1
|
El-Ayoubi A, Klawitter M, Rüttinger J, Wellhäusser G, Holm PS, Danielyan L, Naumann U. Intranasal Delivery of Oncolytic Adenovirus XVir-N-31 via Optimized Shuttle Cells Significantly Extends Survival of Glioblastoma-Bearing Mice. Cancers (Basel) 2023; 15:4912. [PMID: 37894279 PMCID: PMC10605419 DOI: 10.3390/cancers15204912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/05/2023] [Accepted: 10/05/2023] [Indexed: 10/29/2023] Open
Abstract
A glioblastoma (GBM) is an aggressive and lethal primary brain tumor with restricted treatment options and a dismal prognosis. Oncolytic virotherapy (OVT) has developed as a promising approach for GBM treatment. However, reaching invasive GBM cells may be hindered by tumor-surrounding, non-neoplastic cells when the oncolytic virus (OV) is applied intratumorally. Using two xenograft GBM mouse models and immunofluorescence analyses, we investigated the intranasal delivery of the oncolytic adenovirus (OAV) XVir-N-31 via virus-loaded, optimized shuttle cells. Intranasal administration (INA) was selected due to its non-invasive nature and the potential to bypass the blood-brain barrier (BBB). Our findings demonstrate that the INA of XVir-N-31-loaded shuttle cells successfully delivered OAVs to the core tumor and invasive GBM cells, significantly prolonged the survival of the GBM-bearing mice, induced immunogenic cell death and finally reduced the tumor burden, all this highlighting the therapeutic potential of this innovative approach. Overall, this study provides compelling evidence for the effectiveness of the INA of XVir-N-31 via shuttle cells as a promising therapeutic strategy for GBM. The non-invasive nature of the INA of OV-loaded shuttle cells holds great promise for future clinical translation. However, further research is required to assess the efficacy of this approach to ultimately progress in human clinical trials.
Collapse
Affiliation(s)
- Ali El-Ayoubi
- Molecular Neurooncology, Department of Vascular Neurology, Hertie Institute for Clinical Brain Research and Center Neurology, University of Tübingen, D-72076 Tübingen, Germany; (A.E.-A.); (M.K.); (J.R.); (G.W.)
| | - Moritz Klawitter
- Molecular Neurooncology, Department of Vascular Neurology, Hertie Institute for Clinical Brain Research and Center Neurology, University of Tübingen, D-72076 Tübingen, Germany; (A.E.-A.); (M.K.); (J.R.); (G.W.)
| | - Jakob Rüttinger
- Molecular Neurooncology, Department of Vascular Neurology, Hertie Institute for Clinical Brain Research and Center Neurology, University of Tübingen, D-72076 Tübingen, Germany; (A.E.-A.); (M.K.); (J.R.); (G.W.)
| | - Giulia Wellhäusser
- Molecular Neurooncology, Department of Vascular Neurology, Hertie Institute for Clinical Brain Research and Center Neurology, University of Tübingen, D-72076 Tübingen, Germany; (A.E.-A.); (M.K.); (J.R.); (G.W.)
| | - Per Sonne Holm
- Department of Urology, Klinikum Rechts der Isar, Technical University of Munich, D-81675 Munich, Germany;
- Department of Oral and Maxillofacial Surgery, Medical University Innsbruck, A-6020 Innsbruck, Austria
- XVir Therapeutics GmbH, D-80331 Munich, Germany
| | - Lusine Danielyan
- Department of Clinical Pharmacology, University Hospital Tübingen, D-72076 Tübingen, Germany;
- Neuroscience Laboratory and Departments of Biochemistry and Clinical Pharmacology, Yerevan State Medical University, Yerevan 0025, Armenia
| | - Ulrike Naumann
- Molecular Neurooncology, Department of Vascular Neurology, Hertie Institute for Clinical Brain Research and Center Neurology, University of Tübingen, D-72076 Tübingen, Germany; (A.E.-A.); (M.K.); (J.R.); (G.W.)
- Gene and RNA Therapy Center (GRTC), Faculty of Medicine, University of Tübingen, D-72076 Tübingen, Germany
| |
Collapse
|
2
|
Roura AJ, Szadkowska P, Poleszak K, Dabrowski MJ, Ellert-Miklaszewska A, Wojnicki K, Ciechomska IA, Stepniak K, Kaminska B, Wojtas B. Regulatory networks driving expression of genes critical for glioblastoma are controlled by the transcription factor c-Jun and the pre-existing epigenetic modifications. Clin Epigenetics 2023; 15:29. [PMID: 36850002 PMCID: PMC9972689 DOI: 10.1186/s13148-023-01446-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 02/13/2023] [Indexed: 03/01/2023] Open
Abstract
BACKGROUND Glioblastoma (GBM, WHO grade IV) is an aggressive, primary brain tumor. Despite extensive tumor resection followed by radio- and chemotherapy, life expectancy of GBM patients did not improve over decades. Several studies reported transcription deregulation in GBMs, but regulatory mechanisms driving overexpression of GBM-specific genes remain largely unknown. Transcription in open chromatin regions is directed by transcription factors (TFs) that bind to specific motifs, recruit co-activators/repressors and the transcriptional machinery. Identification of GBM-related TFs-gene regulatory networks may reveal new and targetable mechanisms of gliomagenesis. RESULTS We predicted TFs-regulated networks in GBMs in silico and intersected them with putative TF binding sites identified in the accessible chromatin in human glioma cells and GBM patient samples. The Cancer Genome Atlas and Glioma Atlas datasets (DNA methylation, H3K27 acetylation, transcriptomic profiles) were explored to elucidate TFs-gene regulatory networks and effects of the epigenetic background. In contrast to the majority of tumors, c-Jun expression was higher in GBMs than in normal brain and c-Jun binding sites were found in multiple genes overexpressed in GBMs, including VIM, FOSL2 or UPP1. Binding of c-Jun to the VIM gene promoter was stronger in GBM-derived cells than in cells derived from benign glioma as evidenced by gel shift and supershift assays. Regulatory regions of the majority of c-Jun targets have distinct DNA methylation patterns in GBMs as compared to benign gliomas, suggesting the contribution of DNA methylation to the c-Jun-dependent gene expression. CONCLUSIONS GBM-specific TFs-gene networks identified in GBMs differ from regulatory pathways attributed to benign brain tumors and imply a decisive role of c-Jun in controlling genes that drive glioma growth and invasion as well as a modulatory role of DNA methylation.
Collapse
Affiliation(s)
- Adria-Jaume Roura
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Paulina Szadkowska
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Katarzyna Poleszak
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Michal J. Dabrowski
- Institute of Computer Science of the Polish Academy of Sciences, Warsaw, Poland
| | | | - Kamil Wojnicki
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Iwona A. Ciechomska
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Karolina Stepniak
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Bozena Kaminska
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Bartosz Wojtas
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
- Laboratory of Sequencing, Nencki Institute of Experimental Biology, ul. Ludwika Pasteura 3, 02-093 Warsaw, Poland
| |
Collapse
|
3
|
Chen PY, Li XD, Ma WN, Li H, Li MM, Yang XY, Li SY. Comprehensive Transcriptomic Analysis and Experimental Validation Identify lncRNA HOXA-AS2/miR-184/COL6A2 as the Critical ceRNA Regulation Involved in Low-Grade Glioma Recurrence. Onco Targets Ther 2020; 13:4999-5016. [PMID: 32581558 PMCID: PMC7276213 DOI: 10.2147/ott.s245896] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 04/16/2020] [Indexed: 12/18/2022] Open
Abstract
Purpose The recurrence and metastasis of glioma are closely related to complex regulatory networks among protein-coding genes, lncRNAs and microRNAs. The aim of this study was to investigate core genes, lncRNAs, miRNAs and critical ceRNA regulatory mechanisms, which are involved in lower-grade glioma (LGG) recurrence. Materials and Methods We employed multiple datasets from Chinese Glioma Genome Atlas (CGGA) database and The Cancer Genome Atlas (TCGA) to perform comprehensive transcriptomic analysis. Further in vitro experiments including cell proliferation assay, luciferase reporter assay, and Western blot were performed to validate our results. Results Recurrent LGG and glioblastoma (GBM) showed different transcriptome characteristics with less overlap of differentially expressed protein-coding genes (DEPs), lncRNAs (DELs) and miRNAs (DEMs) compared with primary samples. There were no overlapping gene in ontology (GO) terms related to GBM recurrence in the TCGA and CGGA databases, but there were overlaps associated with LGG recurrence. GO analysis and protein–protein interaction (PPI) network analysis identified three core genes: TIMP1, COL1A1 and COL6A2. By hierarchical cluster analysis of them, LGGs could be clustered as Low_risk and High_risk group. The High_risk group with high expression of TIMP1, COL1A1, and COL6A2 showed worse prognosis. By coexpression networks analysis, competing endogenous RNA (ceRNA) network analysis, cell proliferation assay and luciferase reporter assay, we confirmed that lncRNA HOXA-AS2 functioned as a ceRNA for miR-184 to regulate expression of COL6A2, which induced cell proliferation of low-grade glioma. Conclusion In this study, we revealed a 3-hub protein-coding gene signature to improve prognostic prediction in LGG, and identified a critical ceRNA regulation involved in LGG recurrence.
Collapse
Affiliation(s)
- Peng-Yu Chen
- Department of Neurosurgery, Shengjing Hospital Affiliated to China Medical University, Shenyang, People's Republic of China
| | - Xiao-Dong Li
- Department of Neurosurgery, Shengjing Hospital Affiliated to China Medical University, Shenyang, People's Republic of China
| | - Wei-Ning Ma
- Department of Neurosurgery, Shengjing Hospital Affiliated to China Medical University, Shenyang, People's Republic of China
| | - Han Li
- Department of Neurosurgery, Shengjing Hospital Affiliated to China Medical University, Shenyang, People's Republic of China
| | - Miao-Miao Li
- Department of Neurosurgery, Shengjing Hospital Affiliated to China Medical University, Shenyang, People's Republic of China
| | - Xin-Yu Yang
- Department of Neurosurgery, Shengjing Hospital Affiliated to China Medical University, Shenyang, People's Republic of China
| | - Shao-Yi Li
- Department of Neurosurgery, Shengjing Hospital Affiliated to China Medical University, Shenyang, People's Republic of China
| |
Collapse
|
4
|
Rivero-Hinojosa S, Kinney N, Garner HR, Rood BR. Germline microsatellite genotypes differentiate children with medulloblastoma. Neuro Oncol 2020; 22:152-162. [PMID: 31562520 PMCID: PMC6954392 DOI: 10.1093/neuonc/noz179] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The germline genetic events underpinning medulloblastoma (MB) initiation, and therefore the ability to determine who is at risk, are still unknown for the majority of cases. Microsatellites are short repeated sequences that make up ~3% of the genome. Repeat lengths vary among individuals and are often nonrandomly associated with disease, including several cancers such as breast, glioma, lung, and ovarian. Due to their effects on gene function, they have been called the "tuning knobs of the genome." METHODS We have developed a novel approach for identifying a microsatellite-based signature to differentiate MB patients from controls using germline DNA. RESULTS Analyzing germline whole exome sequencing data from a training set of 120 MB subjects and 425 controls, we identified 139 individual microsatellite loci whose genotypes differ significantly between the groups. Using a genetic algorithm, we identified a subset of 43 microsatellites that distinguish MB subjects from controls with a sensitivity and specificity of 92% and 88%, respectively. This microsatellite signature was validated in an independent dataset consisting of 102 subjects and 428 controls, with comparable sensitivity and specificity of 95% and 90%, respectively. Analysis of the allele genotypes of those 139 informative loci demonstrates that their association with MB is a consequence of individual microsatellites' genotypes rather than their hypermutability. Finally, an analysis of the genes harboring these microsatellite loci reveals cellular functions important for tumorigenesis. CONCLUSION This study demonstrates that MB-specific germline microsatellite variations mark those at risk for MB development and suggests mechanisms of predisposition.
Collapse
Affiliation(s)
- Samuel Rivero-Hinojosa
- Center for Cancer and Immunology Research, Children's Research Institute, Children's National Medical Center (CNMC), Washington, DC
| | - Nicholas Kinney
- Center for Bioinformatics and Genetics, Edward Via College of Osteopathic Medicine, Blacksburg, Virginia
- Gibbs Cancer Center and Research Institute, Spartanburg, South Carolina
| | - Harold R Garner
- Center for Bioinformatics and Genetics, Edward Via College of Osteopathic Medicine, Blacksburg, Virginia
- Gibbs Cancer Center and Research Institute, Spartanburg, South Carolina
| | - Brian R Rood
- Center for Cancer and Immunology Research, Children's Research Institute, Children's National Medical Center (CNMC), Washington, DC
| |
Collapse
|
5
|
Zhang X, Lu X, Liu Z, Guan R, Wang J, Kong X, Chen L, Bo C, Tian K, Xu S, Bai M, Zhang H, Li J, Wang L, Shen J, Guo M. Integrating multiple-level molecular data to infer the distinctions between glioblastoma and lower-grade glioma. Int J Cancer 2019; 145:952-961. [PMID: 30694558 DOI: 10.1002/ijc.32174] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 12/28/2018] [Accepted: 01/08/2019] [Indexed: 01/25/2023]
Abstract
Glioblastomas (GBMs) and lower-grade gliomas (LGGs) are the most common malignant brain tumors. Despite extensive studies that have suggested that there are differences between the two in terms of clinical profile and treatment, their distinctions on a molecular level had not been systematically analyzed. Here, we investigated the distinctions between GBM and LGG based on multidimensional data, including somatic mutations, somatic copy number variants (SCNVs), gene expression, lncRNA expression and DNA methylation levels. We found that GBM patients had a higher mutation frequency and SCNVs than LGG patients. Differential mRNAs and lncRNAs between GBM and LGG were identified and a differential mRNA-lncRNA network was constructed and analyzed. We also discovered some differential DNA methylation sites could distinguish between GBM and LGG samples. Finally, we identified some key GBM- and LGG-specific genes featuring multiple-level molecular alterations. These specific genes participate in diverse functions; moreover, GBM-specific genes are enriched in the glioma pathway. Overall, our studies explored the distinctions between GMB and LGG using a comprehensive genomics approach that may provide novel insights into studying the mechanism and treatment of brain tumors.
Collapse
Affiliation(s)
- Xiaoming Zhang
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xiaoyu Lu
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Zhaojun Liu
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Ruoyu Guan
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Jianjian Wang
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xiaotong Kong
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Lixia Chen
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Chunrui Bo
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Kuo Tian
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Si Xu
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Ming Bai
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Huixue Zhang
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Jie Li
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Lihua Wang
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Jia Shen
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA
| | - Mian Guo
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
6
|
Kafka A, Bačić M, Tomas D, Žarković K, Bukovac A, Njirić N, Mrak G, Krsnik Ž, Pećina‐Šlaus N. Different behaviour of DVL1, DVL2, DVL3 in astrocytoma malignancy grades and their association to TCF1 and LEF1 upregulation. J Cell Mol Med 2019; 23:641-655. [PMID: 30468298 PMCID: PMC6307814 DOI: 10.1111/jcmm.13969] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 09/04/2018] [Accepted: 09/27/2018] [Indexed: 01/21/2023] Open
Abstract
Key regulators of the Wnt signalling, DVL1, DVL2 and DVL3, in astrocytomas of different malignancy grades were investigated. Markers for DVL1, DVL2 and DVL3 were used to detect microsatellite instability (MSI) and gross deletions (LOH), while immunohistochemistry and immunoreactivity score were used to determine the signal strengths of the three DVL proteins and transcription factors of the pathway, TCF1 and LEF1. Our findings demonstrated that MSI at all three DVL loci was constantly found across tumour grades with the highest number in grade II (P = 0.008). Collectively, LOHs were more frequent in high-grade tumours than in low grade ones. LOHs of DVL3 gene were significantly associated with grade IV tumours (P = 0.007). The results on protein expressions indicated that high-grade tumours expressed less DVL1 protein as compared with low grade ones. A significant negative correlation was established between DVL1 expression and malignancy grades (P < 0.001). The expression of DVL2 protein was found similar across grades, while DVL3 expression significantly increased with malignancy grades (P < 0.001). The signal strengths of expressed DVL1 and DVL3 were negatively correlated (P = 0.002). However, TCF1 and LEF1 were both significantly upregulated and increasing with astrocytoma grades (P = 0.001). A positive correlation was established between DVL3 and both TCF1 (P = 0.020) and LEF1 (P = 0.006) suggesting their joint involvement in malignant progression. Our findings suggest that DVL1 and DVL2 may be involved during early stages of the disease, while DVL3 may have a role in later phases and together with TCF1 and LEF1 promotes the activation of Wnt signalling.
Collapse
Affiliation(s)
- Anja Kafka
- Laboratory of Neuro‐oncologyCroatian Institute for Brain ResearchSchool of MedicineUniversity of ZagrebZagrebCroatia
- Department of BiologySchool of MedicineUniversity of ZagrebZagrebCroatia
| | | | - Davor Tomas
- Department of PathologySchool of MedicineUniversity of ZagrebZagrebCroatia
- Department of PathologyUniversity Hospital Center “Sisters of Charity”ZagrebCroatia
| | - Kamelija Žarković
- Department of PathologySchool of MedicineUniversity of ZagrebZagrebCroatia
- Division of PathologyUniversity Hospital Center “Zagreb”ZagrebCroatia
| | - Anja Bukovac
- Laboratory of Neuro‐oncologyCroatian Institute for Brain ResearchSchool of MedicineUniversity of ZagrebZagrebCroatia
- Department of BiologySchool of MedicineUniversity of ZagrebZagrebCroatia
| | - Niko Njirić
- Laboratory of Neuro‐oncologyCroatian Institute for Brain ResearchSchool of MedicineUniversity of ZagrebZagrebCroatia
- Department of NeurosurgeryUniversity Hospital Center “Zagreb”School of MedicineUniversity of ZagrebZagrebCroatia
| | - Goran Mrak
- Department of NeurosurgeryUniversity Hospital Center “Zagreb”School of MedicineUniversity of ZagrebZagrebCroatia
| | - Željka Krsnik
- Department of NeuroscienceCroatian Institute for Brain ResearchSchool of MedicineUniversity of ZagrebZagrebCroatia
| | - Nives Pećina‐Šlaus
- Laboratory of Neuro‐oncologyCroatian Institute for Brain ResearchSchool of MedicineUniversity of ZagrebZagrebCroatia
- Department of BiologySchool of MedicineUniversity of ZagrebZagrebCroatia
| |
Collapse
|
7
|
Velmurugan KR, Varghese RT, Fonville NC, Garner HR. High-depth, high-accuracy microsatellite genotyping enables precision lung cancer risk classification. Oncogene 2017; 36:6383-6390. [PMID: 28759038 PMCID: PMC5701090 DOI: 10.1038/onc.2017.256] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 06/01/2017] [Accepted: 06/13/2017] [Indexed: 12/23/2022]
Abstract
There remains a large discrepancy between the known genetic contributions to cancer and that which can be explained by genomic variants, both inherited and somatic. Recently, understudied repetitive DNA regions called microsatellites have been identified as genetic risk markers for a number of diseases including various cancers (breast, ovarian and brain). In this study, we demonstrate an integrated process for identifying and further evaluating microsatellite-based risk markers for lung cancer using data from the cancer genome atlas and the 1000 genomes project. Comparing whole-exome germline sequencing data from 488 TCGA lung cancer samples to germline exome data from 390 control samples from the 1000 genomes project, we identified 119 potentially informative microsatellite loci. These loci were found to be able to distinguish between cancer and control samples with sensitivity and specificity ratios over 0.8. Then these loci, supplemented with additional loci from other cancers and controls, were evaluated using a target enrichment kit and sample-multiplexed nextgen sequencing. Thirteen of the 119 risk markers were found to be informative in a well powered study (>0.99 for a 0.95 confidence interval) using high-depth (579x±315) nextgen sequencing of 30 lung cancer and 89 control samples, resulting in sensitivity and specificity ratios of 0.90 and 0.94, respectively. When 8 loci harvested from the bioinformatic analysis of other cancers are added to the classifier, then the sensitivity and specificity rise to 0.93 and 0.97, respectively. Analysis of the genes harboring these loci revealed two genes (ARID1B and REL) and two significantly enriched pathways (chromatin organization and cellular stress response) suggesting that the process of lung carcinogenesis is linked to chromatin remodeling, inflammation, and tumor microenvironment restructuring. We illustrate that high-depth sequencing enables a high-precision microsatellite-based risk classifier analysis approach. This microsatellite-based platform confirms the potential to create clinically actionable diagnostics for lung cancer.
Collapse
Affiliation(s)
- K R Velmurugan
- Department of Biological Sciences, Center for Bioinformatics and Genetics and the Primary Care Research Network, Edward Via College of Osteopathic Medicine, Blacksburg, VA, USA.,Department of Biological Sciences, Gibbs Cancer Center and Research Institute, Spartanburg, SC, USA
| | - R T Varghese
- Department of Biological Sciences, Center for Bioinformatics and Genetics and the Primary Care Research Network, Edward Via College of Osteopathic Medicine, Blacksburg, VA, USA.,Department of Biological Sciences, Gibbs Cancer Center and Research Institute, Spartanburg, SC, USA
| | - N C Fonville
- Department of Biological Sciences, Riverside Law, LLP Glenhardie Corporate Center, Wayne, PA, USA
| | - H R Garner
- Department of Biological Sciences, Center for Bioinformatics and Genetics and the Primary Care Research Network, Edward Via College of Osteopathic Medicine, Blacksburg, VA, USA.,Department of Biological Sciences, Gibbs Cancer Center and Research Institute, Spartanburg, SC, USA
| |
Collapse
|
8
|
Qiao W, Guo B, Zhou H, Xu W, Chen Y, Liang Y, Dong B. miR-124 suppresses glioblastoma growth and potentiates chemosensitivity by inhibiting AURKA. Biochem Biophys Res Commun 2017; 486:43-48. [DOI: 10.1016/j.bbrc.2017.02.120] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 02/24/2017] [Indexed: 12/12/2022]
|
9
|
Karunasena E, Mciver LJ, Bavarva JH, Wu X, Zhu H, Garner HR. 'Cut from the same cloth': Shared microsatellite variants among cancers link to ectodermal tissues-neural tube and crest cells. Oncotarget 2016; 6:22038-47. [PMID: 26246470 PMCID: PMC4673144 DOI: 10.18632/oncotarget.4194] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 06/05/2015] [Indexed: 11/25/2022] Open
Abstract
The pluripotent cells of the embryonic ectodermal tissues are known to be a precursor for multiple tumor types. The adaptability of these cells is a trait exploited by cancer. We previously described cancer-associated microsatellite loci (CAML) shared between glioblastoma (GBM) and lower-grade gliomas. Therefore, we hypothesized that these variants, identified from germline DNA, are shared by cancers from tissues originating from ectodermal tissues: neural tube cells (NTC) and crest cells (NCC). Using exome sequencing data from four cancers with origins to NTC and NCC, a ‘signature’ of loci significant to each cancer (p-value ≤ 0.01) was created and compared with previously identified CAML from breast cancer. The results of this analysis show that variant loci among the cancers with tissue origins from NTC/NCC were closely linked. Signaling pathways linked to genes with non-coding CAML genotypes revealed enriched connections to hereditary, neurological, and developmental disease or disorders. Thus, variants in genes from tissues initiating from NTC/NCC, if recurrently detected, may indicate a common etiology. Additionally, CAML genotypes from non-tumor DNA may predict cancer phenotypes and are common to shared embryonic tissues of origin.
Collapse
Affiliation(s)
- Enusha Karunasena
- Virginia Bioinformatics Institute, Medical Informatics and Systems Division, Virginia Tech, Blacksburg, VA 24061, USA
| | - Lauren J Mciver
- Virginia Bioinformatics Institute, Medical Informatics and Systems Division, Virginia Tech, Blacksburg, VA 24061, USA
| | - Jasmin H Bavarva
- Virginia Bioinformatics Institute, Medical Informatics and Systems Division, Virginia Tech, Blacksburg, VA 24061, USA
| | - Xiaowei Wu
- Department of Statistics, Virginia Tech, Blacksburg, VA 24061, USA
| | - Hongxiao Zhu
- Department of Statistics, Virginia Tech, Blacksburg, VA 24061, USA
| | - Harold R Garner
- Virginia Bioinformatics Institute, Medical Informatics and Systems Division, Virginia Tech, Blacksburg, VA 24061, USA
| |
Collapse
|
10
|
Garner HR, Waitzkin MB, Bavarva JH. What do the changes in the aging genome mean for pharmacogenomics? Pharmacogenomics 2014; 15:1725-1728. [PMID: 25493565 DOI: 10.2217/pgs.14.131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Harold R Garner
- Virginia Bioinformatics Institute, Virginia Tech, Washington Street, Blacksburg, VA, USA
| | | | | |
Collapse
|