1
|
Mandonnet E, Funck-Brentano T, Hugnot JP, Touat M. Spectrum of IDH-mutant tumors in Ollier-Maffucci disease: the triple interaction theory. Orphanet J Rare Dis 2024; 19:434. [PMID: 39587599 PMCID: PMC11587773 DOI: 10.1186/s13023-024-03457-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 11/11/2024] [Indexed: 11/27/2024] Open
Abstract
We propose to refine our understanding of the pathophysiology underlying the tumor spectrum observed in patients with Ollier disease (OD) and Maffucci syndrome (MS). On one hand, assuming that all IDH-mutated tumors (as well as enchondromas) observed in OD-MS patients derive from one IDH-mutant cell giving rise to different lineages, the observation of different tumors arising in organs deriving from the neuroectoderm, mesoderm and endoderm points towards a very early post-zygotic event for the IDH mutation. To explain then that the spectrum of IDH-mutated tumors is restricted to some types of tumors, we propose the following hypothesis: - First, we posit that not every mutated cell of the lineage will "express" the IDH mutant phenotype. This can be due i/ to the disappearance in some tissue of the IDH-mutated clone due to negative selection pressure later in embryo development ii/ to the lack of expression of the IDH1 protein in specific cell types iii/ to a functional cell state not leading to the accumulation of the oncometabolite D-2-hydroxyglutarate (D-2HG) in that tissue/organ. - Second, generalizing the recent understanding of the gliomagenesis in the general population bearing the rs55705857 G-allele variant at 8q24.21, we postulate that OD-MS patients with an inheritable predisposing single nucleotide polymorphism (SNP) are more likely to develop a malignancy, with a specific SNP for each kind of tumor/organ. In summary, our theory provides a new understanding of IDH-mutated tumors in OD-MS patients, as arising from the triple interaction within the same cell of a developmental defect (the somatic mutation that occurs early during the embryogenesis), an organ-specific functional state "expressing" the IDH mutation and leading to an accumulation of D-2HG, and an inheritable predisposing factor (a risky SNP, also specific to each organ). We discuss how this theory could guide future research in OD-MS patients and, more generally, in patients harboring sporadic IDH-mutated tumors.
Collapse
Affiliation(s)
- Emmanuel Mandonnet
- Department of Neurosurgery, Lariboisière Hospital, AP-HP, 2 rue Ambroise Paré, Paris, 75010, France.
- Frontlab, CNRS UMR 7225, INSERM U1127, Paris Brain Institute (ICM), Paris, France.
- Université Paris Cité, Paris, France.
| | - Thomas Funck-Brentano
- Department of Rheumatology, Lariboisière Hospital, APHP.Nord, Université Paris Cité, Paris, France
- Université Paris Cité, Inserm U1132, BIOSCAR, Paris, F-75010, France
| | - Jean-Philippe Hugnot
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, 34091, France
| | - Mehdi Touat
- Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Service de Neuro-oncologie, Paris, France
- Department of Neurology, Brigham and Women's Hospital, Boston, USA
| |
Collapse
|
2
|
Hammon K, Renner K, Althammer M, Voll F, Babl N, Decking SM, Siska PJ, Matos C, Conejo ZEC, Mendes K, Einwag F, Siegmund H, Iberl S, Berger RS, Dettmer K, Schoenmehl R, Brochhausen C, Herr W, Oefner PJ, Rehli M, Thomas S, Kreutz M. D-2-hydroxyglutarate supports a tolerogenic phenotype with lowered major histocompatibility class II expression in non-malignant dendritic cells and acute myeloid leukemia cells. Haematologica 2024; 109:2500-2514. [PMID: 38235501 PMCID: PMC11290548 DOI: 10.3324/haematol.2023.283597] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 01/11/2024] [Indexed: 01/19/2024] Open
Abstract
D-2-hydroxyglutarate (D-2-HG) accumulates in patients with acute myeloid leukemia (AML) with mutated isocitrate dehydrogenase (IDH) and in other malignancies. D-2-HG suppresses antitumor T-cell immunity but little is known about potential effects on non-malignant myeloid cells. Here we show that D-2-HG impairs human but not murine dendritic cell differentiation, resulting in a tolerogenic phenotype with low major histocompatibility class II expression. In line with this, IDH-mutated AML blasts exhibited lower expression of HLA-DP and were less susceptible to lysis by HLA-DP-specific T cells. Interestingly, besides its expected impact on DNA demethylation, D-2-HG reprogrammed metabolism towards increased lactate production in dendritic cells and AML. Vitamin C accelerated DNA demethylation, but only the combination of vitamin C and glycolytic inhibition lowered lactate levels and supported major histocompatibility complex class II expression. Our results indicate an unexpected link between the immunosuppressive metabolites 2-HG and lactic acid and suggest a potentially novel therapeutic strategy with combinations of anti-glycolytic drugs and epigenetic modulators (hypomethylating agents) or other therapeutics for the treatment of AML.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/pathology
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Dendritic Cells/drug effects
- Glutarates/metabolism
- Glutarates/pharmacology
- Mice
- Animals
- Histocompatibility Antigens Class II/genetics
- Histocompatibility Antigens Class II/metabolism
- Phenotype
- Cell Differentiation/drug effects
- Lactic Acid/metabolism
- Immune Tolerance/drug effects
- Isocitrate Dehydrogenase/genetics
Collapse
Affiliation(s)
- Kathrin Hammon
- Department of Internal Medicine III, University Hospital Regensburg; Regensburg, Germany; LIT - Leibniz Institute for Immunotherapy; Regensburg
| | - Kathrin Renner
- Department of Internal Medicine III, University Hospital Regensburg; Regensburg, Germany; LIT - Leibniz Institute for Immunotherapy; Regensburg, Germany; Department of Otorhinolaryngology, University Hospital Regensburg, Regensburg
| | - Michael Althammer
- Department of Internal Medicine III, University Hospital Regensburg; Regensburg
| | - Florian Voll
- LIT - Leibniz Institute for Immunotherapy; Regensburg
| | - Nathalie Babl
- Department of Internal Medicine III, University Hospital Regensburg; Regensburg
| | - Sonja-Maria Decking
- Department of Otorhinolaryngology, University Hospital Regensburg, Regensburg
| | - Peter J Siska
- Department of Internal Medicine III, University Hospital Regensburg; Regensburg
| | - Carina Matos
- Department of Internal Medicine III, University Hospital Regensburg; Regensburg
| | | | - Karina Mendes
- Department of Internal Medicine III, University Hospital Regensburg; Regensburg, Germany; Present address: Universidade Católica Portuguesa, Center for Interdisciplinary Research in Health (CIIS), Institute of Health Sciences (ICS); Viseu
| | - Friederike Einwag
- Department of Internal Medicine III, University Hospital Regensburg; Regensburg
| | - Heiko Siegmund
- Institute of Pathology, University of Regensburg; Regensburg
| | - Sabine Iberl
- Department of Internal Medicine III, University Hospital Regensburg; Regensburg
| | - Raffaela S Berger
- Institute of Functional Genomics, University of Regensburg; Regensburg
| | - Katja Dettmer
- Institute of Functional Genomics, University of Regensburg; Regensburg
| | - Rebecca Schoenmehl
- Institute of Pathology, University Medical Center Mannheim, University Heidelberg, Mannheim
| | - Christoph Brochhausen
- Institute of Pathology, University of Regensburg; Regensburg, Germany; Institute of Pathology, University Medical Center Mannheim, University Heidelberg, Mannheim
| | - Wolfgang Herr
- Department of Internal Medicine III, University Hospital Regensburg; Regensburg
| | - Peter J Oefner
- Institute of Functional Genomics, University of Regensburg; Regensburg
| | - Michael Rehli
- Department of Internal Medicine III, University Hospital Regensburg; Regensburg, Germany; LIT - Leibniz Institute for Immunotherapy; Regensburg
| | - Simone Thomas
- Department of Internal Medicine III, University Hospital Regensburg; Regensburg, Germany; LIT - Leibniz Institute for Immunotherapy; Regensburg
| | - Marina Kreutz
- Department of Internal Medicine III, University Hospital Regensburg; Regensburg, Germany; LIT - Leibniz Institute for Immunotherapy; Regensburg.
| |
Collapse
|
3
|
Ülgen E, Gerlevik U, Gerlevik S, Oktay Y, Sezerman OU, Turcan Ş, Ozduman K. A microdeletion event at 19q13.43 in IDH-mutant astrocytomas is strongly correlated with MYC overexpression. Acta Neuropathol Commun 2024; 12:95. [PMID: 38877600 PMCID: PMC11177509 DOI: 10.1186/s40478-024-01811-1] [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: 04/06/2024] [Accepted: 06/02/2024] [Indexed: 06/16/2024] Open
Abstract
MYC dysregulation is pivotal in the onset and progression of IDH-mutant gliomas, mostly driven by copy-number alterations, regulatory element alterations, or epigenetic changes. Our pilot analysis uncovered instances of relative MYC overexpression without alterations in the proximal MYC network (PMN), prompting a deeper investigation into potential novel oncogenic mechanisms. Analysing comprehensive genomics profiles of 236 "IDH-mutant 1p/19q non-co-deleted" lower-grade gliomas from The Cancer Genome Atlas, we identified somatic genomic alterations within the PMN. In tumours without PMN-alterations but with MYC-overexpression, genes correlated with MYC-overexpression were identified. Our analyses yielded that 86/236 of astrocytomas exhibited no PMN-alterations, a subset of 21/86 displaying relative MYC overexpression. Within this subset, we discovered 42 genes inversely correlated with relative MYC expression, all on 19q. Further analysis pinpointed a minimal common region at 19q13.43, encompassing 15 genes. The inverse correlations of these 15 genes with relative MYC overexpression were re-confirmed using independent scRNAseq data. Further, the micro-deleted astrocytoma subset displayed significantly higher genomic instability compared to WT cases, but lower instability compared to PMN-hit cases. This newly identified 19q micro-deletion represents a potential novel mechanism underlying MYC dysregulation in astrocytomas. Given the prominence of 19q loss in IDH-mutant gliomas, our findings bear significant implications for understanding gliomagenesis.
Collapse
Affiliation(s)
- Ege Ülgen
- Department of Biostatistics and Medical Informatics, School of Medicine, Acibadem University, Istanbul, Turkey
- Department of Neurosurgery, School of Medicine, Acibadem University, 34752, Istanbul, Turkey
| | - Umut Gerlevik
- Department of Biochemistry, University of Oxford, Oxford, UK
- Faculty of Medicine, Ludwig Maximilian University of Munich, Munich, Germany
| | - Sıla Gerlevik
- Faculty of Life Sciences and Medicine, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, London, UK
| | - Yavuz Oktay
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Department of Medical Biology, Faculty of Medicine, Dokuz Eylül University, Izmir, Turkey
| | - Osman Uğur Sezerman
- Department of Biostatistics and Medical Informatics, School of Medicine, Acibadem University, Istanbul, Turkey
| | - Şevin Turcan
- Neurology Clinic and National Center for Tumor Diseases, Heidelberg University Hospital and Heidelberg University, Heidelberg, Germany
| | - Koray Ozduman
- Department of Neurosurgery, School of Medicine, Acibadem University, 34752, Istanbul, Turkey.
| |
Collapse
|
4
|
Nurminen R, Afyounian E, Paunu N, Katainen R, Isomäki M, Nurminen A, Scaravilli M, Tolppanen J, Fey V, Kivinen A, Helén P, Välimäki N, Kesseli J, Aaltonen LA, Haapasalo H, Nykter M, Rautajoki KJ. Previously reported CCDC26 risk variant and novel germline variants in GALNT13, AR, and MYO10 associated with familial glioma in Finland. Sci Rep 2024; 14:11562. [PMID: 38773237 PMCID: PMC11109329 DOI: 10.1038/s41598-024-62296-5] [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: 12/21/2023] [Accepted: 05/15/2024] [Indexed: 05/23/2024] Open
Abstract
Predisposing factors underlying familial aggregation of non-syndromic gliomas are still to be uncovered. Whole-exome sequencing was performed in four Finnish families with brain tumors to identify rare predisposing variants. A total of 417 detected exome variants and 102 previously reported glioma-related variants were further genotyped in 19 Finnish families with brain tumors using targeted sequencing. Rare damaging variants in GALNT13, MYO10 and AR were identified. Two families carried either c.553C>T (R185C) or c.1214T>A (L405Q) on GALNT13. Variant c.553C>T is located on the substrate-binding site of GALNT13. AR c.2180G>T (R727L), which is located on a ligand-binding domain of AR, was detected in two families, one of which also carried a GALNT13 variant. MYO10 c.4448A>G (N1483S) was detected in two families and c.1511C>T (A504V) variant was detected in one family. Both variants are located on functional domains related to MYO10 activity in filopodia formation. In addition, affected cases in six families carried a known glioma risk variant rs55705857 in CCDC26 and low-risk glioma variants. These novel findings indicate polygenic inheritance of familial glioma in Finland and increase our understanding of the genetic contribution to familial glioma susceptibility.
Collapse
Affiliation(s)
- Riikka Nurminen
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland
- Tays Cancer Center, Tampere University Hospital, Tampere, Finland
| | - Ebrahim Afyounian
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland
- Tays Cancer Center, Tampere University Hospital, Tampere, Finland
| | - Niina Paunu
- Department of Oncology, Tampere University Hospital, Tampere, Finland
| | - Riku Katainen
- Applied Tumor Genomics Research Program, Department of Medical and Clinical Genetics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mari Isomäki
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland
- Tays Cancer Center, Tampere University Hospital, Tampere, Finland
| | - Anssi Nurminen
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland
- Tays Cancer Center, Tampere University Hospital, Tampere, Finland
| | - Mauro Scaravilli
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland
- Tays Cancer Center, Tampere University Hospital, Tampere, Finland
| | - Jenni Tolppanen
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland
- Tays Cancer Center, Tampere University Hospital, Tampere, Finland
| | - Vidal Fey
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland
- Tays Cancer Center, Tampere University Hospital, Tampere, Finland
| | - Anni Kivinen
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland
- Tays Cancer Center, Tampere University Hospital, Tampere, Finland
| | - Pauli Helén
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland
- Tays Cancer Center, Tampere University Hospital, Tampere, Finland
| | - Niko Välimäki
- Applied Tumor Genomics Research Program, Department of Medical and Clinical Genetics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Juha Kesseli
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland
- Tays Cancer Center, Tampere University Hospital, Tampere, Finland
| | - Lauri A Aaltonen
- Applied Tumor Genomics Research Program, Department of Medical and Clinical Genetics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Hannu Haapasalo
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland
- Tays Cancer Center, Tampere University Hospital, Tampere, Finland
- Fimlab Laboratories ltd., Tampere University Hospital, Tampere, Finland
| | - Matti Nykter
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland.
- Tays Cancer Center, Tampere University Hospital, Tampere, Finland.
- Foundation for the Finnish Cancer Institute, Tukholmankatu 8, Helsinki, Finland.
| | - Kirsi J Rautajoki
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland.
- Tays Cancer Center, Tampere University Hospital, Tampere, Finland.
- Tampere Institute for Advanced Study, Tampere University, Tampere, Finland.
| |
Collapse
|
5
|
Vízkeleti L, Spisák S. Rewired Metabolism Caused by the Oncogenic Deregulation of MYC as an Attractive Therapeutic Target in Cancers. Cells 2023; 12:1745. [PMID: 37443779 PMCID: PMC10341379 DOI: 10.3390/cells12131745] [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: 04/10/2023] [Revised: 06/20/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
MYC is one of the most deregulated oncogenes on multiple levels in cancer. As a node transcription factor, MYC plays a diverse regulatory role in many cellular processes, including cell cycle and metabolism, both in physiological and pathological conditions. The relentless growth and proliferation of tumor cells lead to an insatiable demand for energy and nutrients, which requires the rewiring of cellular metabolism. As MYC can orchestrate all aspects of cellular metabolism, its altered regulation plays a central role in these processes, such as the Warburg effect, and is a well-established hallmark of cancer development. However, our current knowledge of MYC suggests that its spatial- and concentration-dependent contribution to tumorigenesis depends more on changes in the global or relative expression of target genes. As the direct targeting of MYC is proven to be challenging due to its relatively high toxicity, understanding its underlying regulatory mechanisms is essential for the development of tumor-selective targeted therapies. The aim of this review is to comprehensively summarize the diverse forms of MYC oncogenic deregulation, including DNA-, transcriptional- and post-translational level alterations, and their consequences for cellular metabolism. Furthermore, we also review the currently available and potentially attractive therapeutic options that exploit the vulnerability arising from the metabolic rearrangement of MYC-driven tumors.
Collapse
Affiliation(s)
- Laura Vízkeleti
- Department of Bioinformatics, Faculty of Medicine, Semmelweis University, 1094 Budapest, Hungary;
| | - Sándor Spisák
- Institute of Enzymology, Research Centre for Natural Sciences, Eötvös Loránd Research Network, 1117 Budapest, Hungary
| |
Collapse
|
6
|
Choi DJ, Armstrong G, Lozzi B, Vijayaraghavan P, Plon SE, Wong TC, Boerwinkle E, Muzny DM, Chen HC, Gibbs RA, Ostrom QT, Melin B, Deneen B, Bondy ML, The Gliogene Consortium, Genomics England Research Consortium, Bainbridge MN. The genomic landscape of familial glioma. SCIENCE ADVANCES 2023; 9:eade2675. [PMID: 37115922 PMCID: PMC10146888 DOI: 10.1126/sciadv.ade2675] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 03/22/2023] [Indexed: 05/03/2023]
Abstract
Glioma is a rare brain tumor with a poor prognosis. Familial glioma is a subset of glioma with a strong genetic predisposition that accounts for approximately 5% of glioma cases. We performed whole-genome sequencing on an exploratory cohort of 203 individuals from 189 families with a history of familial glioma and an additional validation cohort of 122 individuals from 115 families. We found significant enrichment of rare deleterious variants of seven genes in both cohorts, and the most significantly enriched gene was HERC2 (P = 0.0006). Furthermore, we identified rare noncoding variants in both cohorts that were predicted to affect transcription factor binding sites or cause cryptic splicing. Last, we selected a subset of discovered genes for validation by CRISPR knockdown screening and found that DMBT1, HP1BP3, and ZCH7B3 have profound impacts on proliferation. This study performs comprehensive surveillance of the genomic landscape of familial glioma.
Collapse
Affiliation(s)
- Dong-Joo Choi
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Georgina Armstrong
- Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA, USA
| | - Brittney Lozzi
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| | | | - Sharon E. Plon
- Department of Pediatrics/Hematology-Oncology, Baylor College of Medicine, Houston, TX, USA
| | - Terence C. Wong
- Rady Children’s Institute for Genomic Medicine, San Diego, CA, USA
| | - Eric Boerwinkle
- The University of Texas Health Science Center School of Public Health, Houston, TX, USA
| | - Donna M. Muzny
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Hsiao-Chi Chen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Richard A. Gibbs
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Quinn T. Ostrom
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC, USA
| | - Beatrice Melin
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| | - Benjamin Deneen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Melissa L. Bondy
- Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA, USA
| | - The Gliogene Consortium
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
- Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA, USA
- Rady Children’s Institute for Genomic Medicine, San Diego, CA, USA
- Department of Pediatrics/Hematology-Oncology, Baylor College of Medicine, Houston, TX, USA
- The University of Texas Health Science Center School of Public Health, Houston, TX, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC, USA
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| | - Genomics England Research Consortium
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
- Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA, USA
- Rady Children’s Institute for Genomic Medicine, San Diego, CA, USA
- Department of Pediatrics/Hematology-Oncology, Baylor College of Medicine, Houston, TX, USA
- The University of Texas Health Science Center School of Public Health, Houston, TX, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC, USA
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| | | |
Collapse
|
7
|
Yanchus C, Drucker KL, Kollmeyer TM, Tsai R, Winick-Ng W, Liang M, Malik A, Pawling J, De Lorenzo SB, Ali A, Decker PA, Kosel ML, Panda A, Al-Zahrani KN, Jiang L, Browning JWL, Lowden C, Geuenich M, Hernandez JJ, Gosio JT, Ahmed M, Loganathan SK, Berman J, Trcka D, Michealraj KA, Fortin J, Carson B, Hollingsworth EW, Jacinto S, Mazrooei P, Zhou L, Elia A, Lupien M, He HH, Murphy DJ, Wang L, Abyzov A, Dennis JW, Maass PG, Campbell K, Wilson MD, Lachance DH, Wrensch M, Wiencke J, Mak T, Pennacchio LA, Dickel DE, Visel A, Wrana J, Taylor MD, Zadeh G, Dirks P, Eckel-Passow JE, Attisano L, Pombo A, Ida CM, Kvon EZ, Jenkins RB, Schramek D. A noncoding single-nucleotide polymorphism at 8q24 drives IDH1-mutant glioma formation. Science 2022; 378:68-78. [PMID: 36201590 PMCID: PMC9926876 DOI: 10.1126/science.abj2890] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Establishing causal links between inherited polymorphisms and cancer risk is challenging. Here, we focus on the single-nucleotide polymorphism rs55705857, which confers a sixfold greater risk of isocitrate dehydrogenase (IDH)-mutant low-grade glioma (LGG). We reveal that rs55705857 itself is the causal variant and is associated with molecular pathways that drive LGG. Mechanistically, we show that rs55705857 resides within a brain-specific enhancer, where the risk allele disrupts OCT2/4 binding, allowing increased interaction with the Myc promoter and increased Myc expression. Mutating the orthologous mouse rs55705857 locus accelerated tumor development in an Idh1R132H-driven LGG mouse model from 472 to 172 days and increased penetrance from 30% to 75%. Our work reveals mechanisms of the heritable predisposition to lethal glioma in ~40% of LGG patients.
Collapse
Affiliation(s)
- Connor Yanchus
- Centre for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Kristen L. Drucker
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Thomas M. Kollmeyer
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Ricky Tsai
- Centre for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Warren Winick-Ng
- Max-Delbrück Centre for Molecular Medicine, Berlin Institute for Medical Systems Biology, Epigenetic Regulation and Chromatin Architecture Group, 13092 Berlin, Germany
| | - Minggao Liang
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
- Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Ahmad Malik
- Centre for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Judy Pawling
- Centre for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Silvana B. De Lorenzo
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Asma Ali
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Paul A. Decker
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Matt L. Kosel
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Arijit Panda
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Khalid N. Al-Zahrani
- Centre for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Lingyan Jiang
- Centre for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Jared W. L. Browning
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
- Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Chris Lowden
- Centre for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Michael Geuenich
- Centre for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - J. Javier Hernandez
- Centre for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jessica T. Gosio
- Centre for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | | | - Sampath Kumar Loganathan
- Centre for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Jacob Berman
- Centre for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Daniel Trcka
- Centre for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | | | - Jerome Fortin
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada
| | - Brittany Carson
- Centre for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Ethan W. Hollingsworth
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92617, USA
| | - Sandra Jacinto
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92617, USA
| | - Parisa Mazrooei
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Lily Zhou
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada
| | - Andrew Elia
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada
| | - Mathieu Lupien
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
- Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada
| | - Housheng Hansen He
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Daniel J. Murphy
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1BD, Scotland, UK
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, Scotland, UK
| | - Liguo Wang
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Alexej Abyzov
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - James W. Dennis
- Centre for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Philipp G. Maass
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
- Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Kieran Campbell
- Centre for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Michael D. Wilson
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
- Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Daniel H. Lachance
- Departments of Neurology and Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Margaret Wrensch
- Department of Neurological Surgery, University of California, San Francisco, CA 94143, USA
| | - John Wiencke
- Department of Neurological Surgery, University of California, San Francisco, CA 94143, USA
| | - Tak Mak
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Len A. Pennacchio
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94710, USA
- Comparative Biochemistry Program, University of California, Berkeley, CA 94720, USA
- US Department of Energy Joint Genome Institute, Berkeley, CA 94720, USA
| | - Diane E. Dickel
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94710, USA
| | - Axel Visel
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94710, USA
- US Department of Energy Joint Genome Institute, Berkeley, CA 94720, USA
- School of Natural Sciences, University of California, Merced, CA 95343, USA
| | - Jeffrey Wrana
- Centre for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Michael D. Taylor
- Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Gelareh Zadeh
- Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Peter Dirks
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
- Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | | | - Liliana Attisano
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
- Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Ana Pombo
- Max-Delbrück Centre for Molecular Medicine, Berlin Institute for Medical Systems Biology, Epigenetic Regulation and Chromatin Architecture Group, 13092 Berlin, Germany
- Institute of Biology, Humboldt University of Berlin, 10115 Berlin, Germany
| | - Cristiane M. Ida
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Evgeny Z. Kvon
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92617, USA
| | - Robert B. Jenkins
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Daniel Schramek
- Centre for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
8
|
Jarabo P, de Pablo C, González-Blanco A, Casas-Tintó S. Circadian Gene cry Controls Tumorigenesis through Modulation of Myc Accumulation in Glioblastoma Cells. Int J Mol Sci 2022; 23:ijms23042043. [PMID: 35216153 PMCID: PMC8874709 DOI: 10.3390/ijms23042043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/04/2022] [Accepted: 02/08/2022] [Indexed: 12/04/2022] Open
Abstract
Glioblastoma (GB) is the most frequent malignant brain tumor among adults and currently there is no effective treatment. This aggressive tumor grows fast and spreads through the brain causing death in 15 months. GB cells display a high mutation rate and generate a heterogeneous population of tumoral cells that are genetically distinct. Thus, the contribution of genes and signaling pathways relevant for GB progression is of great relevance. We used a Drosophila model of GB that reproduces the features of human GB and describe the upregulation of the circadian gene cry in GB patients and in a Drosophila GB model. We studied the contribution of cry to the expansion of GB cells and the neurodegeneration and premature death caused by GB, and we determined that cry is required for GB progression. Moreover, we determined that the PI3K pathway regulates cry expression in GB cells, and in turn, cry is necessary and sufficient to promote Myc accumulation in GB. These results contribute to understanding the mechanisms underlying GB malignancy and lethality, and describe a novel role of Cry in GB cells.
Collapse
|
9
|
Choi HI, An GY, Yoo E, Baek M, Chai JC, Binas B, Lee YS, Jung KH, Chai YG. Targeting of noncoding RNAs encoded by a novel MYC enhancers inhibits the proliferation of human hepatic carcinoma cells in vitro. Sci Rep 2022; 12:855. [PMID: 35039581 PMCID: PMC8764030 DOI: 10.1038/s41598-022-04869-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/31/2021] [Indexed: 12/13/2022] Open
Abstract
The proto-oncogene MYC is important for development and cell growth, however, its abnormal regulation causes cancer. Recent studies identified distinct enhancers of MYC in various cancers, but any MYC enhancer(s) in hepatocellular carcinoma (HCC) remain(s) elusive. By analyzing H3K27ac enrichment and enhancer RNA (eRNA) expression in cultured HCC cells, we identified six putative MYC enhancer regions. Amongst these, two highly active enhancers, located ~ 800 kb downstream of the MYC gene, were identified by qRT-PCR and reporter assays. We functionally confirmed these enhancers by demonstrating a significantly reduced MYC expression and cell proliferation upon CRISPR/Cas9-based deletion and/or antisense oligonucleotide (ASO)-mediated inhibition. In conclusion, we identified potential MYC enhancers of HCC and propose that the associated eRNAs may be suitable targets for HCC treatment.
Collapse
Affiliation(s)
- Hae In Choi
- Department of Bionanotechnology, Hanyang University, Seoul, 04673, Republic of Korea
| | - Ga Yeong An
- Department of Bionanotechnology, Hanyang University, Seoul, 04673, Republic of Korea
| | - Eunyoung Yoo
- Department of Bionanotechnology, Hanyang University, Seoul, 04673, Republic of Korea
| | - Mina Baek
- Department of Molecular & Life Science, Hanyang University, Ansan, Gyeonggi-do, 15588, Republic of Korea
- Institute of Natural Science and Technology, Hanyang University, Ansan, 15588, Republic of Korea
| | - Jin Choul Chai
- College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Bert Binas
- Department of Molecular & Life Science, Hanyang University, Ansan, Gyeonggi-do, 15588, Republic of Korea
| | - Young Seek Lee
- College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Kyoung Hwa Jung
- Convergence Technology Campus of Korea Polytechnic II, Incheon, 21417, Republic of Korea.
- Department of Biopharmaceutical Systems, Gwangmyeong Convergence Technology Campus of Korea Polytechnic II, Gwangmyeong-si, Gyeonggi-do, 14222, Republic of Korea.
| | - Young Gyu Chai
- Department of Bionanotechnology, Hanyang University, Seoul, 04673, Republic of Korea.
- Department of Molecular & Life Science, Hanyang University, Ansan, Gyeonggi-do, 15588, Republic of Korea.
| |
Collapse
|
10
|
Abstract
Shadow enhancers are seemingly redundant transcriptional cis-regulatory elements that regulate the same gene and drive overlapping expression patterns. Recent studies have shown that shadow enhancers are remarkably abundant and control most developmental gene expression in both invertebrates and vertebrates, including mammals. Shadow enhancers might provide an important mechanism for buffering gene expression against mutations in non-coding regulatory regions of genes implicated in human disease. Technological advances in genome editing and live imaging have shed light on how shadow enhancers establish precise gene expression patterns and confer phenotypic robustness. Shadow enhancers can interact in complex ways and may also help to drive the formation of transcriptional hubs within the nucleus. Despite their apparent redundancy, the prevalence and evolutionary conservation of shadow enhancers underscore their key role in emerging metazoan gene regulatory networks.
Collapse
|
11
|
Ülgen E, Karacan S, Gerlevik U, Can Ö, Bilguvar K, Oktay Y, B. Akyerli C, K. Yüksel Ş, E. Danyeli A, Tihan T, Sezerman OU, Yakıcıer MC, Pamir MN, Özduman K. Mutations and Copy Number Alterations in IDH Wild-Type Glioblastomas Are Shaped by Different Oncogenic Mechanisms. Biomedicines 2020; 8:E574. [PMID: 33297360 PMCID: PMC7762325 DOI: 10.3390/biomedicines8120574] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/27/2020] [Accepted: 11/03/2020] [Indexed: 12/29/2022] Open
Abstract
Little is known about the mutational processes that shape the genetic landscape of gliomas. Numerous mutational processes leave marks on the genome in the form of mutations, copy number alterations, rearrangements or their combinations. To explore gliomagenesis, we hypothesized that gliomas with different underlying oncogenic mechanisms would have differences in the burden of various forms of these genomic alterations. This was an analysis on adult diffuse gliomas, but IDH-mutant gliomas as well as diffuse midline gliomas H3-K27M were excluded to search for the possible presence of new entities among the very heterogenous group of IDH-WT glioblastomas. The cohort was divided into two molecular subsets: (1) Molecularly-defined GBM (mGBM) as those that carried molecular features of glioblastomas (including TERT promoter mutations, 7/10 pattern, or EGFR-amplification), and (2) those who did not (others). Whole exome sequencing was performed for 37 primary tumors and matched blood samples as well as 8 recurrences. Single nucleotide variations (SNV), short insertion or deletions (indels) and copy number alterations (CNA) were quantified using 5 quantitative metrics (SNV burden, indel burden, copy number alteration frequency-wGII, chromosomal arm event ratio-CAER, copy number amplitude) as well as 4 parameters that explored underlying oncogenic mechanisms (chromothripsis, double minutes, microsatellite instability and mutational signatures). Findings were validated in the TCGA pan-glioma cohort. mGBM and "Others" differed significantly in their SNV (only in the TCGA cohort) and CNA metrics but not indel burden. SNV burden increased with increasing age at diagnosis and at recurrences and was driven by mismatch repair deficiency. On the contrary, indel and CNA metrics remained stable over increasing age at diagnosis and with recurrences. Copy number alteration frequency (wGII) correlated significantly with chromothripsis while CAER and CN amplitude correlated significantly with the presence of double minutes, suggesting separate underlying mechanisms for different forms of CNA.
Collapse
Affiliation(s)
- Ege Ülgen
- Department of Biostatistics and Medical Informatics, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey; (E.Ü.); (S.K.); (U.G.); (O.U.S.)
| | - Sıla Karacan
- Department of Biostatistics and Medical Informatics, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey; (E.Ü.); (S.K.); (U.G.); (O.U.S.)
| | - Umut Gerlevik
- Department of Biostatistics and Medical Informatics, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey; (E.Ü.); (S.K.); (U.G.); (O.U.S.)
| | - Özge Can
- Department of Medical Engineering, Faculty of Engineering, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey;
| | - Kaya Bilguvar
- Department of Genetics, School of Medicine, Yale University, New Haven, CT 06520, USA;
- Yale Center for Genome Analysis, West Haven, CT 06516, USA
| | - Yavuz Oktay
- Izmir Biomedicine and Genome Center (IBG), Izmir 35340, Turkey;
- Department of Medical Biology, Faculty of Medicine, Dokuz Eylül University, Izmir 35340, Turkey
| | - Cemaliye B. Akyerli
- Department of Medical Biology, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey; (C.B.A.); (Ş.K.Y.)
| | - Şirin K. Yüksel
- Department of Medical Biology, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey; (C.B.A.); (Ş.K.Y.)
| | - Ayça E. Danyeli
- Department of Pathology, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey;
| | - Tarık Tihan
- Neuropathology Division, Department of Pathology, School of Medicine, University of California San Fransisco (UCSF), San Francisco, CA 94143, USA;
| | - O. Uğur Sezerman
- Department of Biostatistics and Medical Informatics, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey; (E.Ü.); (S.K.); (U.G.); (O.U.S.)
| | - M. Cengiz Yakıcıer
- Department of Molecular Biology, School of Arts and Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey;
| | - M. Necmettin Pamir
- Department of Neurosurgery, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey;
| | - Koray Özduman
- Department of Neurosurgery, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey;
| |
Collapse
|
12
|
Tong Y, Tang Y, Li S, Zhao F, Ying J, Qu Y, Niu X, Mu D. Cumulative evidence of relationships between multiple variants in 8q24 region and cancer incidence. Medicine (Baltimore) 2020; 99:e20716. [PMID: 32590746 PMCID: PMC7328976 DOI: 10.1097/md.0000000000020716] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Genome-wide association studies (GWAS) have identified multiple independent cancer susceptibility loci at chromosome 8q24. We aimed to evaluate the associations between variants in the 8q24 region and cancer susceptibility. A comprehensive research synopsis and meta-analysis was performed to evaluate associations between 28 variants in 8q24 and risk of 7 cancers using data from 103 eligible articles totaling 146,932 cancer cases and 219,724 controls. Results: 20 variants were significantly associated with risk of prostate cancer, colorectal cancer, thyroid cancer, breast cancer, bladder cancer, stomach cancer, and glioma, including 1 variant associated with prostate cancer, colorectal cancer, and thyroid cancer. Cumulative epidemiological evidence of an association was graded as strong for DG8S737 -8 allele, rs10090154, rs7000448 in prostate cancer, rs10808556 in colorectal cancer, rs55705857 in gliomas, rs9642880 in bladder cancer, moderate for rs16901979, rs1447295, rs6983267, rs7017300, rs7837688, rs1016343, rs620861, rs10086908 associated in prostate cancer, rs10505477, rs6983267 in colorectal cancer, rs6983267 in thyroid cancer, rs13281615 in breast cancer, and rs1447295 in stomach cancer, weak for rs6983561, rs13254738, rs7008482, rs4242384 in prostate cancer. Data from ENCODE suggested that these variants with strong evidence and other correlated variants might fall within putative functional regions. Our study provides summary evidence that common variants in the 8q24 are associated with risk of multiple cancers in this large-scale research synopsis and meta-analysis. Further studies are needed to explore the mechanisms underlying variants in the 8q24 involved in various human cancers.
Collapse
Affiliation(s)
- Yu Tong
- Department of Pediatrics
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education
| | - Ying Tang
- Department of Pediatrics
- Department of Diagnostic Ultrasound
| | - Shiping Li
- Department of Pediatrics
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education
| | - Fengyan Zhao
- Department of Pediatrics
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education
| | - Junjie Ying
- Department of Pediatrics
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education
| | - Yi Qu
- Department of Pediatrics
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education
| | - Xiaoyu Niu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Dezhi Mu
- Department of Pediatrics
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education
| |
Collapse
|
13
|
Ülgen E, Can Ö, Bilguvar K, Oktay Y, Akyerli CB, Danyeli AE, Yakıcıer MC, Sezerman OU, Pamir MN, Özduman K. Whole exome sequencing-based analysis to identify DNA damage repair deficiency as a major contributor to gliomagenesis in adult diffuse gliomas. J Neurosurg 2020; 132:1435-1446. [PMID: 30952131 DOI: 10.3171/2019.1.jns182938] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 01/10/2019] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Processes that cause or contribute to cancer, such as aging, exposure to carcinogens, or DNA damage repair deficiency (DDRd), create predictable and traceable nucleotide alterations in one's genetic code (termed "mutational signatures"). Large studies have previously identified various such mutational signatures across cancers that can be attributed to the specific causative processes. To gain further insight into the processes in glioma development, the authors analyzed mutational signatures in adult diffuse gliomas (DGs). METHODS Twenty-five DGs and paired blood samples were whole exome sequenced. Somatic mutational signatures were identified using 2 different methods. Associations of the signatures with age at diagnosis, molecular subset, and mutational load were investigated. As DDRd-related signatures were frequently observed, germline and somatic DDR gene mutations as well as microsatellite instability (MSI) status were determined for all samples. For validation of signature prevalence, publicly available data from The Cancer Genome Atlas (TCGA) were used. RESULTS Each tumor had a unique combination of signatures. The most common signatures were signature 1 (88%, aging related), signature 3 (52%, homologous recombination related), and signature 15 (56%, mismatch repair related). Eighty-four percent of the tumors contained at least 1 DDRd signature. The findings were validated using public TCGA data. The weight of signature 1 positively correlated with age (r = 0.43) while cumulative weight of DDRd signatures negatively correlated with age (r = -0.16). Each subject had at least 1 germline/somatic alteration in a DDR gene, the most common being the risk single nucleotide polymorphism rs1800734 in MLH1. The rs1800734-AA genotype had a higher cumulative DDRd weight as well as higher mutational load; TP53 was the most common somatically altered DDR gene. MSI was observed in 24% of the tumors. No significant associations of MSI status with mutational load, rs1800734, or the cumulative weight of DDRd signatures were identified. CONCLUSIONS Current findings suggest that DDRd may act as a fundamental mechanism in gliomagenesis rather than being a random, secondary event.
Collapse
Affiliation(s)
- Ege Ülgen
- Departments of1Biostatistics and Medical Informatics
| | - Özge Can
- 2Department of Medical Engineering, Acibadem Mehmet Ali Aydınlar University, School of Engineering, Istanbul, Turkey
| | - Kaya Bilguvar
- 3Department of Genetics, Yale Center for Genome Analysis, Yale University, School of Medicine, New Haven, Connecticut
| | - Yavuz Oktay
- 4Izmir International Biomedicine and Genome Institute (IBG), Dokuz Eylül University, Izmir
- 5Department of Medical Biology, Dokuz Eylül University, Izmir; and
| | | | | | - M Cengiz Yakıcıer
- 8Department of Molecular Biology, Acibadem Mehmet Ali Aydınlar University, School of Arts and Sciences, Istanbul, Turkey
| | | | - M Necmettin Pamir
- 9Neurosurgery, Acibadem Mehmet Ali Aydınlar University, School of Medicine, Istanbul
| | - Koray Özduman
- 9Neurosurgery, Acibadem Mehmet Ali Aydınlar University, School of Medicine, Istanbul
| |
Collapse
|
14
|
Lan Y, Lou J, Hu J, Yu Z, Lyu W, Zhang B. Downregulation of SNRPG induces cell cycle arrest and sensitizes human glioblastoma cells to temozolomide by targeting Myc through a p53-dependent signaling pathway. Cancer Biol Med 2020; 17:112-131. [PMID: 32296580 PMCID: PMC7142844 DOI: 10.20892/j.issn.2095-3941.2019.0164] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 08/29/2019] [Indexed: 12/17/2022] Open
Abstract
Objective: Temozolomide (TMZ) is commonly used for glioblastoma multiforme (GBM) chemotherapy. However, drug resistance limits its therapeutic effect in GBM treatment. RNA-binding proteins (RBPs) have vital roles in posttranscriptional events. While disturbance of RBP-RNA network activity is potentially associated with cancer development, the precise mechanisms are not fully known. The SNRPG gene, encoding small nuclear ribonucleoprotein polypeptide G, was recently found to be related to cancer incidence, but its exact function has yet to be elucidated. Methods:SNRPG knockdown was achieved via short hairpin RNAs. Gene expression profiling and Western blot analyses were used to identify potential glioma cell growth signaling pathways affected by SNRPG. Xenograft tumors were examined to determine the carcinogenic effects of SNRPG on glioma tissues. Results: The SNRPG-mediated inhibitory effect on glioma cells might be due to the targeted prevention of Myc and p53. In addition, the effects of SNRPG loss on p53 levels and cell cycle progression were found to be Myc-dependent. Furthermore, SNRPG was increased in TMZ-resistant GBM cells, and downregulation of SNRPG potentially sensitized resistant cells to TMZ, suggesting that SNRPG deficiency decreases the chemoresistance of GBM cells to TMZ via the p53 signaling pathway. Our data confirmed that SNRPG suppression sensitizes GBM cells to TMZ by targeting Myc via the p53 signaling cascade. Conclusions: These results indicated that SNRPG is a probable molecular target of GBM and suggested that suppressing SNRPG in resistant GBM cells might be a substantially beneficial method for overcoming essential drug resistance.
Collapse
Affiliation(s)
- Yulong Lan
- Department of Neurosurgery, Shenzhen People’s Hospital, Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518020, China
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| | - Jiacheng Lou
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| | - Jiliang Hu
- Department of Neurosurgery, Shenzhen People’s Hospital, Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518020, China
| | - Zhikuan Yu
- Department of Neurosurgery, Shenzhen People’s Hospital, Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518020, China
| | - Wen Lyu
- Department of Neurosurgery, Shenzhen People’s Hospital, Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518020, China
| | - Bo Zhang
- Department of Neurosurgery, Shenzhen People’s Hospital, Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518020, China
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| |
Collapse
|
15
|
The Genetic Architecture of Gliomagenesis-Genetic Risk Variants Linked to Specific Molecular Subtypes. Cancers (Basel) 2019; 11:cancers11122001. [PMID: 31842352 PMCID: PMC6966482 DOI: 10.3390/cancers11122001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 12/05/2019] [Accepted: 12/07/2019] [Indexed: 12/19/2022] Open
Abstract
Genome-wide association studies have identified 25 germline genetic loci that increase the risk of glioma. The somatic tumor molecular alterations, including IDH-mutation status and 1p/19q co-deletion, have been included into the WHO 2016 classification system for glioma. To investigate how the germline genetic risk variants correlate with the somatic molecular subtypes put forward by WHO, we performed a meta-analysis that combined findings from 330 Swedish cases and 876 controls with two other recent studies. In total, 5,103 cases and 10,915 controls were included. Three categories of associations were found. First, variants in TERT and TP53 were associated with increased risk of all glioma subtypes. Second, variants in CDKN2B-AS1, EGFR, and RTEL1 were associated with IDH-wildtype glioma. Third, variants in CCDC26 (the 8q24 locus), C2orf80 (close to IDH), LRIG1, PHLDB1, ETFA, MAML2 and ZBTB16 were associated with IDH-mutant glioma. We therefore propose three etiopathological pathways in gliomagenesis based on germline variants for future guidance of diagnosis and potential functional targets for therapies. Future prospective clinical trials of patients with suspicion of glioma diagnoses, using the genetic variants as biomarkers, are necessary to disentangle how strongly they can predict glioma diagnosis.
Collapse
|
16
|
Tong Y, Ye L, Li S, Zhao F, Ying J, Qu Y, Li J, Mu D. The association of 6 variants of 8q24 and the risk of glioma: A meta-analysis. Medicine (Baltimore) 2019; 98:e16205. [PMID: 31277128 PMCID: PMC6635291 DOI: 10.1097/md.0000000000016205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
With the advances in sequencing technologies and genome-wide association studies (GWAS), several inherited variants that increase glioma risk have been identified. Ten studies including 8818 cases and 17,551 controls were collected to conduct a meta-analysis to evaluate the associations between 6 variants in 8q24 and glioma risk. Of the 6 variants located in 8q24, 2 have strong significant associations with the risk of glioma, including rs4295627 (P = .003, odds ratio [OR] = 1.21), rs55705857 (P = 2.31 × 10, OR = 3.54). In particular, both homozygous GG (P = 1.91 × 10, OR1 = 2.01) and heterozygous GT (P = 7.75 × 10, OR2 = 1.35) genotypes of rs4295627 were associated with glioma risk. Further studies are needed to explore the role of the 8q24 variants involved in the etiology of glioma.
Collapse
Affiliation(s)
- Yu Tong
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan Province
| | - Lv Ye
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan Province
| | - Shiping Li
- Chendu Gaoxin-Daan Medical Laboratory Co Ltd Pathology Lab, China
| | - Fengyan Zhao
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan Province
| | - Junjie Ying
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan Province
| | - Yi Qu
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan Province
| | - Jinhui Li
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan Province
| | - Dezhi Mu
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan Province
| |
Collapse
|
17
|
Hummel S, Kohlmann W, Kollmeyer TM, Jenkins R, Sonnen J, Palmer CA, Colman H, Abbott D, Cannon-Albright L, Cohen AL. The contribution of the rs55705857 G allele to familial cancer risk as estimated in the Utah population database. BMC Cancer 2019; 19:190. [PMID: 30823903 PMCID: PMC6397494 DOI: 10.1186/s12885-019-5381-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 02/19/2019] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND IDH1/2 mutated glioma has been associated with a germline risk variant, the rs55705857 G allele. The Utah Population Database (UPDB), a computerized genealogy of people in Utah, is a unique resource to evaluate cancer risk in related individuals. METHODS One hundred and two individuals with IDH1/2 mutant or 1p/19q co-deleted glioma were genotyped and linked to the UPDB. DNA came from blood (21), tumor tissue (43), or both (38). We determined congruence between somatic and germline samples and estimated the relative risk for developing cancer to first and second-degree relatives of G and A allele carriers at rs55705857. RESULTS Somatic (glioma) DNA had 85.7% sensitivity (CI 57.2-98.2%) and 95.8% specificity (CI 78.9-99.89%) for germline rs55705857 G allele. Forty-one patients were linked to pedigrees in the UPDB with at least three generations of data. First-degree relatives of rs55705857 G allele carriers were at significantly increased risk for developing cancer (RR = 1.72, p = 0.045, CI 1.02-2.94), and specifically for oligodendroglioma (RR = 57.61, p = 0.017, CI 2.96-320.98) or prostate cancer (RR = 4.10, p = 0.008, CI 1.62-9.58); relatives of individuals without the G allele were not at increased risk. Second-degree relatives of G allele carriers also had significantly increased risk for developing cancer (RR = 1.50, p = 0.007, CI 1.15-2.01). CONCLUSIONS Tumor DNA may approximate genotype at the rs55705857 locus. We confirmed this locus confers an increased risk of all cancers and especially of oligodendroglioma. No increased cancer or brain tumor risk is seen in family members of individuals without the high-risk G allele.
Collapse
Grants
- P30 CA042014 NCI NIH HHS
- Contract No. HHSN261201300017I Utah Cancer Registry, National Cancer Institute's SEER, Utah State Department of Health, University of Utah
- P30CA42014 Huntsman Cancer Institute, Huntsman Cancer Foundation, University of Utah, and National Cancer Institute of the NIH
- NA/Student Research University of Utah School of Medicine, Department of Human Genetics/Pediatric Division of Medical Genetics, Graduate Program in Genetic Counseling
- Utah Cancer Registry, National Cancer Institute’s SEER, Utah State Department of Health, University of Utah
- University of Utah School of Medicine, Department of Human Genetics/Pediatric Division of Medical Genetics, Graduate Program in Genetic Counseling
Collapse
Affiliation(s)
- Sarah Hummel
- Department of Human Genetics/Pediatric Division of Medical Genetics, Graduate Program in Genetic Counseling, University of Utah School of Medicine, 15 North 2030 East, Salt Lake City, 84112 Utah USA
| | - Wendy Kohlmann
- Department of Population Sciences, University of Utah School of Medicine, Huntsman Cancer Institute, Salt Lake City, Utah USA
| | - Thomas M. Kollmeyer
- The Mayo Clinic, Department of Laboratory Medicine and Pathology, Rochester, Minnesota USA
| | - Robert Jenkins
- The Mayo Clinic, Department of Laboratory Medicine and Pathology, Rochester, Minnesota USA
| | - Joshua Sonnen
- Division of Anatomic Pathology, University of Utah School of Medicine, Salt Lake City, Utah USA
| | - Cheryl A. Palmer
- Division of Anatomic Pathology, University of Utah School of Medicine, Salt Lake City, Utah USA
| | - Howard Colman
- Department of Neurosurgery, University of Utah School of Medicine, Huntsman Cancer Institute, Salt Lake City, Utah USA
| | - Diana Abbott
- Division of Genetic Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah USA
| | - Lisa Cannon-Albright
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah USA
- Division of Genetic Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah USA
| | - Adam L. Cohen
- Division of Oncology, University of Utah School of Medicine, Huntsman Cancer Institute, Salt Lake City, Utah USA
| |
Collapse
|
18
|
González-Castro TB, Juárez-Rojop IE, López-Narváez ML, Tovilla-Zárate CA, Genis-Mendoza AD, Pérez-Hernández N, Martínez-Magaña JJ, Rodríguez-Pérez JM. Genetic Polymorphisms of CCDC26 rs891835, rs6470745, and rs55705857 in Glioma Risk: A Systematic Review and Meta-analysis. Biochem Genet 2019; 57:583-605. [DOI: 10.1007/s10528-019-09911-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 02/07/2019] [Indexed: 01/03/2023]
|
19
|
Lancho O, Herranz D. The MYC Enhancer-ome: Long-Range Transcriptional Regulation of MYC in Cancer. Trends Cancer 2018; 4:810-822. [PMID: 30470303 DOI: 10.1016/j.trecan.2018.10.003] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 10/04/2018] [Accepted: 10/05/2018] [Indexed: 12/20/2022]
Abstract
MYC is one of the most important oncogenes in cancer. Indeed, MYC is upregulated in 50-60% of all tumors. MYC overexpression can be achieved through a variety of mechanisms, including gene duplications, chromosomal translocations, or somatic mutations leading to increased MYC stability. However, recent studies have identified numerous tissue-specific noncoding enhancers of MYC that play major roles in cancer, highlighting long-range transcriptional regulation of MYC as a critical novel mechanism leading to MYC hyperactivation and as a potential target for new therapeutic strategies in the near future. Here we summarize the regions and mechanisms involved in the long-range transcriptional regulation of MYC, underscoring the relevance of MYC enhancers both in normal physiological development and in MYC-driven cancer initiation and progression.
Collapse
Affiliation(s)
- Olga Lancho
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - Daniel Herranz
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA; Department of Pharmacology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA.
| |
Collapse
|
20
|
Rogawski DS, Vitanza NA, Gauthier AC, Ramaswamy V, Koschmann C. Integrating RNA sequencing into neuro-oncology practice. Transl Res 2017; 189:93-104. [PMID: 28746860 PMCID: PMC5659901 DOI: 10.1016/j.trsl.2017.06.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/27/2017] [Accepted: 06/30/2017] [Indexed: 12/22/2022]
Abstract
Malignant tumors of the central nervous system (CNS) cause substantial morbidity and mortality, yet efforts to optimize chemo- and radiotherapy have largely failed to improve dismal prognoses. Over the past decade, RNA sequencing (RNA-seq) has emerged as a powerful tool to comprehensively characterize the transcriptome of CNS tumor cells in one high-throughput step, leading to improved understanding of CNS tumor biology and suggesting new routes for targeted therapies. RNA-seq has been instrumental in improving the diagnostic classification of brain tumors, characterizing oncogenic fusion genes, and shedding light on intratumor heterogeneity. Currently, RNA-seq is beginning to be incorporated into regular neuro-oncology practice in the form of precision neuro-oncology programs, which use information from tumor sequencing to guide implementation of personalized targeted therapies. These programs show great promise in improving patient outcomes for tumors where single agent trials have been ineffective. As RNA-seq is a relatively new technique, many further applications yielding new advances in CNS tumor research and management are expected in the coming years.
Collapse
Affiliation(s)
- David S Rogawski
- Department of Pediatrics, University of Michigan School of Medicine, Ann Arbor, Mich
| | | | | | - Vijay Ramaswamy
- Division of Haematology/Oncology, Department of Pediatrics, Hospital for Sick Children, Toronto, ON, Canada
| | - Carl Koschmann
- Department of Pediatrics, University of Michigan School of Medicine, Ann Arbor, Mich.
| |
Collapse
|
21
|
Remote intracranial recurrence of IDH mutant gliomas is associated with TP53 mutations and an 8q gain. Oncotarget 2017; 8:84729-84742. [PMID: 29156679 PMCID: PMC5689569 DOI: 10.18632/oncotarget.20951] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 08/23/2017] [Indexed: 11/25/2022] Open
Abstract
Most IDH mutant gliomas harbor either 1p/19q co-deletions or TP53 mutation; 1p/19q co-deleted tumors have significantly better prognoses than tumors harboring TP53 mutations. To investigate the clinical factors that contribute to differences in tumor progression of IDH mutant gliomas, we classified recurrent tumor patterns based on MRI and correlated these patterns with their genomic characterization. Accordingly, in IDH mutant gliomas (N = 66), 1p/19 co-deleted gliomas only recurred locally, whereas TP53 mutant gliomas recurred both locally and in remote intracranial regions. In addition, diffuse tensor imaging suggested that remote intracranial recurrence in the astrocytomas, IDH-mutant with TP53 mutations may occur along major fiber bundles. Remotely recurrent tumors resulted in a higher mortality and significantly harbored an 8q gain; astrocytomas with an 8q gain resulted in significantly shorter overall survival than those without an 8q gain. OncoScan® arrays and next-generation sequencing revealed specific 8q regions (i.e., between 8q22 and 8q24) show a high copy number. In conclusion, only tumors with TP53 mutations showed patterns of remote recurrence in IDH mutant gliomas. Furthermore, an 8q gain was significantly associated with remote intracranial recurrence and can be considered a poor prognostic factor in astrocytomas, IDH-mutant.
Collapse
|
22
|
Huang J, Weinstein SJ, Kitahara CM, Karoly ED, Sampson JN, Albanes D. A prospective study of serum metabolites and glioma risk. Oncotarget 2017; 8:70366-70377. [PMID: 29050286 PMCID: PMC5642561 DOI: 10.18632/oncotarget.19705] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Accepted: 06/29/2017] [Indexed: 12/30/2022] Open
Abstract
Malignant glioma is one of the most lethal adult cancers, yet its etiology remains largely unknown. We conducted a prospective serum metabolomic analysis of glioma based on 64 cases and 64 matched controls selected from Alpha-Tocopherol, Beta-Carotene Cancer Prevention (ATBC) Study. Median time from collection of baseline fasting serum to diagnosis was nine years (inter-decile range 3-20 years). LC/MS-MS identified 730 known metabolites, and conditional logistic regression models estimated odds ratios for one-standard deviation differences in log-metabolite signals. Forty-three metabolites were associated with glioma at P<0.05. 2-Oxoarginine, cysteine, alpha-ketoglutarate, chenodeoxycholate and argininate yielded the strongest metabolite signals and were inversely related to overall glioma risk (0.0065≤P<0.0083). Also, seven xanthine metabolites related to caffeine metabolism were higher in cases than in controls (0.017≤P<0.042). Findings were mostly similar in high-grade glioma cases, although prominent inversely associated metabolites included the secondary bile acids glycocholenate sulfate and 3β-hydroxy-5-cholenoic acid, xenobiotic methyl 4-hydroxybenzoate sulfate, sex steroid 5alpha-pregnan-3beta, 20beta-diol-monosulfate, and cofactor/vitamin oxalate (0.0091≤P<0.021). A serum metabolomic profile of glioma identified years in advance of clinical diagnoses is characterized by altered signals in arginine/proline, antioxidant, and coffee-related metabolites. The observed pattern provides new potential leads regarding the molecular basis relevant to etiologic or sub-clinical biomarkers for glioma.
Collapse
Affiliation(s)
- Jiaqi Huang
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Department of Health and Human Services, Bethesda, MD, USA
| | - Stephanie J Weinstein
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Department of Health and Human Services, Bethesda, MD, USA
| | - Cari M Kitahara
- Radiation Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Department of Health and Human Services, Bethesda, MD, USA
| | - Edward D Karoly
- Director of Project Management, Metabolon, Inc., Morrisville, NC, USA
| | - Joshua N Sampson
- Biostatistics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Department of Health and Human Services, Bethesda, MD, USA
| | - Demetrius Albanes
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Department of Health and Human Services, Bethesda, MD, USA
| |
Collapse
|
23
|
Kalkat M, De Melo J, Hickman KA, Lourenco C, Redel C, Resetca D, Tamachi A, Tu WB, Penn LZ. MYC Deregulation in Primary Human Cancers. Genes (Basel) 2017; 8:genes8060151. [PMID: 28587062 PMCID: PMC5485515 DOI: 10.3390/genes8060151] [Citation(s) in RCA: 274] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 05/19/2017] [Accepted: 05/19/2017] [Indexed: 12/12/2022] Open
Abstract
MYC regulates a complex biological program by transcriptionally activating and repressing its numerous target genes. As such, MYC is a master regulator of many processes, including cell cycle entry, ribosome biogenesis, and metabolism. In cancer, the activity of the MYC transcriptional network is frequently deregulated, contributing to the initiation and maintenance of disease. Deregulation often leads to constitutive overexpression of MYC, which can be achieved through gross genetic abnormalities, including copy number alterations, chromosomal translocations, increased enhancer activity, or through aberrant signal transduction leading to increased MYC transcription or increased MYC mRNA and protein stability. Herein, we summarize the frequency and modes of MYC deregulation and describe both well-established and more recent findings in a variety of cancer types. Notably, these studies have highlighted that with an increased appreciation for the basic mechanisms deregulating MYC in cancer, new therapeutic vulnerabilities can be discovered and potentially exploited for the inhibition of this potent oncogene in cancer.
Collapse
Affiliation(s)
- Manpreet Kalkat
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada.
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada.
| | - Jason De Melo
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada.
| | - Katherine Ashley Hickman
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada.
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada.
| | - Corey Lourenco
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada.
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada.
| | - Cornelia Redel
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada.
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada.
| | - Diana Resetca
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada.
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada.
| | - Aaliya Tamachi
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada.
| | - William B Tu
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada.
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada.
| | - Linda Z Penn
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada.
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada.
| |
Collapse
|