Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Charles Chesnelong is active.

Publication


Featured researches published by Charles Chesnelong.


The Journal of Pathology | 2012

Concurrent CIC mutations, IDH mutations, and 1p/19q loss distinguish oligodendrogliomas from other cancers

Stephen Yip; Yaron S N Butterfield; Olena Morozova; Michael D. Blough; Jianghong An; Inanc Birol; Charles Chesnelong; Readman Chiu; Eric Chuah; Richard Corbett; Rod Docking; Marlo Firme; Martin Hirst; Shaun D. Jackman; Aly Karsan; Haiyan Li; David N. Louis; Alexandra Maslova; Richard A. Moore; Annie Moradian; Karen Mungall; Marco Perizzolo; Jenny Q. Qian; Gloria Roldán; Eric E. Smith; Jessica Tamura-Wells; Nina Thiessen; Richard Varhol; Samuel Weiss; Wei Wu

Oligodendroglioma is characterized by unique clinical, pathological, and genetic features. Recurrent losses of chromosomes 1p and 19q are strongly associated with this brain cancer but knowledge of the identity and function of the genes affected by these alterations is limited. We performed exome sequencing on a discovery set of 16 oligodendrogliomas with 1p/19q co‐deletion to identify new molecular features at base‐pair resolution. As anticipated, there was a high rate of IDH mutations: all cases had mutations in either IDH1 (14/16) or IDH2 (2/16). In addition, we discovered somatic mutations and insertions/deletions in the CIC gene on chromosome 19q13.2 in 13/16 tumours. These discovery set mutations were validated by deep sequencing of 13 additional tumours, which revealed seven others with CIC mutations, thus bringing the overall mutation rate in oligodendrogliomas in this study to 20/29 (69%). In contrast, deep sequencing of astrocytomas and oligoastrocytomas without 1p/19q loss revealed that CIC alterations were otherwise rare (1/60; 2%). Of the 21 non‐synonymous somatic mutations in 20 CIC‐mutant oligodendrogliomas, nine were in exon 5 within an annotated DNA‐interacting domain and three were in exon 20 within an annotated protein‐interacting domain. The remaining nine were found in other exons and frequently included truncations. CIC mutations were highly associated with oligodendroglioma histology, 1p/19q co‐deletion, and IDH1/2 mutation (p < 0.001). Although we observed no differences in the clinical outcomes of CIC mutant versus wild‐type tumours, in a background of 1p/19q co‐deletion, hemizygous CIC mutations are likely important. We hypothesize that the mutant CIC on the single retained 19q allele is linked to the pathogenesis of oligodendrogliomas with IDH mutation. Our detailed study of genetic aberrations in oligodendroglioma suggests a functional interaction between CIC mutation, IDH1/2 mutation, and 1p/19q co‐deletion. Copyright


Neuro-oncology | 2012

An in vivo patient-derived model of endogenous IDH1-mutant glioma

H. Artee Luchman; Owen D. Stechishin; N. Ha Dang; Michael D. Blough; Charles Chesnelong; John J. Kelly; Stephanie A. Nguyen; Jennifer A. Chan; Aalim M. Weljie; J. Gregory Cairncross; Samuel Weiss

Somatic mutations in the catalytic domain of isocitrate dehydrogenase (IDH) 1/2 and accumulation of the oncometabolite 2-hydroxyglutarate (2-HG) appear to be among the earliest events in gliomagenesis and may contribute to malignant transformation. The lack of cell lines with endogenous mutations has been one of the major challenges in studying IDH1/2-mutant glioma and developing novel therapeutics for these tumors. Here, we describe the isolation of a glioma brain tumor stem cell line (BT142) with an endogenous R132H mutation in IDH1, aggressive tumor-initiating capacity, and 2-HG production. The neurosphere culture method was used to establish a brain tumor stem cell line from an IDH1-mutant anaplastic oligoastrocytoma sample, and an orthotopic xenograft system was developed to allow its rapid expansion. Production of 2-HG by glioma cells with endogenous IDH1 mutations was confirmed by mass spectrometry. BT142 retained an endogenous R132H IDH1 mutation in culture and possessed aggressive tumor-initiating capacity, allowing it to be readily propagated in orthotopic xenografts of nonobese diabetic/severe combined immune deficiency (NOD SCID) mice. Endogenous 2-HG production by BT142 was detectable in both cell culture medium and xenograft animal serum. BT142 is the first brain tumor cell line with an endogenous IDH1 mutation and detectable 2-HG production both in vitro and in vivo, which thus provides a unique model for studying the biology of IDH1-mutant glioma and in vivo validation of compounds targeting IDH1-mutant cells.


Neuro-oncology | 2014

Lactate dehydrogenase A silencing in IDH mutant gliomas.

Charles Chesnelong; Myriam M. Chaumeil; Michael D. Blough; Mohammad Al-Najjar; Owen D. Stechishin; Jennifer A. Chan; Russell O. Pieper; Sabrina M. Ronen; Samuel Weiss; H. Artee Luchman; J. Gregory Cairncross

BACKGROUND Mutations of the isocitrate dehydrogenase 1 and 2 gene (IDH1/2) were initially thought to enhance cancer cell survival and proliferation by promoting the Warburg effect. However, recent experimental data have shown that production of 2-hydroxyglutarate by IDH mutant cells promotes hypoxia-inducible factor (HIF)1α degradation and, by doing so, may have unexpected metabolic effects. METHODS We used human glioma tissues and derived brain tumor stem cells (BTSCs) to study the expression of HIF1α target genes in IDH mutant ((mt)) and IDH wild-type ((wt)) tumors. Focusing thereafter on the major glycolytic enzyme, lactate dehydrogenase A (LDHA), we used standard molecular methods and pyrosequencing-based DNA methylation analysis to identify mechanisms by which LDHA expression was regulated in human gliomas. RESULTS We found that HIF1α-responsive genes, including many essential for glycolysis (SLC2A1, PDK1, LDHA, SLC16A3), were underexpressed in IDH(mt) gliomas and/or derived BTSCs. We then demonstrated that LDHA was silenced in IDH(mt) derived BTSCs, including those that did not retain the mutant IDH1 allele (mIDH(wt)), matched BTSC xenografts, and parental glioma tissues. Silencing of LDHA was associated with increased methylation of the LDHA promoter, as was ectopic expression of mutant IDH1 in immortalized human astrocytes. Furthermore, in a search of The Cancer Genome Atlas, we found low expression and high methylation of LDHA in IDH(mt) glioblastomas. CONCLUSION To our knowledge, this is the first demonstration of downregulation of LDHA in cancer. Although unexpected findings, silencing of LDHA and downregulation of several other glycolysis essential genes raise the intriguing possibility that IDH(mt) gliomas have limited glycolytic capacity, which may contribute to their slow growth and better prognosis.


Stem cell reports | 2015

Precursor States of Brain Tumor Initiating Cell Lines Are Predictive of Survival in Xenografts and Associated with Glioblastoma Subtypes

Carlo Cusulin; Charles Chesnelong; Pinaki Bose; Misha Bilenky; Karen Kopciuk; Jennifer A. Chan; J. Gregory Cairncross; Steven J.M. Jones; Marco A. Marra; H. Artee Luchman; Samuel Weiss

Summary In glioblastoma multiforme (GBM), brain-tumor-initiating cells (BTICs) with cancer stem cell characteristics have been identified and proposed as primordial cells responsible for disease initiation, recurrence, and therapeutic resistance. However, the extent to which individual, patient-derived BTIC lines reflect the heterogeneity of GBM remains poorly understood. Here we applied a stem cell biology approach and compared self-renewal, marker expression, label retention, and asymmetric cell division in 20 BTIC lines. Through cluster analysis, we identified two subgroups of BTIC lines with distinct precursor states, stem- or progenitor-like, predictive of survival after xenograft. Moreover, stem and progenitor transcriptomic signatures were identified, which showed a strong association with the proneural and mesenchymal subtypes, respectively, in the TCGA cohort. This study proposes a different framework for the study and use of BTIC lines and provides precursor biology insights into GBM.


Annals of Neurology | 2012

DNA Hypermethylation and 1p Loss Silence NHE-1 in Oligodendroglioma

Michael D. Blough; Mohammad Al-Najjar; Charles Chesnelong; Carmen E. Binding; Alexandra D. Rogers; H. Artee Luchman; John J. Kelly; Larry Fliegel; Olena Morozova; Stephen Yip; Marco A. Marra; Samuel Weiss; Jennifer A. Chan; J. Gregory Cairncross

Oligodendroglioma is characterized by mutations of IDH and CIC, 1p/19q loss, and slow growth. We found that NHE‐1 on 1p is silenced in oligodendrogliomas secondary to IDH‐associated hypermethylation and 1p allelic loss. Silencing lowers intracellular pH and attenuates acid load recovery in oligodendroglioma cells. Others have shown that rapid tumor growth cannot occur without NHE‐1–mediated neutralization of the acidosis generated by the Warburg glycolytic shift. Our findings show for the first time that the pH regulator NHE‐1 can be silenced in a human cancer and also suggest that pH deregulation may contribute to the distinctive biology of human oligodendroglioma. Ann Neurol 2012;71:845–849


Proceedings of the National Academy of Sciences of the United States of America | 2017

Clonal expansion and epigenetic reprogramming following deletion or amplification of mutant IDH1

Tali Mazor; Charles Chesnelong; Aleksandr Pankov; Llewellyn E. Jalbert; Chibo Hong; Josie Hayes; Ivan Smirnov; Roxanne Marshall; Camila F. Souza; Yaoqing Shen; Pavithra Viswanath; Houtan Noushmehr; Sabrina M. Ronen; Steven J.M. Jones; Marco A. Marra; J. Gregory Cairncross; Arie Perry; Sarah J. Nelson; Susan M. Chang; Andrew W. Bollen; Annette M. Molinaro; Henrik Bengtsson; Adam B. Olshen; Samuel Weiss; Joanna J. Phillips; H. Artee Luchman; Joseph F. Costello

Significance Identifying the drivers of tumorigenesis provides insight into mechanisms of transformation and can suggest novel therapeutic targets. IDH1 mutations in gliomas are one such promising target. Drivers of tumor initiation may be distinct from those at tumor recurrence, however. Here, we demonstrate that in a subset of initially IDH1 mutant gliomas IDH1 is deleted or amplified at recurrence, yielding a higher grade tumor with a reprogrammed epigenome. We also report systematic selection for cells with IDH1 CNA in vitro and in vivo. Thus, while IDH1 mutation likely initiates gliomagenesis, neither mutant IDH1 nor the oncometabolite 2HG that it produces are required at recurrence. These findings have important implications for emerging therapeutic strategies targeting mutant IDH1. IDH1 mutation is the earliest genetic alteration in low-grade gliomas (LGGs), but its role in tumor recurrence is unclear. Mutant IDH1 drives overproduction of the oncometabolite d-2-hydroxyglutarate (2HG) and a CpG island (CGI) hypermethylation phenotype (G-CIMP). To investigate the role of mutant IDH1 at recurrence, we performed a longitudinal analysis of 50 IDH1 mutant LGGs. We discovered six cases with copy number alterations (CNAs) at the IDH1 locus at recurrence. Deletion or amplification of IDH1 was followed by clonal expansion and recurrence at a higher grade. Successful cultures derived from IDH1 mutant, but not IDH1 wild type, gliomas systematically deleted IDH1 in vitro and in vivo, further suggestive of selection against the heterozygous mutant state as tumors progress. Tumors and cultures with IDH1 CNA had decreased 2HG, maintenance of G-CIMP, and DNA methylation reprogramming outside CGI. Thus, while IDH1 mutation initiates gliomagenesis, in some patients mutant IDH1 and 2HG are not required for later clonal expansions.


NeuroImage: Clinical | 2016

Hyperpolarized 13C MR imaging detects no lactate production in mutant IDH1 gliomas: Implications for diagnosis and response monitoring

Myriam M. Chaumeil; Marina Radoul; Chloe Najac; Pia Eriksson; Pavithra Viswanath; Michael D. Blough; Charles Chesnelong; H. Artee Luchman; J. Gregory Cairncross; Sabrina M. Ronen

Metabolic imaging of brain tumors using 13C Magnetic Resonance Spectroscopy (MRS) of hyperpolarized [1-13C] pyruvate is a promising neuroimaging strategy which, after a decade of preclinical success in glioblastoma (GBM) models, is now entering clinical trials in multiple centers. Typically, the presence of GBM has been associated with elevated hyperpolarized [1-13C] lactate produced from [1-13C] pyruvate, and response to therapy has been associated with a drop in hyperpolarized [1-13C] lactate. However, to date, lower grade gliomas had not been investigated using this approach. The most prevalent mutation in lower grade gliomas is the isocitrate dehydrogenase 1 (IDH1) mutation, which, in addition to initiating tumor development, also induces metabolic reprogramming. In particular, mutant IDH1 gliomas are associated with low levels of lactate dehydrogenase A (LDHA) and monocarboxylate transporters 1 and 4 (MCT1, MCT4), three proteins involved in pyruvate metabolism to lactate. We therefore investigated the potential of 13C MRS of hyperpolarized [1-13C] pyruvate for detection of mutant IDH1 gliomas and for monitoring of their therapeutic response. We studied patient-derived mutant IDH1 glioma cells that underexpress LDHA, MCT1 and MCT4, and wild-type IDH1 GBM cells that express high levels of these proteins. Mutant IDH1 cells and tumors produced significantly less hyperpolarized [1-13C] lactate compared to GBM, consistent with their metabolic reprogramming. Furthermore, hyperpolarized [1-13C] lactate production was not affected by chemotherapeutic treatment with temozolomide (TMZ) in mutant IDH1 tumors, in contrast to previous reports in GBM. Our results demonstrate the unusual metabolic imaging profile of mutant IDH1 gliomas, which, when combined with other clinically available imaging methods, could be used to detect the presence of the IDH1 mutation in vivo.


Cancer Research | 2016

Abstract 2524: STAT3 is a key regulator of an “EMT-like” process mediated by Slug in GBM

Charles Chesnelong; H. Artee Luchman; J. Gregory Cairncross; Samuel Weiss

Glioblastoma Multiforme (GBM) is the most aggressive subtype of adult brain tumor with a median survival of 15 months. Despite a combination of maximal safe resection, radiation and chemotherapy, GBM invariably recurs, highlighting the need to better delineate the basis of recurrent disease and develop novel, more targeted and effective therapies. The Signal Transducer and Activator of Transcription 3 (STAT3) has been implicated in a proneural to mesenchymal shift associated with the emergence of a more aggressive, more resistant GBM phenotype at recurrence. Brain Tumor Initiating Cells (BTICs), defined by the key features of self-renewal, multipotency and tumorigenic potential, are integral players of recurrence post-treatment and represent a “reservoir of disease” that needs to be specifically targeted if GBM outcome is to be improved. Analysis of GBM patient transcriptomic data from The Cancer Genome Atlas (TCGA) shows that an Epithelial to Mesenchymal Transition (EMT) gene signature is particularly enriched in the mesenchymal subtype, tightly correlates with STAT3 activity and is associated with shorter survival. The key EMT transcription factor Slug is also highly expressed in mesenchymal samples and associated with poorer prognosis. Interestingly, we found stronger expression of EMT transcription factors Snail, Slug and Twist in faster proliferating, more aggressive BTIC lines. Noteworthy, Slug was more highly expressed in both BTICs and parental tumors compared to Snail and Twist and positively correlated with pSTAT3. We also found that Slug expression correlates with faster growth in vitro and shorter survival in orthotopic xenografts. Conversely, higher E-cadherin expression correlates with slower growth and longer survival of xenografted mice. While Slug is not a known STAT3 target gene (unlike Twist and Snail), we show that Slug expression is decreased after pharmacological inhibition of STAT3 signaling in BTICs. In contrast, activation of the STAT3 pathway via growth factor/cytokine treatment (EGF, OSM), as well as expression of a constitutively active form of STAT3 promotes Slug expression. We have identified a potential STAT3 consensus binding site in the Slug promoter and preliminary Chromatin Immuno Precipitation (ChIP) experiments suggest that Slug is a novel direct transcriptional target of STAT3. Over-expression of Slug in BTIC lines triggered down-regulation of E-cadherin and resulted in increased Cyclin D1 and pRB protein levels. However, we found that Cyclin D1 RNA levels remain unchanged suggesting that overexpression of Slug leads to the post-transcriptional stabilization of Cyclin D1 potentially via repression of UbcH5C. To conclude, STAT3 is a key regulator of an EMT-like process in GBM BTICs, mediated at least in part by Slug. Our results suggest that STAT3 and the key regulator Slug may be involved in the promotion of a more aggressive GBM phenotype and represent interesting therapeutic targets in GBM. Citation Format: Charles Chesnelong, H. Artee Luchman, J. Gregory Cairncross, Samuel Weiss. STAT3 is a key regulator of an “EMT-like” process mediated by Slug in GBM. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 2524.


Archive | 2019

Isolation and Culture of Glioblastoma Brain Tumor Stem Cells

Charles Chesnelong; Ian Restall; Samuel Weiss

Cancer stem cells (CSCs) have been identified in glioblastoma (GBM) and are proposed to be the main actors of post-treatment recurrence contributing to the very dismal prognosis of this devastating disease. Consequently, this important population of cells needs to be further studied to uncover potential vulnerabilities, identify novel therapeutic targets, and develop drugs that can be translated to the clinic. One obstacle preventing progress in understanding the biology of GBM and the development of novel therapies has arguably been the absence of biologically relevant in vitro models representative of the CSC population in GBM. Adherent and non-adherent serum-free culture methods, initially developed for culturing neural stem cells, have been adapted to identify, isolate, maintain, and expand brain tumor stem cells (BTSCs) from GBM. In this chapter, we describe a method to isolate and culture these BTSCs from fresh GBM patient samples.


Journal of Visualized Experiments | 2018

Live-Cell Imaging Assays to Study Glioblastoma Brain Tumor Stem Cell Migration and Invasion

Ian Restall; Danielle Bozek; Charles Chesnelong; Samuel Weiss; H. Artee Luchman

Glioblastoma (GBM) is an aggressive brain tumor that is poorly controlled with the currently available treatment options. Key features of GBMs include rapid proliferation and pervasive invasion into the normal brain. Recurrence is thought to result from the presence of radio- and chemo-resistant brain tumor stem cells (BTSCs) that invade away from the initial cancerous mass and, thus, evade surgical resection. Hence, therapies that target BTSCs and their invasive abilities may improve the otherwise poor prognosis of this disease. Our group and others have successfully established and characterized BTSC cultures from GBM patient samples. These BTSC cultures demonstrate fundamental cancer stem cell properties such as clonogenic self-renewal, multi-lineage differentiation, and tumor initiation in immune-deficient mice. In order to improve on the current therapeutic approaches for GBM, a better understanding of the mechanisms of BTSC migration and invasion is necessary. In GBM, the study of migration and invasion is restricted, in part, due to the limitations of existing techniques which do not fully account for the in vitro growth characteristics of BTSCs grown as neurospheres. Here, we describe rapid and quantitative live-cell imaging assays to study both the migration and invasion properties of BTSCs. The first method described is the BTSC migration assay which measures the migration toward a chemoattractant gradient. The second method described is the BTSC invasion assay which images and quantifies a cellular invasion from neurospheres into a matrix. The assays described here are used for the quantification of BTSC migration and invasion over time and under different treatment conditions.

Collaboration


Dive into the Charles Chesnelong's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marco A. Marra

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge