | - Zan Liu
- 1Department of Pediatric Surgery, Hunan Children's Hospital, Changsha, 410004 Hunan, China
| |
Collapse
|
36
|
Li Y, Zhuo ZJ, Zhou H, Liu J, Zhang J, Cheng J, Zhou H, Li S, Li M, He J, Xiao Z, He J, Xiao Y. H19 gene polymorphisms and neuroblastoma susceptibility in Chinese children: a six-center case-control study. J Cancer 2019; 10:6358-6363. [PMID: 31772668 PMCID: PMC6856749 DOI: 10.7150/jca.37564] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 09/21/2019] [Indexed: 02/07/2023] Open
Abstract
Neuroblastoma is the most common seen solid tumor in children less than one year old. Given that polymorphisms in the lncRNA H19 gene are observed in several types of human malignancies, there likely to be similar events that contribute to the pathogenesis of neuroblastoma. We hypothesize that single nucleotide polymorphisms (SNPs) in the H19 gene might predispose to neuroblastoma. Here, we genotyped three SNPs (rs2839698 G>A, rs3024270 C>G, rs217727 G>A) from H19 gene in a Chinese population (700 subjects with neuroblastoma and 1516 control subjects) enrolled from six hospitals and examined the effect of individual and combined SNPs on the risk of neuroblastoma. Odds ratios (ORs) and 95% confidence intervals (CIs) calculated from logistic regression were adopted to assess such association, adjusted for age and gender. Among them, 700 controls and 1514 cases were successfully genotyped. None of these three SNPs were found to be relevant to the risk of neuroblastoma, either in overall analysis or stratification analysis. Findings from this study excluded the participation of lncRNA H19 gene SNPs in the risk of neuroblastoma. More independent case-control studies are encouraged to better elucidate this relationship.
Collapse
Affiliation(s)
- Yong Li
- Department of Pediatric Surgery, Hunan Children's Hospital, Changsha 410004, Hunan, China
| | - Zhen-Jian Zhuo
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Haiyan Zhou
- Department of Pathology, Xiang-ya School of Medicine, Central South University, Changsha 410013, Hunan, China
| | - Jiabin Liu
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Jiao Zhang
- Department of Pediatric Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Jiwen Cheng
- Department of Pediatric Surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi, China
| | - Haixia Zhou
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
| | - Suhong Li
- Department of Pathology, Children Hospital and Women Health Center of Shanxi, Taiyuan 030013, Shannxi, China
| | - Ming Li
- Department of Pediatric Surgery, Hunan Children's Hospital, Changsha 410004, Hunan, China
| | - Jun He
- Department of Pediatric Surgery, Hunan Children's Hospital, Changsha 410004, Hunan, China
| | - Zhenghui Xiao
- Emergency center of Hunan Children's Hospital, Changsha 410004, Hunan, China
| | - Jing He
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
- ✉ Corresponding authors: Yaling Xiao, Department of Pediatric Surgery, Hunan Children's Hospital, 86 Ziyuan Road, Changsha 410004, Hunan, China, ; or Jing He, Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Guangzhou 510623, Guangdong, China,
| | - Yaling Xiao
- Department of Pediatric Surgery, Hunan Children's Hospital, Changsha 410004, Hunan, China
- ✉ Corresponding authors: Yaling Xiao, Department of Pediatric Surgery, Hunan Children's Hospital, 86 Ziyuan Road, Changsha 410004, Hunan, China, ; or Jing He, Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Guangzhou 510623, Guangdong, China,
| |
Collapse
|
37
|
Novel Therapies for Relapsed and Refractory Neuroblastoma. CHILDREN-BASEL 2018; 5:children5110148. [PMID: 30384486 PMCID: PMC6262328 DOI: 10.3390/children5110148] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 10/23/2018] [Accepted: 10/23/2018] [Indexed: 12/17/2022]
Abstract
While recent increases in our understanding of the biology of neuroblastoma have allowed for more precise risk stratification and improved outcomes for many patients, children with high-risk neuroblastoma continue to suffer from frequent disease relapse, and despite recent advances in our understanding of neuroblastoma pathogenesis, the outcomes for children with relapsed neuroblastoma remain poor. These children with relapsed neuroblastoma, therefore, continue to need novel treatment strategies based on a better understanding of neuroblastoma biology to improve outcomes. The discovery of new tumor targets and the development of novel antibody- and cell-mediated immunotherapy agents have led to a large number of clinical trials for children with relapsed neuroblastoma, and additional clinical trials using molecular and genetic tumor profiling to target tumor-specific aberrations are ongoing. Combinations of these new therapeutic modalities with current treatment regimens will likely be needed to improve the outcomes of children with relapsed and refractory neuroblastoma.
Collapse
|
38
|
Genetic Predisposition to Neuroblastoma. CHILDREN-BASEL 2018; 5:children5090119. [PMID: 30200332 PMCID: PMC6162470 DOI: 10.3390/children5090119] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 08/22/2018] [Accepted: 08/28/2018] [Indexed: 02/07/2023]
Abstract
Neuroblastoma is the most common solid tumor in children under the age of one. It displays remarkable phenotypic heterogeneity, resulting in differences in outcomes that correlate with clinical and biologic features at diagnosis. While neuroblastoma accounts for approximately 5% of all cancer diagnoses in pediatrics, it disproportionately results in about 9% of all childhood deaths. Research advances over the decades have led to an improved understanding of neuroblastoma biology. However, the initiating events that lead to the development of neuroblastoma remain to be fully elucidated. It has only been recently that advances in genetics and genomics have allowed researchers to unravel the predisposing factors enabling the development of neuroblastoma and fully appreciate the interplay between the genetics of tumor and host. In this review, we outline the current understanding of familial neuroblastoma and highlight germline variations that predispose children to sporadic disease. We also discuss promising future directions in neuroblastoma genomic research and potential clinical applications for these advances.
Collapse
|