1
|
Sharma R, Yadav J, Bhat SA, Musayev A, Myrzagulova S, Sharma D, Padha N, Saini M, Tuli HS, Singh T. Emerging Trends in Neuroblastoma Diagnosis, Therapeutics, and Research. Mol Neurobiol 2025; 62:6423-6466. [PMID: 39804528 DOI: 10.1007/s12035-024-04680-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 12/20/2024] [Indexed: 03/29/2025]
Abstract
This review explores the current understanding and recent advancements in neuroblastoma, one of the most common extracranial solid pediatric cancers, accounting for ~ 15% of childhood cancer-related mortality. The hallmarks of NBL, including angiogenesis, metastasis, apoptosis resistance, cell cycle dysregulation, drug resistance, and responses to hypoxia and ROS, underscore its complex biology. The tumor microenvironment's significance in disease progression is acknowledged in this study, along with the pivotal role of cancer stem cells in sustaining tumor growth and heterogeneity. A number of molecular signatures are being studied in order to better understand the disease, with many of them serving as targets for the development of new therapeutics. This includes inhibitor therapies for NBL patients, which notably concentrate on ALK signaling, MDM2, PI3K/Akt/mTOR, Wnt, and RAS-MAPK pathways, along with regulators of epigenetic mechanisms. Additionally, this study offers an extensive understanding of the molecular therapies used, such as monoclonal antibodies and CAR-T therapy, focused on both preclinical and clinical studies. Radiation therapy's evolving role and the promise of stem cell transplantation-mediated interventions underscore the dynamic landscape of NBL treatment. This study has also emphasized the recent progress in the field of diagnosis, encompassing the adoption of artificial intelligence and liquid biopsy as a non-intrusive approach for early detection and ongoing monitoring of NBL. Furthermore, the integration of innovative treatment approaches such as CRISPR-Cas9, and cancer stem cell therapy has also been emphasized in this review.
Collapse
Affiliation(s)
- Rishabh Sharma
- Translational Oncology Laboratory, Department of Zoology, Hansraj College, Delhi University, New Delhi, 110007, India
- Amity Stem Cell Institute, Amity Medical School, Amity University, Haryana, 122412, India
| | - Jaya Yadav
- Translational Oncology Laboratory, Department of Zoology, Hansraj College, Delhi University, New Delhi, 110007, India
- Amity Stem Cell Institute, Amity Medical School, Amity University, Haryana, 122412, India
| | - Sajad Ahmad Bhat
- Asfendiyarov Kazakh National Medical University, Almaty, 050000, Kazakhstan
- Department of Biochemistry, NIMS University, Rajasthan, Jaipur, 303121, India
| | - Abdugani Musayev
- Asfendiyarov Kazakh National Medical University, Almaty, 050000, Kazakhstan
| | | | - Deepika Sharma
- Translational Oncology Laboratory, Department of Zoology, Hansraj College, Delhi University, New Delhi, 110007, India
| | - Nipun Padha
- Translational Oncology Laboratory, Department of Zoology, Hansraj College, Delhi University, New Delhi, 110007, India
- Department of Zoology, Cluster University of Jammu, Jammu, 180001, India
| | - Manju Saini
- Translational Oncology Laboratory, Department of Zoology, Hansraj College, Delhi University, New Delhi, 110007, India
- Amity Stem Cell Institute, Amity Medical School, Amity University, Haryana, 122412, India
| | - Hardeep Singh Tuli
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, Haryana, 133207, India
| | - Tejveer Singh
- Translational Oncology Laboratory, Department of Zoology, Hansraj College, Delhi University, New Delhi, 110007, India.
- Division of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences, (INMAS-DRDO), New Delhi, Delhi, 110054, India.
| |
Collapse
|
2
|
Yin H, Liu T, Wu D, Li X, Li G, Song W, Wang X, Xin S, Liu Y, Pan J. Exploring FAM13A-N-Myc interactions to uncover potential targets in MYCN-amplified neuroblastoma: a study of protein interactions and molecular dynamics simulations. BMC Cancer 2025; 25:470. [PMID: 40087586 PMCID: PMC11907995 DOI: 10.1186/s12885-025-13903-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 03/10/2025] [Indexed: 03/17/2025] Open
Abstract
Neuroblastoma (NB), a common infantile neuroendocrine tumor, presents a substantial therapeutic challenge when MYCN is amplified. Given that the protein structure of N-Myc is disordered, we utilized Alphafold for prediction and GROMACS for optimization of the N-Myc structure, thereby improving the reliability of the predicted structure. The publicly available datasets GSE49710 and GSE73517 were adopted, which contain the transcriptome data of clinical samples from 598 NB patients. Through various machine learning algorithms, FAM13A was identified as a characteristic gene of MYCN. Cell functional experiments, including those on cell proliferation, apoptosis, and cell cycle, also indicate that FAM13A is a potential risk factor. Additionally, Alphafold and GROMACS were employed to predict and optimize the structure of FAM13A. Protein-protein docking and molecular dynamic modeling techniques were then used to validate the enhanced protein stability resulting from the interaction between N-Myc and FAM13A. Consequently, targeting FAM13A holds the potential to reduce the stability of N-Myc, hinder the proliferation of NB cells, and increase the infiltration of immune cells. This multi-faceted approach effectively combats tumor cells, making FAM13A a prospective therapeutic target for MYCN-amplified NB.
Collapse
Affiliation(s)
- Hongli Yin
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215003, China.
| | - Tianyi Liu
- Department of Pharmaceutics, Dalian Women and Children's Medical Group, Dalian, Liaoning, 116012, China
| | - Di Wu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Xiaolu Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Gen Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Weiwei Song
- Department of Pharmacy, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Xiaodong Wang
- Department of Orthopaedics, Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Shan Xin
- Genetics and Cellular Engineering Group, Research Unit Signaling and Translation, Helmholtz Zentrum Munich, Ingolstaedter Landstr. 1, Neuherberg, 85764, Germany.
| | - Yisu Liu
- China College of Life Sciences and Chemistry, Hunan University of Technology, Zhuzhou, 412007, China.
| | - Jian Pan
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215003, China.
| |
Collapse
|
3
|
Murra N, Pommert NS, Schmidt B, Issa RS, Kaehler M, Bruckmueller H, Tim V, Cascorbi I, Waetzig V. Regulation and Function of CCL2 and N-Myc in Retinoic Acid-treated Neuroblastoma Cells. Cancer Genomics Proteomics 2025; 22:90-102. [PMID: 39730182 PMCID: PMC11696317 DOI: 10.21873/cgp.20490] [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: 09/29/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 12/29/2024] Open
Abstract
BACKGROUND/AIM Treatment with retinoic acid (RA) often promotes neuroblastoma differentiation and growth inhibition, including the suppression of the expression of the MYCN oncogene. However, RA also targets protumoral chemokines, such as CCL2, which may contribute to the development of resistance. The present study aimed to investigate the regulation and function of CCL2 and N-Myc in RA-treated neuroblastoma cells. MATERIALS AND METHODS In Kelly or SH-SY5Y cells, viability was quantified by cell fitness assays. Expression was analyzed using quantitative PCR and the regulation of proteins using enzyme-linked immunoabsorbent assays (ELISA) or western blots. RESULTS In MYCN-amplified Kelly cells, endogenous CCL2 levels were significantly lower compared to MYCN non-amplified SH-SY5Y cells. Treatment with 5 μM RA increased CCL2 release in both cell lines, but reduced N-Myc levels and cell numbers in Kelly cells. Over-expression of MYCN enhanced viability in SH-SY5Y cells, but did not affect RA-induced CCL2 release, while supplementation of CCL2 in Kelly cells did not prevent RA-mediated growth reduction. Impaired N-Myc or CCL2 signaling reduced the survival of all RA-treated cells and inhibition of N-Myc also decreased CCL2 levels. However, attenuated survival signaling was not generally associated with reduced levels of N-Myc or CCL2. Co-application of RA and the growth factor receptor inhibitors cediranib or crizotinib decreased N-Myc levels only in Kelly cells, while CCL2 release was dependent on the cell type and stimulus. CONCLUSION CCL2 and N-Myc promote the viability of RA-treated cells, although the levels of these mediators were not consistently correlated with cellular outcomes, especially during apoptotic signaling.
Collapse
Affiliation(s)
- Nanke Murra
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Nina Sophie Pommert
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Berit Schmidt
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Reema Sami Issa
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Meike Kaehler
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Henrike Bruckmueller
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Vera Tim
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Ingolf Cascorbi
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Vicki Waetzig
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| |
Collapse
|
4
|
Ustjanzew A, Nedwed AS, Sandhoff R, Faber J, Marini F, Paret C. Unraveling the glycosphingolipid metabolism by leveraging transcriptome-weighted network analysis on neuroblastic tumors. Cancer Metab 2024; 12:29. [PMID: 39449099 PMCID: PMC11515559 DOI: 10.1186/s40170-024-00358-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 10/11/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Glycosphingolipids (GSLs) are membrane lipids composed of a ceramide backbone linked to a glycan moiety. Ganglioside biosynthesis is a part of the GSL metabolism, which involves sequential reactions catalyzed by specific enzymes that in part have a poor substrate specificity. GSLs are deregulated in cancer, thus playing a role as potential biomarkers for personalized therapy or subtype classification. However, the analysis of GSL profiles is complex and requires dedicated technologies, that are currently not included in the commonly utilized high-throughput assays adopted in contexts such as molecular tumor boards. METHODS In this study, we developed a method to discriminate the enzyme activity among the four series of the ganglioside metabolism pathway by incorporating transcriptome data and topological information of the metabolic network. We introduced three adjustment options for reaction activity scores (RAS) and demonstrated their application in both exploratory and comparative analyses by applying the method on neuroblastic tumors (NTs), encompassing neuroblastoma (NB), ganglioneuroblastoma (GNB), and ganglioneuroma (GN). Furthermore, we interpreted the results in the context of earlier published GSL measurements in the same tumors. RESULTS By adjusting RAS values using a weighting scheme based on network topology and transition probabilities (TPs), the individual series of ganglioside metabolism can be differentiated, enabling a refined analysis of the GSL profile in NT entities. Notably, the adjustment method we propose reveals the differential engagement of the ganglioside series between NB and GNB. Moreover, MYCN gene expression, a well-known prognostic marker in NTs, appears to correlate with the expression of therapeutically relevant gangliosides, such as GD2. Using unsupervised learning, we identified subclusters within NB based on altered GSL metabolism. CONCLUSION Our study demonstrates the utility of adjusting RAS values in discriminating ganglioside metabolism subtypes, highlighting the potential for identifying novel cancer subgroups based on sphingolipid profiles. These findings contribute to a better understanding of ganglioside dysregulation in NT and may have implications for stratification and targeted therapeutic strategies in these tumors and other tumor entities with a deregulated GSL metabolism.
Collapse
Affiliation(s)
- Arsenij Ustjanzew
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, 55131, Germany.
| | - Annekathrin Silvia Nedwed
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, 55131, Germany
| | - Roger Sandhoff
- Lipid Pathobiochemistry, German Cancer Research Center, Heidelberg, 69120, Germany
| | - Jörg Faber
- Department of Pediatric Hematology/Oncology, Center for Pediatric and Adolescent Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, 55131, Germany
- University Cancer Center (UCT), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, 55131, Germany
| | - Federico Marini
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, 55131, Germany
- Research Center for Immunotherapy (FZI), Mainz, 55131, Germany
| | - Claudia Paret
- Department of Pediatric Hematology/Oncology, Center for Pediatric and Adolescent Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, 55131, Germany
- University Cancer Center (UCT), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, 55131, Germany
- Research Center for Immunotherapy (FZI), Mainz, 55131, Germany
| |
Collapse
|
5
|
Maher S, Wynne K, Zhernovkov V, Halasz M. A temporal (phospho-)proteomic dataset of neurotrophic receptor tyrosine kinase signalling in neuroblastoma. Sci Data 2024; 11:1111. [PMID: 39389992 PMCID: PMC11467210 DOI: 10.1038/s41597-024-03965-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 10/02/2024] [Indexed: 10/12/2024] Open
Abstract
Neurotrophic receptor tyrosine kinases (TrkA, TrkB, TrkC), despite their homology, contribute to the clinical heterogeneity of the childhood cancer neuroblastoma. TrkA expression is associated with low-stage disease and is often seen with spontaneous tumour regression. Conversely, TrkB is present in unfavourable neuroblastomas that often harbour amplification of the MYCN oncogene. The role of TrkC is less clearly defined, although some studies suggest its association with a favourable outcome. Understanding the differences in activity of Trk receptors that drive divergent clinical phenotypes as well as the influence of MYCN amplification on downstream Trk receptor signalling remains poorly understood. Here, we present a comprehensive label-free mass spectrometry-based total proteomics and phosphoproteomics dataset (432 raw files with FragPipe search outputs; available on PRIDE with accession number PXD054441) where we identified and quantified 4,907 proteins, 16,744 phosphosites and 5,084 phosphoproteins, derived from NGF/BDNF/NT-3 treated TrkA/B/C-overexpressing neuroblastoma cells with differential MYCN status. Analysing our dataset offers valuable insights into TrkA/B/C receptor signalling in neuroblastoma and its modulation by MYCN status; and holds potential for advancing therapeutic strategies in this challenging childhood cancer.
Collapse
Affiliation(s)
- Stephanie Maher
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin, Ireland
| | - Kieran Wynne
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Vadim Zhernovkov
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin, Ireland
| | - Melinda Halasz
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin, Ireland.
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland.
| |
Collapse
|
6
|
Ryl T, Afanasyeva E, Hartmann T, Schwermer M, Schneider M, Schröder C, Wagemanns M, Bister A, Kanber D, Steenpass L, Schramm K, Jones B, Jones DTW, Biewald E, Astrahantseff K, Hanenberg H, Rahmann S, Lohmann DR, Schramm A, Ketteler P. A MYCN-driven de-differentiation profile identifies a subgroup of aggressive retinoblastoma. Commun Biol 2024; 7:919. [PMID: 39079981 PMCID: PMC11289481 DOI: 10.1038/s42003-024-06596-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 07/17/2024] [Indexed: 08/02/2024] Open
Abstract
Retinoblastoma are childhood eye tumors arising from retinal precursor cells. Two distinct retinoblastoma subtypes with different clinical behavior have been described based on gene expression and methylation profiling. Using consensus clustering of DNA methylation analysis from 61 retinoblastomas, we identify a MYCN-driven cluster of subtype 2 retinoblastomas characterized by DNA hypomethylation and high expression of genes involved in protein synthesis. Subtype 2 retinoblastomas outside the MYCN-driven cluster are characterized by high expression of genes from mesodermal development, including NKX2-5. Knockdown of MYCN expression in retinoblastoma cell models causes growth arrest and reactivates a subtype 1-specific photoreceptor signature. These molecular changes suggest that removing the driving force of MYCN oncogenic activity rescues molecular circuitry driving subtype 1 biology. The MYCN-RB gene signature generated from the cell models better identifies MYCN-driven retinoblastoma than MYCN amplification and can identify cases that may benefit from MYCN-targeted therapy. MYCN drives tumor progression in a molecularly defined retinoblastoma subgroup, and inhibiting MYCN activity could restore a more differentiated and less aggressive tumor biology.
Collapse
Affiliation(s)
- Tatsiana Ryl
- Department of Pediatric Hematology and Oncology, University Hospital Essen, Essen, Germany
| | - Elena Afanasyeva
- Department of Pediatric Hematology and Oncology, University Hospital Essen, Essen, Germany
| | - Till Hartmann
- Algorithms for Reproducible Bioinformatics, Genome Informatics, Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Melanie Schwermer
- Department of Pediatric Hematology and Oncology, University Hospital Essen, Essen, Germany
| | - Markus Schneider
- Department of Pediatric Hematology and Oncology, University Hospital Essen, Essen, Germany
| | - Christopher Schröder
- Algorithms for Reproducible Bioinformatics, Genome Informatics, Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Maren Wagemanns
- Department of Pediatric Hematology and Oncology, University Hospital Essen, Essen, Germany
| | - Arthur Bister
- Department of Pediatric Hematology and Oncology, University Hospital Essen, Essen, Germany
| | - Deniz Kanber
- Institute of Human Genetics, University Hospital Essen, University Duisburg Essen, Essen, Germany
| | - Laura Steenpass
- Human and Animal Cell Lines, Leibniz Institute DSMZ German Collection of Microorganisms and Cell Cultures, 38124, Braunschweig, Germany
| | - Kathrin Schramm
- Division of Pediatric Glioma Research, Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, University of Heidelberg, Heidelberg, Germany
| | - Barbara Jones
- Division of Pediatric Glioma Research, Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, University of Heidelberg, Heidelberg, Germany
| | - David T W Jones
- Division of Pediatric Glioma Research, Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, University of Heidelberg, Heidelberg, Germany
| | - Eva Biewald
- Department of Ophthalmology, Medical Faculty, University of Duisburg-Essen, 45147, Essen, Germany
| | - Kathy Astrahantseff
- Department of Pediatric Oncology and Hematology, Charité - University Medicine Berlin, Berlin, Germany
| | - Helmut Hanenberg
- Department of Pediatric Hematology and Oncology, University Hospital Essen, Essen, Germany
| | - Sven Rahmann
- Algorithmic Bioinformatics, Center for Bioinformatics Saar and Saarland University, Saarland Informatics Campus, Saarbrücken, Germany
| | - Dietmar R Lohmann
- Institute of Human Genetics, University Hospital Essen, University Duisburg Essen, Essen, Germany
| | - Alexander Schramm
- Laboratory for Molecular Oncology, Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Petra Ketteler
- Department of Pediatric Hematology and Oncology, University Hospital Essen, Essen, Germany.
- Institute of Human Genetics, University Hospital Essen, University Duisburg Essen, Essen, Germany.
| |
Collapse
|
7
|
Romiani A, Simonsson K, Pettersson D, Al-Awar A, Rassol N, Bakr H, Lind D, Umapathy G, Spetz J, Palmer R, Hallberg B, Helou K, Forssell-Aronsson E. Comparison of 177Lu-octreotate and 177Lu-octreotide for treatment in human neuroblastoma-bearing mice. Heliyon 2024; 10:e31409. [PMID: 38826727 PMCID: PMC11141386 DOI: 10.1016/j.heliyon.2024.e31409] [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: 02/16/2024] [Revised: 05/01/2024] [Accepted: 05/15/2024] [Indexed: 06/04/2024] Open
Abstract
Background Patients with high-risk neuroblastoma (NB) have a 5-year event-free survival of less than 50 %, and novel and improved treatment options are needed. Radiolabeled somatostatin analogs (SSTAs) could be a treatment option. The aims of this work were to compare the biodistribution and the therapeutic effects of 177Lu-octreotate and 177Lu-octreotide in mice bearing the human CLB-BAR NB cell line, and to evaluate their regulatory effects on apoptosis-related genes. Methods The biodistribution of 177Lu-octreotide in mice bearing CLB-BAR tumors was studied at 1, 24, and 168 h after administration, and the absorbed dose was estimated to tumor and normal tissues. Further, animals were administered different amounts of 177Lu-octreotate or 177Lu-octreotide. Tumor volume was measured over time and compared to a control group given saline. RNA was extracted from tumors, and the expression of 84 selected genes involved in apoptosis was quantified with qPCR. Results The activity concentration was generally lower in most tissues for 177Lu-octreotide compared to 177Lu-octreotate. Mean absorbed dose per administered activity to tumor after injection of 1.5 MBq and 15 MBq was 0.74 and 0.03 Gy/MBq for 177Lu-octreotide and 2.9 and 0.45 Gy/MBq for 177Lu-octreotate, respectively. 177Lu-octreotide treatment resulted in statistically significant differences compared to controls. Fractionated administration led to a higher survival fraction than after a single administration. The pro-apoptotic genes TNSFS8, TNSFS10, and TRADD were regulated after administration with 177Lu-octreotate. Treatment with 177Lu-octreotide yielded regulation of the pro-apoptotic genes CASP5 and TRADD, and of the anti-apoptotic gene IL10 as well as the apoptosis-related gene TNF. Conclusion 177Lu-octreotide gave somewhat better anti-tumor effects than 177Lu-octreotate. The similar effect observed in the treated groups with 177Lu-octreotate suggests saturation of the somatostatin receptors. Pronounced anti-tumor effects following fractionated administration merited receptor saturation as an explanation. The gene expression analyses suggest apoptosis activation through the extrinsic pathway for both radiopharmaceuticals.
Collapse
Affiliation(s)
- A. Romiani
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - K. Simonsson
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - D. Pettersson
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - A. Al-Awar
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - N. Rassol
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - H. Bakr
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Medical Physics and Biomedical Engineering, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - D.E. Lind
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - G. Umapathy
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - J. Spetz
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - R.H. Palmer
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - B. Hallberg
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - K. Helou
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - E. Forssell-Aronsson
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Medical Physics and Biomedical Engineering, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
8
|
Hussein R, Abou-Shanab AM, Badr E. A multi-omics approach for biomarker discovery in neuroblastoma: a network-based framework. NPJ Syst Biol Appl 2024; 10:52. [PMID: 38760476 PMCID: PMC11101461 DOI: 10.1038/s41540-024-00371-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 04/16/2024] [Indexed: 05/19/2024] Open
Abstract
Neuroblastoma (NB) is one of the leading causes of cancer-associated death in children. MYCN amplification is a prominent genetic marker for NB, and its targeting to halt NB progression is difficult to achieve. Therefore, an in-depth understanding of the molecular interactome of NB is needed to improve treatment outcomes. Analysis of NB multi-omics unravels valuable insight into the interplay between MYCN transcriptional and miRNA post-transcriptional modulation. Moreover, it aids in the identification of various miRNAs that participate in NB development and progression. This study proposes an integrated computational framework with three levels of high-throughput NB data (mRNA-seq, miRNA-seq, and methylation array). Similarity Network Fusion (SNF) and ranked SNF methods were utilized to identify essential genes and miRNAs. The specified genes included both miRNA-target genes and transcription factors (TFs). The interactions between TFs and miRNAs and between miRNAs and their target genes were retrieved where a regulatory network was developed. Finally, an interaction network-based analysis was performed to identify candidate biomarkers. The candidate biomarkers were further analyzed for their potential use in prognosis and diagnosis. The candidate biomarkers included three TFs and seven miRNAs. Four biomarkers have been previously studied and tested in NB, while the remaining identified biomarkers have known roles in other types of cancer. Although the specific molecular role is yet to be addressed, most identified biomarkers possess evidence of involvement in NB tumorigenesis. Analyzing cellular interactome to identify potential biomarkers is a promising approach that can contribute to optimizing efficient therapeutic regimens to target NB vulnerabilities.
Collapse
Affiliation(s)
- Rahma Hussein
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Ahmed M Abou-Shanab
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Eman Badr
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12578, Egypt.
- Faculty of Computers and Artificial Intelligence, Cairo University, Giza, 12613, Egypt.
| |
Collapse
|
9
|
Hu X, Zhou Y, Hill C, Chen K, Cheng C, Liu X, Duan P, Gu Y, Wu Y, Ewing RM, Li Z, Wu Z, Wang Y. Identification of MYCN non-amplified neuroblastoma subgroups points towards molecular signatures for precision prognosis and therapy stratification. Br J Cancer 2024; 130:1841-1854. [PMID: 38553589 PMCID: PMC7616008 DOI: 10.1038/s41416-024-02666-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 03/13/2024] [Accepted: 03/19/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUND Despite the extensive study of MYCN-amplified neuroblastomas, there is a significant unmet clinical need in MYCN non-amplified cases. In particular, the extent of heterogeneity within the MYCN non-amplified population is unknown. METHODS A total of 1566 samples from 16 datasets were identified in Gene Expression Omnibus (GEO) and ArrayExpress. Characterisation of the subtypes was analysed by ConsensusClusterPlus. Independent predictors for subgrouping were constructed from the single sample predictor based on the multiclassPairs package. Findings were verified using immunohistochemistry and CIBERSORTx analysis. RESULTS We demonstrate that MYCN non-amplified neuroblastomas are heterogeneous and can be classified into 3 subgroups based on their transcriptional signatures. Within these groups, subgroup_2 has the worst prognosis and this group shows a 'MYCN' signature that is potentially induced by the overexpression of Aurora Kinase A (AURKA); whilst subgroup_3 is characterised by an 'inflamed' gene signature. The clinical implications of this subtype classification are significant, as each subtype demonstrates a unique prognosis and vulnerability to investigational therapies. A total of 420 genes were identified as independent subgroup predictors with average balanced accuracy of 0.93 and 0.84 for train and test datasets, respectively. CONCLUSION We propose that transcriptional subtyping may enhance precision prognosis and therapy stratification for patients with MYCN non-amplified neuroblastomas.
Collapse
Affiliation(s)
- Xiaoxiao Hu
- Department of Paediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200092, China
- Department of Paediatric Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Division of Paediatric Oncology, Shanghai Institute of Paediatric Research, Shanghai, 200092, China
| | - Yilu Zhou
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
- Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Charlotte Hill
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Kai Chen
- Department of Paediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200092, China
- Division of Paediatric Oncology, Shanghai Institute of Paediatric Research, Shanghai, 200092, China
| | - Cheng Cheng
- Department of Paediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200092, China
- Division of Paediatric Oncology, Shanghai Institute of Paediatric Research, Shanghai, 200092, China
| | - Xiaowei Liu
- Department of Paediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200092, China
- Division of Paediatric Oncology, Shanghai Institute of Paediatric Research, Shanghai, 200092, China
| | - Peiwen Duan
- Department of Paediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200092, China
- Division of Paediatric Oncology, Shanghai Institute of Paediatric Research, Shanghai, 200092, China
| | - Yaoyao Gu
- Department of Paediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200092, China
- Division of Paediatric Oncology, Shanghai Institute of Paediatric Research, Shanghai, 200092, China
| | - Yeming Wu
- Department of Paediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200092, China
- Division of Paediatric Oncology, Shanghai Institute of Paediatric Research, Shanghai, 200092, China
- Department of Paediatric Surgery, Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Rob M Ewing
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
- Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Zhongrong Li
- Department of Paediatric Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Zhixiang Wu
- Department of Paediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200092, China.
- Division of Paediatric Oncology, Shanghai Institute of Paediatric Research, Shanghai, 200092, China.
- Department of Paediatric Surgery, Children's Hospital of Soochow University, Suzhou, 215003, China.
| | - Yihua Wang
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK.
- Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK.
| |
Collapse
|
10
|
Zugbi S, Aschero R, Ganiewich D, Cancela MB, Winter U, Ottaviani D, Sampor C, Dinardi M, Torbidoni AV, Mena M, Balaguer-Lluna L, Lamas G, Sgroi M, Lagomarsino E, Lubieniecki F, Fandiño A, Radvanyi F, Abramson DH, Podhajcer O, Llera AS, Cafferata EG, Chantada G, Carcaboso AM, Schaiquevich P. Establishment and Comprehensive Characterization of a Novel Preclinical Platform of Metastatic Retinoblastoma for Therapeutic Developments. Invest Ophthalmol Vis Sci 2023; 64:27. [PMID: 38117242 PMCID: PMC10741097 DOI: 10.1167/iovs.64.15.27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 11/20/2023] [Indexed: 12/21/2023] Open
Abstract
Purpose Although there have been improvements in the management of metastatic retinoblastoma, most patients do not survive, and all patients suffer from multiple short- and long-term treatment toxicities. Reliable and informative models to assist clinicians are needed. Thus we developed and comprehensively characterized a novel preclinical platform of primary cell cultures and xenograft models of metastatic retinoblastoma to provide insights into the molecular biology underlying metastases and to perform drug screening for the identification of hit candidates with the highest potential for clinical translation. Methods Orbital tumor, bone marrow, cerebrospinal fluid, and lymph node tumor infiltration specimens were obtained from seven patients with metastatic retinoblastoma at diagnosis, disease progression, or relapse. Tumor specimens were engrafted in immunodeficient animals, and primary cell lines were established. Genomic, immunohistochemical/immunocytochemical, and pharmacological analysis were performed. Results We successfully established five primary cell lines: two derived from leptomeningeal, two from orbital, and one from lymph node tumor dissemination. After the intravitreal or intraventricular inoculation of these cells, we established cell-derived xenograft models. Both primary cell lines and xenografts accurately retained the histological and genomic features of the tumors from which they were derived and faithfully recapitulated the dissemination patterns and pharmacological sensitivity observed in the matched patients. Conclusions Ours is an innovative and thoroughly characterized preclinical platform of metastatic retinoblastoma developed for the understanding of tumor biology of this highly aggressive tumor and has the potential to identify drug candidates to treat patients who currently lack effective treatment options.
Collapse
Affiliation(s)
- Santiago Zugbi
- Innovative Treatments Unit, Hospital de Pediatría JP Garrahan, Buenos Aires, Argentina
- National Scientific and Technical Research Council, CONICET, Buenos Aires, Argentina
| | - Rosario Aschero
- National Scientific and Technical Research Council, CONICET, Buenos Aires, Argentina
- Pathology Service, Hospital de Pediatría JP Garrahan, Buenos Aires, Argentina
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Deu, Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| | - Daiana Ganiewich
- Laboratory of Molecular and Cellular Therapy, Instituto Leloir – Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA), Buenos Aires, Argentina
| | - María B. Cancela
- Innovative Treatments Unit, Hospital de Pediatría JP Garrahan, Buenos Aires, Argentina
- National Scientific and Technical Research Council, CONICET, Buenos Aires, Argentina
| | - Ursula Winter
- Innovative Treatments Unit, Hospital de Pediatría JP Garrahan, Buenos Aires, Argentina
- Pathology Service, Hospital de Pediatría JP Garrahan, Buenos Aires, Argentina
| | - Daniela Ottaviani
- Institut Curie; PSL Research University, Centre National de la Recherche Scientifique (CNRS); Equipe Ligue contre le cancer, Paris, France
| | - Claudia Sampor
- Hematology-Oncology Service, Hospital de Pediatría JP Garrahan, Buenos Aires, Argentina
| | - Milagros Dinardi
- Innovative Treatments Unit, Hospital de Pediatría JP Garrahan, Buenos Aires, Argentina
| | - Ana V. Torbidoni
- Innovative Treatments Unit, Hospital de Pediatría JP Garrahan, Buenos Aires, Argentina
- National Scientific and Technical Research Council, CONICET, Buenos Aires, Argentina
| | - Marcela Mena
- Laboratory of Molecular and Cellular Therapy, Instituto Leloir – Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA), Buenos Aires, Argentina
| | - Leire Balaguer-Lluna
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Deu, Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| | - Gabriela Lamas
- Pathology Service, Hospital de Pediatría JP Garrahan, Buenos Aires, Argentina
| | - Mariana Sgroi
- Ophthalmology Service, Hospital de Pediatría JP Garrahan, Buenos Aires, Argentina
| | - Eduardo Lagomarsino
- Pharmacy Service, Hospital de Pediatría JP Garrahan, Buenos Aires, Argentina
| | - Fabiana Lubieniecki
- Pathology Service, Hospital de Pediatría JP Garrahan, Buenos Aires, Argentina
| | - Adriana Fandiño
- Ophthalmology Service, Hospital de Pediatría JP Garrahan, Buenos Aires, Argentina
| | - François Radvanyi
- Institut Curie; PSL Research University, Centre National de la Recherche Scientifique (CNRS); Equipe Ligue contre le cancer, Paris, France
| | - David H. Abramson
- Ophthalmic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York, United States
| | - Osvaldo Podhajcer
- National Scientific and Technical Research Council, CONICET, Buenos Aires, Argentina
- Laboratory of Molecular and Cellular Therapy, Instituto Leloir – Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA), Buenos Aires, Argentina
| | - Andrea S. Llera
- National Scientific and Technical Research Council, CONICET, Buenos Aires, Argentina
- Laboratory of Molecular and Cellular Therapy, Instituto Leloir – Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA), Buenos Aires, Argentina
| | - Eduardo G. Cafferata
- National Scientific and Technical Research Council, CONICET, Buenos Aires, Argentina
- Laboratory of Molecular and Cellular Therapy, Instituto Leloir – Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA), Buenos Aires, Argentina
| | - Guillermo Chantada
- National Scientific and Technical Research Council, CONICET, Buenos Aires, Argentina
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Deu, Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| | - Angel M. Carcaboso
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Deu, Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| | - Paula Schaiquevich
- Innovative Treatments Unit, Hospital de Pediatría JP Garrahan, Buenos Aires, Argentina
- National Scientific and Technical Research Council, CONICET, Buenos Aires, Argentina
| |
Collapse
|
11
|
Chaudhry KA, Jacobi JJ, Gillard BM, Karasik E, Martin JC, da Silva Fernandes T, Hurley E, Feltri ML, Attwood KM, Twist CJ, Smiraglia DJ, Long MD, Bianchi-Smiraglia A. Aryl hydrocarbon receptor is a tumor promoter in MYCN-amplified neuroblastoma cells through suppression of differentiation. iScience 2023; 26:108303. [PMID: 38026169 PMCID: PMC10654598 DOI: 10.1016/j.isci.2023.108303] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/25/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Neuroblastoma is the most common extracranial solid tumor in children. MYCN amplification is detected in almost half of high-risk cases and is associated with poorly differentiated tumors, poor patient prognosis and poor response to therapy, including retinoids. We identify the aryl hydrocarbon receptor (AhR) as a transcription factor promoting the growth and suppressing the differentiation of MYCN-amplified neuroblastoma. A neuroblastoma specific AhR transcriptional signature reveals an inverse correlation of AhR activity with patients' outcome, suggesting AhR activity is critical for disease progression. AhR modulates chromatin structures, reducing accessibility to regions responsive to retinoic acid. Genetic and pharmacological inhibition of AhR results in induction of differentiation. Importantly, AhR antagonism with clofazimine synergizes with retinoic acid in inducing differentiation both in vitro and in vivo. Thus, we propose AhR as a target for MYCN-amplified neuroblastoma and that its antagonism, combined with current standard-of-care, may result in a more durable response in patients.
Collapse
Affiliation(s)
- Kanita A. Chaudhry
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Justine J. Jacobi
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Bryan M. Gillard
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Ellen Karasik
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Jeffrey C. Martin
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | | | - Edward Hurley
- Department of Biochemistry and Neurology, Institute for Myelin and Glia Exploration, State University of New York at Buffalo, Buffalo, NY, USA
| | - Maria Laura Feltri
- Department of Biochemistry and Neurology, Institute for Myelin and Glia Exploration, State University of New York at Buffalo, Buffalo, NY, USA
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Foundation I.R.C.C.S. Carlo Besta Neurological Institute Milan, Italy
| | - Kristopher M. Attwood
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Clare J. Twist
- Department of Pediatric Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Dominic J. Smiraglia
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Mark D. Long
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Anna Bianchi-Smiraglia
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| |
Collapse
|
12
|
Obata H, Tsuji AB, Sudo H, Sugyo A, Hashiya K, Ikeda H, Itoh M, Minegishi K, Nagatsu K, Ogawa M, Bando T, Sugiyama H, Zhang MR. Novel Auger-Electron-Emitting 191Pt-Labeled Pyrrole-Imidazole Polyamide Targeting MYCN Increases Cytotoxicity and Cytosolic dsDNA Granules in MYCN-Amplified Neuroblastoma. Pharmaceuticals (Basel) 2023; 16:1526. [PMID: 38004392 PMCID: PMC10675227 DOI: 10.3390/ph16111526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/24/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023] Open
Abstract
Auger electrons can cause nanoscale physiochemical damage to specific DNA sites that play a key role in cancer cell survival. Radio-Pt is a promising Auger-electron source for damaging DNA efficiently because of its ability to bind to DNA. Considering that the cancer genome is maintained under abnormal gene amplification and expression, here, we developed a novel 191Pt-labeled agent based on pyrrole-imidazole polyamide (PIP), targeting the oncogene MYCN amplified in human neuroblastoma, and investigated its targeting ability and damaging effects. A conjugate of MYCN-targeting PIP and Cys-(Arg)3-coumarin was labeled with 191Pt via Cys (191Pt-MYCN-PIP) with a radiochemical purity of >99%. The binding potential of 191Pt-MYCN-PIP was evaluated via the gel electrophoretic mobility shift assay, suggesting that the radioagent bound to the DNA including the target sequence of the MYCN gene. In vitro assays using human neuroblastoma cells showed that 191Pt-MYCN-PIP bound to DNA efficiently and caused DNA damage, decreasing MYCN gene expression and MYCN signals in in situ hybridization analysis, as well as cell viability, especially in MYCN-amplified Kelly cells. 191Pt-MYCN-PIP also induced a substantial increase in cytosolic dsDNA granules and generated proinflammatory cytokines, IFN-α/β, in Kelly cells. Tumor uptake of intravenously injected 191Pt-MYCN-PIP was low and its delivery to tumors should be improved for therapeutic application. The present results provided a potential strategy, targeting the key oncogenes for cancer survival for Auger electron therapy.
Collapse
Affiliation(s)
- Honoka Obata
- Department of Advanced Nuclear Medicine Sciences, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan; (H.O.)
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
- Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-ku, Sapporo 060-0812, Japan
| | - Atsushi B. Tsuji
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Hitomi Sudo
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Aya Sugyo
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Kaori Hashiya
- Department of Chemistry, Graduate School of Science, Kyoto University, Kitashirakawa-oiwakecho, Sakyo-ku, Kyoto 606-8502, Japan
| | - Hayato Ikeda
- Cyclotron and Radioisotope Center (CYRIC), Tohoku University, Sendai 980-8578, Japan
- Research Center for Electron Photon Science (ELPH), Tohoku University, Sendai 982-0826, Japan
| | - Masatoshi Itoh
- Cyclotron and Radioisotope Center (CYRIC), Tohoku University, Sendai 980-8578, Japan
| | - Katsuyuki Minegishi
- Department of Advanced Nuclear Medicine Sciences, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan; (H.O.)
| | - Kotaro Nagatsu
- Department of Advanced Nuclear Medicine Sciences, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan; (H.O.)
| | - Mikako Ogawa
- Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-ku, Sapporo 060-0812, Japan
| | - Toshikazu Bando
- Department of Chemistry, Graduate School of Science, Kyoto University, Kitashirakawa-oiwakecho, Sakyo-ku, Kyoto 606-8502, Japan
| | - Hiroshi Sugiyama
- Institute for Integrated Cell-Material Science (iCeMS), Kyoto University, Yoshida-ushinomiyacho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan; (H.O.)
| |
Collapse
|
13
|
Zhang X, Cong X, Jin X, Liu Y, Zhang T, Fan X, Shi X, Zhang X, Wang X, Yang YG, Dai X. Deficiency of BAP1 inhibits neuroblastoma tumorigenesis through destabilization of MYCN. Cell Death Dis 2023; 14:504. [PMID: 37543638 PMCID: PMC10404282 DOI: 10.1038/s41419-023-06030-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 07/17/2023] [Accepted: 07/31/2023] [Indexed: 08/07/2023]
Abstract
The transcription factor MYCN is frequently amplified and overexpressed in a variety of cancers including high-risk neuroblastoma (NB) and promotes tumor cell proliferation, survival, and migration. Therefore, MYCN is being pursued as an attractive therapeutic target for selective inhibition of its upstream regulators because MYCN is considered a "undruggable" target. Thus, it is important to explore the upstream regulators for the transcription and post-translational modification of MYCN. Here, we report that BRCA1-associated protein-1 (BAP1) promotes deubiquitination and subsequent stabilization of MYCN by directly binding to MYCN protein. Furthermore, BAP1 knockdown inhibits NB tumor cells growth and migration in vitro and in vivo, which can be rescued partially by ectopic expression of MYCN. Importantly, depletion of BAP1 confers cellular resistance to bromodomain and extraterminal (BET) protein inhibitor JQ1 and Aurora A kinase inhibitor Alisertib. Furthermore, IHC results of NB tissue array confirmed the positive correlation between BAP1 and MYCN protein. Altogether, our work not only uncovers an oncogenic function of BAP1 by stabilizing MYCN, but also reveals a critical mechanism for the post-translational regulation of MYCN in NB. Our findings further indicate that BAP1 could be a potential therapeutic target for MYCN-amplified neuroblastoma.
Collapse
Affiliation(s)
- Xiaoling Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, China.
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital, Jilin University, Changchun, China.
| | - Xianling Cong
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xiangting Jin
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital, Jilin University, Changchun, China
| | - Yu'e Liu
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai, China
| | - Tong Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital, Jilin University, Changchun, China
| | - Xinyuan Fan
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital, Jilin University, Changchun, China
| | - Xiyao Shi
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital, Jilin University, Changchun, China
| | - Xiaoying Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital, Jilin University, Changchun, China
| | - Xue Wang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital, Jilin University, Changchun, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, China.
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital, Jilin University, Changchun, China.
- International Center of Future Science, Jilin University, Changchun, China.
| | - Xiangpeng Dai
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, China.
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital, Jilin University, Changchun, China.
| |
Collapse
|
14
|
Obata H, Ogawa M, Zalutsky MR. DNA Repair Inhibitors: Potential Targets and Partners for Targeted Radionuclide Therapy. Pharmaceutics 2023; 15:1926. [PMID: 37514113 PMCID: PMC10384049 DOI: 10.3390/pharmaceutics15071926] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
The present review aims to explore the potential targets/partners for future targeted radionuclide therapy (TRT) strategies, wherein cancer cells often are not killed effectively, despite receiving a high average tumor radiation dose. Here, we shall discuss the key factors in the cancer genome, especially those related to DNA damage response/repair and maintenance systems for escaping cell death in cancer cells. To overcome the current limitations of TRT effectiveness due to radiation/drug-tolerant cells and tumor heterogeneity, and to make TRT more effective, we propose that a promising strategy would be to target the DNA maintenance factors that are crucial for cancer survival. Considering their cancer-specific DNA damage response/repair ability and dysregulated transcription/epigenetic system, key factors such as PARP, ATM/ATR, amplified/overexpressed transcription factors, and DNA methyltransferases have the potential to be molecular targets for Auger electron therapy; moreover, their inhibition by non-radioactive molecules could be a partnering component for enhancing the therapeutic response of TRT.
Collapse
Affiliation(s)
- Honoka Obata
- Department of Advanced Nuclear Medicine Sciences, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
- Departments of Radiology and Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
- Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-ku, Sapporo 060-0812, Japan
| | - Mikako Ogawa
- Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-ku, Sapporo 060-0812, Japan
| | - Michael R Zalutsky
- Departments of Radiology and Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
15
|
Cuesta-Rubio O, Monzote L, Fernández-Acosta R, Pardo-Andreu GL, Rastrelli L. A review of nemorosone: Chemistry and biological properties. PHYTOCHEMISTRY 2023; 210:113674. [PMID: 37044362 DOI: 10.1016/j.phytochem.2023.113674] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 06/19/2023]
Abstract
Nemorosone is a bicyclic polyprenylated acylphloroglucinol derivative originally isolated from Clusia spp. and it can be obtained through chemical synthesis employing different synthetic strategies. Since its discovery, it has attracted great attention both from a biological and chemical viewpoint. In the present article, we attempted to review various chemical and biological topics around nemorosone, with an emphasis on its antiproliferative activities. For this purpose, relevant data was collected from different scientific databases including Google Scholar, PubMed, Scopus and ISI Web of Knowledge. This natural compound has shown activity against several types of malignancies such as leukemia, human colorectal, pancreatic, and breast cancer because it modulates multiple molecular pathways. Nemorosone has both cytostatic and cytotoxic activity and it also seems to induce apoptosis and ferroptosis. Additionally, it has antimicrobial capabilities against Gram-positive bacteria and parasites belonging to genus Leishmania. Its promising antiproliferative pre-clinical effects deserve further attention for anticancer and anti-parasitic drug development and translation to the clinic.
Collapse
Affiliation(s)
- Osmany Cuesta-Rubio
- Universidad Técnica de Machala, Facultad de Ciencias Químicas y de la Salud, Ave. Panamericana km 5½, 070101, Machala, Ecuador.
| | - Lianet Monzote
- Departamento de Parasitología, Instituto de Medicina Tropical Pedro Kourí, Autopista Novia del Mediodía Km 6 1/2, 11400, La Habana, Cuba.
| | - Roberto Fernández-Acosta
- Department of Pharmacy, Institute of Pharmaceutical and Food Sciences, University of Havana, 222 St. # 2317, La Coronela, 13600, Havana, Cuba.
| | - Gilberto Lázaro Pardo-Andreu
- Center for Research and Biological Evaluation, Institute of Pharmaceutical and Food Sciences, University of Havana, 222 St. # 2317, 13600, Havana, Cuba.
| | - Luca Rastrelli
- Universitá degli Studi di Salerno, Dipartimento di Farmacia, Via Giovanni Paolo II, 84084, Fisciano, SA, Italy; NBFC, National Biodiversity Future Center, Palermo, 90133, Italy.
| |
Collapse
|
16
|
Seven ES, Kirbas Cilingir E, Bartoli M, Zhou Y, Sampson R, Shi W, Peng Z, Ram Pandey R, Chusuei CC, Tagliaferro A, Vanni S, Graham RM, Seven YB, Leblanc RM. Hydrothermal vs microwave nanoarchitechtonics of carbon dots significantly affects the structure, physicochemical properties, and anti-cancer activity against a specific neuroblastoma cell line. J Colloid Interface Sci 2023; 630:306-321. [PMID: 36244103 DOI: 10.1016/j.jcis.2022.10.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/15/2022] [Accepted: 10/03/2022] [Indexed: 11/11/2022]
Abstract
Carbon dots (CDs) from glucose were synthesized using two of the most common bottom-up methods, namely, microwave assisted (MW) and hydrothermal carbonization (HT). Synthetic parameters such as reaction time, temperature, and precursor concentration were changed to study the effects of each parameter on CD size, structure, surface functionalities, charge, photoluminescence behavior, quantum yield, cytotoxicity, blood-brain barrier (BBB) crossing ability and bioimaging. A detailed analysis is performed to compare the structure and properties of the CDs synthesized in ten different conditions. We show that the synthesis route drastically changes the structure, properties, and related functions of glucose-derived CDs yielding two different subtypes of CDs. Surprisingly, CDs that was synthesized via HT method showed specific anticancer activity against a neuroblastoma cell line while being non-toxic towards healthy cell lines, indicating significant potential for therapeutic applications. CDs synthesized via MW crosses the BBB in zebrafish and rat models, and accumulates in neurons. CDs synthesized via MW method showed high biocompatibility and a great potential to be used for bioimaging applications in vitro and in vivo targeting neurons. Finally, a formation mechanism of CDs is proposed for both HT and MW synthesis routes.
Collapse
Affiliation(s)
- Elif S Seven
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, FL 33146, United States; C-Dots Nanotec, LLC, United States
| | - Emel Kirbas Cilingir
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, FL 33146, United States
| | - Mattia Bartoli
- Department of Applied Science and Technology, Politecnico di Torino, Italy
| | - Yiqun Zhou
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, FL 33146, United States; C-Dots Nanotec, LLC, United States
| | | | - Wenquan Shi
- School of Materials Science and Engineering, Yunnan Key Laboratory for Micro/Nano Materials & Technology, Yunnan University, Kunming, Yunnan 650091, China
| | - Zhili Peng
- School of Materials Science and Engineering, Yunnan Key Laboratory for Micro/Nano Materials & Technology, Yunnan University, Kunming, Yunnan 650091, China
| | - Raja Ram Pandey
- Department of Chemistry, Middle Tennessee State University, Murfreesboro, TN 37132, United States
| | - Charles C Chusuei
- Department of Chemistry, Middle Tennessee State University, Murfreesboro, TN 37132, United States
| | | | - Steven Vanni
- Department of Neurological Surgery, Miller School of Medicine, University of Miami, 1095 NW 14th Terrace, Miami, FL 33136, United States; HCA Florida University Hospital, 3476 S University Dr., Davie, FL 33328, United States; Department of Medicine, Dr. Kiran C. Patel College of Allopathic Medicine, Davie, FL, 33328, United States
| | - Regina M Graham
- Department of Neurological Surgery, Miller School of Medicine, University of Miami, 1095 NW 14th Terrace, Miami, FL 33136, United States; Sylvester Comprehensive Cancer Center, University of Miami Health System, Miami, FL 33136, United States
| | - Yasin B Seven
- Department of Physical Therapy, University of Florida, 101 Newell Dr., Gainesville, FL 32603, United States; McKnight Brain Institute, University of Florida, 1149 Newell Dr., Gainesville, FL 32610, United States
| | - Roger M Leblanc
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, FL 33146, United States
| |
Collapse
|
17
|
Shi X, Wang Y, Zhang L, Zhao W, Dai X, Yang YG, Zhang X. Targeting bromodomain and extra-terminal proteins to inhibit neuroblastoma tumorigenesis through regulating MYCN. Front Cell Dev Biol 2022; 10:1021820. [PMID: 36187481 PMCID: PMC9523081 DOI: 10.3389/fcell.2022.1021820] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 09/01/2022] [Indexed: 11/13/2022] Open
Abstract
Bromodomain and extra-terminal domain (BET) family proteins play important roles in regulating the expression of multiple proto-oncogenes by recognizing acetylation of histones and non-histone proteins including transcription factors, which subsequently promote tumor cell proliferation, survival, metastasis and immune escape. Therefore, BET family proteins are considered attractive therapeutic targets in various cancers. Currently, blocking of the BET proteins is a widely used therapeutic strategy for MYCN amplified high-risk neuroblastoma. Here, we summarized and reviewed the recent research progresses for the critical function of BET proteins, as an epigenetic reader, on tumorigenesis and the therapeutic potential of the BET/BRD4 inhibitors on MYCN amplified neuroblastoma. We also discussed the combined therapeutic strategies for BET inhibitor-resistant neuroblastoma.
Collapse
|
18
|
Bartolucci D, Montemurro L, Raieli S, Lampis S, Pession A, Hrelia P, Tonelli R. MYCN Impact on High-Risk Neuroblastoma: From Diagnosis and Prognosis to Targeted Treatment. Cancers (Basel) 2022; 14:4421. [PMID: 36139583 PMCID: PMC9496712 DOI: 10.3390/cancers14184421] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/05/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022] Open
Abstract
Among childhood cancers, neuroblastoma is the most diffuse solid tumor and the deadliest in children. While to date, the pathology has become progressively manageable with a significant increase in 5-year survival for its less aggressive form, high-risk neuroblastoma (HR-NB) remains a major issue with poor outcome and little survivability of patients. The staging system has also been improved to better fit patient needs and to administer therapies in a more focused manner in consideration of pathology features. New and improved therapies have been developed; nevertheless, low efficacy and high toxicity remain a staple feature of current high-risk neuroblastoma treatment. For this reason, more specific procedures are required, and new therapeutic targets are also needed for a precise medicine approach. In this scenario, MYCN is certainly one of the most interesting targets. Indeed, MYCN is one of the most relevant hallmarks of HR-NB, and many studies has been carried out in recent years to discover potent and specific inhibitors to block its activities and any related oncogenic function. N-Myc protein has been considered an undruggable target for a long time. Thus, many new indirect and direct approaches have been discovered and preclinically evaluated for the interaction with MYCN and its pathways; a few of the most promising approaches are nearing clinical application for the investigation in HR-NB.
Collapse
Affiliation(s)
| | - Luca Montemurro
- Pediatric Oncology and Hematology Unit, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | | | | | - Andrea Pession
- Pediatric Unit, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Patrizia Hrelia
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Roberto Tonelli
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| |
Collapse
|
19
|
Belhajova M, Podhorska N, Vicha A, Eckschlager T. KDM5B expression in cisplatin resistant neuroblastoma cell lines. Oncol Lett 2022; 24:365. [DOI: 10.3892/ol.2022.13485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 07/21/2022] [Indexed: 11/06/2022] Open
Affiliation(s)
- Marie Belhajova
- Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University and Motol University Hospital, 15006 Prague, Czech Republic
| | - Natalia Podhorska
- Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University and Motol University Hospital, 15006 Prague, Czech Republic
| | - Ales Vicha
- Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University and Motol University Hospital, 15006 Prague, Czech Republic
| | - Tomas Eckschlager
- Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University and Motol University Hospital, 15006 Prague, Czech Republic
| |
Collapse
|
20
|
Abstract
"De novo" genes evolve from previously non-genic DNA. This strikes many of us as remarkable, because it seems extraordinarily unlikely that random sequence would produce a functional gene. How is this possible? In this two-part review, I first summarize what is known about the origins and molecular functions of the small number of de novo genes for which such information is available. I then speculate on what these examples may tell us about how de novo genes manage to emerge despite what seem like enormous opposing odds.
Collapse
Affiliation(s)
- Caroline M Weisman
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
21
|
Ponzoni M, Bachetti T, Corrias MV, Brignole C, Pastorino F, Calarco E, Bensa V, Giusto E, Ceccherini I, Perri P. Recent advances in the developmental origin of neuroblastoma: an overview. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:92. [PMID: 35277192 PMCID: PMC8915499 DOI: 10.1186/s13046-022-02281-w] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 02/06/2022] [Indexed: 02/04/2023]
Abstract
Neuroblastoma (NB) is a pediatric tumor that originates from neural crest-derived cells undergoing a defective differentiation due to genomic and epigenetic impairments. Therefore, NB may arise at any final site reached by migrating neural crest cells (NCCs) and their progeny, preferentially in the adrenal medulla or in the para-spinal ganglia. NB shows a remarkable genetic heterogeneity including several chromosome/gene alterations and deregulated expression of key oncogenes that drive tumor initiation and promote disease progression. NB substantially contributes to childhood cancer mortality, with a survival rate of only 40% for high-risk patients suffering chemo-resistant relapse. Hence, NB remains a challenge in pediatric oncology and the need of designing new therapies targeted to specific genetic/epigenetic alterations become imperative to improve the outcome of high-risk NB patients with refractory disease or chemo-resistant relapse. In this review, we give a broad overview of the latest advances that have unraveled the developmental origin of NB and its complex epigenetic landscape. Single-cell RNA sequencing with spatial transcriptomics and lineage tracing have identified the NCC progeny involved in normal development and in NB oncogenesis, revealing that adrenal NB cells transcriptionally resemble immature neuroblasts or their closest progenitors. The comparison of adrenal NB cells from patients classified into risk subgroups with normal sympatho-adrenal cells has highlighted that tumor phenotype severity correlates with neuroblast differentiation grade. Transcriptional profiling of NB tumors has identified two cell identities that represent divergent differentiation states, i.e. undifferentiated mesenchymal (MES) and committed adrenergic (ADRN), able to interconvert by epigenetic reprogramming and to confer intra-tumoral heterogeneity and high plasticity to NB. Chromatin immunoprecipitation sequencing has disclosed the existence of two super-enhancers and their associated transcription factor networks underlying MES and ADRN identities and controlling NB gene expression programs. The discovery of NB-specific regulatory circuitries driving oncogenic transformation and maintaining the malignant state opens new perspectives on the design of innovative therapies targeted to the genetic and epigenetic determinants of NB. Remodeling the disrupted regulatory networks from a dysregulated expression, which blocks differentiation and enhances proliferation, toward a controlled expression that prompts the most differentiated state may represent a promising therapeutic strategy for NB.
Collapse
Affiliation(s)
- Mirco Ponzoni
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, Via G. Gaslini 5, 16147, Genoa, Italy
| | - Tiziana Bachetti
- U.O. Proteomica e Spettrometria di Massa, IRCSS Ospedale Policlinico San Martino, Genoa, Italy
| | - Maria Valeria Corrias
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, Via G. Gaslini 5, 16147, Genoa, Italy
| | - Chiara Brignole
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, Via G. Gaslini 5, 16147, Genoa, Italy
| | - Fabio Pastorino
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, Via G. Gaslini 5, 16147, Genoa, Italy
| | - Enzo Calarco
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, Via G. Gaslini 5, 16147, Genoa, Italy
| | - Veronica Bensa
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, Via G. Gaslini 5, 16147, Genoa, Italy
| | - Elena Giusto
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, Via G. Gaslini 5, 16147, Genoa, Italy
| | - Isabella Ceccherini
- Laboratory of Genetics and Genomics of Rare Diseases, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Patrizia Perri
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, Via G. Gaslini 5, 16147, Genoa, Italy.
| |
Collapse
|
22
|
CO 2 pneumoperitoneum effects on proliferation and apoptosis in two different neuroblastoma cell lines. Pediatr Surg Int 2022; 38:457-464. [PMID: 35083505 DOI: 10.1007/s00383-022-05063-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/05/2022] [Indexed: 10/19/2022]
Abstract
PURPOSE The proto-oncogene MYCN is considered a transcription factor involved in the regulation of neuroblastoma (NB) cell biology. Since minimally invasive-surgery represents a debated treatment of NB, we investigated CO2 effects on proliferative activity and apoptotic pathway in two NB cell lines, SH-SY5Y (MYCN-non-amplified) and IMR-32 (MYCN-amplified). METHODS SH-SY5Y and IMR-32 were exposed to CO2 (100%) at a pressure of 15 mmHg for 4 h and then moved to normal condition for 24 h. Cell proliferation, caspase 3 activity and transcript levels of BAX, BCL-2, cyclin B, cyclin D and MMP-2 were evaluated. RESULTS CO2 exposure caused a decrease in cell proliferation associated to increases in BAX/BCL-2 ratio and caspase 3 activity in SH-SY5Y, while opposite effects have been found in IMR-32. CO2 exposure induced a decrease of cyclin B1 in SH-SY5Y, while an increase in cyclin B1 and D1 was observed in IMR-32. A slight up-regulation of MMP-2 expression in SH-SY5Y and a significant increase of 2.2 folds in IMR-32 was observed (p < 0.05). CONCLUSIONS Our results suggest that CO2 exposure may cause different effects on various NB cell lines, likely due to MYCN amplification status. Further in vitro and in vivo studies are needed to highlight the role of laparoscopy on NB behaviour.
Collapse
|
23
|
Hattori N, Asada K, Miyajima N, Mori A, Nakanishi Y, Kimura K, Wakabayashi M, Takeshima H, Nitani C, Hara J, Ushijima T. Combination of a synthetic retinoid and a DNA demethylating agent induced differentiation of neuroblastoma through retinoic acid signal reprogramming. Br J Cancer 2021; 125:1647-1656. [PMID: 34635821 DOI: 10.1038/s41416-021-01571-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 09/16/2021] [Accepted: 09/29/2021] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND The CpG island methylator phenotype of neuroblastoma (NBL) is strongly associated with poor prognosis and can be targeted by 5-aza-2'-deoxycytidine (5-aza-dC). Differentiation therapy is a standard maintenance therapy for high-risk NBLs. However, the in vivo effect of tamibarotene, a synthetic retinoic acid, and the efficacy of its combination with 5-aza-dC have not been studied. Here, we conducted a preclinical study to assess the in vivo tamibarotene effect and the combination. METHODS Treatment effects were analysed by in vitro cell growth and differentiation state and by in vivo xenograft suppression. Demethylated genes were analysed by DNA methylation microarrays and geneset enrichment. RESULTS Tamibarotene monotherapy induced neural extension and upregulation of differentiation markers of NBL cells in vitro, and tumour regression without severe side effects in vivo. 5-Aza-dC monotherapy suppressed tumour growth both in vitro and in vivo, and induced demethylation of genes related to nervous system development and function. Pre-treatment with 5-aza-dC in vitro enhanced upregulation of differentiation markers and genes involved in retinoic acid signaling. Pre-treatment with 5-aza-dC in vivo significantly suppressed tumour growth and reduced the variation in tumour sizes. CONCLUSIONS Epigenetic drug-based differentiation therapy using 5-aza-dC and TBT is a promising strategy for refractory NBLs.
Collapse
Affiliation(s)
- Naoko Hattori
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan.
| | - Kiyoshi Asada
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Nozomu Miyajima
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Akiko Mori
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Yoko Nakanishi
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Kana Kimura
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Mika Wakabayashi
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Hideyuki Takeshima
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Chika Nitani
- Department of Pediatric Hematology and Oncology, Osaka City General Hospital, Osaka, Japan
| | - Junichi Hara
- Department of Pediatric Hematology and Oncology, Osaka City General Hospital, Osaka, Japan
| | - Toshikazu Ushijima
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan.
| |
Collapse
|
24
|
Tagalakis AD, Jayarajan V, Maeshima R, Ho KH, Syed F, Wu L, Aldossary AM, Munye MM, Mistry T, Ogunbiyi OK, Sala A, Standing JF, Moghimi SM, Stoker AW, Hart SL. Integrin-Targeted, Short Interfering RNA Nanocomplexes for Neuroblastoma Tumor-Specific Delivery Achieve MYCN Silencing with Improved Survival. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2104843. [PMID: 35712226 PMCID: PMC9178728 DOI: 10.1002/adfm.202104843] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Indexed: 06/15/2023]
Abstract
The authors aim to develop siRNA therapeutics for cancer that can be administered systemically to target tumors and retard their growth. The efficacy of systemic delivery of siRNA to tumors with nanoparticles based on lipids or polymers is often compromised by their rapid clearance from the circulation by the liver. Here, multifunctional cationic and anionic siRNA nanoparticle formulations are described, termed receptor-targeted nanocomplexes (RTNs), that comprise peptides for siRNA packaging into nanoparticles and receptor-mediated cell uptake, together with lipids that confer nanoparticles with stealth properties to enhance stability in the circulation, and fusogenic properties to enhance endosomal release within the cell. Intravenous administration of RTNs in mice leads to predominant accumulation in xenograft tumors, with very little detected in the liver, lung, or spleen. Although non-targeted RTNs also enter the tumor, cell uptake appears to be RGD peptide-dependent indicating integrin-mediated uptake. RTNs with siRNA against MYCN (a member of the Myc family of transcription factors) in mice with MYCN-amplified neuroblastoma tumors show significant retardation of xenograft tumor growth and enhanced survival. This study shows that RTN formulations can achieve specific tumor-targeting, with minimal clearance by the liver and so enable delivery of tumor-targeted siRNA therapeutics.
Collapse
Affiliation(s)
- Aristides D. Tagalakis
- Department of Genetics and Genomic MedicineUCL Great Ormond Street Institute of Child HealthUniversity College London30 Guilford StreetLondonWC1N 1EHUK
- Present address:
Department of BiologyEdge Hill UniversityOrmskirkL39 4QPUK
| | - Vignesh Jayarajan
- Department of Genetics and Genomic MedicineUCL Great Ormond Street Institute of Child HealthUniversity College London30 Guilford StreetLondonWC1N 1EHUK
| | - Ruhina Maeshima
- Department of Genetics and Genomic MedicineUCL Great Ormond Street Institute of Child HealthUniversity College London30 Guilford StreetLondonWC1N 1EHUK
| | - Kin H. Ho
- Department of InflammationInfection and ImmunityUCL Great Ormond Street Institute of Child HealthUniversity College London30 Guilford StreetLondonWC1N 1EHUK
| | - Farhatullah Syed
- Department of InflammationInfection and ImmunityUCL Great Ormond Street Institute of Child HealthUniversity College London30 Guilford StreetLondonWC1N 1EHUK
| | - Lin‐Ping Wu
- Centre for Pharmaceutical Nanotechnology and NanotoxicologyFaculty of Health and Medical SciencesUniversity of CopenhagenUniversitetsparken 2Copenhagen2100Denmark
- Present address:
Guangzhou institute of Biomedicine and HealthChinese Academy of SciencesGuangzhou510530People's Republic of China
| | - Ahmad M. Aldossary
- Department of Genetics and Genomic MedicineUCL Great Ormond Street Institute of Child HealthUniversity College London30 Guilford StreetLondonWC1N 1EHUK
- Present address:
National Center for BiotechnologyKing Abdulaziz City for Science and TechnologyRiyadh11442Saudi Arabia
| | - Mustafa M. Munye
- Department of Genetics and Genomic MedicineUCL Great Ormond Street Institute of Child HealthUniversity College London30 Guilford StreetLondonWC1N 1EHUK
- Present address:
Cell and Gene Therapy Catapult12th Floor Tower Wing, Guy's Hospital, Great Maze PondLondonSE1 9RTUK
| | - Talisa Mistry
- Department of HistopathologyGreat Ormond Street Hospital for ChildrenNHS Foundation TrustLondonWC1N 3JHUK
| | - Olumide Kayode Ogunbiyi
- Department of HistopathologyGreat Ormond Street Hospital for ChildrenNHS Foundation TrustLondonWC1N 3JHUK
| | - Arturo Sala
- Department of Life SciencesBrunel University LondonKingston LaneMiddlesexUB8 3PHUK
| | - Joseph F. Standing
- Department of InflammationInfection and ImmunityUCL Great Ormond Street Institute of Child HealthUniversity College London30 Guilford StreetLondonWC1N 1EHUK
| | - Seyed M. Moghimi
- Centre for Pharmaceutical Nanotechnology and NanotoxicologyFaculty of Health and Medical SciencesUniversity of CopenhagenUniversitetsparken 2Copenhagen2100Denmark
- Present address:
School of Pharmacy, and Translational and Clinical Research Institute, the Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneNE1 7RUUK
- Present address:
Colorado Center for Nanomedicine and Nanosafety, Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of Colorado Anschutz Medical CampusAuroraCO80045USA
| | - Andrew W. Stoker
- Department of Developmental Biology and CancerUCL Great Ormond Street Institute of Child HealthUniversity College London30 Guilford StreetLondonWC1N 1EHUK
| | - Stephen L. Hart
- Department of Genetics and Genomic MedicineUCL Great Ormond Street Institute of Child HealthUniversity College London30 Guilford StreetLondonWC1N 1EHUK
| |
Collapse
|
25
|
El-Soussi S, Hanna R, Semaan H, Khater AR, Abdallah J, Abou-Kheir W, Abou-Antoun T. A Novel Therapeutic Mechanism of Imipridones ONC201/ONC206 in MYCN-Amplified Neuroblastoma Cells via Differential Expression of Tumorigenic Proteins. Front Pediatr 2021; 9:693145. [PMID: 34422720 PMCID: PMC8373200 DOI: 10.3389/fped.2021.693145] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 06/22/2021] [Indexed: 01/07/2023] Open
Abstract
Neuroblastoma is the most common extracranial nervous system tumor in children. It presents with a spectrum of clinical prognostic measures ranging from benign growths that regress spontaneously to highly malignant, treatment evasive tumors affiliated with increased mortality rates. MYCN amplification is commonly seen in high-risk neuroblastoma, rendering it highly malignant and recurrence prone. In our current study, we investigated the therapeutic potential of small molecule inducers of TRAIL, ONC201, and ONC206 in MYCN-amplified IMR-32 and non-MYCN-amplified SK-N-SH human neuroblastoma cell lines. Our results exhibit potent antitumor activity of ONC201 and ONC206 via a novel inhibition of EGF-induced L1CAM and PDGFRβ phosphorylation in both cell lines. Drug treatment significantly reduced cellular proliferation, viability, migration, invasion, tumorsphere formation potential, and increased apoptosis in both cell lines. The protein expression of tumorigenic NMYC, Sox-2, Oct-4, FABP5, and HMGA1 significantly decreased 48 h post-drug treatment, whereas cleaved PARP1/caspase-3 and γH2AX increased 72 h post-drug treatment, compared with vehicle-treated cells in the MYCN-amplified IMR-32 cell line. We are the first to report this novel differential protein expression after ONC201 or ONC206 treatment in human neuroblastoma cells, demonstrating an important multitarget effect which may yield added therapeutic benefits in treating this devastating childhood cancer.
Collapse
Affiliation(s)
- Sarra El-Soussi
- Shool of Pharmacy, Lebanese American University, Byblos, Lebanon
| | - Reine Hanna
- Shool of Pharmacy, Lebanese American University, Byblos, Lebanon
| | - Hanna Semaan
- Shool of Pharmacy, Lebanese American University, Byblos, Lebanon
- Faculty of Sciences, Lebanese University, Fanar, Lebanon
| | | | - Jad Abdallah
- Shool of Pharmacy, Lebanese American University, Byblos, Lebanon
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | | |
Collapse
|
26
|
Guo YF, Duan JJ, Wang J, Li L, Wang D, Liu XZ, Yang J, Zhang HR, Lv J, Yang YJ, Yang ZY, Cai J, Liao XM, Tang T, Huang TT, Wu F, Yang XY, Wen Q, Bian XW, Yu SC. Inhibition of the ALDH18A1-MYCN positive feedback loop attenuates MYCN-amplified neuroblastoma growth. Sci Transl Med 2021; 12:12/531/eaax8694. [PMID: 32075946 DOI: 10.1126/scitranslmed.aax8694] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 01/28/2020] [Indexed: 12/14/2022]
Abstract
MYCN-amplified neuroblastoma (NB) is characterized by poor prognosis, and directly targeting MYCN has proven challenging. Here, we showed that aldehyde dehydrogenase family 18 member A1 (ALDH18A1) exerts profound impacts on the proliferation, self-renewal, and tumorigenicity of NB cells and is a potential risk factor in patients with NB, especially those with MYCN amplification. Mechanistic studies revealed that ALDH18A1 could both transcriptionally and posttranscriptionally regulate MYCN expression, with MYCN reciprocally transactivating ALDH18A1 and thus forming a positive feedback loop. Using molecular docking and screening, we identified an ALDH18A1-specific inhibitor, YG1702, and demonstrated that pharmacological inhibition of ALDH18A1 was sufficient to induce a less proliferative phenotype and confer tumor regression and prolonged survival in NB xenograft models, providing therapeutic insights into the disruption of this reciprocal regulatory loop in MYCN-amplified NB.
Collapse
Affiliation(s)
- Yu-Feng Guo
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jiang-Jie Duan
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jun Wang
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Lin Li
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Di Wang
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xun-Zhou Liu
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jing Yang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Hua-Rong Zhang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jing Lv
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yong-Jun Yang
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Ze-Yu Yang
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jiao Cai
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xue-Mei Liao
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Tao Tang
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Ting-Ting Huang
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Feng Wu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xian-Yan Yang
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Qian Wen
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xiu-Wu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China. .,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Shi-Cang Yu
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China. .,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| |
Collapse
|
27
|
Zhang R, Fang YF, Wu DM, Lin Y, Zhang B, Liu MK, Bai JX, Chen F. Comparison of the Efficacy of Minimally Invasive and Open Surgery on Children with Neuroblastoma: A Meta-Analysis. J Laparoendosc Adv Surg Tech A 2021; 31:829-838. [PMID: 34076504 DOI: 10.1089/lap.2020.0618] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Purpose: Evaluate the clinical efficacy and safety of minimally invasive surgery (MIS) and open surgery in the treatment of neuroblastoma (NB) in children by a meta-analysis. Materials and Methods: This is a meta-analysis. We searched for random or nonrandomized controlled study of MIS group and OPEN surgery group for the treatment of childhood NB included in PubMed, ClinicalTrials, EMBASE, and Cochrane library before January 31, 2020. Data extraction was performed in a standard format for the included studies, including tumor diameter, operation time, intraoperative bleeding, length of hospital stay (LOHS), complications, recurrence, and MYCN. Results: Seven retrospective studies were finally included, with a total of 571 children, including 162 in MIS group and 409 in the OPEN surgery group. Compared with the OPEN surgery group, the MIS group had reduced intraoperative bleeding (mean difference [MD] = -12.72, 95% CI: -24.84 to -0.61, P < .05), and reduced l LOHS (MD = -3.35, 95% CI: -5.55 to -1.15, P < .05) and decreased postoperative recurrence (MD = 0.20, 95% CI: 0.05-0.75, P < .05). The differences between the groups were statistically significant. There was no significant difference between groups in tumor diameter (MD = -18.84, 95% CI: -48.12 to 10.43, P > .05), operation time (MD = -21.7, 95% CI: -97.52 to 54.13, P > .05), and MYCN results (odds ratio = 2.27, 95% CI: 0.56-9.18, P > .05). Conclusions: Preliminary evidence indicates that the treatment of NB with MIS has the advantages of less intraoperative bleeding, shorter LOHS, and less postoperative recurrence compared with open surgery.
Collapse
Affiliation(s)
- Rong Zhang
- Department of Pediatric Surgery, Fujian Province Maternal and Child Health Hospital, Fuzhou, China
| | - Yi Fan Fang
- Department of Pediatric Surgery, Fujian Province Maternal and Child Health Hospital, Fuzhou, China
| | - Dian Ming Wu
- Department of Pediatric Surgery, Fujian Province Maternal and Child Health Hospital, Fuzhou, China
| | - Yu Lin
- Department of Pediatric Surgery, Fujian Province Maternal and Child Health Hospital, Fuzhou, China
| | - Bing Zhang
- Department of Pediatric Surgery, Fujian Province Maternal and Child Health Hospital, Fuzhou, China
| | - Ming Kun Liu
- Department of Pediatric Surgery, Fujian Province Maternal and Child Health Hospital, Fuzhou, China
| | - Jiang Xi Bai
- Department of Pediatric Surgery, Fujian Province Maternal and Child Health Hospital, Fuzhou, China
| | - Fei Chen
- Department of Pediatric Surgery, Fujian Province Maternal and Child Health Hospital, Fuzhou, China
| |
Collapse
|
28
|
Malone CF, Dharia NV, Kugener G, Forman AB, Rothberg MV, Abdusamad M, Gonzalez A, Kuljanin M, Robichaud AL, Conway AS, Dempster JM, Paolella BR, Dumont N, Hovestadt V, Mancias JD, Younger ST, Root DE, Golub TR, Vazquez F, Stegmaier K. Selective Modulation of a Pan-Essential Protein as a Therapeutic Strategy in Cancer. Cancer Discov 2021; 11:2282-2299. [PMID: 33883167 DOI: 10.1158/2159-8290.cd-20-1213] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 02/12/2021] [Accepted: 03/26/2021] [Indexed: 12/26/2022]
Abstract
Cancer dependency maps, which use CRISPR/Cas9 depletion screens to profile the landscape of genetic dependencies in hundreds of cancer cell lines, have identified context-specific dependencies that could be therapeutically exploited. An ideal therapy is both lethal and precise, but these depletion screens cannot readily distinguish between gene effects that are cytostatic or cytotoxic. Here, we use a diverse panel of functional genomic screening assays to identify NXT1 as a selective and rapidly lethal in vivo relevant genetic dependency in MYCN-amplified neuroblastoma. NXT1 heterodimerizes with NXF1, and together they form the principal mRNA nuclear export machinery. We describe a previously unrecognized mechanism of synthetic lethality between NXT1 and its paralog NXT2: their common essential binding partner NXF1 is lost only in the absence of both. We propose a potential therapeutic strategy for tumor-selective elimination of a protein that, if targeted directly, is expected to cause widespread toxicity. SIGNIFICANCE: We provide a framework for identifying new therapeutic targets from functional genomic screens. We nominate NXT1 as a selective lethal target in neuroblastoma and propose a therapeutic approach where the essential protein NXF1 can be selectively eliminated in tumor cells by exploiting the NXT1-NXT2 paralog relationship.See related commentary by Wang and Abdel-Wahab, p. 2129.This article is highlighted in the In This Issue feature, p. 2113.
Collapse
Affiliation(s)
- Clare F Malone
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Neekesh V Dharia
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts.,Harvard Medical School, Boston, Massachusetts.,Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts
| | | | - Alexandra B Forman
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | | | - Mai Abdusamad
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | | | - Miljan Kuljanin
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Amanda L Robichaud
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Amy Saur Conway
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | | | - Nancy Dumont
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Volker Hovestadt
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts.,Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts
| | - Joseph D Mancias
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Scott T Younger
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - David E Root
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Todd R Golub
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts.,Harvard Medical School, Boston, Massachusetts.,Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts
| | | | - Kimberly Stegmaier
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. .,Broad Institute of MIT and Harvard, Cambridge, Massachusetts.,Harvard Medical School, Boston, Massachusetts.,Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts
| |
Collapse
|
29
|
Abstract
Neuroblastoma (NB) is a pediatric cancer of the sympathetic nervous system and one of the most common solid tumors in infancy. Amplification of MYCN, copy number alterations, numerical and segmental chromosomal aberrations, mutations, and rearrangements on a handful of genes, such as ALK, ATRX, TP53, RAS/MAPK pathway genes, and TERT, are attributed as underlying causes that give rise to NB. However, the heterogeneous nature of the disease-along with the relative paucity of recurrent somatic mutations-reinforces the need to understand the interplay of genetic factors and epigenetic alterations in the context of NB. Epigenetic mechanisms tightly control gene expression, embryogenesis, imprinting, chromosomal stability, and tumorigenesis, thereby playing a pivotal role in physio- and pathological settings. The main epigenetic alterations include aberrant DNA methylation, disrupted patterns of posttranslational histone modifications, alterations in chromatin composition and/or architecture, and aberrant expression of non-coding RNAs. DNA methylation and demethylation are mediated by DNA methyltransferases (DNMTs) and ten-eleven translocation (TET) proteins, respectively, while histone modifications are coordinated by histone acetyltransferases and deacetylases (HATs, HDACs), and histone methyltransferases and demethylases (HMTs, HDMs). This article focuses predominately on the crosstalk between the epigenome and NB, and the implications it has on disease diagnosis and treatment.
Collapse
Affiliation(s)
- Irfete S Fetahu
- St. Anna Children's Cancer Research Institute, Zimmermannplatz 10, 1090, Vienna, Austria.
| | - Sabine Taschner-Mandl
- St. Anna Children's Cancer Research Institute, Zimmermannplatz 10, 1090, Vienna, Austria.
| |
Collapse
|
30
|
Custers L, Paassen I, Drost J. In vitro Modeling of Embryonal Tumors. Front Cell Dev Biol 2021; 9:640633. [PMID: 33718380 PMCID: PMC7952537 DOI: 10.3389/fcell.2021.640633] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 01/25/2021] [Indexed: 01/06/2023] Open
Abstract
A subset of pediatric tumors affects very young children and are thought to arise during fetal life. A common theme is that these embryonal tumors hijack developmental programs, causing a block in differentiation and, as a consequence, unrestricted proliferation. Embryonal tumors, therefore typically maintain an embryonic gene signature not found in their differentiated progeny. Still, the processes underpinning malignant transformation remain largely unknown, which is hampering therapeutic innovation. To gain more insight into these processes, in vitro and in vivo research models are indispensable. However, embryonic development is an extremely dynamic process with continuously changing cellular identities, making it challenging to define cells-of-origin. This is crucial for the development of representative models, as targeting the wrong cell or targeting a cell within an incorrect developmental time window can result in completely different phenotypes. Recent innovations in in vitro cell models may provide more versatile platforms to study embryonal tumors in a scalable manner. In this review, we outline different in vitro models that can be explored to study embryonal tumorigenesis and for therapy development.
Collapse
Affiliation(s)
- Lars Custers
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Irene Paassen
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Jarno Drost
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| |
Collapse
|
31
|
Huo Z, Bilang R, Brantner B, von der Weid N, Holland-Cunz SG, Gros SJ. Perspective on Similarities and Possible Overlaps of Congenital Disease Formation-Exemplified on a Case of Congenital Diaphragmatic Hernia and Neuroblastoma in a Neonate. CHILDREN-BASEL 2021; 8:children8020163. [PMID: 33671521 PMCID: PMC7926624 DOI: 10.3390/children8020163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/17/2021] [Accepted: 02/17/2021] [Indexed: 11/25/2022]
Abstract
The coincidence of two rare diseases such as congenital diaphragmatic hernia (CDH) and neuroblastoma is exceptional. With an incidence of around 2–3:10,000 and 1:8000 for either disease occurring on its own, the chance of simultaneous presentation of both pathologies at birth is extremely low. Unfortunately, the underlying processes leading to congenital malformation and neonatal tumors are not yet thoroughly understood. There are several hypotheses revolving around the formation of CDH and neuroblastoma. The aim of our study was to put the respective hypotheses of disease formation as well as known factors in this process into perspective regarding their similarities and possible overlaps of congenital disease formation. We present the joint occurrence of these two rare diseases based on a patient presentation and immunochemical prognostic marker evaluation. The aim of this manuscript is to elucidate possible similarities in the pathogeneses of both disease entities. Discussed are the role of toxins, cell differentiation, the influence of retinoic acid and NMYC as well as of hypoxia. The detailed discussion reveals that some of the proposed pathophysiological mechanisms of both malformations have common aspects. Especially disturbances of the retinoic acid pathway and NMYC expression can influence and disrupt cell differentiation in either disease. Due to the rarity of both diseases, interdisciplinary efforts and multi-center studies are needed to investigate the reasons for congenital malformations and their interlinkage with neonatal tumor disease.
Collapse
Affiliation(s)
- Zihe Huo
- Department of Pediatric Surgery, University Children’s Hospital Basel, 4031 Basel, Switzerland; (Z.H.); (R.B.); (B.B.); (S.G.H.-C.)
- Department of Clinical Research, University of Basel, 4001 Basel, Switzerland;
| | - Remo Bilang
- Department of Pediatric Surgery, University Children’s Hospital Basel, 4031 Basel, Switzerland; (Z.H.); (R.B.); (B.B.); (S.G.H.-C.)
- Department of Clinical Research, University of Basel, 4001 Basel, Switzerland;
| | - Benedikt Brantner
- Department of Pediatric Surgery, University Children’s Hospital Basel, 4031 Basel, Switzerland; (Z.H.); (R.B.); (B.B.); (S.G.H.-C.)
| | - Nicolas von der Weid
- Department of Clinical Research, University of Basel, 4001 Basel, Switzerland;
- Department of Hematology and Oncology, University Children’s Hospital Basel, 4056 Basel, Switzerland
| | - Stefan G. Holland-Cunz
- Department of Pediatric Surgery, University Children’s Hospital Basel, 4031 Basel, Switzerland; (Z.H.); (R.B.); (B.B.); (S.G.H.-C.)
- Department of Clinical Research, University of Basel, 4001 Basel, Switzerland;
| | - Stephanie J. Gros
- Department of Pediatric Surgery, University Children’s Hospital Basel, 4031 Basel, Switzerland; (Z.H.); (R.B.); (B.B.); (S.G.H.-C.)
- Department of Clinical Research, University of Basel, 4001 Basel, Switzerland;
- Correspondence:
| |
Collapse
|
32
|
Pini N, Huo Z, Kym U, Holland-Cunz S, Gros SJ. AQP1-Driven Migration Is Independent of Other Known Adverse Factors but Requires a Hypoxic Undifferentiated Cell Profile in Neuroblastoma. CHILDREN-BASEL 2021; 8:children8010048. [PMID: 33467498 PMCID: PMC7829990 DOI: 10.3390/children8010048] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/30/2020] [Accepted: 01/12/2021] [Indexed: 12/14/2022]
Abstract
Neuroblastoma is a biologically very heterogeneous tumor with its clinical manifestation ranging from spontaneous regression to highly aggressive metastatic disease. Several adverse factors have been linked to oncogenesis, tumor progression and metastases of neuroblastoma including NMYC amplification, the neural adhesion molecule NCAM, as well as CXCR4 as a promoter of metastases. In this study, we investigate to what extent the expression of AQP1 in neuroblastoma correlates with changing cellular factors such as the hypoxic status, differentiation, expression of known adverse factors such as NMYC and NCAM, and CXCR4-related metastatic spread. Our results show that while AQP1 expression leads to an increased migratory behavior of neuroblastoma cells under hypoxic conditions, we find that hypoxia is associated with a reduction of NMYC in the same cells. A similar effect can be observed when using the tetracycline driven mechanism of SH-EP/Tet cells. When NMYC is not expressed, the expression of AQP1 is increased together with an increased expression of HIF-1α and HIF-2α. We furthermore show that when growing cells in different cell densities, they express AQP1, HIF-1α, HIF-2α, NMYC and NCAM to different degrees. AQP1 expression correlates with a hypoxic profile of these cells with increased HIF-1α and HIF-2α expression, as well as with NMYC and NCAM expression in two out of three neuroblastoma cell lines. When investigating cell properties of the cells that actually migrate, we find that the increased APQ1 expression in the migrated cells correlates with an increased NMYC and NCAM expression again in two out of three cell lines. Expression of the tumor cell homing marker CXCR4 varies between different tumor areas and between cell lines. While some migrated tumor cells highly express CXCR4, cells of other origin do not. In the initial phase of migration, we determined a dominant role of AQP1 expression of migrating cells in the scratch assay.
Collapse
Affiliation(s)
- Nicola Pini
- Department of Pediatric Surgery, University Children’s Hospital Basel, 4031 Basel, Switzerland; (N.P.); (Z.H.); (U.K.); (S.H.-C.)
| | - Zihe Huo
- Department of Pediatric Surgery, University Children’s Hospital Basel, 4031 Basel, Switzerland; (N.P.); (Z.H.); (U.K.); (S.H.-C.)
| | - Urs Kym
- Department of Pediatric Surgery, University Children’s Hospital Basel, 4031 Basel, Switzerland; (N.P.); (Z.H.); (U.K.); (S.H.-C.)
| | - Stefan Holland-Cunz
- Department of Pediatric Surgery, University Children’s Hospital Basel, 4031 Basel, Switzerland; (N.P.); (Z.H.); (U.K.); (S.H.-C.)
| | - Stephanie J. Gros
- Department of Pediatric Surgery, University Children’s Hospital Basel, 4031 Basel, Switzerland; (N.P.); (Z.H.); (U.K.); (S.H.-C.)
- Department of Clinical Research, University of Basel, 4031 Basel, Switzerland
- Correspondence:
| |
Collapse
|
33
|
Current and future applications of induced pluripotent stem cell-based models to study pathological proteins in neurodegenerative disorders. Mol Psychiatry 2021; 26:2685-2706. [PMID: 33495544 PMCID: PMC8505258 DOI: 10.1038/s41380-020-00999-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/02/2020] [Accepted: 12/09/2020] [Indexed: 12/13/2022]
Abstract
Neurodegenerative disorders emerge from the failure of intricate cellular mechanisms, which ultimately lead to the loss of vulnerable neuronal populations. Research conducted across several laboratories has now provided compelling evidence that pathogenic proteins can also contribute to non-cell autonomous toxicity in several neurodegenerative contexts, including Alzheimer's, Parkinson's, and Huntington's diseases as well as Amyotrophic Lateral Sclerosis. Given the nearly ubiquitous nature of abnormal protein accumulation in such disorders, elucidating the mechanisms and routes underlying these processes is essential to the development of effective treatments. To this end, physiologically relevant human in vitro models are critical to understand the processes surrounding uptake, release and nucleation under physiological or pathological conditions. This review explores the use of human-induced pluripotent stem cells (iPSCs) to study prion-like protein propagation in neurodegenerative diseases, discusses advantages and limitations of this model, and presents emerging technologies that, combined with the use of iPSC-based models, will provide powerful model systems to propel fundamental research forward.
Collapse
|
34
|
Maeshima R, Moulding D, Stoker AW, Hart SL. MYCN Silencing by RNAi Induces Neurogenesis and Suppresses Proliferation in Models of Neuroblastoma with Resistance to Retinoic Acid. Nucleic Acid Ther 2020; 30:237-248. [PMID: 32240058 PMCID: PMC7415885 DOI: 10.1089/nat.2019.0831] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 03/02/2020] [Indexed: 12/14/2022] Open
Abstract
Neuroblastoma (NB) is the most common solid tumor in childhood. Twenty percent of patients display MYCN amplification, which indicates a very poor prognosis. MYCN is a highly specific target for an NB tumor therapy as MYCN expression is absent or very low in most normal cells, while, as a transcription factor, it regulates many essential cell activities in tumor cells. We aim to develop a therapy for NB based on MYCN silencing by short interfering RNA (siRNA) molecules, which can silence target genes by RNA interference (RNAi), a naturally occurring method of gene silencing. It has been shown previously that MYCN silencing can induce apoptosis and differentiation in MYCN amplified NB. In this article, we have demonstrated that siRNA-mediated silencing of MYCN in MYCN-amplified NB cells induced neurogenesis in NB cells, whereas retinoic acid (RA) treatment did not. RA can differentiate NB cells and is used for treatment of residual disease after surgery or chemotherapy, but resistance can develop. In addition, MYCN siRNA treatment suppressed growth in a MYCN-amplified NB cell line more than that by RA. Our result suggests that gene therapy using RNAi targeting MYCN can be a novel therapy toward MYCN-amplified NB that have complete or partial resistance toward RA.
Collapse
Affiliation(s)
- Ruhina Maeshima
- Genetics and Genomic Medicine Department, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Dale Moulding
- UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Andrew W. Stoker
- Developmental Biology & Cancer Department, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Stephen L. Hart
- Genetics and Genomic Medicine Department, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| |
Collapse
|
35
|
Asthana A, Ndyabawe K, Mendez D, Douglass M, Haidekker MA, Kisaalita WS. Calcium Oscillation Frequency Is a Potential Functional Complex Physiological Relevance Indicator for a Neuroblastoma-Based 3D Culture Model. ACS Biomater Sci Eng 2020; 6:4314-4323. [PMID: 33463347 DOI: 10.1021/acsbiomaterials.9b01988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
In vitro screening for drugs that affect neural function in vivo is still primitive. It primarily relies on single cellular responses from 2D monolayer cultures that have been shown to be exaggerations of the in vivo response. For the 3D model to be physiologically relevant, it should express characteristics that not only differentiate it from 2D but also closely emulate those seen in vivo. These complex physiologically relevant (CPR) outcomes can serve as a standard for determining how close a 3D culture is to its native tissue or which out of a given number of 3D platforms is better suited for a given application. In this study, Fluo-4-based calcium fluorescence imaging was performed followed by automated image data processing to quantify the calcium oscillation frequency of SHSY5Y cells cultured in 2D and 3D formats. It was found that the calcium oscillation frequency is upregulated in traditional 2D cultures while it was comparable to in vivo in spheroid and microporous polymer scaffold-based 3D models, suggesting calcium oscillation frequency as a potential functional CPR indicator for neural cultures.
Collapse
|
36
|
Zhao Z, Shelton SD, Oviedo A, Baker AL, Bryant CP, Omidvarnia S, Du L. The PLAGL2/MYCN/miR-506-3p interplay regulates neuroblastoma cell fate and associates with neuroblastoma progression. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:41. [PMID: 32087738 PMCID: PMC7036248 DOI: 10.1186/s13046-020-1531-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 01/21/2020] [Indexed: 12/17/2022]
Abstract
Background The oncogene MYCN is critical for tumorigenesis of several types of cancers including neuroblastoma. We previously reported that miR-506-3p repressed MYCN expression in neuroblastoma cells. However, the mechanism underlying such regulation was undetermined since there is no miR-506-3p target site in MYCN 3’UTR. Methods By a systematic investigation combining microarray, informatics and luciferase reporter assay, we identified that the transcriptional factor pleiomorphic adenoma gene-like 2 (PLAGL2) is a direct target of miR-506-3p that mediates its regulation on MYCN expression. Using CHIP-PCR and luciferase reporter assay, we validated the transcriptional regulation of MYCN by PLAGL2 and we further demonstrated the transcriptional regulation of PLAGL2 by MYCN. We examined the function of PLAGL2 in regulating neuroblastoma cell fate by cell viability assay, colony formation and Western blotting of differentiation markers. We examined the effect of retinoic acid, the differentiation agent used in neuroblastoma therapy, on miR-506-3p, PLAGL2 and MYCN expressions by quantitative PCR and Western blots. We investigated the clinical relevance of PLAGL2 expression by examining the correlation of tumor PLAGL2 mRNA levels with MYCN mRNA expression and patient survival using public neuroblastoma patient datasets. Results We found that miR-506-3p directly down-regulated PLAGL2 expression, and we validated a PLAGL2 binding site in the MYCN promoter region responsible for promoting MYCN transcription, thereby establishing a mechanism through which miR-506-3p regulates MYCN expression. Conversely, we discovered that MYCN regulated PLAGL2 transcription through five N-Myc-binding E-boxes in the PLAGL2 promoter region. We further confirmed the reciprocal regulation between endogenous PLAGL2 and MYCN in multiple neuroblastoma cell lines. Moreover, we found that PLAGL2 knockdown induced neuroblastoma cell differentiation and reduced cell proliferation, and combined knockdown of PLAGL2 and MYCN showed a synergistic effect. More strikingly, we found that high tumor PLAGL2 mRNA levels were significantly correlated with high MYCN mRNA levels and poor patient survival in neuroblastoma patients. Furthermore, we found that retinoic acid increased expression of miR-506-3p and repressed expression of MYCN and PLAGL2. Conclusions Our findings altogether suggest that the interplay network formed by PLAGL2, MYCN and miR-506-3p is an important mechanism in regulating neuroblastoma cell fate, determining neuroblastoma prognosis, and mediating the therapeutic function of retinoic acid.
Collapse
Affiliation(s)
- Zhenze Zhao
- Department of Chemistry and Biochemistry, Texas State University, 601 University Drive, San Marcos, TX, 78666, USA
| | - Spencer D Shelton
- Department of Chemistry and Biochemistry, Texas State University, 601 University Drive, San Marcos, TX, 78666, USA
| | - Alejandro Oviedo
- Department of Chemistry and Biochemistry, Texas State University, 601 University Drive, San Marcos, TX, 78666, USA
| | - Amy L Baker
- Department of Chemistry and Biochemistry, Texas State University, 601 University Drive, San Marcos, TX, 78666, USA
| | - Collin P Bryant
- Department of Chemistry and Biochemistry, Texas State University, 601 University Drive, San Marcos, TX, 78666, USA
| | - Soroush Omidvarnia
- Department of Chemistry and Biochemistry, Texas State University, 601 University Drive, San Marcos, TX, 78666, USA
| | - Liqin Du
- Department of Chemistry and Biochemistry, Texas State University, 601 University Drive, San Marcos, TX, 78666, USA.
| |
Collapse
|
37
|
Almstedt E, Elgendy R, Hekmati N, Rosén E, Wärn C, Olsen TK, Dyberg C, Doroszko M, Larsson I, Sundström A, Arsenian Henriksson M, Påhlman S, Bexell D, Vanlandewijck M, Kogner P, Jörnsten R, Krona C, Nelander S. Integrative discovery of treatments for high-risk neuroblastoma. Nat Commun 2020; 11:71. [PMID: 31900415 PMCID: PMC6941971 DOI: 10.1038/s41467-019-13817-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 11/22/2019] [Indexed: 12/22/2022] Open
Abstract
Despite advances in the molecular exploration of paediatric cancers, approximately 50% of children with high-risk neuroblastoma lack effective treatment. To identify therapeutic options for this group of high-risk patients, we combine predictive data mining with experimental evaluation in patient-derived xenograft cells. Our proposed algorithm, TargetTranslator, integrates data from tumour biobanks, pharmacological databases, and cellular networks to predict how targeted interventions affect mRNA signatures associated with high patient risk or disease processes. We find more than 80 targets to be associated with neuroblastoma risk and differentiation signatures. Selected targets are evaluated in cell lines derived from high-risk patients to demonstrate reversal of risk signatures and malignant phenotypes. Using neuroblastoma xenograft models, we establish CNR2 and MAPK8 as promising candidates for the treatment of high-risk neuroblastoma. We expect that our method, available as a public tool (targettranslator.org), will enhance and expedite the discovery of risk-associated targets for paediatric and adult cancers. We lack effective treatment for half of children with high-risk neuroblastoma. Here, the authors introduce an algorithm that can predict the effect of interventions on gene expression signatures associated with high disease processes and risk, and identify and validate promising drug targets.
Collapse
Affiliation(s)
- Elin Almstedt
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Ramy Elgendy
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Neda Hekmati
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Emil Rosén
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Caroline Wärn
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Thale Kristin Olsen
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, SE-17176, Stockholm, Sweden
| | - Cecilia Dyberg
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, SE-17176, Stockholm, Sweden
| | - Milena Doroszko
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Ida Larsson
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Anders Sundström
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Marie Arsenian Henriksson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, SE-171 77, Stockholm, Sweden
| | - Sven Påhlman
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, SE-223 81, Lund, Sweden
| | - Daniel Bexell
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, SE-223 81, Lund, Sweden
| | - Michael Vanlandewijck
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-751 85, Uppsala, Sweden.,Department of Medicine, Integrated Cardio-Metabolic Centre Single Cell Facility, Karolinska Institutet, SE-17177, Stockholm, Sweden
| | - Per Kogner
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, SE-17176, Stockholm, Sweden
| | - Rebecka Jörnsten
- Mathematical Sciences, Chalmers University of Technology, Gothenburg, SE-41296, Sweden
| | - Cecilia Krona
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Sven Nelander
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-751 85, Uppsala, Sweden.
| |
Collapse
|
38
|
Alshangiti AM, Tuboly E, Hegarty SV, McCarthy CM, Sullivan AM, O'Keeffe GW. 4-Hydroxychalcone Induces Cell Death via Oxidative Stress in MYCN-Amplified Human Neuroblastoma Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:1670759. [PMID: 31885773 PMCID: PMC6915131 DOI: 10.1155/2019/1670759] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/21/2019] [Accepted: 11/20/2019] [Indexed: 12/26/2022]
Abstract
Neuroblastoma is an embryonal malignancy that arises from cells of sympathoadrenal lineage during the development of the nervous system. It is the most common pediatric extracranial solid tumor and is responsible for 15% of childhood deaths from cancer. Fifty percent of cases are diagnosed as high-risk metastatic disease with a low overall 5-year survival rate. More than half of patients experience disease recurrence that can be refractory to treatment. Amplification of the MYCN gene is an important prognostic indicator that is associated with rapid disease progression and a poor prognosis, highlighting the need for new therapeutic approaches. In recent years, there has been an increasing focus on identifying anticancer properties of naturally occurring chalcones, which are secondary metabolites with variable phenolic structures. Here, we report that 4-hydroxychalcone is a potent cytotoxin for MYCN-amplified IMR-32 and SK-N-BE (2) neuroblastoma cells, when compared to non-MYCN-amplified SH-SY5Y neuroblastoma cells and to the non-neuroblastoma human embryonic kidney cell line, HEK293t. Moreover, 4-hydroxychalcone treatment significantly decreased cellular levels of the antioxidant glutathione and increased cellular reactive oxygen species. In addition, 4-hydroxychalcone treatment led to impairments in mitochondrial respiratory function, compared to controls. In support of this, the cytotoxic effect of 4-hydroxychalcone was prevented by co-treatment with either the antioxidant N-acetyl-L-cysteine, a pharmacological inhibitor of oxidative stress-induced cell death (IM-54) or the mitochondrial reactive oxygen species scavenger, Mito-TEMPO. When combined with the anticancer drugs cisplatin or doxorubicin, 4-hydroxychalcone led to greater reductions in cell viability than was induced by either anti-cancer agent alone. In summary, this study identifies a cytotoxic effect of 4-hydroxychalcone in MYCN-amplified human neuroblastoma cells, which rationalizes its further study in the development of new therapies for pediatric neuroblastoma.
Collapse
Affiliation(s)
- Amnah M. Alshangiti
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- Cork Neuroscience Centre, University College Cork, Cork, Ireland
| | - Eszter Tuboly
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - Shane V. Hegarty
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- Cork Neuroscience Centre, University College Cork, Cork, Ireland
| | - Cathal M. McCarthy
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - Aideen M. Sullivan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- Cork Neuroscience Centre, University College Cork, Cork, Ireland
| | - Gerard W. O'Keeffe
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- Cork Neuroscience Centre, University College Cork, Cork, Ireland
| |
Collapse
|
39
|
Yoda H, Nakayama T, Miura M, Toriyama M, Motohashi S, Suzuki T. Vitamin K3 derivative induces apoptotic cell death in neuroblastoma via downregulation of MYCN expression. Biochem Biophys Rep 2019; 20:100701. [PMID: 31844686 PMCID: PMC6895568 DOI: 10.1016/j.bbrep.2019.100701] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 10/15/2019] [Accepted: 10/22/2019] [Indexed: 11/22/2022] Open
Abstract
Neuroblastoma is a pediatric malignant tumor arising from the sympathetic nervous system. The patients with high-risk neuroblastomas frequently exhibit amplification and high expression of the MYCN gene, resulting in worse clinical outcomes. Vitamin K3 (VK3) is a synthetic VK-like compound that has been known to have antitumor activity against various types of cancers. In the present study, we have asked whether VK3 and its derivative, VK3-OH, could have the antitumor activity against neuroblastoma-derived cells. Based on our results, VK3-OH strongly inhibited cell proliferation and induced apoptotic cell death compared to VK3. Treatment of MYCN-driven neuroblastoma cells with VK3-OH potentiated tumor suppressor p53 accompanied by downregulation of anti-apoptotic Bcl-2 and Mcl-1. Interestingly, VK3-OH also suppressed the MYCN at mRNA and protein levels. Furthermore, we found downregulation of LIN28B following VK3-OH treatment in MYCN-amplified and overexpressed neuroblastoma cells. Collectively, our current findings strongly suggest that VK3-OH provides a potential therapeutic strategy for patients with MYCN-driven neuroblastomas.
Collapse
Affiliation(s)
- Hiroyuki Yoda
- Laboratory of Clinical Medicine, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi, Chiba, 274-8555, Japan
| | - Toshimitsu Nakayama
- Center for Pharmacist Education, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi, Chiba, 274-8555, Japan
| | - Motofumi Miura
- Laboratory of Molecular Chemistry, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi, Chiba, 274-8555, Japan
| | - Masaharu Toriyama
- Laboratory of Molecular Chemistry, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi, Chiba, 274-8555, Japan
| | - Shigeyasu Motohashi
- Laboratory of Molecular Chemistry, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi, Chiba, 274-8555, Japan
| | - Takashi Suzuki
- Laboratory of Clinical Medicine, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi, Chiba, 274-8555, Japan
- Department of Pediatrics and Child Health, School of Medicine, Nihon University, 30-1 Oyaguchikami, Itabashi-ku, Tokyo, 173-0032, Japan
| |
Collapse
|
40
|
Taylor JS, Zeki J, Ornell K, Coburn J, Shimada H, Ikegaki N, Chiu B. Down-regulation of MYCN protein by CX-5461 leads to neuroblastoma tumor growth suppression. J Pediatr Surg 2019; 54:1192-1197. [PMID: 30879743 PMCID: PMC6545249 DOI: 10.1016/j.jpedsurg.2019.02.028] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 02/21/2019] [Indexed: 11/19/2022]
Abstract
PURPOSE MYCN oncogene amplification is an independent predictor of poor prognosis in neuroblastoma. CX-5461 is a small molecular inhibitor that prevents initiation of ribosomal RNA (rRNA) synthesis by RNA Pol I, down-regulating MYCN/MYC proteins. We hypothesize that neuroblastoma tumor growth can be suppressed by CX-5461. METHODS MYCN-amplified (KELLY, IMR5) and nonamplified (SY5Y, SKNAS) neuroblastoma cells were treated with CX-5461. MYCN/MYC expression after 24-48 h was determined by Western blot. Orthotopic neuroblastoma tumors created in mice using KELLY cells were treated with CX-5461-loaded silk films implanted locally. Tumor growth was monitored using ultrasound. Histologic evaluation of tumors was performed. RESULTS IC50 for KELLY, IMR5, SY5Y, and SKNAS cells to CX-5461 was 0.75 μM, 0.02 μM, 0.8 μM, and 1.7 μM, respectively. CX-5461 down-regulated MYCN and MYC proteins at 0.25-1.0 μM on Western blot analysis. CX-5461-loaded silk film released 23.7±3 μg of the drug in 24 h and 48.2±3.9 μg at 120 h. KELLY tumors treated with CX-5461-loaded film reached 800 mm3 after 7.8±1.4 days, while those treated with control film reached the same size on 5.1±0.6 days (p=0.03). CX-5461-treated tumors showed collapse of nucleolar hypertrophy and MYCN protein downregulation. CONCLUSION We demonstrated that local delivery of CX-5461 via sustained release platform can suppress orthotopic neuroblastoma tumor growth, especially those with MYCN/MYC overexpression.
Collapse
Affiliation(s)
| | - Jasmine Zeki
- Department of Surgery, Stanford University, Stanford, CA
| | - Kimberly Ornell
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA
| | - Jeannine Coburn
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA
| | - Hiroyuki Shimada
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, University of Southern California, Los Angeles, CA
| | - Naohiko Ikegaki
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL
| | - Bill Chiu
- Department of Surgery, Stanford University, Stanford, CA; Department of Surgery, University of Illinois at Chicago, Chicago, IL.
| |
Collapse
|
41
|
Abdullah A, Talwar P, d'Hellencourt CL, Ravanan P. IRE1α is critical for Kaempferol-induced neuroblastoma differentiation. FEBS J 2019; 286:1375-1392. [PMID: 30719816 DOI: 10.1111/febs.14776] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 01/13/2019] [Accepted: 01/31/2019] [Indexed: 12/21/2022]
Abstract
Neuroblastoma is an embryonic malignancy that arises out of the neural crest cells of the sympathetic nervous system. It is the most common childhood tumor known for its spontaneous regression via the process of differentiation. The induction of differentiation using small molecules such as retinoic acid is one of the therapeutic strategies to treat the residual disease. In this study, we have reported the effect of kaempferol (KFL) in inducing differentiation of neuroblastoma cells in vitro. Treatment of neuroblastoma cells with KFL reduced the proliferation and enhanced apoptosis along with the induction of neuritogenesis. Analysis of the expression of neuron-specific markers such as β-III tubulin, neuron-specific enolase, and N-myc downregulated gene 1 revealed the process of differentiation accompanying KFL-induced apoptosis. Further analysis to understand the molecular mechanism of action showed that the effect of KFL is mediated by the activation of the endoribonuclease activity of inositol-requiring enzyme 1 alpha (IRE1α), an endoplasmic reticulum-resident transmembrane protein. In silico docking analysis and biochemical assays using recombinant human IRE1α confirm the binding of KFL to the ATP-binding site of IRE1α, which thereby activates IRE1α ribonuclease activity. Treatment of cells with the small molecule STF083010, which specifically targets and inhibits the endoribonuclease activity of IRE1α, showed reduced expression of neuron-specific markers and curtailed neuritogenesis. The knockdown of IRE1α using plasmid-based shRNA lentiviral particles also showed diminished changes in the morphology of the cells upon KFL treatment. Thus, our study suggests that KFL induces differentiation of neuroblastoma cells via the IRE1α -XBP1 pathway.
Collapse
Affiliation(s)
- Ahmad Abdullah
- Apoptosis and Cell Survival Research Lab, Department of Biosciences, School of Biosciences and Technology, VIT University, Vellore, India
| | - Priti Talwar
- Apoptosis and Cell Survival Research Lab, Department of Biosciences, School of Biosciences and Technology, VIT University, Vellore, India
| | - Christian Lefebvre d'Hellencourt
- Institut National de la Santé et de la Recherche Médicale, UMR Diabète Athérothombose Thérapies Réunion Océan Indien, Université de La Réunion, Saint-Denis de La Réunion, France
| | - Palaniyandi Ravanan
- Apoptosis and Cell Survival Research Lab, Department of Biosciences, School of Biosciences and Technology, VIT University, Vellore, India
| |
Collapse
|
42
|
Lee MW, Kim DS, Kim HR, Park HJ, Lee JW, Sung KW, Koo HH, Yoo KH. Inhibition of N-myc expression sensitizes human neuroblastoma IMR-32 cells expressing caspase-8 to TRAIL. Cell Prolif 2019; 52:e12577. [PMID: 30724400 PMCID: PMC6536445 DOI: 10.1111/cpr.12577] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 12/21/2018] [Accepted: 12/28/2018] [Indexed: 12/13/2022] Open
Abstract
Objectives This study aims to explore the roles of N‐myc and caspase‐8 in TRAIL‐resistant IMR‐32 cells which exhibit MYCN oncogene amplification and lack caspase‐8 expression. Materials and methods We established N‐myc–downregulated IMR‐32 cells using shRNA lentiviral particles targeting N‐myc and examined the effect the N‐myc inhibition on TRAIL susceptibility in human neuroblastoma IMR‐32 cells expressing caspase‐8. Results Cisplatin treatment in IMR‐32 cells increased the expression of death receptor 5 (DR5; TRAIL‐R2), but not other receptors, via downregulation of NF‐κB activity. However, the cisplatin‐mediated increase in DR5 failed to induce cell death following TRAIL treatment. Furthermore, interferon (IFN)‐γ pretreatment increased caspase‐8 expression in IMR‐32 cells, but cisplatin failed to trigger TRAIL cytotoxicity. We downregulated N‐myc expression in IMR‐32 cells using N‐myc–targeting shRNA. These cells showed decreased growth rate and Bcl‐2 expression accompanied by a mild collapse in the mitochondrial membrane potential as compared with those treated with scrambled shRNA. TRAIL treatment in N‐myc–negative cells expressing caspase‐8 following IFN‐γ treatment significantly triggered apoptotic cell death. Concurrent treatment with cisplatin enhanced TRAIL‐mediated cytotoxicity, which was abrogated by an additional pretreatment with DR5:Fc chimera protein. Conclusions N‐myc and caspase‐8 expressions are involved in TRAIL susceptibility in IMR‐32 cells, and the combination of treatment with cisplatin and TRAIL may serve as a promising strategy for the development of therapeutics against neuroblastoma that is controlled by N‐myc and caspase‐8 expression.
Collapse
Affiliation(s)
- Myoung Woo Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Dae Seong Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hye Ryung Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hyun Jin Park
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ji Won Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ki Woong Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hong Hoe Koo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
| | - Keon Hee Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
| |
Collapse
|
43
|
Otsuka K, Sasada M, Iyoda T, Nohara Y, Sakai S, Asayama T, Suenaga Y, Yokoi S, Higami Y, Kodama H, Fukai F. Combining peptide TNIIIA2 with all- trans retinoic acid accelerates N-Myc protein degradation and neuronal differentiation in MYCN-amplified neuroblastoma cells. Am J Cancer Res 2019; 9:434-448. [PMID: 30906641 PMCID: PMC6405964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 01/07/2019] [Indexed: 06/09/2023] Open
Abstract
Neuroblastoma is one of the common solid tumors of childhood. Nearly half of neuroblastoma patients are classified into the high-risk group, and their 5-year event-free survival (EFS) rates remain unsatisfactory in the range of 30-40%. High-risk neuroblastoma is characterized by amplification of the MYCN gene and excessive expression of its protein product, N-Myc. Because N-Myc is a transcription factor for various pro-proliferative proteins, the excessive expression causes aberrant or blocked neuronal differentiation during development of sympathetic nervous system, which is a central aspect of neuroblastoma genesis. The current main treatment for high-risk neuroblastoma is intensive chemotherapy using anti-cancer drugs that induce apoptosis in tumor cells, but intensive chemotherapy has another serious risk of long-lasting side effects, so-called "late effects", that occur many years after chemotherapy has ended. As a solution for such situation, differentiation therapy has been expected as a mild chemotherapy with a low risk of late effects, and an application of retinoic acid (RA) and its derivatives as treatment for high-risk neuroblastoma has long been attempted. However, the clinical outcome has not been sufficient with the use of retinoids, including all-trans retinoic acid (ATRA), mainly because of the inhibition of differentiation caused by N-Myc. In the present study, we succeeded in synergistically accelerating the ATRA-induced neuronal differentiation of MYCN-amplified neuroblastoma cells by combining a peptide derived from tenascin-C, termed TNIIIA2, which has a potent ability to activate β1-integrins. Accelerated differentiation was caused by a decrease in N-Myc protein level in neuroblastoma cells after the combined treatment of TNIIIA2 with ATRA. That is, combination treatment using ATRA with TNIIIA2 induced proteasomal degradation in the N-Myc oncoprotein of neuroblastoma cells with MYCN gene amplification, and this caused acceleration of neuronal differentiation and attenuation of malignant properties. Furthermore, an in vivo experiment using a xenograft mouse model showed a therapeutic potential of the combination administration of ATRA and TNIIIA2 for high-risk neuroblastoma. These results provide a new insight into differentiation therapy for high-risk neuroblastoma based on N-Myc protein degradation.
Collapse
Affiliation(s)
- Kazuki Otsuka
- Department of Molecular Patho-Physiology, Faculty of Pharmaceutical Sciences, Tokyo University of ScienceNoda, Chiba, Japan
| | - Manabu Sasada
- Department of Molecular Patho-Physiology, Faculty of Pharmaceutical Sciences, Tokyo University of ScienceNoda, Chiba, Japan
- Translational Research Center, Research Institutes for Science and Technology, Tokyo University of ScienceNoda, Chiba, Japan
- Cancer Genome Center, Chiba Cancer Center Research InstituteChiba, Japan
| | - Takuya Iyoda
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City UniversitySanyo-Onoda, Yamaguchi, Japan
| | - Yusuke Nohara
- Department of Molecular Patho-Physiology, Faculty of Pharmaceutical Sciences, Tokyo University of ScienceNoda, Chiba, Japan
| | - Shunsuke Sakai
- Department of Molecular Patho-Physiology, Faculty of Pharmaceutical Sciences, Tokyo University of ScienceNoda, Chiba, Japan
| | - Tatsufumi Asayama
- Department of Molecular Patho-Physiology, Faculty of Pharmaceutical Sciences, Tokyo University of ScienceNoda, Chiba, Japan
| | - Yusuke Suenaga
- Cancer Genome Center, Chiba Cancer Center Research InstituteChiba, Japan
| | - Sana Yokoi
- Cancer Genome Center, Chiba Cancer Center Research InstituteChiba, Japan
| | - Yoshikazu Higami
- Translational Research Center, Research Institutes for Science and Technology, Tokyo University of ScienceNoda, Chiba, Japan
- Department of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of ScienceNoda, Chiba, Japan
| | - Hiroaki Kodama
- Department of Biochemistry, Faculty of Science and Engineering, Saga UniversitySaga, Japan
| | - Fumio Fukai
- Department of Molecular Patho-Physiology, Faculty of Pharmaceutical Sciences, Tokyo University of ScienceNoda, Chiba, Japan
- Translational Research Center, Research Institutes for Science and Technology, Tokyo University of ScienceNoda, Chiba, Japan
| |
Collapse
|
44
|
Zhang Q, Zhang Q, Jiang X, Ye Y, Liao H, Zhu F, Yan J, Luo L, Tian L, Jiang C, Chen Y, Liang X, Sun Y. Collaborative ISL1/GATA3 interaction in controlling neuroblastoma oncogenic pathways overlapping with but distinct from MYCN. Theranostics 2019; 9:986-1000. [PMID: 30867811 PMCID: PMC6401405 DOI: 10.7150/thno.30199] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 01/14/2019] [Indexed: 12/12/2022] Open
Abstract
Background: Transcription factor ISL1 plays a critical role in sympathetic neurogenesis. Expression of ISL1 has been associated with neuroblastoma, a pediatric tumor derived from sympatho-adrenal progenitors, however the role of ISL1 in neuroblastoma remains unexplored. Method: Here, we knocked down ISL1 (KD) in SH-SY5Y neuroblastoma cells and performed RNA-seq and ISL1 ChIP-seq analyses. Results: Analyses of these data revealed that ISL1 acts upstream of multiple oncogenic genes and pathways essential for neuroblastoma proliferation and differentiation, including LMO1 and LIN28B. ISL1 promotes expression of a number of cell cycle associated genes, but represses differentiation associated genes including RA receptors and the downstream target genes EPAS1 and CDKN1A. Consequently, Knockdown of ISL1 inhibits neuroblastoma cell proliferation and migration in vitro and impedes tumor growth in vivo, and enhances neuronal differentiation by RA treatment. Furthermore, genome-wide mapping revealed a substantial co-occupancy of binding regions by ISL1 and GATA3, and ISL1 physically interacts with GATA3, and together they synergistically regulate the aforementioned oncogenic pathways. In addition, analyses of the roles of ISL1 and MYCN in MYCN-amplified and MYCN non-amplified neuroblastoma cells revealed an epistatic relationship between ISL1 and MYCN. ISL1 and MYCN function in parallel to regulate common yet distinct oncogenic pathways in neuroblastoma. Conclusion: Our study has demonstrated that ISL1 plays an essential role in neuroblastoma regulatory networks and may serve as a potential therapeutic target in neuroblastoma.
Collapse
|
45
|
Lee ACL, Shih YY, Zhou F, Chao TC, Lee H, Liao YF, Hsu WM, Hong JH. Calreticulin regulates MYCN expression to control neuronal differentiation and stemness of neuroblastoma. J Mol Med (Berl) 2019; 97:325-339. [PMID: 30612140 DOI: 10.1007/s00109-018-1730-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 11/15/2018] [Accepted: 12/05/2018] [Indexed: 11/28/2022]
Abstract
Oncogenic N-MYC (MYCN) is widely used as a biomarker in clinics for neuroblastoma (NB) patients; nevertheless, mechanism that underlines MYCN regulation remains elusive. In the present study, we identified calreticulin (CRT) as a novel MYCN suppressor that downregulated MYCN promoter activity and protein expression to modulate neuronal differentiation and stemness. Our data showed that CRT-mediated MYCN suppression led to increased neurite length and commensurate elevation in differentiation marker GAP-43. We examined effect of radiotherapy and discovered that ionizing radiation (IR) was able to augment CRT expression dose-dependently in NB. Interestingly, neuronal differentiation and neurosphere formation (NSF) of NB were not only co-modulated by IR and CRT but were also dependent on Ca2+-buffering domain (C-domain) of CRT. Mutagenesis analysis showed that C-domain was indispensable for CRT-mediated MYCN regulation in NB differentiation and NSF. Of note, IR-induced formation of neural stem-like neurospheres (NS) was significantly impaired in CRT-overexpressed NB cells. The occupancy of CRT on MYCN 5' proximal promoter was confirmed by chromatin immunoprecipitation assays, revealing potential CRT binding sites that coincided with transcription factor E2F1 binding elements. In addition, we identified a physical interaction between CRT and E2F1, and demonstrated that CRT occupancy on MYCN promoter prevented E2F1-mediated MYCN upregulation. In line with in vitro findings, hampered tumor latency and retarded tumor growth in xenograft model corroborated IR and CRT co-mediated neuronal differentiation of NB. Together, our data delineated a novel mechanism of CRT-mediated MYCN regulation and warranted further preclinical investigation towards new therapeutic strategy for NB. CRT suppresses MYCN expression and promotes neuronal differentiation in NB. CRT regulates MYCN via interaction with E2F1 and direct binding to MYCN promoter. Ca2+-buffering domain of CRT is critical in MYCN regulation and NB differentiation. CRT-MYCN axis impacts on NB stemness by modulating neurosphere formation. Xenograft model corroborates in vitro NB differentiation mediated by CRT and IR.
Collapse
Affiliation(s)
- Andy Chi-Lung Lee
- Department of Radiation Oncology, Chang Gung Memorial Hospital, Linkou, Taiwan.,Radiation Biology Research Center, Institute for Radiological Research, Chang Gung Memorial Hospital/Chang Gung University, Taoyuan, Taiwan
| | - Yu-Yin Shih
- Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Fanfan Zhou
- Faculty of Pharmacy, University of Sydney, Sydney, NSW, Australia
| | - Tsi-Chian Chao
- Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Hsinyu Lee
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan.,Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Yung-Feng Liao
- Department of Life Science, National Taiwan University, Taipei, Taiwan.,Laboratory of Molecular Neurobiology, Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Wen-Ming Hsu
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan. .,Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.
| | - Ji-Hong Hong
- Department of Radiation Oncology, Chang Gung Memorial Hospital, Linkou, Taiwan. .,Radiation Biology Research Center, Institute for Radiological Research, Chang Gung Memorial Hospital/Chang Gung University, Taoyuan, Taiwan. .,Proton and Radiation Therapy Center, Chang Gung Memorial Hospital, No. 5, Fuxing 1st Rd., Guishan Dist., Taoyuan, 333, Taiwan.
| |
Collapse
|
46
|
Atluri R, Atmaramani R, Tharaka G, McCallister T, Peng J, Diercks D, GhoshMitra S, Ghosh S. Photo-Magnetic Irradiation-Mediated Multimodal Therapy of Neuroblastoma Cells Using a Cluster of Multifunctional Nanostructures. NANOMATERIALS 2018; 8:nano8100774. [PMID: 30274306 PMCID: PMC6215308 DOI: 10.3390/nano8100774] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 08/13/2018] [Accepted: 08/25/2018] [Indexed: 02/07/2023]
Abstract
The use of high intensity chemo-radiotherapies has demonstrated only modest improvement in the treatment of high-risk neuroblastomas. Moreover, undesirable drug specific and radiation therapy-incurred side effects enhance the risk of developing into a second cancer at a later stage. In this study, a safer and alternative multimodal therapeutic strategy involving simultaneous optical and oscillating (AC, Alternating Current) magnetic field stimulation of a multifunctional nanocarrier system has successfully been implemented to guide neuroblastoma cell destruction. This novel technique permitted the use of low-intensity photo-magnetic irradiation and reduced the required nanoparticle dose level. The combination of released cisplatin from the nanodrug reservoirs and photo-magnetic coupled hyperthermia mediated cytotoxicity led to the complete ablation of the B35 neuroblastoma cells in culture. Our study suggests that smart nanostructure-based photo-magnetic hybrid irradiation is a viable approach to remotely guide neuroblastoma cell destruction, which may be adopted in clinical management post modification to treat aggressive cancers.
Collapse
Affiliation(s)
- Rohini Atluri
- Nano-Bio Engineering Laboratory, Southeast Missouri State University, Cape Girardeau, MO 63701, USA.
- Mechanical and Energy Engineering Department, University of North Texas, Denton, TX 76207, USA.
| | - Rahul Atmaramani
- Nano-Bio Engineering Laboratory, Southeast Missouri State University, Cape Girardeau, MO 63701, USA.
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX 75080, USA.
| | - Gamage Tharaka
- Department of Physics and Engineering Physics, Southeast Missouri State University, Cape Girardeau, MO 63701, USA.
| | - Thomas McCallister
- Nano-Bio Engineering Laboratory, Southeast Missouri State University, Cape Girardeau, MO 63701, USA.
| | - Jian Peng
- Department of Physics and Engineering Physics, Southeast Missouri State University, Cape Girardeau, MO 63701, USA.
| | - David Diercks
- Department of Metallurgical and Materials Engineering, Colorado School of Mines, Golden, CO 80439, USA.
| | - Somesree GhoshMitra
- Nano-Bio Engineering Laboratory, Southeast Missouri State University, Cape Girardeau, MO 63701, USA.
| | - Santaneel Ghosh
- Nano-Bio Engineering Laboratory, Southeast Missouri State University, Cape Girardeau, MO 63701, USA.
- Department of Physics and Engineering Physics, Southeast Missouri State University, Cape Girardeau, MO 63701, USA.
| |
Collapse
|
47
|
Burkholderia Lethal Factor 1, a Novel Anti-Cancer Toxin, Demonstrates Selective Cytotoxicity in MYCN-Amplified Neuroblastoma Cells. Toxins (Basel) 2018; 10:toxins10070261. [PMID: 29954071 PMCID: PMC6071135 DOI: 10.3390/toxins10070261] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 06/15/2018] [Accepted: 06/20/2018] [Indexed: 11/16/2022] Open
Abstract
Immunotoxins are being investigated as anti-cancer therapies and consist of a cytotoxic enzyme fused to a cancer targeting antibody. All currently used toxins function via the inhibition of protein synthesis, making them highly potent in both healthy and transformed cells. This non-specific cell killing mechanism causes dose-limiting side effects that can severely limit the potential of immunotoxin therapy. In this study, the recently characterised bacterial toxin Burkholderia lethal factor 1 (BLF1) is investigated as a possible alternative payload for targeted toxin therapy in the treatment of neuroblastoma. BLF1 inhibits translation initiation by inactivation of eukaryotic initiation translation factor 4A (eIF4A), a putative anti-cancer target that has been shown to regulate a number of oncogenic proteins at the translational level. We show that cellular delivery of BLF1 selectively induces apoptosis in neuroblastoma cells that display MYCN amplification but has little effect on non-transformed cells. Future immunotoxins based on this enzyme may therefore have higher specificity towards MYCN-amplified cancer cells than more conventional ribosome-inactivating proteins, leading to an increased therapeutic window and decreased side effects.
Collapse
|
48
|
Zeid R, Lawlor MA, Poon E, Reyes JM, Fulciniti M, Lopez MA, Scott TG, Nabet B, Erb MA, Winter GE, Jacobson Z, Polaski DR, Karlin KL, Hirsch RA, Munshi NP, Westbrook TF, Chesler L, Lin CY, Bradner JE. Enhancer invasion shapes MYCN-dependent transcriptional amplification in neuroblastoma. Nat Genet 2018; 50:515-523. [PMID: 29379199 PMCID: PMC6310397 DOI: 10.1038/s41588-018-0044-9] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 12/18/2017] [Indexed: 11/08/2022]
Abstract
Amplification of the locus encoding the oncogenic transcription factor MYCN is a defining feature of high-risk neuroblastoma. Here we present the first dynamic chromatin and transcriptional landscape of MYCN perturbation in neuroblastoma. At oncogenic levels, MYCN associates with E-box binding motifs in an affinity-dependent manner, binding to strong canonical E-boxes at promoters and invading abundant weaker non-canonical E-boxes clustered at enhancers. Loss of MYCN leads to a global reduction in transcription, which is most pronounced at MYCN target genes with the greatest enhancer occupancy. These highly occupied MYCN target genes show tissue-specific expression and are linked to poor patient survival. The activity of genes with MYCN-occupied enhancers is dependent on the tissue-specific transcription factor TWIST1, which co-occupies enhancers with MYCN and is required for MYCN-dependent proliferation. These data implicate tissue-specific enhancers in defining often highly tumor-specific 'MYC target gene signatures' and identify disruption of the MYCN enhancer regulatory axis as a promising therapeutic strategy in neuroblastoma.
Collapse
Affiliation(s)
- Rhamy Zeid
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Matthew A Lawlor
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Evon Poon
- Division of Clinical Studies, The Institute of Cancer Research, London, UK
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, UK
| | - Jaime M Reyes
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Mariateresa Fulciniti
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Michael A Lopez
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Thomas G Scott
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Behnam Nabet
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Michael A Erb
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Georg E Winter
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Zoe Jacobson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Donald R Polaski
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kristen L Karlin
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Rachel A Hirsch
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Nikhil P Munshi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Thomas F Westbrook
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Louis Chesler
- Division of Clinical Studies, The Institute of Cancer Research, London, UK
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, UK
| | - Charles Y Lin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
| | - James E Bradner
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
- Novartis Institute for Biomedical Research, Cambridge, MA, USA.
| |
Collapse
|
49
|
Blancas S, Medina-Berlanga R, Ortíz-García L, Loredo-Ramírez A, Santos L. Protein Expression Analysis in Uterine Cervical Cancer for Potential Targets in Treatment. Pathol Oncol Res 2018. [PMID: 29532409 DOI: 10.1007/s12253-018-0401-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Specific markers in lesions of the human uterine cervix cancer (UCC) are still needed for prognostic, diagnostic and/or therapeutic purposes. In this study we evaluated key molecules at protein level between normal epithelium, cervical intraepithelial neoplasia (CIN1-3) and invasive cancer of a group of molecules previously reported at mRNA level. For that purpose, human formalin-fixed paraffin embedded tissue microarrays (TMAs) were constructed containing 205 Mexican tissue core specimens. Immunohistochemistry and quantitative analysis of histological staining was performed against twenty-two distinct proteins for each core and the processing platform ImageJ. In the progression of the disease we found key statistical differences for the proteins SEL1, Notch3 and SOCS3. High expressions of SEL1L, Notch3 and SOCS3 have potential value to increase the prognostic of UCC in combination with markers such as p16INK4a. This study identified key drivers in cervical carcinogenesis that should be evaluated for the development of UCC therapies.
Collapse
Affiliation(s)
- Sugela Blancas
- División de Biología Molecular, Instituto Potosino de Investigación Científica y Tecnológica, A.C. (IPICYT), San Luis Potosí, Mexico.,Centro de Ciencias de la Salud, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| | - Rogelio Medina-Berlanga
- División de Biología Molecular, Instituto Potosino de Investigación Científica y Tecnológica, A.C. (IPICYT), San Luis Potosí, Mexico
| | - Liliana Ortíz-García
- Facultad de Ingeniería en Biotecnología, Universidad Politécnica de Pénjamo, Pénjamo, Guanajuato, Mexico
| | - Alfredo Loredo-Ramírez
- Laboratorio de Patología Quirúrgica, Mariano Arista 743, Interior 208, San Luis Potosí, Mexico
| | - Leticia Santos
- División de Biología Molecular, Instituto Potosino de Investigación Científica y Tecnológica, A.C. (IPICYT), San Luis Potosí, Mexico.
| |
Collapse
|
50
|
Abstract
Neuroblastoma (NB) is the most common solid childhood tumor outside the brain and causes 15% of childhood cancer-related mortality. The main drivers of NB formation are neural crest cell-derived sympathoadrenal cells that undergo abnormal genetic arrangements. Moreover, NB is a complex disease that has high heterogeneity and is therefore difficult to target for successful therapy. Thus, a better understanding of NB development helps to improve treatment and increase the survival rate. One of the major causes of sporadic NB is known to be MYCN amplification and mutations in ALK (anaplastic lymphoma kinase) are responsible for familial NB. Many other genetic abnormalities can be found; however, they are not considered as driver mutations, rather they support tumor aggressiveness. Tumor cell elimination via cell death is widely accepted as a successful technique. Therefore, in this review, we provide a thorough overview of how different modes of cell death and treatment strategies, such as immunotherapy or spontaneous regression, are or can be applied for NB elimination. In addition, several currently used and innovative approaches and their suitability for clinical testing and usage will be discussed. Moreover, significant attention will be given to combined therapies that show more effective results with fewer side effects than drugs targeting only one specific protein or pathway.
Collapse
|